ArticlePDF Available

Peripherally administered antibodies against amyloid ??-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease

Authors:

Abstract and Figures

One hallmark of Alzheimer disease is the accumulation of amyloid beta-peptide in the brain and its deposition as plaques. Mice transgenic for an amyloid beta precursor protein (APP) mini-gene driven by a platelet-derived (PD) growth factor promoter (PDAPP mice), which overexpress one of the disease-linked mutant forms of the human amyloid precursor protein, show many of the pathological features of Alzheimer disease, including extensive deposition of extracellular amyloid plaques, astrocytosis and neuritic dystrophy. Active immunization of PDAPP mice with human amyloid beta-peptide reduces plaque burden and its associated pathologies. Several hypotheses have been proposed regarding the mechanism of this response. Here we report that peripheral administration of antibodies against amyloid beta-peptide, was sufficient to reduce amyloid burden. Despite their relatively modest serum levels, the passively administered antibodies were able to enter the central nervous system, decorate plaques and induce clearance of preexisting amyloid. When examined in an ex vivo assay with sections of PDAPP or Alzheimer disease brain tissue, antibodies against amyloid beta-peptide triggered microglial cells to clear plaques through Fc receptor-mediated phagocytosis and subsequent peptide degradation. These results indicate that antibodies can cross the blood-brain barrier to act directly in the central nervous system and should be considered as a therapeutic approach for the treatment of Alzheimer disease and other neurological disorders.
Content may be subject to copyright.
916 NATURE MEDICINE VOLUME 6 NUMBER 8 AUGUST 2000
ARTICLES
One hallmark of Alzheimer disease is the accumulation of amy-
loid β-peptide in the brain and its deposition as plaques. Mice
transgenic for an amyloid β precursor protein (APP) mini-gene
driven by a platelet-derived (PD) growth factor promoter
(PDAPP mice), which overexpress one of the disease-linked mu-
tant forms of the human amyloid precursor protein, show many
of the pathological features of Alzheimer disease, including ex-
tensive deposition of extracellular amyloid plaques, astrocytosis
and neuritic dystrophy
1,2
. Active immunization of PDAPP mice
with human amyloid β-peptide reduces plaque burden and its
associated pathologies
3
. Several hypotheses have been pro-
posed regarding the mechanism of this response
4,5
. Here we re-
port that peripheral administration of antibodies against
amyloid β-peptide, was sufficient to reduce amyloid burden.
Despite their relatively modest serum levels, the passively ad-
ministered antibodies were able to enter the central nervous
system, decorate plaques and induce clearance of preexisting
amyloid. When examined in an ex vivo assay with sections of
PDAPP or Alzheimer disease brain tissue, antibodies against
amyloid β-peptide triggered microglial cells to clear plaques
through Fc receptor-mediated phagocytosis and subsequent
peptide degradation. These results indicate that antibodies can
cross the blood–brain barrier to act directly in the central ner-
vous system and should be considered as a therapeutic ap-
proach for the treatment of Alzheimer disease and other
neurological disorders.
We administered antibodies against amyloid β-peptide (Aβ) by
intraperitoneal injection to mice transgenic for an Aβ precursor
protein (APP) mini-gene driven by a platelet-derived (PD)
growth factor promoter (PDAPP mice). In the first experiment,
8- to 10-month-old heterozygous PDAPP mice (n = 8) received
either PBS, one of two different mouse monoclonal antibodies
against Aβ (10D5 or 21F12) or a polyclonal immunoglobulin
(Ig) fraction obtained from mice immunized with the 42-
amino-acid form of Aβ (pabAβ
1–42
). The mice received weekly
injections of antibody for six months, maintaining a constant
antibody serum concentration throughout the course of the ex-
periment. We used quantitative image analysis to determine
amyloid burden and enzyme-linked immunoassay (ELISA) to
determine Aβ
42
levels in the cortex, as described
3
. Relative to
control-treated mice, the polyclonal immunoglobulin fraction
against Aβ and one of the monoclonal antibodies (10D5) re-
duced plaque burden by 93% and 81%, respectively (Fig. 1a, P <
0.005). There were similar reductions in cortical levels of Aβ
42
by ELISA measurements (6200, 4890 and 13800 ng/g tissue for
10D5, pabAβ
1–42
and PBS, respectively). Although 21F12 seemed
to have a relatively modest effect on plaque burden, there was
no substantial reduction as determined by ELISA measure-
ments (13,580 ng/g). The effect of pabAβ
1-42
on plaque burden
was demonstrated in micrographs of brain sections obtained
from mice having the median level of plaque burden within
their respective groups. Most of the diffuse deposits and many
of the larger compacted plaques were absent in mice treated
with pabAβ
1–42
compared to those in the control groups.
In a second study, we repeated 10D5 treatment and tested two
additional antibodies against Aβ, 3D6 and 16C11 (Fig.1c).
Control groups received either PBS or an irrelevant isotype-
matched antibody (TM2a). The experimental design was the
same as in the previous study except for the age of the mice
(11.5–12 months old). Once again, after 6 months of treatment,
10D5 reduced plaque burden by greater than 80% compared
with that of either the PBS or isotype-matched antibody controls
(P = 0.003). Treatment with 3D6 was equally effective, producing
an 86% reduction in plaque burden (P = 0.003). In contrast,
16C11 failed to have any effect on plaque burden. Unlike ac-
tively immunized mice, mice receiving exogenous antibodies
against Aβ did not demonstrate a T-cell proliferative response to
Aβ when their splenocytes were examined in vitro, indicating
that a T-cell response is not required for amyloid plaque clear-
ance (data not shown).These results indicate that in the absence
of T-cell immunity, antibodies against Aβ peptide are sufficient
to decrease amyloid deposition in PDAPP mice.
