Sei sulla pagina 1di 16

The n e w e ng l a n d j o u r na l of m e dic i n e

review article

Mechanisms of Disease

Alzheimers Disease
Henry W. Querfurth, M.D., Ph.D., and Frank M. LaFerla, Ph.D.

M
ore than 35 million people worldwide 5.5 million in the From the Department of Neurology, Car-
itas St. Elizabeths Medical Center, Brigh-
United States have Alzheimers disease, a deterioration of memory and
ton, MA (H.W.Q.); the Department of
other cognitive domains that leads to death within 3 to 9 years after diag- Neurology, Tufts Medical Center, Boston
nosis. Alzheimers disease is the most common form of dementia, accounting for (H.W.Q.); the Department of Neurology,
Rhode Island Hospital and the Warren
50 to 56% of cases at autopsy and in clinical series. Alzheimers disease combined
Alpert Medical School at Brown Univer-
with intracerebral vascular disease accounts for another 13 to 17% of cases. sity, Providence (H.W.Q.); and the Depart-
The principal risk factor for Alzheimers disease is age. The incidence of the ment of Neurobiology and Behavior, Uni-
versity of California, Irvine, Irvine (F.M.L.).
disease doubles every 5 years after 65 years of age, with the diagnosis of 1275 new
Address reprint requests to Dr. Querfurth
cases per year per 100,000 persons older than 65 years of age.1 Data on centenarians at the Department of Neurology, Rhode
show that Alzheimers disease is not necessarily the outcome of aging2; neverthe- Island Hospital, 563 Eddy St., Providence,
RI 02903-4923, or at henry_querfurth@
less, the odds of receiving the diagnosis of Alzheimers disease after 85 years of
brown.edu.
age exceed one in three. As the aging population increases, the prevalence will
approach 13.2 to 16.0 million cases in the United States by mid-century.3 N Engl J Med 2010;362:329-44.
Many molecular lesions have been detected in Alzheimers disease, but the over- Copyright 2010 Massachusetts Medical Society.

arching theme to emerge from the data is that an accumulation of misfolded


proteins in the aging brain results in oxidative and inflammatory damage, which
in turn leads to energy failure and synaptic dysfunction.

Pro tein A bnor m a l i t ie s in A l zheimers Dise a se

-Amyloid
Cerebral plaques laden with -amyloid peptide (A) and dystrophic neurites in
neocortical terminal fields as well as prominent neurofibrillary tangles in medial
temporal-lobe structures are important pathological features of Alzheimers dis-
ease. Loss of neurons and white matter, congophilic (amyloid) angiopathy, in-
flammation, and oxidative damage are also present.
A peptides are natural products of metabolism consisting of 36 to 43 amino
acids. Monomers of A40 are much more prevalent than the aggregation-prone and
damaging A42 species. -amyloid peptides originate from proteolysis of the amy-
loid precursor protein by the sequential enzymatic actions of beta-site amyloid
precursor proteincleaving enzyme 1 (BACE-1), a -secretase, and -secretase, a pro-
tein complex with presenilin 1 at its catalytic core4 (Fig. 1). An imbalance between
production and clearance, and aggregation of peptides, causes A to accumulate,
and this excess may be the initiating factor in Alzheimers disease. This idea,
called the amyloid hypothesis, is based on studies of genetic forms of Alzheimers
disease, including Downs syndrome,5 and evidence that A42 is toxic to cells.6,7
A spontaneously self-aggregates into multiple coexisting physical forms. One
form consists of oligomers (2 to 6 peptides), which coalesce into intermediate as-
semblies8,9 (Fig. 1). -amyloid can also grow into fibrils, which arrange themselves
into -pleated sheets to form the insoluble fibers of advanced amyloid plaques.
Soluble oligomers and intermediate amyloids are the most neurotoxic forms of

n engl j med 362;4 nejm.org january 28, 2010 329


The New England Journal of Medicine
Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

A
Nonamyloidogenic Amyloidogenic

sAPP
sAPP
A
p3
-Secretase -Secretase BACE-1 -Secretase

GPI
Cholesterol

Cellular
membrane

Cytosol
C83 APP C99 AICD
C4950 Gene expression
A in nucleus
Non-Raft Raft

Figure 1. Processing of Amyloid Precursor Protein.


In Panel A, cleavage by -secretase interior to the -amyloid peptide (A) sequence initiates nonamyloidogenic processing. A large am-
yloid precursor protein (sAPP) ectodomain is released, leaving behind an 83-residue carboxy-terminal fragment. C83 is then digested by
-secretase, liberating extracellular p3 and the amyloid intracellular domain (AICD). Amyloidogenic processing is initiated by -secretase
beta-site amyloid precursor proteincleaving enzyme 1 (BACE-1), releasing a shortened sAPP. The retained C99 is also a -secretase
substrate, generating A and AICD. -Secretase cleavage occurs within the cell membrane in a unique process termed regulated intra-
membranous proteolysis. sAPP and sAPP are secreted APP fragments after -secretase and -secretase cleavages, respectively. AICD
is a short tail (approximately 50 amino acids) that is released into the cytoplasm after progressive -to- cleavages by -secretase. AICD
is targeted to the nucleus, signaling transcription activation. Lipid rafts are tightly packed membrane micro- environments enriched in
sphingomylelin, cholesterol, and glycophosphatidylinositol (GPI)anchored proteins. Soluble A is prone to aggregation. In Panel B, left
inset, protofibrils (upper) and annular or porelike profiles (lower) are intermediate aggregates. (Photomicrographs courtesy of Hilal
Lashuel, Ph.D.) In the right inset, self-association of 2 to 14 A monomers into oligomers is dependent on concentration (left immuno-
blot). In the right immunoblot, oligomerization is promoted by oxidizing conditions (lane 2) and divalent metal conditions (lane 3).
COLOR FIGURE
(Immuno blots courtesy of Hongwei Zhou, Ph.D.)
Draft 4 01/12/10
Author Querfurth
Fig # 1 A.10 In brain-slice preparations, dimers and brain, not the total A burden.13 Neuronal activa-
Title trimers of A are toxic to synapses.11,12 The se- tion rapidly increases A secretion at the syn-
ME
verity of the cognitive defect in Alzheimers dis- apse, a process tied to the normal release of vesi-
DE
Artist SBL ease correlates with levels of oligomers in the cles containing neurotransmitters. Physiologic
AUTHOR PLEASE NOTE:
Figure has been redrawn and type has been reset
Please check carefully
330
Issue date n engl j med 362;4 nejm.org january 28, 2010

The New England Journal of Medicine


Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

levels of synaptic A may dampen excitatory trans- may be inert, however, since decreases in axonal
mission and prevent neuronal hyperactivity.14 transport and neuron number are independent
The proteases neprilysin and insulin-degrad- of the burden of neurofibrillary tangles.30 These
ing enzyme regulate steady-state levels of A. helical filaments sequester toxic intermediate tau
Neprilysin, a membrane-anchored zinc endopep- species, a process that may be protective.31
tidase, degrades A monomers and oligomers.15 More than 30 mutations of Tau on chromo-
A reduction in neprilysin causes accumulation of some 17 have been detected in frontotemporal
cerebral A.16 Insulin-degrading enzyme, a thiol dementia with parkinsonism.32 By contrast, Tau
metalloendopeptidase, degrades small peptides mutations do not occur in Alzheimers disease,
such as insulin and monomeric A.17 In mice, and the extent of neuron loss is out of propor-
deletion of insulin-degrading enzyme reduces A tion to the number of neurofibrillary tangles.33
degradation by more than 50%.18 Conversely, Nevertheless, increased levels of phosphorylated
overexpression of neprilysin or insulin-degrading and total tau in the cerebrospinal fluid correlate
enzyme prevents plaque formation.19 with reductions in scores on cognitive examina-
Clinical trials of a -secretase inhibitor (LY450139) tions.34 Elevated levels of phosphotau amino acids
(ClinicalTrials.gov number, NCT00765115),20 aggre- T181, T231, and total tau in the cerebrospinal
gation blockers, vaccination with A, and mono- fluid together constitute a biomarker test with
clonal antibodies against various A epitopes are good accuracy for predicting incipient Alzhei-
in progress. The antibodies bind A, thereby trig- mers disease in patients with mild cognitive im-
gering complement and Fc-receptormediated pairment.35 Experimental evidence indicates that
phagocytosis by microglia, or enhance clearance A accumulation precedes and drives tau aggre-
of A, or both.21 Vaccination in a phase 2a trial gation.36-38 Moreover, A-induced degeneration
(NCT00021723)22 resulted in encephalitis,23 and of cultured neurons and cognitive deficits in mice
follow-up of immunized patients showed no cog- with an Alzheimers diseaselike illness require
nitive or survival benefit despite diminution of the presence of endogenous tau.39,40
plaques.24 A phase 2 trial of passive immunization Increased oxidative stress, the impaired pro-
resulted in vasogenic cerebral edema in some pa- tein-folding function of the endoplasmic reticu-
tients (NCT00112073). Phase 3 trials of two mono- lum, and deficient proteasome-mediated and au-
clonal antibodies against A (NCT00574132 and tophagic-mediated clearance of damaged proteins
NCT00904683) and of 10% intravenous immune all of which are also associated with aging
globulin are under way (NCT00818662). accelerate the accumulation of amyloid and
tau proteins in Alzheimers disease.41,42 Agents
Tau capable of counteracting these changes are not
Neurofibrillary tangles, which are filamentous in- available, but trials of small-molecule inhibitors
clusions in pyramidal neurons, occur in Alzhei- of -amyloid (e.g., scylloinositol) (NCT00568776)
mers disease and other neurodegenerative dis- and tau oxidation and aggregation (e.g., methyl-
orders termed tauopathies.25 The number of ene blue) (NCT00568776) are under way.43 Poly-
neurofibrillary tangles is a pathologic marker of phenolic extracts from grape seeds (e.g., resvera-
the severity of Alzheimers disease. The major trol), which stimulate aging-suppressor genes,
component of the tangles is an abnormally hyper- also show promise as therapeutic agents.44
phosphorylated and aggregated form of tau.
Normally an abundant soluble protein in axons, The S y na pse in A l zheimers
tau promotes assembly and stability of microtu- Dise a se
bules and vesicle transport. Hyperphosphorylated
tau is insoluble, lacks affinity for microtubules, Synaptic Failure
and self-associates into paired helical filament Alzheimers disease may be primarily a disorder
structures (Fig. 2). Enzymes that add and those of synaptic failure.45 Hippocampal synapses be-
that remove phosphate residues regulate the ex- gin to decline in patients with mild cognitive im-
tent of tau phosphorylation.26 pairment (a limited cognitive deficit often pre-
Like A oligomers, intermediate aggregates of ceding dementia) in whom remaining synaptic
abnormal tau molecules are cytotoxic27 and im- profiles show compensatory increases in size.46
pair cognition.28,29 Insoluble helical filaments In mild Alzheimers disease, there is a reduction

n engl j med 362;4 nejm.org january 28, 2010 331


The New England Journal of Medicine
Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Fyn GSK-3 Akt PKA CaMKII GSK-3


cdk5 MARK cdk5
MAPK MAPK
Tau and Microtubule
microtubule-associated P P P P P P P
proteins KXGS
Y SP SP S SP SP
18 46 184-202 214 235 262-356 394,404
E2 E3 R1 R2 R3 R4
1 441
50,69 181, 205-231 403
TP TP MBD T
P P P

R1
R3
Cdk5/p25 _ R2 +
GSK-3
Hyperphosphorylation R4
of tau

Neurofibrillary
tangles

Paired helical
filaments

Destabilized microtubules
(impaired axonal transport)

Hyperphosphorylated
tau proteins

Figure 2. Tau Structure and Function.


