You are on page 1of 12

REVIEWS

A systemic view of Alzheimer disease


— insights from amyloid‑β metabolism
beyond the brain
Jun Wang1*, Ben J. Gu2*, Colin L. Masters2 and Yan-Jiang Wang1
Abstract | Alzheimer disease (AD) is the most common type of dementia, and is currently
incurable; existing treatments for AD produce only a modest amelioration of symptoms. Research
into this disease has conventionally focused on the CNS. However, several peripheral and
systemic abnormalities are now understood to be linked to AD, and our understanding of how
these alterations contribute to AD is becoming more clearly defined. This Review focuses on
amyloid‑β (Aβ), a major hallmark of AD. We review emerging findings of associations between
systemic abnormalities and Aβ metabolism, and describe how these associations might interact
with or reflect on the central pathways of Aβ production and clearance. On the basis of these
findings, we propose that these abnormal systemic changes might not only develop secondary to
brain dysfunction but might also affect AD progression, suggesting that the interactions between
the brain and the periphery have a crucial role in the development and progression of AD. Such
a systemic view of the molecular pathogenesis of AD could provide a novel perspective for
understanding this disease and present new opportunities for its early diagnosis and treatment.

The global burden of Alzheimer disease (AD), already abnormalities and AD pathogenesis in the brain. We
the most common type of dementia, is expected to review emerging findings of associations between sys-
increase still further owing to population ageing. AD not temic abnormalities and Aβ metabolism, and describe
only causes severe distress for patients and caregivers, how these associations might interact with or reflect on
but also results in a large economic burden on society. the central pathways of Aβ production and clearance.
Current major challenges in AD include the lack of reli- On the basis of these findings, we suggest that inter­
able biomarkers for its early diagnosis, as well as the lack actions between the brain and the periphery might have
of effective preventive strategies and treatments1,2. Thus, a crucial role in the development and progression of AD.
increased understanding of the molecular patho­genesis
1
Department of Neurology of AD could lead to the development of improved Aβ biogenesis and catabolism
and Centre for Clinical
­diagnostic and therapeutic strategies. A steady accrual of data from laboratories and clinics
Neuroscience, Daping
Hospital, Third Military AD is conventionally regarded as a CNS disorder. is providing increasing support for the concept that an
Medical University, However, increasing experimental, epidemiological and imbalance between the production and clearance of Aβ is
10 Changjiang branch road, clinical evidence has suggested that manifestations of a very early (and often initiating) factor in AD3. Normal
Daping, Chongqing, 400042, AD extend beyond the brain. These systemic alterations metabolism of Aβ and maintenance of the homeostatic
China.
2
The Florey Institute,
might not be simply secondary effects of the cerebral balance between Aβ production and clearance is, there-
The University of Melbourne, degeneration seen in AD, but could reflect under­lying fore, essential to maintain brain health. In fact, physio-
30 Royal Parade, Parkville, processes linked to progression of the disease. AD logical metabolism of Aβ occurs not only in the brain
Victoria 3052, Australia. pathogenesis is complex, involving abnormal amyloid-β but also in the periphery, and c­ ommunication between
*These authors contributed
(Aβ) metabolism, tau hyperphosphorylation, oxidative these regions is possible (FIG. 1).
equally to this work.
stress, reactive glial and microglial changes, and other
Correspondence to
Y.-J.W. and C.L.M. 
pathological events. Given that Aβ is a major hallmark Central and peripheral production of Aβ
yanjiang_wang@tmmu.edu.cn; of AD and a fertile area of research in this disease, this Aβ is derived from the proteolytic cleavage of amyloid
c.masters@florey.edu.au Review focuses on the systemic role of Aβ in AD. We dis- precursor protein (APP), which is expressed not only in
doi:10.1038/nrneurol.2017.111 cuss the communication between peripheral and central brain cells, including neurons, astrocytes and microglia,
Published online 29 Sep 2017 pools of Aβ, and describe interactions between systemic but also in peripheral organs and tissues, such as the

612 | OCTOBER 2017 | VOLUME 13 www.nature.com/nrneurol


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points These factors might also help to explain why Aβ