To determine whether the peripherally administered antibod-
ies against Aβ had entered the central nervous system (CNS) to
act directly on plaques, we examined brain sections taken from
saline-perfused mice at the end of the second study. We exposed
unfixed cryostat brain sections to a fluorochrome-tagged reagent
against mouse immunoglobulin. Plaques within the brains of
the 10D5- and 3D6-treated groups were strongly ‘decorated’ with
antibody, whereas there was no staining in the 16C11-treated
group (Fig. 2a, top). To show the full extent of plaque deposi-
tion, we immunostained serial sections of each brain with an ex-
ogenous antibody against Aβ, followed by the secondary reagent
(Fig. 2a, bottom). After peripheral administration, 10D5 and 3D6
had gained access to most plaques within the CNS where they
Peripherally administered antibodies against amyloid
β-peptide enter the central nervous system and reduce
pathology in a mouse model of Alzheimer disease
FRÉDÉRIQUE BARD, CATHERINE CANNON, ROBIN BARBOUR, RAE-LYN BURKE, DORA GAMES,
H
ENRY GRAJEDA
, TERESA GUIDO, KANG HU, JIPING HUANG, KELLY JOHNSON-WOOD,
K
AREN KHAN
, DORA KHOLODENKO, MIKE LEE
, IVAN LIEBERBURG, RUTH MOTTER,
M
INH NGUYEN
, FERDIE SORIANO, NICKI VASQUEZ
, KIM WEISS, BRENT WELCH
, PETER SEUBERT,
D
ALE SCHENK & TED YEDNOCK
Elan Pharmaceuticals, 800 Gateway Boulevard, South San Francisco, California 94080, USA
Correspondence should be addressed to F.B.; email: fbard@elanpharma.com
© 2000 Nature America Inc. • http://medicine.nature.com
© 2000 Nature America Inc. • http://medicine.nature.com
NATURE MEDICINE VOLUME 6 NUMBER 8 AUGUST 2000 917
ARTICLES
may have directly triggered amyloid clearance. It is likely that
16C11 also had access to the plaques but was unable to bind (de-
scribed below).
We undertook a separate study to determine if antibody treat-
ment resulted in the clearance of preexisting amyloid or simply
prevented formation of new plaques. We administered 3D6 or
control antibody to 13-month-old heterozygous PDAPP mice,
then examined the brains for total plaque burden after 3 and 35
days of treatment. Although there was no obvious change in the
number of large plaques over the short treatment period (Fig.
2b), there seemed to be a reduction in diffuse amyloid and small
aggregates of Aβ (Fig. 2b, inset). By image analysis of the frontal
cortex, nearly 60% of the small plaques and diffuse amyloid, cor-
responding to pixels with intermediate to low intensity, had
been eliminated during the intervening 32 days of treatment
(Fig. 2c; P = 0.001). These results confirm that the antibody treat-
ment triggered clearance of preexisting amyloid.
To further examine the effect of antibodies on plaque clear-
ance, we established an ex vivo assay in which primary microglial
cells were cultured with unfixed cryostat sections of either
PDAPP mouse or human Alzheimer disease (AD) brains. After 24
hours, we fixed, permeabilized and immunostained the cultures
to follow the fate of amyloid β-peptide, and visualized the exoge-
nous microglial cells with a nuclear stain. In PDAPP brain sec-
tions assayed in the presence of 16C11 (one of the antibodies
against Aβ that was not efficacious in vivo), amyloid β-protein
plaques remained intact and there was no phagocytosis. In con-
trast, after culture of adjacent sections in the presence of 10D5,
amyloid deposits were mostly eliminated and the microglial cells
showed many phagocytic vesicles containing Aβ (Fig. 3a, top).
We obtained identical results with AD brain sections: 10D5 in-
duced phagocytosis of AD plaques, whereas 16C11 was inactive
(Fig. 3a, bottom). Furthermore, the assay was equally effective
with either mouse or human microglial cells, and with mouse,
rabbit or primate antibodies against Aβ (data not shown).
By comparison of six different antibodies tested in both sys-
tems, the ex vivo assay was predictive of in vivo efficacy (Table
1). Antibodies 10D5, 3D6 and pabAβ
1–42
were all active in the ex
vivo assay and demonstrated efficacy in vivo. In contrast,
16C11, 21F12 and the control antibody TM2a were inactive in
Fig. 2 After peripheral administration, 10D5 and 3D6 enter the CNS, bind to Aβ plaques and
trigger amyloid clearance.
a
, Cryostat sections of brains from treated mice were exposed directly
to fluorescently conjugated goat anitbody against mouse immunoglobulin (top). Serial sections
from the same brains were exposed to 10D5 before GaM-Cy3 reagent to show full extent of
plaque burden (bottom). Scale bar represents 100 µm. Anti-Aβ, antibody against Aβ.
b
, Total
plaque burden in the hippocampus and cortex in mice treated for 3 and 35 d with 3D6. The
plaque burden is greatly reduced by 3D6 and 10D5 compared with that of 16C11.Scale bar rep-
resents 120 µm. Insets, high-powered magnification of individual large plaques within the cortex
of the corresponding section (scale bar represents 20µm). There is a lack of amyloid fibrils and
small aggregates surrounding the plaque in the day-35 section.