Four repeat sequences (R1-R4) make up the microtubule-binding domain (MBD) of tau. Normal phosphorylation of tau occurs on serine
(S; inset, above horizontal bar) and threonine (T; inset, below horizontal bar) residues, numbered according to their position in the full tau
sequence.COL When
O R Ffollowed
I G U R E by proline (P), these amino acids are phosphorylated by glycogen synthase kinase 3 (GSK-3), cyclin-depen-
dent kinase (cdk5) and its activator subunit p25, or mitogen-activated protein kinase (MAPK). Nonproline-directed kinases Akt, Fyn,
Draft 3 01/11/10
protein kinase A (PKA), calciumcalmodulin protein kinase 2 (CaMKII), and microtubule affinity-regulating kinase (MARK) are also
shown. Querfurth
Author
KXGS (denoting lysine, an unknown or other amino acid, glycine, and serine) is a target motif. Hyperphosphorylated sites specif-
Fig # 2
ic to paired helical filament tau in Alzheimers disease tend to flank the MBD. Tau binding promotes microtubule assembly and stability.
Title
ME
Excessive kinase, reduced phosphatase activities, or both cause hyperphosphorylated tau to detach and self-aggregate and microtubules
DE
to stabilize.
Artist SBL
AUTHOR PLEASE NOTE:
Figure has been redrawn and type has been reset
Please check carefully

Issue date

332 n engl j med 362;4 nejm.org january 28, 2010

The New England Journal of Medicine


Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

Paired helical APP


filaments
APP
nAChr ACh TrkAr

Glutamate
BDNF

VGCC TrkBr
p75NTr
AMPAr nAChr NMDAr NMDAr

Ca2+ leak
Ca2+ influx (influx)
Calpain
+Calcineurin

+pCaMKII pCREB

NGF

LTP LTD

Figure 3. Synaptic Dysfunction in Alzheimers Disease.


Synaptic loss correlates best with cognitive decline in Alzheimers disease. A control synapse is shown at the top of the
figure. At the bottom of the figure, an Alzheimers disease synapse depicting the pleiotropic effects of the -amyloid
peptide (A) is shown. Rings represent synaptic vesicles. Experimental application and expression of A, especially
oligomers, impair synaptic plasticity by altering the balance between long-term potentiation (LTP) and long-term de-
pression (LTD) and reducing the numbers of dendritic spines. At high concentrations, oligomers may suppress basal
synaptic transmission. A facilitates endocytosis of receptors of N-methyl-d-aspartate (NMDAr) and -amino-3-
hydroxy-5-methyl-4-isoxazole propionic acid (AMPAr). A also binds to the receptors of p75 neurotrophin (p75NTr)
and brain-derived neurotrophic factor (the BDNF receptor, also known as the tyrosine kinase B receptor [trkBr]), ex-
acerbating a situation in which levels of BDNF and nerve growth factor (NGF) are already suppressed. A impairs
nicotinic acetylcholine (ACh) receptor (nAChr) signaling and ACh release from the presynaptic terminal. Numbers of
COLOR FIGURE
hippocampal synapses decrease in mild cognitive impairment in which remaining synaptic profiles show compensa-
Draft 3 01/11/10
tory increases in size. APP denotes amyloid precursor protein, pCaMKII phosphorylated calciumcalmodulindepen-
thor Querfurth dent protein kinase 2, pCREB phosphorylated cyclic AMP response-element-binding protein, trkAr tyrosine kinase A
# 3 receptor, and VGCC voltage-gated calcium channel.
e
E
of about 25% in the presynaptic vesicle protein Basal transmission of single impulses and
st SBL
AUTHOR PLEASE NOTE:
synaptophysin.47 With advancing disease, synaps- long-term potentiation, an experimental indica-
eshasare
Figure has been redrawn and type
Please check carefully
beendisproportionately
reset lost relative to neurons, tor of memory formation at synapses, are im-
ue date and this loss is the best correlate with demen- paired in plaque-bearing mice with Alzheimers
tia.48-50 Aging itself causes synaptic loss,51 which disease and after A peptide has been applied to
particularly affects the dentate region of the hip- brain slices.11,53 Subsequent to this impairment,
pocampus.52 signaling molecules important to memory are in-

n engl j med 362;4 nejm.org january 28, 2010 333


The New England Journal of Medicine
Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

hibited. Disruptions of the release of presynaptic of nicotinic acetylcholine receptors or M1 receptors


neurotransmitters and postsynaptic glutamate- limits tau phosphorylation.69,70 Although cholin-
receptor ion currents54,55 occur partially as a result esterase inhibitors improve neurotransmission and
of endocytosis of N-methyl-D-aspartate (NMDA) provide mild palliative relief in Alzheimers dis-
surface receptors56 and endocytosis of -amino- ease, they lose efficacy over time. The use of
3-hydroxy-5-methyl-4-isoxazole propionic acid agonists and modulators of -7 nicotinic acetyl-
surface receptors57 (Fig. 3). The latter further choline receptors is under investigation. Clinical
weakens synaptic activity by inducing a lasting trials of selective M1 agonists have shown im-
reduction in currents after a high-frequency stim- provements in cognition71 and reduced A levels
ulus train. A similar shift in the balance be- in the cerebrospinal fluid,72 but these agents are
tween potentiation and depression in synapses toxic.
occurs with normal aging. Intraneuronal A can
trigger these synaptic deficits even earlier.58 Mi t o chondr i a l Dysf unc t ion

Depletion of Neurotrophin A is a potent mitochondrial poison, especially


and Neurotransmitters affecting the synaptic pool.73 In Alzheimers dis-
Neurotrophins promote proliferation, differentia- ease, exposure to A inhibits key mitochondrial
tion, and survival of neurons and glia, and they enzymes in the brain and in isolated mitochon-
mediate learning, memory, and behavior. The dria.74,75 Cytochrome c oxidase is specifically at-
normally high levels of neurotrophin receptors in tacked.76 Consequently, electron transport, ATP
cholinergic neurons in the basal forebrain are se- production, oxygen consumption, and mitochon-
verely reduced in late-stage Alzheimers disease drial membrane potential all become impaired.
(Fig. 3). Injection of nerve growth factor can res- The increase in mitochondrial superoxide radical
cue basal neurons in animal models,59 and a formation and conversion into hydrogen peroxide
phase 1 trial of treatment with the NGF gene in cause oxidative stress, release of cytochrome c,
Alzheimers disease showed improvement in cog- and apoptosis (Fig. 4).
nition and brain metabolism.60 In Alzheimers The accumulation of A within structurally
disease and mild cognitive impairment, levels of damaged mitochondria isolated from the brains
brain-derived neurotrophic factor (BDNF), a mem- of patients with Alzheimers disease77 and trans-
ber of the neurotrophin family, are depressed,61 genic brains76 is consistent with other evidence
a finding reproduced experimentally with A42 of intraneuronal A in Alzheimers disease.78
oligomers.62 BDNF treatment in rodents and non- Alcohol dehydrogenase is one such mitochondrial-
human primates supports neuronal survival, syn- binding target of A.79 Similar changes occur
aptic function, and memory,63 suggesting that in normal cells that have been repopulated with
BDNF replacement is another option for the treat- mitochondrial DNA (mtDNA) from patients
ment of Alzheimers disease.64 with sporadic Alzheimers disease.80 Both in Alz-
The deficiency of cholinergic projections in heimers disease and in the normal aging pro-
Alzheimers disease has been linked to the build- cess, mtDNA sustains high levels of oxidative
up of A and tau. Presynaptic -7 nicotinic ace- damage.77 This instability and the irreparability
tylcholine receptors are essential for cognitive of the brains mitochondrial genome allow the
processing, and their levels increase in early Alz- gradual accumulation of mtDNA mutations.81
heimers disease,65 before decreasing later.66 Ex- Fragmentation (or fission) of mitochondria from
perimental studies show that A binds to -7 the oxidation of a dynamin-like transporter pro-
nicotinic acetylcholine receptors, impairing the tein may cause synapse loss in Alzheimers
release of acetylcholine and maintenance of long- disease.82 The antihistamine dimebolin hydro-
term potentiation.67 The level of muscarinic ace- chloride, a putative mitochondrial stimulant, has
tylcholine receptors, or receptor coupling, is re- been reported to improve cognition and behav-
duced in the brains of patients with Alzheimers ior in patients with mild-to-moderate Alzhei-
disease. Pharmacologic stimulation of the post- mers disease.83
synaptic muscarinic type 1 (M1) acetylcholine
receptors activates protein kinase C, favoring Oxidative Stress
processing of amyloid precursor protein that Dysfunctional mitochondria release oxidizing free
does not yield amyloid.68 Furthermore, activation radicals, and in Alzheimers disease and the nor-

334 n engl j med 362;4 nejm.org january 28, 2010

The New England Journal of Medicine


Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

NMDAr MAC

Ca2+
A Na+/K+ GLUT1, 4
ATPases
HNE

Ca2+ ROS RNS


Na2+ Glucose O2s NO iNOS Ca2+
Ca2+ A Ca2+ Na2+
OH NOOs Ca2+
Tau phosphorylation Lipid peroxidation products:
and aggregation HNE, isoprostanes
Aging and
oxidation

A
OH Complex I

Complex III
mtDNA
Complex IV
damage

MPP -Ketoglutarate and


m pyruvate dehydrogenases

Cytochrome c
JNK Apoptosis-
O2 s Proteolysis and
NO p38 initiating factor
cell death

Caspase 3
p53

Figure 4. Oxidative Stress and Mitochondrial Failure.