aggregates are mainly deposited in the brain and cere­
• An imbalance between the production and clearance of amyloid‑β (Aβ) is an early, bral vessel walls, and only rarely in peripheral organs
often initiating, factor in Alzheimer disease (AD) (although detection of Aβ aggregates has been claimed
• Peripheral systems are suggested to be involved in Aβ production and clearance in skin, subcutaneous tissue, intestinal tissues, and
• The central and peripheral pathways of Aβ metabolism communicate with each other, heart)13–15. The aggregation (oligomerization and fibrillo­
and work synergistically to clear Aβ from the brain genesis) of Aβ peptides is determined by the relative
• Increasing experimental, epidemiologic and clinical evidence suggests that AD proportions of Aβ species, their concentrations, the pH,
manifestations extend beyond the brain, and that AD pathogenesis is closely temperature and ionic strength of solution, and incuba-
associated with systemic abnormalities tion time16. In the periphery, therefore, an increased pro-
• The systemic abnormalities in patients with AD might not be secondary to the portion of Aβ40 might lead to sequestration of Aβ42 in a
cerebral degeneration; instead, they might reflect underlying disease processes stable mixed formation, thereby preventing its oligomer-
• A systemic view of AD provides a novel perspective for understanding the role of Aβ ization and aggregation17, whereas an increased propor-
in AD pathogenesis and offers opportunities for the development of new treatments tion of Aβ42 in the brain might render Aβ42 ­susceptible
and diagnostic biomarkers for AD to aggregation.

Central and peripheral clearance of Aβ


adrenal gland, kidney, heart, liver, spleen, pancreas, mus- The failure to clear Aβ (especially Aβ42) is an important
cles, and various blood and endothelial cells4,5. Aβ levels cause of sporadic AD, which accounts for 99% of AD
(in both peripheral tissues and the brain) are known to cases. Understanding how Aβ is physiologically cleared
be lower in cognitively normal elderly individuals than from the brain is, therefore, essential. Several poten-
in patients with AD (TABLE 1). The accumulation of Aβ tial pathways could clear Aβ from the brain: phago-
aggregates in elderly patients with and without AD is an cytosis, endocytosis and macropinocytosis by various
important factor that could influence the ratio of Aβ42 cells (such as microglia, perivascular macrophages,
to Aβ40 in both brain and periphery. Given that skeletal astrocytes, oligo­dendroglia and neurons); proteolytic
muscle represents about one-quarter of body weight in degradation by various enzymes (including neprilysin,
humans and is just one of many peripheral sources of Aβ, insulin-­degrading enzyme (IDE) and matrix metallo-
peripherally derived Aβ is likely to represent a substantial proteinases); and efflux of Aβ to the peripheral circula-
proportion of the total. However, levels and profiles of the tion, via transportation across the blood–brain barrier
dominant Aβ species in brain and periphery still need to (BBB) and blood–CSF barrier, interstitial fluid bulk flow
be measured in young people without Aβ deposition in and CSF egress pathways, including arachnoid villi and
future studies, for comparison purposes. glymphatic–­lymphatic pathways18 (FIG. 1). Some endo­
Important differences have been found between the genous inhibitors of Aβ aggregation, such as the secreted
central and peripheral pools of Aβ. First, Aβ42, which is ectodomain of tumour necrosis factor receptor super-
the most aggregation-prone and most neurotoxic form family member 16 (also known as low affinity neuro-
of Aβ, is the dominant molecular species in the brain, trophin receptor p75NTR)19 and the N‑terminal domain
whereas Aβ40 is dominant in the periphery, although the of myelin basic protein20, prevent Aβ deposition in the
mechanism underlying this difference is still unclear. brain and f­ acilitate its efflux into the circulation.
Differential expression of APP isoforms in the brain and How Aβ is cleared in the periphery is poorly under-
periphery is one possible explanation. APP695 is the domi­ stood. Previous studies have suggested that ~60% of
nant species produced by neurons, whereas APP751 and brain Aβ is cleared via transportation to the periph-
APP770 are the dominant species produced by peripheral ery 21,22. Our group has demonstrated, in a mouse model
cells, including platelets and leukocytes6. Another possi- of AD, that brain-derived Aβ can be physiologically
ble explanation is that the tissue microenvironment dif- cleared in the periphery, and that a singular peripheral
fers between the CNS and periphery, which could result system can remove ~40% of the Aβ produced in the
in differential processing of APP by γ‑secretase and gen- brain23. These findings indicate that peripheral clearance
eration of different Aβ species4,5. Second, levels of Aβ in has a crucial role in removing brain-derived Aβ, and sug-
the central pool are higher than those in the peripheral gest that effective peripheral Aβ clearance can improve
pool. Concentrations of Aβ in cerebrospinal fluid (CSF) the efficacy of Aβ efflux from the brain. In fact, several
are at least 5–15 times higher than those in plasma7,8. One peripheral tissues or organs participate in Aβ catabolism
possible interpretation is that APP processing in periph- and constitute potential Aβ clearance pathways. These
eral cells probably occurs via α‑cleavage (rather than the include uptake and phagocytosis or endocytosis by
β‑cleavage used in neurons)9,10, resulting in decreased monocytes, macrophages, neutrophils, lymphocytes, and
peripheral production of Aβ. Another explanation for hepatocytes24,25; excretion via bile or urine26,27; proteo-
the lower Aβ levels in the periphery is that the periphery lytic degradation by Aβ‑degrading enzymes28; and clear-
contains abundant Aβ‑binding proteins (lipoproteins and ance from blood mediated by Aβ‑binding proteins and
albumin)11 and Aβ‑binding cells, such as erythrocytes12, cells, such as erythrocytes, albumin, antithrombin III
which all contribute to Aβ transportation and clearance. and lipoproteins, including apolipoprotein E (ApoE) and
Additionally, the high volume of the circulatory system apolipoprotein J (ApoJ)11,12. These central and periph-
and the blood-dilution effect efficiently reduce systemic eral pathways might interact with each other and work
Aβ concentrations. ­synergistically to clear Aβ from the brain.