c
, Quantification of diffuse amy-
loid and small plaques shows a 60% reduction between the 3- and 35-day treatment groups (n =
10/group; P = 0.001).
a b
c
Fig. 1 Aβ burden in the frontal cortex of PDAPP mice is reduced after 6
months of treatment with antibodies against Aβ. a and c, Percentage of
frontal cortex area occupied by Aβ deposits shown in individual mice sorted
by treatment group (n = 8). Horizontal lines indicate median values. b, Brain
sections of the cortex and hippocampus obtained from mice with the median
level of plaque burden in their respective groups. Scale bar represents 250 µm.
a cb
© 2000 Nature America Inc. • http://medicine.nature.com
© 2000 Nature America Inc. • http://medicine.nature.com
918 NATURE MEDICINE VOLUME 6 NUMBER 8 AUGUST 2000
ARTICLES
both assays. Comparison of other antibody characteristics
showed that whereas 16C11 and 21F12 bound to aggregated
synthetic Aβ peptide with high avidity, they were unable to
bind to plaques in unfixed brain sections. This result is consis-
tent with the inability of these two antibodies to decorate
plaques after in vivo administration (as shown for 16C11 in Fig.
2a, top) and explains their inability to trigger plaque clearance.
We found no correlation between antibody efficacy and affin-
ity for soluble Aβ: 3D6 and the inactive antibodies captured
soluble Aβ with moderate to high affinity, whereas 10D5 could
not capture the soluble peptide. These results indicate that
recognition and clearance of deposited Aβ is an important
component of antibody efficacy in vivo and may be more im-
portant than a mechanism involving the prevention of plaque
deposition by recognition of soluble Aβ.
We then used confocal microscopy to confirm that Aβ was in-
ternalized during the course of the ex vivo assay (Fig. 3b). In the
presence of control antibody, the exogenous microglial cells
(red) remained in a confocal plane above the tissue section and
contained no phagocytic vesicles, whereas Aβ (green) remained
in plaques within the tissue plane (Fig. 3b, top). In the presence
of 10D5, nearly all plaque amyloid was contained in vesicles
within the exogenous microglial cells (Fig. 3b, bottom). This re-
sult is consistent with the earlier finding of intracellular Aβ im-
munoreactivity within macrophage and microglial cells in the
CNS of immunized mice
3
. To determine the fate of the internal-
ized peptide, we assessed 10D5-treated cultures by western blot
analysis (Fig. 3b, bottom right). At 1 hour, when no phagocytosis
had yet occurred, reaction with a polyclonal antibody against Aβ
showed a strong 4-kDa band, corresponding to Aβ peptide. Aβ
immunoreactivity decreased at day 1 and was absent by day 3.
There was no degradation of Aβ staining in cultures with control
IgG1. Thus, in contrast to what has been reported for Aβ with
other microglial scavenging pathways
6,7
, antibody-mediated
phagocytosis of Aβ led to its degradation.
To determine if phagocytosis in the ex vivo assay was mediated
by Fc, we prepared F(ab)
2
fragments of 3D6. Although the anti-
body fragments retained their full ability to react with plaques
(Fig. 3c, bottom), they were unable to trigger microglial cell
phagocytosis (Fig. 3c, top). In addition, phagocytosis induced by
the whole 3D6 antibody was in-
hibited by antibodies specific for
Fc receptors on either human or
mouse microglial cells (data not
shown). These results indicate that
in vivo clearance of Aβ occurred
through Fc receptor-mediated
phagocytosis.
In summary, we have shown
that passively administered anti-
bodies against Aβ peptide reduced
the extent of plaque deposition in
a mouse model of Alzheimer dis-
Fig. 3 Fc receptor-mediated phagocytosis of Aβ in an ex vivo assay.
a
,
Mouse microglia cultured with unfixed cryostat sections of PDAPP (top)
or AD (bottom) brain in the presence of antibodies against Aβ. In the
presence of 10D5, Aβ (red) localizes to vesicles within the membrane
boundaries of microglial cells; in the presence of 16C11, plaques remain
intact and there is no evidence of cellular staining. Scale bar represents
10 µm.
b
, Confocal microscopy shows that in the presence of control
IgG1 (top), microglial cells (red) are in a confocal plane above the tissue
section and Aβ (green) remain in plaques within the tissue plane (differ-
ent planes of the same field are shown here). In the presence of 10D5
(bottom left), nearly all Aβ is localized within the microglial cells. Bottom
right, western blot analysis with a polyclonal antibody against Aβ shows
complete degradation of Aβ within 3 d (left margin, molecular size
marker).
c
, Fc receptor-mediated phagocytosis of Aβ, shown by ex vivo
assay in the presence of intact or F(ab)
2
fragments of 3D6 (top). Scale
bar represents 50 µm. Aβ plaques in consecutive sections are equally
‘decorated’ with intact 3D6 and its F(ab)
2
fragments (bottom). Scale bar
represents 300 µm.
Table 1 The ex vivo assay as predictor of in vivo efficacy.
Antibody Isotype Avidity for Affinity for Binding to Ex vivo In vivo
aggregated soluble Aβ efficacy efficacy
Aβ (pM) Aβ (nM) plaques
monoclonal
3D6 IgG2b 470 < 30 + + +
10D5 IgG1 43 no capture + + +
16C11 IgG1 90 110
21F12 IgG2a 500 80
TM2a IgG1
PabAβ
1–42
mix 600 nd + + +
The affinity of 10D5 was too low to capture soluble Aβ and could not be measured in this assay. nd, not done.