A -amyloid peptide (A)centric scheme depicts production of reactive oxygen species (ROS) and reactive nitrogen species (RNS). Their
peroxidative attack on cell and organelle membrane lipids yields the mitochondrial toxins hydroxynonenal (HNE) and malondialdehyde.
Oxidative damage to membrane-bound, ion-specific ATPases and stimulation of calcium (Ca2+) entry mechanisms for example, glu-
tamate (N-methyl-d-aspartate [NMDA]) receptors (NMDAr), membrane-attack complex (MAC) of complement, and ion-selective amy-
loid pore formation cause cytosolic and mitochondrial Ca2+ overload. Cellular A directly attacks electron transport complex IV (cyto-
chrome c oxidase) and key Krebs-cycle enzymes (-ketoglutarate and pyruvate dehydrogenase) and damages mitochondrial DNA (mtDNA),
leading to fragmentation. Lipid peroxidation products also promote tau phosphorylation and aggregation, which in turn inhibit complex I.
Exaggerated amounts of ROS and RNS are generated at complexes I and III. As the mitochondrial membrane potential (MPP) collapses
andCpermeability-transition
OLOR FIGURE pores (m) open, caspases are activated. A also induces the stress-activated protein kinases p38 and c-jun
N-terminal kinase (JNK), as well as p53, which are further linked with apoptosis. Substrate deficiencies, notably NADH and glucose,
Draft 4 01/12/10
combine with electron transport uncoupling to further diminish ATP production. Alcohol dehydrogenase was recently identified as the
Author Querfurth
mitochondrial-binding target for A. Endoplasmic reticulum contributions are shown. GLUT1, 4 denotes glucose transporter 1, 4.
Fig # 4
Title
ME
mal aging brain, they cause considerable oxida- is a prime initiator of this damage. The receptor
DE 84,85
Artist tive stress.
SBL Experimental models show that for advanced glycation end products mediates
markers AUTHORof oxidative
PLEASE NOTE: damage precede pathologi- As pro-oxidant effects on neural, microglial,
Figure has been redrawn and type has been reset
cal changes. 86 A, a potent generator of reactive
Please check carefully and cerebrovascular cells.89 Mitochondrial hydro-
Issue oxygen
date species87 and reactive nitrogen species,88 gen peroxide readily diffuses into the cytosol to

n engl j med 362;4 nejm.org january 28, 2010 335


The New England Journal of Medicine
Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

participate in metal ioncatalyzed hydroxyl radi- protein kinaseextracellular signal-regulated ki-


cal formation. Stimulated microglia are a major nase pathway,103 but it is unclear whether signal-
source of the highly diffusible nitric oxide radi- ing is up-regulated (compensatory) or down-reg-
cal. These reactive oxygen species and reactive ulated (pathologic) in Alzheimers disease. Aging
nitrogen species damage several molecular tar- and life span are also influenced by insulin.104
gets. Peroxidation of membrane lipids yields toxic Resistance to insulin signaling renders neurons
aldehydes,90 which impair critical mitochondrial energy-deficient and vulnerable to oxidizing or oth-
enzymes.77,91 Other essential proteins are direct- er metabolic insults and impairs synaptic plastic-
ly oxidized, yielding carbonyl and nitrated deriv- ity. Moreover, the higher serum glucose levels that
atives.92 Subsequently, increases in membrane are common in normal aging directly damage hip-
permeability to calcium, other ionic imbalances, pocampal structures,105 up-regulate the tau kinase,
and impaired glucose transport93 aggravate the glycogen synthase kinase 3,106 and reduce levels
energy imbalance. of insulin-degrading enzyme in the brain in Alz-
Elevated levels of free divalent transition metal heimers disease.107 Treatment with thiazolidine
ions (iron, copper, and zinc) and aluminum are drugs (peroxisome-proliferator-activated receptor
linked with reactive oxygen speciesmediated [PPAR] agonists, which activate insulin-respon-
damage and neurodegeneration in several sive gene transcription) prevented Alzheimers dis-
ways.94-100 These metal ions also promote aggre- easeassociated changes and cognitive decline in
gation of tau and changes in its conformation or transgenic mice103,108 and had significant effects
phosphorylation.95 Zinc, typically thought to be in subpopulations of patients with Alzheimers
a toxin in Alzheimers disease, might at lower disease.109
concentrations actually protect cells by blocking
A channels96 or compete with copper for A Vascular Effects
binding.97 In Alzheimers disease, vascular injury and pa-
Although animal models and most cross-sec- renchymal inflammation perpetuate the cycle of
tional studies in aging populations show an as- protein aggregation and oxidation in the brain;
sociation between antioxidant intake and cognitive damage from strokes and white-matter lesions
performance, randomized trials of antioxidants contribute greatly to cognitive decline. Ischemic
have generally failed.98 Therapeutic chelation of disease affects 60 to 90% of patients with Alz-
divalent metals is potentially harmful because heimers disease, with major infarctions repre-
essential enzymes rely on coordination with them. senting one third of vascular lesions in autopsy
In a pilot phase 2 trial (NCT00471211), PBT2, a cases. Conversely, one third of putative cases of
safe compound derived from clioquinol that atten- vascular dementia have coincidental pathological
uates metal proteins,99 showed some efficacy. features of Alzheimers disease. Although clini-
cally and radiographically pure cases of vascu-
Insulin-Signaling Pathway lar dementia are recognized,110,111 most cases of
Another metabolic disturbance of emerging im- dementia are in fact mixed. Pervasive pathologi-
portance in Alzheimers disease and tied into cal changes include cerebral amyloid angiopathy,112
synaptic and energy homeostasis involves insulin affecting more than 90% of patients with Alz-
signaling in the brain. Subgroups of patients heimers disease, capillary abnormalities, disrup-
with advanced Alzheimers disease have high fast- tion of the bloodbrain barrier, and large-vessel
ing insulin levels and low rates of glucose dis- atheroma.113 None of these changes alone ex-
posal (peripheral resistance).100 Glucose intoler- plain the symmetric reductions of cerebral blood
ance and type 2 diabetes are considered to be flow in patients with Alzheimers disease, which
risk factors for dementia.101 Levels of insulin re- are more likely to reflect regional energy under-
ceptors, glucose-transport proteins, and other utilization.114,115
insulin-pathway components in the brain are re- Another hypothesis holds that clearance of A
duced in some studies of Alzheimers disease102 along diseased perivascular channels and through
(central resistance). Insulin (mostly bloodborne) the bloodbrain barrier is impeded in Alzhei-
and brain-derived insulin-like growth factor I ini- mers disease. The source of vascular A (mostly
tiate signals in the brain by activating the phos- 40 amino acid form) is heterogeneous, compris-
phatidylinositol-3-kinaseAkt (also known as pro- ing neurons, degenerating myocytes, and the
tein kinase B) pathway and the mitogen-activated circulation. Amyloid deposition in the arteriolar

336 n engl j med 362;4 nejm.org january 28, 2010

The New England Journal of Medicine


Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

wall enhances vasoconstriction in ex vivo stud- autolytic attack are under way.126 Stimulated as-
ies.116 A is also cytotoxic to endothelial117 and troglia also release acute-phase reactants, alpha1-
smooth-muscle118 cells, conferring a predisposi- antichymotrypsin, alpha2-macroglobulin, and
tion to lobar hemorrhage in advanced age. The C-reactive protein, which can both aggravate and
neurovascular uncoupling hypothesis proposes ameliorate Alzheimers disease. Although inflam-
that deregulation of A transport across the matory (and oxidative) events are implicated in a
capillary bloodbrain barrier is caused by the breakdown of the vascular bloodbrain barrier
imbalanced expression of low-density lipopro- in Alzheimers disease, it is not certain that this
tein receptorrelated proteins and receptors for leads to monocyte or amyloid influx from the
advanced glycation end products, which mediate circulation in humans.130,131
A efflux and influx, respectively119 (Fig. 5). The contradictory roles of microglia elim-
Short of prophylaxis against stroke, there are inating A and releasing proinflammatory mole-
few specific therapies for the vascular changes cules complicate treatment.132 Nonsteroidal
in Alzheimers disease. Centrally acting angio- antiinflammatory agents have been reported to
tensin-convertingenzyme inhibitors were asso- lower the risk of Alzheimers disease and slow
ciated with reductions in yearly cognitive decline progression of the disease, but only in prospec-
in one observational study.120 Patients with hyper- tive observational studies.133,134 Their mechanisms
tension who are receiving medication have fewer of action include selective reduction of A42,135,136
neuropathologic features of Alzheimers dis- inhibition of cyclooxygenase-2 or the prostaglan-
ease.121 Folic acid reduces homocysteine levels din E2 receptor, stimulation of phagocytosis by
and may lower the risk of Alzheimers disease, microglia, and activation of PPAR-. Recent ran-
but it does not improve cognition in established domized trials of nonsteroidal antiinflammatory
Alzheimers disease.122,123 A phase 2 study of in- agents137 and a trial of a derivative, tarenflurbil
hibitors of receptors for advanced glycation end (Flurizan) (NCT00105547), did not show evidence
products in mild-to-moderate Alzheimers dis- of reducing the risk of Alzheimers disease or
ease (NCT00566397) is under way. Concern has slowing cognitive decline. In addition to the A-
been expressed about the safety of A immuno- immunization efforts, various TNF- and com-
therapy because of the possibilities of increased plement factor blockers and agents that promote
vascular amyloid, microhemorrhages, and vaso- phagocytosis are being investigated.138
genic edema as the efflux of A into vascular
compartments is stimulated.124 Calcium
Loss of calcium regulation is common to several
Inflammation neurodegenerative disorders. In Alzheimers dis-
Activated microglia and reactive astrocytes local- ease, elevated concentrations of cytosolic calcium
ize to fibrillar plaques, and their biochemical stimulate A aggregation and amyloidogene-
markers are elevated in the brains of patients sis.139,140 The presenilins modulate calcium bal-
with Alzheimers disease.125 Initially, the phago- ance. Presenilin mutations cause about one half
cytic microglia engulf and degrade A. However, of the few cases of Alzheimers disease (<1%)
chronically activated microglia release chemo- that are of the early-onset, familial type. These
kines and a cascade of damaging cytokines mutations might disrupt calcium homeostasis in
notably, interleukin-1, interleukin-6, and tumor endoplasmic reticulum.141,142 However, the main
necrosis factor (TNF-)126 (Fig. 5). In common effect of the mutations is to increase A42 levels,
with vascular cells, microglia express receptors which in turn increases calcium stores in the en-
for advanced glycation end products, which bind doplasmic reticulum and the release of calcium
A, thereby amplifying the generation of cyto- into the cytoplasm.143 The relevance of these
kines, glutamate, and nitric oxide.89,127 In exper- mechanisms to sporadic Alzheimers disease is
imental studies, chemokines promote the migra- unclear.
tion of monocytes from the peripheral blood into A chronic state of excitatory amino acid (glu-
plaque-bearing brain.128 taminergic) receptor activation is thought to ag-
Fibrillar A and glial activation also stimulate gravate neuronal damage in late-stage Alzhei-
the classic complement pathway.129 Tangles and mers disease.144 Glutamate increases cytosolic
plaques contain complement cleavage products, calcium, which in turn stimulates calcium-release
C1q and C5b-9, indicating that opsonization and channels in the endoplasmic reticulum. How-

n engl j med 362;4 nejm.org january 28, 2010 337


The New England Journal of Medicine
Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Capillary