NATURE REVIEWS | NEUROLOGY VOLUME 13 | OCTOBER 2017 | 613


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Brain
Aβ production Aβ clearance

Degradation clearance ISF bulk flow clearance


(by microglia, astrocytes, (CSF sink, perivascular
neurons, etc., or by drainage or glymphatic
Aβ-degrading enzymes) pathways)

CNS clearance
Neuron
CNS pool

Aβ in ISF Aβ in CSF

Astrocyte
Aβ Aggregation and deposition
(mainly Aβ42)
CSF absorption clearance
BBB clearance (arachnoid villi, BCSFB,
(LRP1, P-glycoprotein, etc) lymphatic pathway)

Microglia
Influx Efflux
Blood
Aβ production Aβ clearance

BBB entrance
(RAGE)
Degradation clearance
(phagocytosis by blood
cells and degradation
Monocytes Neutrophils Aβ-degrading by enzymes)
enzymes

Blood flow clearance


Aβ (by RBCs, lipoproteins,
(mainly Aβ42) albumin, etc)

Peripheral clearance
RBC Lipoproteins Albumin
Peripheral pool

Platelets

Peripheral organs or tissues


Aβ production Aβ clearance

Osteoblasts

Aβ Macrophages Liver Kidney


Skin fibroblasts (mainly Aβ42)
Degradation clearance Degradation clearance
(by macrophages in tissues) (by hepatocytes) Excretion with urine
Excretion with bile

Skeletal muscle cells

Figure 1 | Physiological metabolism of Aβ in the brain and periphery. Amyloid‑β (Aβ) is generated by neurons,
Nature Reviews | Neurology
microglia and astrocytes in the brain, and by platelets, skin fibroblasts, osteoblasts, and skeletal muscle cells in the
periphery. The CNS and peripheral pools of Aβ can interact; some Aβ peptides in the CNS are cleared via phagocytosis or
proteolytic degradation, whereas others are released into the blood via the blood–brain barrier (BBB), interstitial fluid (ISF)
bulk flow or cerebrospinal fluid (CSF) egress pathways. Some Aβ peptides in blood are phagocytosed, including by
monocytes or neutrophils, some are degraded by Aβ-degrading enzymes, and some are transported by carriers (such as
erythrocytes, albumin and lipoproteins) to peripheral organs or tissues, where they are degraded by macrophages or
hepatocytes, or excreted via the liver or kidney. BCSFB, blood–CSF barrier; RAGE, receptor for advanced glycation end
products; RBC, red blood cell.

Communication between Aβ pools BBB, including LDL-related protein 1 (LRP1) and ATP-
Brain-derived Aβ can be transported into the periph- dependent efflux transporter P-glycoprotein29. The
eral pool via the BBB, blood–CSF barrier, arachnoid arachnoid villi absorb Aβ in the CSF and mediate its
villi or glymphatic–lymphatic pathway. Several trans- release into the circulation30. The glymphatic–lymphatic
porters mediate Aβ flow out of the brain across the pathway, which consists of the glymphatic pathway in