PDAPP
mouse brain
AD
Brain
16C11 10D5
a b
IgG110D5
c
Intact antibody F (ab)
2
Ex vivo
Immunostaining
1 hr
1 day
3 days
3.4kD
© 2000 Nature America Inc. • http://medicine.nature.com
© 2000 Nature America Inc. • http://medicine.nature.com
NATURE MEDICINE VOLUME 6 NUMBER 8 AUGUST 2000 919
ARTICLES
ease. Antibody entry into the CNS was not due to abnormal leak-
age of the blood–brain barrier, as there was no increase in vascu-
lar permeability in PDAPP mice. In addition, the concentration
of endogenous immunoglobulins in the brain parenchyma of
aged PDAPP mice was the same as in nontransgenic mice, repre-
senting 0.1% of the antibody concentration in serum (regardless
of isotype; data not shown). Similar findings have been pub-
lished for antibody levels in human cerebrospinal fluid
8
. These
studies indicate that monoclonal antibodies are able to enter the
CNS at therapeutically relevant levels, and that antibodies may
be considered not only for the treatment of Alzheimer disease,
but possibly for other CNS disorders as well.
Methods
Immunization procedures. Monoclonal antibodies against Aβ were raised
against synthetic peptide fragments derived from different regions of Aβ,
and coupled to a carrier protein as described
9,10
. Polyclonal immune sera
were pooled from 100 mice that had been immunized with full length
Aβ
1–42
, and the immunoglobulin fraction was isolated by standard ammo-
nium sulfate precipitation. After purification, all antibodies were dialyzed
against PBS and had a final endotoxin level of < 1 endotoxin unit (EU), as-
measured by the limulus amoebocyte gel clot assay (Associates of Cape
Cod, Cape Cod, Massachusetts). Antibody titers in serum were determined
weekly as described
3
.
Ex vivo assay. Cryostat sections (10 µm in thickness) of PDAPP mouse or
human AD brains (postmortem interval, less than 3 h) were ‘thaw-
mounted’ onto polylysine-coated, round glass coverslips and placed in
wells of 24-well tissue culture plates. The coverslips were washed twice with
assay medium consisting of hybridoma-serum free medium (H-SFM; Life
Technologies) plus 1% FBS, glutamine, penicillin/streptomycin, and 5
ng/ml recombinant mouse GM-CSF granulocyte–monocyte colony-stimu-
lating factor)(R&D Systems, Minneapolis, Minnesota). Antibodies (control
or against Aβ) were added at a 2X concentration (5 µg/ml final) for 1 h.
Microglial cells were then seeded at a density of 0.8 × 10
6
cells/ml assay
medium. In some experiments, medium contained 10 µg/ml soybean
trypsin inhibitor (Life Technologies). The cultures were maintained in a hu-
midified incubator at 37 °C in an atmosphere of 5% CO
2
for 24 h or longer.
After incubation, cultures were fixed with 4% paraformaldehyde and per-
meabilized with 0.1% Triton-X100. Sections were stained with biotinylated
3D6, followed by streptavidin/Cy3 conjugate (Jackson ImmunoResearch,
West Grove, Pennsylvania). Cultures were observed with an inverted fluo-
rescent microscope (TE300; Nikon, Melville, New York) and photomicro-
graphs were taken with a SPOT digital camera using SPOT software
(Diagnostic Instruments, Sterling Heights, Michigan). For western blot
analysis, cultures were extracted with 8 M urea, diluted 1:1 in reducing
tricine sample buffer, and loaded onto a 16% tricine gel (Novex, San Diego,
California). After transfer onto Immobilon, blots were exposed to 5 µg/ml
pabAβ
42
, followed by horseradish peroxidase-conjugated antibody against
mouse, and were developed with an ECL kit (Amersham).
Microglia culture. Microglial cells were obtained from cerebral cortices of
neonate DBA/2N mice (1–3 days old). Cortices were mechanically dissoci-
ated in Hanks’ balanced salt solution with 50 µg/ml DNase I (both from
Sigma). Dissociated cells were filtered through a 100 µm cell strainer
(Falcon, Heidelberg, Germany) and centrifuged at 200g for 5 min. Pellets
were resuspended in growth medium (high-glucose DMEM, 10% FBS and
25 ng/ml recombinant mouse granulocyte–monocyte colony-stimulating
factor), and cells were plated at a density of two brains per T-75 plastic cul-
ture flask. After 7–9 d, the flasks were rotated at 200 rpm using a Lab-Line
orbital shaker with a 19-mm orbit for 2 h at 37 °C. Cell suspensions were
centrifuged at 200g and resuspended in assay medium.
Acknowledgments
We thank K. Hoenow for technical support.
RECEIVED 5 JUNE; ACCEPTED 20 JUNE 2000
1. Games, D. et al. Alzheimer-type neuropathology in transgenic mice overexpressing
V717F β-amyloid precursor protein. Nature 373, 523–527 (1995).
2. Masliah, E. et al. Comparison of neurodegenerative pathology in transgenic mice
overexpressing V717F beta-amyloid precursor protein and Alzheimer’s disease. J.
Neurosci. 16, 5795–57811 (1996).