Apolipoprotein J LRP-1
Phagocytosis of Monocyte
Microglia 2M
A plaque A

Microglia RAGE
A A

RAGE
A
APP

2M
A LRP-1 A
Inflammatory
cytokines RAGE
Endosome

MVB
Neurotoxicity
A
Lysosome
APOE LRP-1
APOE APOE
LRP-1 A Aggresome
A MTOC

Tau Neuron
IDE
Degradation
Proteasome
Cell
Astrocyte IDE
death
Plasmin
Neuritic MMP Degradation
plaque pathways
Apolipoprotein J Acute-phase Nep
MMP reactants

Figure 5. Inflammation and Mechanisms of A Clearance.


-amyloid peptide (A) is formed within intracellular compartments (the endoplasmic reticulum, Golgi apparatus, and endosomes) or it
can enter multiple cell types through the low-density lipoprotein receptorrelated protein. The ubiquitous apolipoprotein E (APOE) and 2-
macroglobulins (2M) are chaperones in this process and in the genesis of extracellular plaques. Microglia directly engulf A through
phagocytosis. Astrocytes also participate in A clearance through receptor-mediated internalization and facilitation of its transfer out of the
central nervous system and into the circulation. Microglia and astrocytes are recruited and stimulated in Alzheimers disease to release
proinflammatory cytokines and acute-phase reactants. Receptors for advanced glycation end products (RAGE) molecules transduce extra-
cellular A toxic and inflammatory effects and mediate influx of vascular A. The inflammatory milieu provokes neuritic changes and break-
down of the vascular bloodbrain barrier. In addition to cell-mediated reactions, A clearance occurs through enzymatic proteolysis, mainly
through neprilysin (Nep) and insulin-degrading enzyme (IDE). A oligomers block proteasome function, facilitating the buildup of intracel-
lular tau and accumulation of A into aggresomes. APP denotes amyloid precursor protein, MMP matrix metalloproteinase, MOTC micro-
tubule-organizing center, and MVB multivesicular body.
COLOR FIGURE

Draft 3 01/11/10
uthor Querfurth ever, the evidence of excessive excitatory amino blocker, MEM 1003, is in a phase 3 trial, and
g# 5 acid mechanisms in Alzheimers disease is mod- memantine, an NMDA-receptor blocker, is ap-
tle
E
est. A forms voltage-independent, cation chan- proved by the Food and Drug Administration.
E nels in lipid membranes,145 resulting in calcium
tist SBL uptake and degeneration of neuritis.146 Indirectly, Axonal-Transport Deficits
AUTHOR PLEASE NOTE:
glutamate activates voltage-gated calcium chan-
Figure has been redrawn and type has been reset Another internal derangement that is probably
Please check carefully
nels. The L-type voltage-gated calcium-channel an effect rather than a cause of Alzheimers dis-
sue date

338 n engl j med 362;4 nejm.org january 28, 2010

The New England Journal of Medicine


Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

ease is a reduction in the transport of critical naseinhibitor proteins, which maintain cell-cycle
protein cargoes to the synapse. Molecular mo- exit, are also deranged in Alzheimers disease.165
tors of the kinesin family drive vesicles and mito- Oxidative stress and DNA-damaging agents, in-
chondria destined for the synaptic terminal along cluding A and the carboxyl-terminal 99 amino
axonal microtubules. The kinesin superfamily acid BACE-1 product C99, all initiate DNA repli-
heavy-chain protein 5 and its associated kinesin cation and death in cultured neurons.166 The
light chain 1 facilitate fast anterograde trans- event inciting cell-cycle reentry in Alzheimers dis-
port. Tau forms the cross-bridges that maintain ease is unknown. Furthermore, whether it is patho-
the critical spacing between microtubules. genic or just reflects a survival response to repair
The riddle of Alzheimers disease is entwined damaged DNA167 is unclear.
with the elusive goal of finding the biologic func-
tion of amyloid precursor protein. It was exciting Cholesterol Metabolism
when amyloid precursor protein, BACE-1, and A defect in cholesterol metabolism is an appeal-
presenilin 1 were reported to undergo fast antero- ing hypothesis because it ties together the apo-
grade transport147 into terminal fields where A lipoprotein E (APOE) genetic risk, amyloid pro-
and other proteolytic derivatives are released.148 duction and aggregation, and vasculopathy of
Impairment of transport causes amyloid precur- Alzheimers disease. However, proof is also lack-
sor protein, vesicle, and kinesin accumulations ing for this hypothesis. Cholesterol is an essential
in axonal swelling, local A deposition, and neu- component of neuronal membranes and is con-
rodegeneration.149,150 However, whether amyloid centrated in sphingolipid islands termed lipid
precursor protein functions as the critical cargo rafts. Rafts are ordered platforms for the assem-
vesicle receptor for the motor protein complex bly of -secretases and -secretases and process-
remains unclear.151 Furthermore, an essential role ing of amyloid precursor protein into A168 (Fig.
is not evident from studies of amyloid precursor 1 and 2). A generation and aggregation are pro-
proteindeficient mice, which are viable, with moted and clearance from the brain is reduced
only subtle synaptic and learning defects.152,153 when an overabundance of esterified cholesterol
The anatomical distribution of pathological decreases membrane lipid turnover. Glial-derived
features in Alzheimers disease nonetheless sug- APOE is the primary cholesterol transporter in
gests that microtubules are dysfunctional, since the brain. A major determinant of the risk of
tau is primarily deranged in the source of corti- late-onset Alzheimers disease is the APOE iso-
cal projections.154 In addition, defects in the form inheritance pattern (APOE2, APOE3, or
white-matter tract are observed in patients at all APOE4)169; a single E4 allele increases the risk by
stages of Alzheimers disease155 and in animal a factor of 4, and two E4 alleles increase the risk
models.156 Pharmacologic disruption of micro- by a factor of 19.170 APOE4 is not only a patho-
tubules and inhibition of tau phosphatases cause logical chaperone, promoting A deposition171
similar axonal swelling and synaptic failure.157 and tau phosphorylation,172 but it is also the least
Since paclitaxel reverses these defects in mouse effective of the three in promoting healthy mem-
models,158 inhibitors of tau polymerization, brane lipid turnover and the uptake of lipopro-
phospho tau peptide vaccines,159 and other micro- tein particles.
tubule stabilizers160 are being investigated. High serum cholesterol levels in midlife in-
crease the risk of Alzheimers disease.173 In ob-
Aberrant Cell-Cycle Reentry servational studies, use of statins was shown to
In league with secondary deregulations of cal- be associated with a reduced risk. Statins appear
cium and transport, a failure in the normal sup- to reduce the membrane pool of free cholester-
pression of the cell cycle in Alzheimers disease ol.174 Other actions of statins that are not depen-
has been hypothesized.161 Markers of aberrant dent on cholesterol include reductions in inflam-
cell-cycle reentry are detected in all stages of Alz- mation175 and isoprenoids and up-regulation of
heimers disease and in mild cognitive impair- both -secretase176 and vascular function. One
ment,162 but they are most prominent at the G1 prospective trial of statins showed cognitive im-
S-phase boundary.163 This may progress to provements in patients with mild Alzheimers dis-
completion of DNA replication, resulting in tetra- ease,177 but a recent multicenter trial did not.178
ploid neurons and activation of mitotic cyclins, Thus, the benefit of statins remains controversial.
but mitoses are absent.164 Cyclin-dependent ki- An alternative pharmacologic approach is to

n engl j med 362;4 nejm.org january 28, 2010 339


The New England Journal of Medicine
Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

limit the esterification of cholesterol.179 Improve- position and tauopathy in humans is unknown.
ment of membrane biophysics and function Prospective studies also show that cognitive lei-
through ingestion of n3 fatty acid supplements sure activity and training can lower the risk of
has also been studied (NCT00440050).180 dementia190; findings from these studies provide
support for the concept of building a cognitive
C onclusions reserve. The figure in the Supplementary Ap-
pendix (available with the full text of this article
An effective treatment for sporadic Alzheimers at NEJM.org) summarizes the heterogeneity of
disease rests on the translation of the disease pathways that could initiate and drive Alzhei-
pathways we have discussed, as well as additional mers disease. There is no single linear chain of
molecular mechanisms or new risk genes (e.g., events. Complicating matters, some changes are
apolipoprotein J) defined by gene-expression pro- not pathologic but reactionary or protective.
filing and whole-genome association studies,181,182 Thus, the development of a multitargeted ap-
into specific pharmacologic targets. Examples of proach to prevent or symptomatically treat Alz-
recently discovered proteins encoded by these heimers disease, as used in current practice for
risk genes and mechanisms include apolipopro- other multigenic disorders, is needed.191 Recent
tein J (clusterin), another A chaperone,183 studies point to brain atrophy and other patho-
TOMM40, a transporter of proteins across the mi- logic conditions, not severe amyloid or tangle
tochondrial membrane, and Sortillin-related re- load, in accounting for dementia in the oldest old
ceptor, which functions to partition amyloid pre- (persons 80 years of age or older).192 It remains
cursor protein away from -secretase and possible that many of these mechanisms, includ-
-secretase; this is consistent with observations ing the amyloid hypothesis, are minor or wrong
that levels are reduced in the brains of patients and that some critical aging-related process is
with Alzheimers disease and mild cognitive im- the disease trigger.
pairment.184,185 Another potential risk factor for Dr. Querfurth reports receiving consulting and lecture fees
sporadic Alzheimers disease, general anesthe- from Novartis and Forest Pharmaceuticals and holding a provi-
sia, promotes tau insolubility and A oligomer- sional patent on a drug-screening assay for Alzheimers disease;
and Dr. LaFerla, receiving consulting fees from Sonexa Therapeu-
ization,186,187 deficiency of estrogen in the brains tics, Forest Pharmaceuticals, and Abbott. No other potential
of postmenopausal women,188 and chronic acti- conflict of interest relevant to this article was reported. Financial
vation of the glucocorticoid axis.189 However, and other disclosures provided by the authors are available with
the full text of this article at NEJM.org.
their underlying mechanisms are diverse, and We thank Donna-Marie Mironchuk and David Cheng for as-
whether any of these factors lead to amyloid de- sistance in preparation of earlier drafts of the figures.