3. Schenk, D. et al. Immunization with amyloid-β attenuates Alzheimer-disease-like
pathology in the PDAPP mouse. Nature 400, 173–177 (1999).
4. St. George-Hyslop, P.H. & Westaway, D.A. Alzheimer’s disease: antibody clears se-
nile plaques. Nature 400, 116–117 (1999).
5. Duff, K. Curing amyloidosis: will it work in humans? Trends Neurosci. 22, 485-486
(1999).
6. Paresce, D.M., Chung, H. & Maxfield, F.R. Slow degradation of aggregates of the
Alzheimer’s disease amyloid β-protein by microglial cells. J. Biol. Chem. 272,
29390–29397 (1997).
7. Chung, H., Brazil, M.I., Soe, T.T. & Maxfield, F.R. Uptake, degradation, and release
of fibrillar and soluble forms of Alzheimer’s amyloid β-peptide by microglial cells. J.
Biol. Chem. 274, 32301–32308 (1999).
8. Thompson, E.J. & Keir, G. Laboratory investigation of cerebrospinal fluid proteins.
Ann. Clin. Biochem. 27, 425–435 (1990).
9. Johnson-Wood K. et al. Amyloid precursor protein processing and Aβ42 deposition
in a transgenic mouse model of Alzheimer disease. Proc. Natl. Acad. Sci. USA 94,
1550–1555 (1997).
10. Hyman, B.T. et al. Kunitz protease inhibitor-containing amyloid beta protein pre-
cursor immunoreactivity in Alzheimer’s disease. J. Neuropathol. Exp. Neurol. 51,
76–83 (1992).
© 2000 Nature America Inc. • http://medicine.nature.com
© 2000 Nature America Inc. • http://medicine.nature.com
... It is well-known that neuroinflammation contributes to the pathogenesis of AD. Specifically, microglial activation is induced by the presence of amyloid-beta plaques as an attempt to phagocytose and clear these plaques [469,470]. Proinflammatory signaling molecules, such as TNF-α, IL-1β, and IL-6, play central roles in AD pathology [471,472]. TNF-α and IL-1β levels have been reported to be enriched in the brains and blood of AD patients [473][474][475]. ...
Article
Full-text available
Pregnane neuroactive steroids, notably allopregnanolone and pregnenolone, exhibit efficacy in mitigating inflammatory signals triggered by toll-like receptor (TLR) activation, thus attenuating the production of inflammatory factors. Clinical studies highlight their therapeutic potential, particularly in conditions like postpartum depression (PPD), where the FDA-approved compound brexanolone, an intravenous formulation of allopregnanolone, effectively suppresses TLR-mediated inflammatory pathways, predicting symptom improvement. Additionally, pregnane neurosteroids exhibit trophic and anti-inflammatory properties, stimulating the production of vital trophic proteins and anti-inflammatory factors. Androstane neuroactive steroids, including estrogens and androgens, along with dehydroepiandrosterone (DHEA), display diverse effects on TLR expression and activation. Notably, androstenediol (ADIOL), an androstane neurosteroid, emerges as a potent anti-inflammatory agent, promising for therapeutic interventions. The dysregulation of immune responses via TLR signaling alongside reduced levels of endogenous neurosteroids significantly contributes to symptom severity across various neuropsychiatric disorders. Neuroactive steroids, such as allopregnanolone, demonstrate efficacy in alleviating symptoms of various neuropsychiatric disorders and modulating neuroimmune responses, offering potential intervention avenues. This review emphasizes the significant therapeutic potential of neuroactive steroids in modulating TLR signaling pathways, particularly in addressing inflammatory processes associated with neuropsychiatric disorders. It advances our understanding of the complex interplay between neuroactive steroids and immune responses, paving the way for personalized treatment strategies tailored to individual needs and providing insights for future research aimed at unraveling the intricacies of neuropsychiatric disorders.
... PDAPP was the first transgenic AD mouse model with Aβ deposition expressing the mutated form of the human amyloid precursor protein (APP) gene with the Indiana mutation (V717F) under control of the plateletderived growth factor-β promoter [25]. PDAPP transgenic mouse models showed a significant reduction of brain Aβ burden with improved cognition on different murine analogs of the humanized monoclonal antibodies [26,27]. ...
... Aβ is cleared through proteolytic degradation by enzymes [14,15] or internalization into cells via phagocytosis or endocytosis via receptors such as LDL Receptor-Related Protein-1 (LRP-1). Microglia primarily undertake phagocytosis [16], but endocytosis has also been demonstrated in cells such as astrocytes and neurons [17,18]. It has also demonstrated that soluble Aβ is actively transported into the bloodstream through transporters present in the blood-brain barrier (BBB) [19]. ...
Article
Full-text available
Background Alzheimer’s disease (AD) is pathologically characterized by the abnormal accumulation of Aβ and tau proteins. There has long been a keen interest among researchers in understanding how Aβ and tau are ultimately cleared in the brain. The discovery of this glymphatic system introduced a novel perspective on protein clearance and it gained recognition as one of the major brain clearance pathways for clearing these pathogenic proteins in AD. This finding has sparked interest in exploring the potential contribution of the glymphatic/meningeal lymphatic system in AD. Furthermore, there is a growing emphasis and discussion regarding the possibility that activating the glymphatic/meningeal lymphatic system could serve as a novel therapeutic strategy against AD. Objectives Given this current research trend, the primary focus of this comprehensive review is to highlight the role of the glymphatic/meningeal lymphatic system in the pathogenesis of AD. The discussion will encompass future research directions and prospects for treatment in relation to the glymphatic/meningeal lymphatic system.