References
1. Hirtz D, Thurman DJ, Gwinn-Hardy K, proteins, and therapy. Physiol Rev 2001; Amyloid beta protein dimer-containing
Mohamed M, Chaudhuri AR, Zalutsky R. 81:741-66. human CSF disrupts synaptic plasticity:
How common are the common neuro- 7. Tanzi RE, Bertram L. Twenty years of prevention by systemic passive immuni-
logic disorders? Neurology 2007;68:326- the Alzheimers disease amyloid hypoth- zation. J Neurosci 2008;28:4231-7.
37. esis: a genetic perspective. Cell 2005;120: 13. Lue LF, Kuo YM, Roher AE, et al. Sol-
2. den Dunnen WF, Brouwer WH, Bijlard 545-55. uble amyloid beta peptide concentration
E, et al. No disease in the brain of a 8. Kayed R, Head E, Thompson JL, et al. as a predictor of synaptic change in Alz-
115-year-old woman. Neurobiol Aging Common structure of soluble amyloid heimers disease. Am J Pathol 1999;155:
2008;29:1127-32. oligomers implies common mechanism 853-62.
3. Hebert LE, Scherr PA, Bienias JL, Ben- of pathogenesis. Science 2003;300:486-9. 14. Kamenetz F, Tomita T, Hsieh H, et al.
nett DA, Evans DA. Alzheimer disease in 9. Klein WL, Krafft GA, Finch CE. Tar- APP processing and synaptic function.
the US population: prevalence estimates geting small Abeta oligomers: the solu- Neuron 2003;37:925-37.
using the 2000 census. Arch Neurol tion to an Alzheimers disease conundrum? 15. Kanemitsu H, Tomiyama T, Mori H.
2003;60:1119-22. Trends Neurosci 2001;24:219-24. Human neprilysin is capable of degrading
4. Haass C, Selkoe DJ. Soluble protein 10. Walsh DM, Selkoe DJ. A beta oligo- amyloid beta peptide not only in the mono-
oligomers in neurodegeneration: lessons mers a decade of discovery. J Neuro- meric form but also the pathological oligo-
from the Alzheimers amyloid beta-pep- chem 2007;101:1172-84. meric form. Neurosci Lett 2003;350:113-6.
tide. Nat Rev Mol Cell Biol 2007;8:101-12. 11. Walsh DM, Townsend M, Podlisny 16. Iwata N, Tsubuki S, Takaki Y, et al.
5. Busciglio J, Pelsman A, Wong C, et al. MB, et al. Certain inhibitors of synthetic Metabolic regulation of brain Abeta by
Altered metabolism of the amyloid beta amyloid beta-peptide (Abeta) fibrillogen- neprilysin. Science 2001;292:1550-2.
precursor protein is associated with mito- esis block oligomerization of natural 17. Qiu WQ, Walsh DM, Ye Z, et al. Insu-
chondrial dysfunction in Downs syn- Abeta and thereby rescue long-term poten- lin-degrading enzyme regulates extracel-
drome. Neuron 2002;33:677-88. tiation. J Neurosci 2005;25:2455-62. lular levels of amyloid beta-protein by deg-
6. Selkoe DJ. Alzheimers disease: genes, 12. Klyubin I, Betts V, Welzel AT, et al. radation. J Biol Chem 1998;273:32730-8.

340 n engl j med 362;4 nejm.org january 28, 2010

The New England Journal of Medicine


Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

18. Farris W, Mansourian S, Chang Y, et al. mers disease. Ann Neurol 1997;41:17- 49. Terry RD, Masliah E, Salmon DP, et al.
Insulin-degrading enzyme regulates the 24. Physical basis of cognitive alterations in
levels of insulin, amyloid beta-protein, and 34. Wallin AK, Blennow K, Andreasen N, Alzheimers disease: synapse loss is the
the beta-amyloid precursor protein intra- Minthon L. CSF biomarkers for Alzhei- major correlate of cognitive impairment.
cellular domain in vivo. Proc Natl Acad mers disease: levels of beta-amyloid, tau, Ann Neurol 1991;30:572-80.
Sci U S A 2003;100:4162-7. phosphorylated tau relate to clinical symp- 50. Davies CA, Mann DM, Sumpter PQ,
19. Leissring MA, Farris W, Chang AY, et toms and survival. Dement Geriatr Cogn Yates PO. A quantitative morphometric
al. Enhanced proteolysis of beta-amyloid Disord 2006;21:131-8. analysis of the neuronal and synaptic con-
in APP transgenic mice prevents plaque 35. Mattsson N, Zetterberg H, Hansson tent of the frontal and temporal cortex in
formation, secondary pathology, and pre- O, et al. CSF biomarkers and incipient patients with Alzheimers disease. J Neu-
mature death. Neuron 2003;40:1087-93. Alzheimer disease in patients with mild rol Sci 1987;78:151-64.
20. Siemers ER, Quinn JF, Kaye J, et al. cognitive impairment. JAMA 2009;302: 51. Masliah E, Crews L, Hansen L. Synap-
Effects of a gamma-secretase inhibitor in 385-93. tic remodeling during aging and in Alz-
a randomized study of patients with Alz- 36. Oddo S, Caccamo A, Shepherd JD, et al. heimers disease. J Alzheimers Dis 2006;9:
heimer disease. Neurology 2006;66:602-4. Triple-transgenic model of Alzheimers Suppl:91-9.
21. McGeer PL, McGeer E. Is there a fu- disease with plaques and tangles: intra- 52. Lister JP, Barnes CA. Neurobiological
ture for vaccination as a treatment for Alz- cellular Abeta and synaptic dysfunction. changes in the hippocampus during nor-
heimers disease? Neurobiol Aging 2003; Neuron 2003;39:409-21. mative aging. Arch Neurol 2009;66:829-33.
24:391-5. 37. Gotz J, Chen F, van Dorpe J, Nitsch 53. Larson J, Lynch G, Games D, Seubert
22. Hock C, Konietzko U, Streffer JR, et al. RM. Formation of neurofibrillary tangles P. Alterations in synaptic transmission
Antibodies against beta-amyloid slow cog- in P301l tau transgenic mice induced by and long-term potentiation in hippocam-
nitive decline in Alzheimers disease. Abeta 42 fibrils. Science 2001;293:1491- pal slices from young and aged PDAPP
Neuron 2003;38:547-54. 5. mice. Brain Res 1999;840:23-35.
23. Gilman S, Koller M, Black RS, et al. 38. Lewis J, Dickson DW, Lin WL, et al. 54. Chapman PF, White GL, Jones MW, et
Clinical effects of Abeta immunization Enhanced neurofibrillary degeneration in al. Impaired synaptic plasticity and learn-
(AN1792) in patients with AD in an inter- transgenic mice expressing mutant tau ing in aged amyloid precursor protein
rupted trial. Neurology 2005;64:1553-62. and APP. Science 2001;293:1487-91. transgenic mice. Nat Neurosci 1999;2:
24. Holmes C, Boche D, Wilkinson D, et 39. Roberson ED, Scearce-Levie K, Palop 271-6.
al. Long-term effects of Abeta42 immuni- JJ, et al. Reducing endogenous tau amelio- 55. Shankar GM, Bloodgood BL, Townsend
sation in Alzheimers disease: follow-up rates amyloid beta-induced deficits in an M, Walsh DM, Selkoe DJ, Sabatini BL.
of a randomised, placebo-controlled phase Alzheimers disease mouse model. Science Natural oligomers of the Alzheimer amy-
I trial. Lancet 2008;372:216-23. 2007;316:750-4. loid-beta protein induce reversible synapse
25. Lee VM, Goedert M, Trojanowski JQ. 40. Rapoport M, Dawson HN, Binder LI, loss by modulating an NMDA-type gluta-
Neurodegenerative tauopathies. Annu Rev Vitek MP, Ferreira A. Tau is essential to mate receptor-dependent signaling path-
Neurosci 2001;24:1121-59. beta-amyloid-induced neurotoxicity. Proc way. J Neurosci 2007;27:2866-75.
26. Iqbal K, Alonso Adel C, Chen S, et al. Natl Acad Sci U S A 2002;99:6364-9. 56. Snyder EM, Nong Y, Almeida CG, et al.
Tau pathology in Alzheimer disease and 41. Lpez Salon M, Morelli L, Castao Regulation of NMDA receptor trafficking
other tauopathies. Biochim Biophys Acta EM, Soto EF, Pasquini JM. Defective by amyloid-beta. Nat Neurosci 2005;8:
2005;1739:198-210. ubiquitination of cerebral proteins in Alz- 1051-8.
27. Khlistunova I, Biernat J, Wang Y, et al. heimers disease. J Neurosci Res 2000;62: 57. Hsieh H, Boehm J, Sato C, et al. AMPAR
Inducible expression of Tau repeat domain 302-10. removal underlies Abeta-induced synaptic
in cell models of tauopathy: aggregation 42. Hoozemans JJ, Veerhuis R, Van Haas- depression and dendritic spine loss. Neu-
is toxic to cells but can be reversed by in- tert ES, et al. The unfolded protein re- ron 2006;52:831-43.
hibitor drugs. J Biol Chem 2006;281:1205- sponse is activated in Alzheimers disease. 58. Mucke L, Masliah E, Yu GQ, et al.
14. Acta Neuropathol 2005;110:165-72. High-level neuronal expression of abeta
28. Santacruz K, Lewis J, Spires T, et al. 43. McLaurin J, Kierstead ME, Brown ME, 1-42 in wild-type human amyloid protein
Tau suppression in a neurodegenerative et al. Cyclohexanehexol inhibitors of Abeta precursor transgenic mice: synaptotoxic-
mouse model improves memory function. aggregation prevent and reverse Alzhei- ity without plaque formation. J Neurosci
Science 2005;309:476-81. mer phenotype in a mouse model. Nat 2000;20:4050-8.
29. Oddo S, Vasilevko V, Caccamo A, Kita- Med 2006;12:801-8. 59. Cooper JD, Salehi A, Delcroix JD, et al.
zawa M, Cribbs DH, LaFerla FM. Reduc- 44. Ono K, Condron MM, Ho L, et al. Ef- Failed retrograde transport of NGF in a
tion of soluble Abeta and tau, but not fects of grape seed-derived polyphenols mouse model of Downs syndrome: rever-
soluble Abeta alone, ameliorates cognitive on amyloid beta-protein self-assembly and sal of cholinergic neurodegenerative phe-
decline in transgenic mice with plaques cytotoxicity. J Biol Chem 2008;283:32176- notypes following NGF infusion. Proc Natl
and tangles. J Biol Chem 2006;281:39413- 87. Acad Sci U S A 2001;98:10439-44.
23. 45. Selkoe DJ. Alzheimers disease is a syn- 60. Tuszynski MH. Nerve growth factor
30. Andorfer C, Kress Y, Espinoza M, et al. aptic failure. Science 2002;298:789-91. gene therapy in Alzheimer disease. Alz-
Hyperphosphorylation and aggregation of 46. Scheff SW, Price DA, Schmitt FA, heimer Dis Assoc Disord 2007;21:179-89.
tau in mice expressing normal human tau DeKosky ST, Mufson EJ. Synaptic altera- 61. Connor B, Young D, Yan Q, Faull RL,
isoforms. J Neurochem 2003;86:582-90. tions in CA1 in mild Alzheimer disease Synek B, Dragunow M. Brain-derived neu-
31. Lee HG, Perry G, Moreira PI, et al. Tau and mild cognitive impairment. Neurol- rotrophic factor is reduced in Alzheimers
phosphorylation in Alzheimers disease: ogy 2007;68:1501-8. disease. Brain Res Mol Brain Res 1997;49:
pathogen or protector? Trends Mol Med 47. Masliah E, Mallory M, Alford M, et al. 71-81.
2005;11:164-9. Altered expression of synaptic proteins oc- 62. Garzon DJ, Fahnestock M. Oligomeric
32. Goedert M, Jakes R. Mutations caus- curs early during progression of Alzhei- amyloid decreases basal levels of brain-
ing neurodegenerative tauopathies. Bio- mers disease. Neurology 2001;56:127-9. derived neurotrophic factor (BDNF) mRNA
chim Biophys Acta 2005;1739:240-50. 48. DeKosky ST, Scheff SW. Synapse loss via specific downregulation of BDNF tran-
33. Gmez-Isla T, Hollister R, West H, et in frontal cortex biopsies in Alzheimers scripts IV and V in differentiated human
al. Neuronal loss correlates with but ex- disease: correlation with cognitive sever- neuroblastoma cells. J Neurosci 2007;27:
ceeds neurofibrillary tangles in Alzhei- ity. Ann Neurol 1990;27:457-64. 2628-35.