... Recently, the FDA has conditionally approved two monoclonal antibody medications, lecanemab and aducanumab (FDA Approves Aducanumab, 2021) (FDA Approves Lecanemab, 2023). These are disease modifying treatments that bind to brain amyloid allowing the brain's immune system to remove the amyloid deposits (Bard et al., 2000). Aducanumab works by tagging the beta-amyloid plaques found surrounding brain cells while lecanemab works by preferentially binding amyloid protein protofibrils and toxic oligomers which are the precursors to amyloid plaque formation. ...
Article
Full-text available
This paper covers recent breakthroughs in diagnosis and treatment of neurodegenerative disorders, specifically diagnostic biomarkers and disease modifying treatments for Alzheimer's Disease (AD), Parkinson's Disease (PD) and Amyotrophic Lateral Sclerosis (ALS). Biomarkers in the brain, spinal fluid and blood are helping identify the pathologic misfolded proteins that specifically characterize these neurodegenerative disorders. The signature proteins include beta-amyloid and hyperphosphorylated tau (P-tau) in AD, alpha-synuclein in PD, and recently discovered in ALS, TDP-43. As new biomarkers are validated, there is increasing optimism that innovative disease modifying treatments will prevent, reverse or delay the progression of these neurodegenerative diseases. The growing trend of FDA approval of medications through the Accelerated Approval program based on biomarker indicators is expediting drug development for AD, PD, and ALS. Using this pathway, the FDA has granted conditional approval for an AD treatment that lowers brain amyloid and another AD treatment that is associated in studies with slowing of both cognitive and functional loss. The FDA also granted conditional approval for an ALS treatment demonstrating reduction in a neurodegeneration biomarker. The FDA granted Breakthrough Device designation for a blood test as a diagnostic biomarker for AD and the recent breakthrough in the ability to diagnose PD with an accurate biomarker is a major milestone. Continued collaboration among the drug discovery and clinical research communities is needed to accelerate our understanding of the underlying biology and develop disease modifying treatments for these neurodegenerative disorders.
Chapter
This handbook is currently in development, with individual articles publishing online in advance of print publication. At this time, we cannot add information about unpublished articles in this handbook, however the table of contents will continue to grow as additional articles pass through the review process and are added to the site. Please note that the online publication date for this handbook is the date that the first article in the title was published online. For more information, please read the site FAQs.
Article
Alzheimer’s disease (AD) is an extremely complex and heterogeneous pathology influenced by many factors contributing to its onset and progression, including aging, amyloid-beta (Aβ) plaques, tau fibril accumulation, inflammation, etc. Despite promising advances in drug development, there is no cure for AD. Although there have been substantial advancements in understanding the pathogenesis of AD, there have been over 200 unsuccessful clinical trials in the past decade. In recent years, immunotherapies have been at the forefront of these efforts. Immunotherapy alludes to the immunological field that strives to identify disease treatments via the enhancement, suppression, or induction of immune responses. Interestingly, immunotherapy in AD is a relatively new approach for non-infectious disease. At present, antibody therapy (passive immunotherapy) that targets anti-Aβ aimed to prevent the fibrillization of Aβ peptides and disrupt pre-existing fibrils is a predominant AD immunotherapy due to the continuous failure of active immunotherapy for AD. The most rational and safe strategies will be those targeting the toxic molecule without triggering an abnormal immune response, offering therapeutic advantages, thus making clinical trial design more efficient. This review offers a concise overview of immunotherapeutic strategies, including active and passive immunotherapy for AD. Our review encompasses approved methods and those presently under investigation in clinical trials, while elucidating the recent challenges, complications, successes, and potential treatments. Thus, immunotherapies targeting Aβ throughout the disease progression using a mutant oligomer-Aβ stimulated dendritic cell vaccine may offer a promising therapy in AD.
Article
Full-text available
Overexpression of mutated human amyloid precursor protein (hAPP717V→F) under control of platelet-derived growth factor promoter (PDAPP minigene) in transgenic (tg) mice results in neurodegenerative changes similar to Alzheimer’s disease (AD). To clarify the pathology of these mice, we studied images derived from laser scanning confocal and electron microscopy and performed comparisons between PDAPP tg mice and AD. Similar to AD, neuritic plaques in PDAPP tg mouse contained a dense amyloid core surrounded by anti-hAPP- and anti-neurofilament-immunoreactive dystrophic neurites and astroglial cells. Neurons were found in close proximity to plaques in PDAPP tg mice and, to a lesser extent, in AD. In PDAPP tg mice, and occasionally in AD, neuronal processes contained fine intracellular amyloid fibrils in close proximity to the rough endoplasmic reticulum, coated vesicles, and electron-dense material. Extracellular amyloid fibrils (9–11 nm in diameter) were abundant in PDAPP tg and were strikingly similar to those observed in AD. Dystrophic neurites in plaques of PDAPP tg mouse and AD formed synapses and contained many dense multilaminar bodies and neurofilaments (10 nm). Apoptotic-like figures were present in the tg mice. No paired helical filaments have yet been observed in the heterozygote PDAPP tg mice. In summary, this study shows that PDAPP tg mice develop massive neuritic plaque formation and neuronal degeneration similar to AD. These findings show that overproduction of hAPP717V→F in tg mice is sufficient to cause not only amyloid deposition, but also many of the complex subcellular degenerative changes associated with AD.