n engl j med 362;4 nejm.org january 28, 2010 341


The New England Journal of Medicine
Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

63. Ernfors P, Bramham CR. The coupling in Alzheimers disease. FASEB J 2005;19: oxynitrite-mediated damage in Alzhei-
of a trkB tyrosine residue to LTP. Trends 2040-1. mers disease. J Neurosci 1997;17:2653-7.
Neurosci 2003;26:171-3. 77. Hirai K, Aliev G, Nunomura A, et al. 93. Mark RJ, Pang Z, Geddes JW, Uchida
64. Nagahara AH, Merrill DA, Coppola G, Mitochondrial abnormalities in Alzhei- K, Mattson MP. Amyloid beta-peptide im-
et al. Neuroprotective effects of brain- mers disease. J Neurosci 2001;21:3017- pairs glucose transport in hippocampal
derived neurotrophic factor in rodent and 23. and cortical neurons: involvement of mem-
primate models of Alzheimers disease. 78. Gouras GK, Almeida CG, Takahashi brane lipid peroxidation. J Neurosci 1997;
Nat Med 2009;15:331-7. RH. Intraneuronal Abeta accumulation 17:1046-54.
65. Ikonomovic MD, Wecker L, Abraham- and origin of plaques in Alzheimers dis- 94. Lovell MA, Robertson JD, Teesdale
son EE, et al. Cortical alpha7 nicotinic ease. Neurobiol Aging 2005;26:1235-44. WJ, Campbell JL, Markesbery WR. Copper,
acetylcholine receptor and beta-amyloid 79. Lustbader JW, Cirilli M, Lin C, et al. iron and zinc in Alzheimers disease se-
levels in early Alzheimer disease. Arch ABAD directly links Abeta to mitochon- nile plaques. J Neurol Sci 1998;158:47-52.
Neurol 2009;66:646-51. drial toxicity in Alzheimers disease. Sci- 95. Yamamoto A, Shin RW, Hasegawa K,
66. Maelicke A, Samochocki M, Jostock R, ence 2004;304:448-52. et al. Iron (III) induces aggregation of
et al. Allosteric sensitization of nicotinic 80. Cardoso SM, Santana I, Swerdlow RH, hyperphosphorylated tau and its reduc-
receptors by galantamine, a new treat- Oliveira CR. Mitochondria dysfunction of tion to iron (II) reverses the aggregation:
ment strategy for Alzheimers disease. Alzheimers disease cybrids enhances Abeta implications in the formation of neuro-
Biol Psychiatry 2001;49:279-88. toxicity. J Neurochem 2004;89:1417-26. fibrillary tangles of Alzheimers disease.
67. Wang HY, Lee DH, DAndrea MR, Pe- 81. Wallace DC. Mitochondrial diseases in J Neurochem 2002;82:1137-47.
terson PA, Shank RP, Reitz AB. beta-Amy- man and mouse. Science 1999;283:1482-8. 96. Arispe N, Pollard HB, Rojas E. Zn2+
loid(1-42) binds to alpha7 nicotinic ace- 82. Cho DH, Nakamura T, Fang J, et al. interaction with Alzheimer amyloid beta
tylcholine receptor with high affinity: S-nitrosylation of Drp1 mediates beta- protein calcium channels. Proc Natl Acad
implications for Alzheimers disease pa- amyloid-related mitochondrial fission and Sci U S A 1996;93:1710-5.
thology. J Biol Chem 2000;275:5626-32. neuronal injury. Science 2009;324:102-5. 97. Cuajungco MP, Fagt KY. Zinc takes
68. Nitsch RM. From acetylcholine to am- 83. Doody RS, Gavrilova SI, Sano M, et al. the center stage: its paradoxical role in
yloid: neurotransmitters and the pathol- Effect of dimebon on cognition, activities Alzheimers disease. Brain Res Brain Res
ogy of Alzheimers disease. Neurodegen- of daily living, behaviour, and global func- Rev 2003;41:44-56.
eration 1996;5:477-82. tion in patients with mild-to-moderate 98. Pratic D. Oxidative stress hypothe-
69. Caccamo A, Oddo S, Billings LM, et al. Alzheimers disease: a randomised, double- sis in Alzheimers disease: a reappraisal.
M1 receptors play a central role in modu- blind, placebo-controlled study. Lancet Trends Pharmacol Sci 2008;29:609-15.
lating AD-like pathology in transgenic 2008;372:207-15. 99. Ritchie CW, Bush AI, Mackinnon A,
mice. Neuron 2006;49:671-82. 84. Good PF, Werner P, Hsu A, Olanow et al. Safety, efficacy, and biomarker find-
70. Bitner RS, Nikkel AL, Markosyan S, CW, Perl DP. Evidence of neuronal oxida- ings of PBT2 in targeting abeta as a mod-
Otte S, Puttfarcken P, Gopalakrishnan M. tive damage in Alzheimers disease. Am J ifying therapy for Alzheimers disease:
Selective alpha7 nicotinic acetylcholine re- Pathol 1996;149:21-8. a phase IIa, double-blind, randomised,
ceptor activation regulates glycogen syn- 85. Smith MA, Perry G, Richey PL, et al. placebo-controlled trial. Lancet Neurol
thase kinase3beta and decreases tau phos- Oxidative damage in Alzheimers. Nature 2008;7:779-86.
phorylation in vivo. Brain Res 2009;1265: 1996;382:120-1. 100. Craft S, Peskind E, Schwartz MW,
65-74. 86. Nunomura A, Perry G, Aliev G, et al. Schellenberg GD, Raskind M, Porte D Jr.
71. Bodick NC, Offen WW, Levey AI, et al. Oxidative damage is the earliest event in Cerebrospinal fluid and plasma insulin
Effects of xanomeline, a selective musca- Alzheimer disease. J Neuropathol Exp levels in Alzheimers disease: relationship
rinic receptor agonist, on cognitive func- Neurol 2001;60:759-67. to severity of dementia and apolipopro-
tion and behavioral symptoms in Alz- 87. Hensley K, Carney JM, Mattson MP, tein E genotype. Neurology 1998;50:164-8.
heimer disease. Arch Neurol 1997;54: et al. A model for beta-amyloid aggrega- 101. Arvanitakis Z, Wilson RS, Bienias JL,
465-73. tion and neurotoxicity based on free radi- Evans DA, Bennett DA. Diabetes mellitus
72. Nitsch RM, Deng M, Tennis M, Schoen- cal generation by the peptide: relevance to and risk of Alzheimer disease and decline
feld D, Growdon JH. The selective musca- Alzheimer disease. Proc Natl Acad Sci U S A in cognitive function. Arch Neurol 2004;
rinic M1 agonist AF102B decreases levels 1994;91:3270-4. 61:661-6.
of total Abeta in cerebrospinal fluid of 88. Combs CK, Karlo JC, Kao SC, Lan- 102. Messier C, Teutenberg K. The role of
patients with Alzheimers disease. Ann dreth GE. beta-Amyloid stimulation of insulin, insulin growth factor, and insu-
Neurol 2000;48:913-8. microglia and monocytes results in lin-degrading enzyme in brain aging and
73. Mungarro-Menchaca X, Ferrera P, TNFalpha-dependent expression of induc- Alzheimers disease. Neural Plast 2005;
Moran J, Arias C. beta-Amyloid peptide ible nitric oxide synthase and neuronal 12:311-28.
induces ultrastructural changes in synapto- apoptosis. J Neurosci 2001;21:1179-88. 103. de la Monte SM, Tong M, Lester-Coll
somes and potentiates mitochondrial dys- 89. Yan SD, Chen X, Fu J, et al. RAGE and N, Plater M Jr, Wands JR. Therapeutic res-
function in the presence of ryanodine. amyloid-beta peptide neurotoxicity in Alz- cue of neurodegeneration in experimental
J Neurosci Res 2002;68:89-96. heimers disease. Nature 1996;382:685-91. type 3 diabetes: relevance to Alzheimers
74. Hauptmann S, Keil U, Scherping I, 90. Keller JN, Mark RJ, Bruce AJ, et al. disease. J Alzheimers Dis 2006;10:89-109.
Bonert A, Eckert A, Muller WE. Mitochon- 4-Hydroxynonenal, an aldehydic product 104. Cohen E, Bieschke J, Perciavalle RM,
drial dysfunction in sporadic and genetic of membrane lipid peroxidation, impairs Kelly JW, Dillin A. Opposing activities
Alzheimers disease. Exp Gerontol 2006; glutamate transport and mitochondrial protect against age-onset proteotoxicity.
41:668-73. function in synaptosomes. Neuroscience Science 2006;313:1604-10.
75. Reddy PH, Beal MF. Amyloid beta, 1997;80:685-96. 105. Wu W, Brickman AM, Luchsinger J,
mitochondrial dysfunction and synaptic 91. Humphries KM, Szweda LI. Selective et al. The brain in the age of old: the hip-
damage: implications for cognitive decline inactivation of alpha-ketoglutarate dehy- pocampal formation is targeted differen-
in aging and Alzheimers disease. Trends drogenase and pyruvate dehydrogenase: tially by diseases of late life. Ann Neurol
Mol Med 2008;14:45-53. reaction of lipoic acid with 4-hydroxy-2- 2008;64:698-706.
76. Caspersen C, Wang N, Yao J, et al. nonenal. Biochemistry 1998;37:15835-41. 106. Takashima A. GSK-3 is essential in
Mitochondrial Abeta: a potential focal 92. Smith MA, Richey Harris PL, Sayre LM, the pathogenesis of Alzheimers disease.
point for neuronal metabolic dysfunction Beckman JS, Perry G. Widespread per- J Alzheimers Dis 2006;9:Suppl:309-17.