Article
Full-text available
ALZHEIMER'S disease (AD) is the most common cause of progressive intellectual failure in aged humans. AD brains contain numerous amyloid plaques surrounded by dystrophic neurites, and show profound synaptic loss, neurofibrillary tangle formation and gliosis. The amyloid plaques are composed of amyloid beta-peptide (A beta), a 40-42-amino-acid fragment of the beta-amyloid precursor protein (APP)(1). A primary pathogenic role for APP/A beta is suggested by missense mutations in APP that are tightly linked to autosomal dominant forms of AD(2,3). A major obstacle to elucidating and treating AD has been the lack of an animal model. Animals transgenic for APP have previously failed to show extensive AD-type neuropathology(4-10), but we now report the production of transgenic mice that express high levels of human mutant APP (with valine at residue 717 substituted by phenylalanine) and which progressively develop many of the pathological hallmarks of AD, including numerous extracellular thioflavin S-positive AP deposits, neuritic plaques, synaptic loss, astrocytosis and microgliosis. These mice support a primary role for APP/A beta in the genesis of AD and could provide a preclinical model for testing therapeutic drugs.
Article
Full-text available
The PDAPP transgenic mouse, which overexpresses human amyloid precursor protein (APP717V-->F), has been shown to develop much of the pathology associated with Alzheimer disease. In this report, levels of APP and its amyloidogenic metabolites were measured in brain regions of transgenic mice between 4 and 18 months of age. While absolute levels of APP expression likely contribute to the rate of amyloid beta-peptide (Abeta) deposition, regionally specific factors also seem important, as homozygotic mice express APP levels in pathologically unaffected regions in excess of that measured in certain amyloid plaque-prone regions of heterozygotic mice. Regional levels of APP and APP-beta were nearly constant at all ages, while A beta levels dramatically and predictably increased in brain regions undergoing histochemically confirmed amyloidosis, most notably in the cortex and hippocampus. In hippocampus, A beta concentrations increase 17-fold between the ages of 4 and 8 months, and by 18 months of age are over 500-fold that at 4 months, reaching an average level in excess of 20 nmol of A beta per g of tissue. A beta1-42 constitutes the vast majority of the depositing A beta species. The similarities observed between the PDAPP mouse and human Alzheimer disease with regard to A beta42 deposition occurring in a temporally and regionally specific fashion further validate the use of the model in understanding processes related to the disease.
Article
Full-text available
Microglia are immune system cells associated with senile plaques containing β-amyloid (Aβ) in Alzheimer’s disease. Although microglia are an integral part of senile plaques, their role in the development of Alzheimer’s disease is not known. Because microglia are phagocytic cells, it has been suggested that microglia may function as plaque-attacking scavenger cells. Microglia bind and internalize microaggregates of Aβ that resemble those present in dense Alzheimer’s disease plaques. In this study, we compared the degradation by microglia of Aβ microaggregates with the degradation of two other proteins, acetylated low density lipoprotein and α2-macroglobulin. We found that the majority of the internalized Aβ in microaggregates was undegraded 72 h after uptake, whereas 70–80% of internalized acetylated low density lipoprotein or α2-macroglobulin was degraded and released from cells in trichloroacetic acid-soluble form after 4 h. In the continued presence of fluorescent Aβ microaggregates for 4 days, microglia took up huge amounts of Aβ and became engorged with undigested material. These data suggest that microglia can slowly degrade limited amounts of Aβ plaque material, but the degradation mechanisms can be overwhelmed by larger amounts of Aβ.
Article
The amyloid beta-protein (beta/A4) that is deposited in senile plaques and in cerebral vessels in Alzheimer's disease (AD) is derived from a larger membrane-associated glycoprotein, the amyloid beta-protein precursor (APP). The gene encoding APP produces at least four major transcripts. Three of the four transcripts contain an alternatively-spliced exon encoding a Kunitz protease inhibitor domain (KPI). We now report the results of a series of experiments using novel immunohistochemical reagents to anatomically localize beta/A4, APP, and KPI-containing forms of APP (APP-KPI) in the hippocampal formation and temporal neocortex. A new monoclonal antibody against beta/A4 recognized senile plaques and vascular amyloid, but no cellular elements. Anti-APP and anti-KPI monoclonal antibodies stained neurons, including proximal axons and dendrites. The neuritic component of some plaques in patients with AD and in elderly control individuals were also immunoreactive for both APP and APP-KPI. Quantitative assessment of senile plaques in temporal neocortex showed that, on average, about one-third of beta/A4 immunoreactive plaques stained with either anti-APP or anti-KPI. Amyloid beta-protein precursor and APP-KPI immunoreactivity were also found in the white and grey matter vessels of both AD patients and control individuals. These results suggest that KPI-containing forms of APP are present in dystrophic neurites of senile plaques, and normally in neurons, neuronal processes, and in the vascular compartment in the brain. Thus, APP-KPI is in a position to be intimately associated with beta/A4 deposition in the neuropil, in plaques and in amyloid angiopathy.
Article
Analysis of CSF proteins is useful in the diagnosis and management of neurological diseases in the following situations: In inflammatory conditions when there is breakdown of blood-CSF barrier integrity. Meningitis is a medical emergency, with CSF total protein measurement being only a screening test. In the detection of immune responses within the CNS. This is by far the most important application in a routine clinical setting, as it is now a firmly established criterion in the diagnosis of multiple sclerosis. Oligoclonal bands restricted to the CSF are the only reliable indicators of intrathecal immunoglobulin G synthesis and are practically always associated with inflammatory disease of the CNS. The method of choice for detecting oligoclonal bands is isoelectric focusing with immunofixation. Quantitative measurement of IgG in the CSF is of no value in diagnostic pathology. In destructive brain diseases when brain-specific proteins are released into the CSF, measurement of these proteins can give prognostic information.