342 n engl j med 362;4 nejm.org january 28, 2010

The New England Journal of Medicine


Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

107. Cook DG, Leverenz JB, McMillan PJ, Green R, Mayeux R. Relation of higher tory drugs decrease amyloid beta42 pro-
et al. Reduced hippocampal insulin-degrad- folate intake to lower risk of Alzheimer duction by direct modulation of gamma-
ing enzyme in late-onset Alzheimers dis- disease in the elderly. Arch Neurol 2007; secretase activity. J Biol Chem 2003;278:
ease is associated with the apolipoprotein 64:86-92. 31831-7.
E-epsilon4 allele. Am J Pathol 2003;162: 123. Aisen PS, Schneider LS, Sano M, et 137. Szekely CA, Breitner JC, Fitzpatrick
313-9. al. High-dose B vitamin supplementation AL, et al. NSAID use and dementia risk in
108. Pedersen WA, McMillan PJ, Kulstad and cognitive decline in Alzheimer dis- the Cardiovascular Health Study: role of
JJ, Leverenz JB, Craft S, Haynatzki GR. ease: a randomized controlled trial. JAMA APOE and NSAID type. Neurology 2008;
Rosiglitazone attenuates learning and 2008;300:1774-83.124. 70:17-24.
memory deficits in Tg2576 Alzheimer 124. Wilcock DM, Jantzen PT, Li Q, Mor- 138. Shen Y, Meri S. Yin and Yang: com-
mice. Exp Neurol 2006;199:265-73. gan D, Gordon MN. Amyloid-beta vacci- plement activation and regulation in Alz-
109. Risner ME, Saunders AM, Altman JF, nation, but not nitro-nonsteroidal anti- heimers disease. Prog Neurobiol 2003;
et al. Efficacy of rosiglitazone in a geneti- inflammatory drug treatment, increases 70:463-72.
cally defined population with mild-to- vascular amyloid and microhemorrhage 139. Isaacs AM, Senn DB, Yuan M, Shine
moderate Alzheimers disease. Pharmaco- while both reduce parenchymal amyloid. JP, Yankner BA. Acceleration of amyloid
genomics J 2006;6:246-54. Neuroscience 2007;144:950-60. beta-peptide aggregation by physiological
110. OBrien JT, Erkinjuntti T, Reisberg B, 125. Wyss-Coray T, Mucke L. Inflamma- concentrations of calcium. J Biol Chem
et al. Vascular cognitive impairment. Lan- tion in neurodegenerative disease a 2006;281:27916-23.
cet Neurol 2003;2:89-98. double-edged sword. Neuron 2002;35:419- 140. Pierrot N, Ghisdal P, Caumont AS,
111. Hachinski V, Iadecola C, Petersen RC, 32. Octave JN. Intraneuronal amyloid-beta1-42
et al. National Institute of Neurological 126. Akiyama H, Barger S, Barnum S, et production triggered by sustained increase
Disorders and StrokeCanadian Stroke al. Inflammation and Alzheimers dis- of cytosolic calcium concentration induc-
Network vascular cognitive impairment ease. Neurobiol Aging 2000;21:383-421. es neuronal death. J Neurochem 2004;88:
harmonization standards. Stroke 2006;37: 127. Li Y, Liu L, Barger SW, Griffin WS. 1140-50.
2220-41. Interleukin-1 mediates pathological ef- 141. Leissring MA, Akbari Y, Fanger CM,
112. Greenberg SM, Gurol ME, Rosand J, fects of microglia on tau phosphorylation Cahalan MD, Mattson MP, LaFerla FM.
Smith EE. Amyloid angiopathy-related vas- and on synaptophysin synthesis in corti- Capacitative calcium entry deficits and
cular cognitive impairment. Stroke 2004; cal neurons through a p38-MAPK path- elevated luminal calcium content in mu-
35:Suppl 1:2616-9. way. J Neurosci 2003;23:1605-11. tant presenilin-1 knockin mice. J Cell Biol
113. Roher AE, Esh C, Rahman A, Kok- 128. Simard AR, Soulet D, Gowing G, Ju- 2000;149:793-8.
john TA, Beach TG. Atherosclerosis of lien JP, Rivest S. Bone marrow-derived 142. Nelson O, Tu H, Lei T, Bentahir M,
cerebral arteries in Alzheimer disease. microglia play a critical role in restricting de Strooper B, Bezprozvanny I. Familial
Stroke 2004;35:Suppl 1:2623-7. senile plaque formation in Alzheimers Alzheimer disease-linked mutations spe-
114. Ruitenberg A, den Heijer T, Bakker disease. Neuron 2006;49:489-502. cifically disrupt Ca2+ leak function of pre-
SL, et al. Cerebral hypoperfusion and 129. McGeer EG, Yasojima K, Schwab C, senilin 1. J Clin Invest 2007;117:1230-9.
clinical onset of dementia: the Rotterdam McGeer PL. The pentraxins: possible role 143. LaFerla FM. Calcium dyshomeosta-
Study. Ann Neurol 2005;57:789-94. in Alzheimers disease and other innate sis and intracellular signalling in Alz-
115. Jagust WJ. Neuroimaging in demen- inflammatory diseases. Neurobiol Aging heimers disease. Nat Rev Neurosci 2002;
tia. Neurol Clin 2000;18:885-902. 2001;22:843-8. 3:862-72.
116. Price JM, Chi X, Hellermann G, Sut- 130. Matsumoto Y, Yanase D, Noguchi- 144. Rothman SM, Olney JW. Excitotoxic-
ton ET. Physiological levels of beta-amy- Shinohara M, Ono K, Yoshita M, Yamada ity and the NMDA re4ptor still lethal
loid induce cerebral vessel dysfunction and M. Blood-brain barrier permeability cor- after eight years. Trends Neurosci 1995;
reduce endothelial nitric oxide production. relates with medial temporal lobe atrophy 18:57-8.
Neurol Res 2001;23:506-12. but not with amyloid-beta protein trans- 145. Arispe N, Pollard HB, Rojas E. Giant
117. Paris D, Patel N, DelleDonne A, port across the blood-brain barrier in Alz- multilevel cation channels formed by Alz-
Quadros A, Smeed R, Mullan M. Impaired heimers disease. Dement Geriatr Cogn heimer disease amyloid beta-protein [A beta
angiogenesis in a transgenic mouse mod- Disord 2007;23:241-5. P-(1-40)] in bilayer membranes. Proc Natl
el of cerebral amyloidosis. Neurosci Lett 131. Clifford PM, Zarrabi S, Siu G, et al. Acad Sci U S A 1993;90:10573-7.
2004;366:80-5. Abeta peptides can enter the brain 146. Lin H, Bhatia R, Lal R. Amyloid beta
118. Van Nostrand WE, Melchor JP, through a defective blood-brain barrier protein forms ion channels: implications
Ruffini L. Pathologic amyloid beta-pro- and bind selectively to neurons. Brain Res for Alzheimers disease pathophysiology.
tein cell surface fibril assembly on cultured 2007;1142:223-36. FASEB J 2001;15:2433-44. [Erratum,
human cerebrovascular smooth muscle 132. Fiala M, Lin J, Ringman J, et al. Inef- FASEB J 2002;16:759.]
cells. J Neurochem 1998;70:216-23. fective phagocytosis of amyloid-beta by 147. Koo EH, Sisodia SS, Archer DR, et al.
119. Deane R, Zlokovic BV. Role of the macrophages of Alzheimers disease pa- Precursor of amyloid protein in Alzheimer
blood-brain barrier in the pathogenesis of tients. J Alzheimers Dis 2005;7:221-32. disease undergoes fast anterograde axonal
Alzheimers disease. Curr Alzheimer Res 133. McGeer PL, McGeer EG. NSAIDs and transport. Proc Natl Acad Sci U S A 1990;
2007;4:191-7. Alzheimer disease: epidemiological, ani- 87:1561-5.
120. Sink KM, Leng X, Williamson J, et al. mal model and clinical studies. Neurobiol 148. Lazarov O, Lee M, Peterson DA, Si-
Angiotensin-converting enzyme inhibitors Aging 2007;28:639-47. sodia SS. Evidence that synaptically re-
and cognitive decline in older adults with 134. Vlad SC, Miller DR, Kowall NW, Fel- leased beta-amyloid accumulates as extra-
hypertension: results from the Cardiovas- son DT. Protective effects of NSAIDs on cellular deposits in the hippocampus of
cular Health Study. Arch Intern Med the development of Alzheimer disease. transgenic mice. J Neurosci 2002;22:
2009;169:1195-202. Neurology 2008;70:1672-7. 9785-93.
121. Hoffman LB, Schmeidler J, Lesser 135. Lle A, Berezovska O, Herl L, et al. 149. Kamal A, Almenar-Queralt A, LeBlanc
GT, et al. Less Alzheimer disease neuro- Nonsteroidal anti-inflammatory drugs JF, Roberts EA, Goldstein LS. Kinesin-
pathology in medicated hypertensive than lower Abeta42 and change presenilin 1 mediated axonal transport of a membrane
nonhypertensive persons. Neurology 2009; conformation. Nat Med 2004;10:1065-6. compartment containing beta-secretase
72:1720-6. 136. Weggen S, Eriksen JL, Sagi SA, et al. and presenilin-1 requires APP. Nature
122. Luchsinger JA, Tang MX, Miller J, Evidence that nonsteroidal anti-inflamma- 2001;414:643-8.