Article
The amyloid beta protein (beta/A4) that is deposited in senile plaques and in cerebral vessels in Alzheimer's disease (AD) is derived from a larger membrane-associated glycoprotein, the amyloid beta protein precursor (APP). The gene encoding APP produces at least four major transcripts. Three of the four transcripts contain an alternatively-spliced exon encoding a Kunitz protease inhibitor domain (KPI). We now report the results of a series of experiments using novel immunohistochemical reagents to anatomically localize beta/A4, APP, and KPI-containing forms of APP (APP-KPI) in the hippocampal formation and temporal neocortex. A new monoclonal antibody against beta/A4 recognized senile plaques and vascular amyloid, but no cellular elements. Anti-APP and anti-KPI monoclonal antibodies stained neurons, including proximal axons and dendrites. The neuritic component of some plaques in patients with AD and in elderly control individuals were also immunoreactive for both APP and APP-KPI. Quantitative assessment of senile plaques in temporal neocortex showed that, on average, about one-third of beta/A4 immunoreactive plaques stained with either anti-APP or anti-KPI. Amyloid beta protein precursor and APP-KPI immunoreactivity were also found in the white and grey matter vessels of both AD patients and control individuals. These results suggest that KPI-containing forms of APP are present in dystrophic neurites of senile plaques, and normally in neurons, neuronal processes, and in the vascular compartment in the brain. Thus, APP-KPI is in a position to be intimately associated with beta/A4 deposition in the neuropil, in plaques and in amyloid angiopathy.
Article
Overexpression of mutated human amyloid precursor protein (hAPP717V-->F) under control of platelet-derived growth factor promoter (PDAPP minigene) in transgenic (tg) mice results in neurodegenerative changes similar to Alzheimer's disease (AD). To clarify the pathology of these mice, we studied images derived from laser scanning confocal and electron microscopy and performed comparisons between PDAPP tg mice and AD. Similar to AD, neuritic plaques in PDAPP tg mouse contained a dense amyloid core surrounded by anti-hAPP- and antineurofilament-immunoreactive dystrophic neurites and astroglial cells. Neurons were found in close proximity to plaques in PDAPP tg mice and, to a lesser extent, in AD. In PDAPP tg mice, and occasionally in AD, neuronal processes contained fine intracellular amyloid fibrils in close proximity to the rough endoplasmic reticulum, coated vesicles, and electron-dense material. Extracellular amyloid fibrils (9-11 nm in diameter) were abundant in PDAPP tg and were strikingly similar to those observed in AD. Dystrophic neurites in plaques of PDAPP tg mouse and AD formed synapses and contained many dense multilaminar bodies and neurofilaments (10 nm). Apoptotic-like figures were present in the tg mice. No paired helical filaments have yet been observed in the heterozygote PDAPP tg mice. In summary, this study shows that PDAPP tg mice develop massive neuritic plaque formation and neuronal degeneration similar to AD. These findings show that overproduction of hAPP717V-->F in tg mice is sufficient to cause not only amyloid deposition, but also many of the complex subcellular degenerative changes associated with AD.
Article
Amyloid-beta peptide (Abeta) seems to have a central role in the neuropathology of Alzheimer's disease (AD). Familial forms of the disease have been linked to mutations in the amyloid precursor protein (APP) and the presenilin genes. Disease-linked mutations in these genes result in increased production of the 42-amino-acid form of the peptide (Abeta42), which is the predominant form found in the amyloid plaques of Alzheimer's disease. The PDAPP transgenic mouse, which overexpresses mutant human APP (in which the amino acid at position 717 is phenylalanine instead of the normal valine), progressively develops many of the neuropathological hallmarks of Alzheimer's disease in an age- and brain-region-dependent manner. In the present study, transgenic animals were immunized with Abeta42, either before the onset of AD-type neuropathologies (at 6 weeks of age) or at an older age (11 months), when amyloid-beta deposition and several of the subsequent neuropathological changes were well established. We report that immunization of the young animals essentially prevented the development of beta-amyloid-plaque formation, neuritic dystrophy and astrogliosis. Treatment of the older animals also markedly reduced the extent and progression of these AD-like neuropathologies. Our results raise the possibility that immunization with amyloid-beta may be effective in preventing and treating Alzheimer's disease.
Article
People suffering from Alzheimer's disease develop a progressive dementia in adulthood, accompanied by three main structural changes in the brain: diffuse loss of neurons in the hippocampus and neocortex; accumulation of intracellular protein deposits termed neurofibrillary tangles; and accumulation of extracellular protein deposits termed amyloid or senile plaques, surrounded by misshapen nerve terminals (dystrophic neurites). A main constituent of these amyloid plaques is the amyloid- peptide (A), a 40-42-amino-acid protein that is produced through cleavage of the -amyloid precursor protein (APP). Although Alzheimer's disease can be treated, we can currently neither prevent nor cure it. On page 173 of this issue, however, Schenk et al.1 show that, in a mouse model of Alzheimer's disease, immunization with A inhibits the formation of amyloid plaques and the associated dystrophic neurites.