n engl j med 362;4 nejm.org january 28, 2010 343


The New England Journal of Medicine
Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

150. Stokin GB, Lillo C, Falzone TL, et al. 164. Mosch B, Morawski M, Mittag A, 179. Hutter-Paier B, Huttunen HJ, Pugliel-
Axonopathy and transport deficits early Lenz D, Tarnok A, Arendt T. Aneuploidy li L, et al. The ACAT inhibitor CP-113,818
in the pathogenesis of Alzheimers dis- and DNA replication in the normal human markedly reduces amyloid pathology in a
ease. Science 2005;307:1282-8. brain and Alzheimers disease. J Neurosci mouse model of Alzheimers disease.
151. Lazarov O, Morfini GA, Lee EB, et al. 2007;27:6859-67. Neuron 2004;44:227-38.
Axonal transport, amyloid precursor pro- 165. Ogawa O, Lee HG, Zhu X, et al. In- 180. Quinn JF, Raman R, Thomas RG, et
tein, kinesin-1, and the processing appa- creased p27, an essential component of al. A clinical trial of docosahexanoic acid
ratus: revisited. J Neurosci 2005;25:2386- cell cycle control, in Alzheimers disease. (DHA) for the treatment of Alzheimers
95. Aging Cell 2003;2:105-10. disease. Alzheimers and Dement 2009;5:
152. Ring S, Weyer SW, Kilian SB, et al. 166. Klein JA, Ackerman SL. Oxidative Suppl:84. abstract.
The secreted beta-amyloid precursor pro- stress, cell cycle, and neurodegeneration. 181. Lu T, Pan Y, Kao SY, et al. Gene regu-
tein ectodomain APPs alpha is sufficient J Clin Invest 2003;111:785-93. lation and DNA damage in the ageing hu-
to rescue the anatomical, behavioral, and 167. Yang Y, Herrup K. Cell division in man brain. Nature 2004;429:883-91.
electrophysiological abnormalities of APP- the CNS: protective response or lethal 182. Bertram L, Lange C, Mullin K, et al.
deficient mice. J Neurosci 2007;27:7817- event in post-mitotic neurons? Biochim Genome-wide association analysis reveals
26. Biophys Acta 2007;1772:457-66. putative Alzheimers disease susceptibili-
153. Dawson GR, Seabrook GR, Zheng H, 168. Ehehalt R, Keller P, Haass C, Thiele ty loci in addition to APOE. Am J Hum
et al. Age-related cognitive deficits, im- C, Simons K. Amyloidogenic processing Genet 2008;83:623-32.
paired long-term potentiation and reduc- of the Alzheimer beta-amyloid precursor 183. Lambert JC, Heath S, Even G, et al.
tion in synaptic marker density in mice protein depends on lipid rafts. J Cell Biol Genome-wide association study identifies
lacking the beta-amyloid precursor pro- 2003;160:113-23. variants at CLU and CR1 associated with
tein. Neuroscience 1999;90:1-13. 169. St George-Hyslop PH. Molecular ge- Alzheimers disease. Nat Genet 2009;41:
154. Arriagada PV, Marzloff K, Hyman netics of Alzheimers disease. Biol Psy- 1094-9.
BT. Distribution of Alzheimer-type patho- chiatry 2000;47:183-99. 184. Sager KL, Wuu J, Leurgans SE, et al.
logic changes in nondemented elderly in- 170. Strittmatter WJ, Roses AD. Apolipo- Neuronal LR11/sorLA expression is re-
dividuals matches the pattern in Alzhei- protein E and Alzheimers disease. Annu duced in mild cognitive impairment. Ann
mers disease. Neurology 1992;42:1681-8. Rev Neurosci 1996;19:53-77. Neurol 2007;62:640-7.
155. Roher AE, Weiss N, Kokjohn TA, et 171. Reiman EM, Chen K, Liu X, et al. 185. Rogaeva E, Meng Y, Lee JH, et al. The
al. Increased A beta peptides and reduced Fibrillar amyloid-beta burden in cogni- neuronal sortilin-related receptor SORL1
cholesterol and myelin proteins charac- tively normal people at 3 levels of genetic is genetically associated with Alzheimer
terize white matter degeneration in Alz- risk for Alzheimers disease. Proc Natl disease. Nat Genet 2007;39:168-77.
heimers disease. Biochemistry 2002;41: Acad Sci U S A 2009;106:6820-5. 186. Planel E, Bretteville A, Liu L, et al.
11080-90. 172. Holtzman DM, Bales KR, Tenkova T, Acceleration and persistence of neurofi-
156. Sisodia SS. Biomedicine: a cargo re- et al. Apolipoprotein E isoform-dependent brillary pathology in a mouse model of
ceptor mystery APParently solved? Science amyloid deposition and neuritic degener- tauopathy following anesthesia. FASEB J
2002;295:805-7. ation in a mouse model of Alzheimers 2009;23:2595-604.
157. Butler D, Bendiske J, Michaelis ML, disease. Proc Natl Acad Sci U S A 2000; 187. Xie Z, Culley DJ, Dong Y, et al. The
Karanian DA, Bahr BA. Microtubule-sta- 97:2892-7. common inhalation anesthetic isoflurane
bilizing agent prevents protein accumula- 173. Kivipelto M, Helkala EL, Laakso MP, induces caspase activation and increases
tion-induced loss of synaptic markers. et al. Midlife vascular risk factors and Alz- amyloid beta-protein level in vivo. Ann
Eur J Pharmacol 2007;562:20-7. heimers disease in later life: longitudi- Neurol 2008;64:618-27.
158. Li G, Faibushevich A, Turunen BJ, et nal, population based study. BMJ 2001; 188. Henderson VW. Estrogens, episodic
al. Stabilization of the cyclin-dependent 322:1447-51. memory, and Alzheimers disease: a criti-
kinase 5 activator, p35, by paclitaxel de- 174. Vega GL, Weiner MF, Lipton AM, et cal update. Semin Reprod Med 2009;
creases beta-amyloid toxicity in cortical al. Reduction in levels of 24S-hydroxycho- 27:283-93.
neurons. J Neurochem 2003;84:347-62. lesterol by statin treatment in patients 189. Csernansky JG, Dong H, Fagan AM,
159. Sigurdsson EM. Immunotherapy tar- with Alzheimer disease. Arch Neurol 2003; et al. Plasma cortisol and progression of
geting pathological tau protein in Alz- 60:510-5. dementia in subjects with Alzheimer-type
heimers disease and related tauopathies. 175. Cordle A, Landreth G. 3-Hydroxy-3- dementia. Am J Psychiatry 2006;163:
J Alzheimers Dis 2008;15:157-68. methylglutaryl-coenzyme A reductase in- 2164-9.
160. Matsuoka Y, Jouroukhin Y, Gray AJ, hibitors attenuate beta-amyloid-induced 190. Hall CB, Lipton RB, Sliwinski M,
et al. A neuronal microtubule-interacting microglial inflammatory responses. J Neu- Katz MJ, Derby CA, Verghese J. Cognitive
agent, NAPVSIPQ, reduces tau pathology rosci 2005;25:299-307. activities delay onset of memory decline
and enhances cognitive function in a 176. Pedrini S, Carter TL, Prendergast G, in persons who develop dementia. Neu-
mouse model of Alzheimers disease. Petanceska S, Ehrlich ME, Gandy S. Mod- rology 2009;73:356-61.
J Pharmacol Exp Ther 2008;325:146-53. ulation of statin-activated shedding of 191. Chobanian AV, Bakris GL, Black HR,
161. Busser J, Geldmacher DS, Herrup K. Alzheimer APP ectodomain by ROCK. et al. Seventh report of the Joint National
Ectopic cell cycle proteins predict the sites PLoS Med 2005;2(1):e18. Committee on Prevention, Detection, Eval-
of neuronal cell death in Alzheimers dis- 177. Sparks DL, Sabbagh MN, Connor DJ, uation, and Treatment of High Blood
ease brain. J Neurosci 1998;18:2801-7. et al. Atorvastatin for the treatment of mild Pressure. Hypertension 2003;42:1206-52.
162. Yang Y, Mufson EJ, Herrup K. Neu- to moderate Alzheimer disease: prelimi- 192. Savva GM, Wharton SB, Ince PG,
ronal cell death is preceded by cell cycle nary results. Arch Neurol 2005;62:753-7. Forster G, Matthews FE, Brayne C. Age,
events at all stages of Alzheimers dis- 178. Jones RW, Kivipelto M, Feldman H, neuropathology, and dementia. N Engl J
ease. J Neurosci 2003;23:2557-63. et al. The Atorvastatin/Donepezil in Alz- Med 2009;360:2302-9.
163. Liu DX, Greene LA. Neuronal apop- heimers Disease Study (LEADe): design Copyright 2010 Massachusetts Medical Society.
tosis at the G1/S cell cycle checkpoint. and baseline characteristics. Alzheimers
Cell Tissue Res 2001;305:217-28. Dement 2008;4:145-53.

344 n engl j med 362;4 nejm.org january 28, 2010

The New England Journal of Medicine


Downloaded from www.nejm.org on September 21, 2010. For personal use only. No other uses without permission.
Copyright 2010 Massachusetts Medical Society. All rights reserved.

Potrebbero piacerti anche