614 | OCTOBER 2017 | VOLUME 13 www.nature.com/nrneurol


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 | Amounts of Aβ in the CNS and periphery processes in the AD brain can also drive these systemic
disorders, forming feedback loops. Mechanisms that
Source of Aβ Cognitively normal Patients with Alzheimer Refs might underlie the effects of systemic abnormalities or
elderly individuals disease
alterations on Aβ metabolism are outlined in TABLE 2.
Brain grey matter Here, we discuss the interactions between Aβ metabo­
Total Aβ* 3.2 μg/g 13.1 μg/g 5,168 lism in the brain and periphery, and place them in a
systemic context.
Aβ40 0.2 μg/g 0.6 μg/g 5
Aβ42 0.78 μg/g 6.1 μg/g 5 Disorders of systemic immunity
Packed quiescent platelets ‡ One of the primary pathways of Aβ clearance in the
84.0 ng/ml Not available 5
brain is phagocytosis or endocytosis by professional
Aβ40
phagocytes and microglia, as well as by astrocytes,
Aβ42 1.7 ng/ml Not available 5 oligodendrocytes and neurons. Accumulations of
Packed activated platelets Aβ in the periphery can similarly be phagocytosed
Aβ40 56.8 ng/ml Not available 5 by monocytes and neutrophils in the blood, and by
macrophages in tissues41. Of note, in transgenic mice
Aβ42 1.6 ng/ml Not available 5 with AD, expression of Aβ scavenger receptors and
Skeletal muscle§ Aβ‑degrading enzymes in circulating mononuclear
Aβ40 29.8 ng/g 37.8 ng/g 5 phagocytes decreases substantially as these mice age42,
and the phagocytic functions of these cells are impaired
Aβ42 10.2 ng/g 15.7 ng/g 5
in both mice and humans with AD43–45. Infusion of
*Includes Aβ40, Aβ42 and shorter peptides (Aβn–40, Aβn–42).‡Suggested to be the primary source of monocytes derived from peripheral human umbilical
soluble Aβ (which is mainly Aβ40) in the circulation169,170. §Aβ might also be generated by skin
fibroblasts and osteoblasts5,171–173, although these findings have not yet been confirmed. cord blood reduces the Aβ burden and improves cog-
Aβ, amyloid‑β. nitive deficits in a mouse model of AD46, implying that
peripheral mononuclear phagocytes have an important
role in Aβ clearance. Promoting the phagocytic func-
the brain and the CNS lymphatic vessels (discovered in tion of peripheral blood monocytes or promoting the
2015)31,32, might also transport Aβ from the brain to the recruitment of peripheral macrophages into the brain
periphery for clearance18,33. However, the glymphatic–­ might, therefore, improve Aβ clearance in the brain47,
lymphatic pathway and arachnoid villi are unidirec- although the existence of conflicting data48 renders this
tional; they only mediate Aβ efflux from the CNS to approach controversial.
the periphery 18. In this regard, a cluster of genes associated with the
Whether peripherally generated Aβ can enter the risk of sporadic AD (including CD33, CR1, MS4A6A,
brain and exert neurotoxic effects there remains poorly MS4A4E, ABCA7 and TREM2)49,50 encode proteins that
understood. In the absence of a relevant transport are involved in innate immunity. Variants in these genes,
mechanism, systemic amyloidosis might not neces­ especially in TREM2 and CD33, are associated with
sarily lead to AD. However, peripheral inoculation compromised phagocytic function of peripheral mono-
of Aβ‑containing brain extracts induces cerebral Aβ cytes or macrophages and altered Aβ accumulation in
deposition in both mice and humans, suggesting that AD brains24,51. Interestingly, CR1 (encoding complement
peripherally generated Aβ is able to enter the brain and receptor‑1, also known as CD35) is expressed primarily
participate in the pathogenesis of AD34–37. Receptor for in peripheral leukocytes and erythrocytes, but not in any
advanced glycation end products (RAGE) has been sug- brain cells.
gested to transport Aβ across the BBB, from the blood In regard to adaptive immunity, much attention
into the brain38. Expression of the AGER gene (encoding has been focused on autoimmunity and autoreactive
RAGE) is upregulated in the AD brain vasculature39,40, antibodies related to the pathogenesis of AD, includ-
indicating that influx of peripheral Aβ into the brain ing naturally occurring antibodies and autoantibodies.
is increased in AD. The contribution of peripherally These autoreactive antibodies are ubiquitous in human
derived Aβ to amyloidosis in the AD brain needs to be blood and CSF, and profiles of these antibodies are
determined in future studies. altered in patients with AD52–56. Identification of the
A decline in peripheral Aβ clearance might also most antigenic epitopes targeted by human antibodies
impede efflux of Aβ from the brain to the periphery, against Aβ aggregates could lead to development of an
and thereby attenuate central clearance of Aβ. Moreover, effective immunotherapy for AD. Aducanumab, derived
the influx and efflux of Aβ might result in equilib- from a naturally occurring human autoantibody against
rium between the central and peripheral pools of Aβ. Cu 2+‑modified Aβ aggregates (which are the most
Mechanisms that might regulate this equilibrium need neuro­toxic Aβ species in the AD brain), showed prom-
to be understood. ise in clearing brain Aβ deposits and improving cogni-
tion in a 2016 phase Ib trial57. Lymphocytes (including
Systemic abnormalities in AD B cells, T cells and natural killer cells) also participate
An increasing number of studies indicate that a series in Aβ clearance via immunoglobulin-mediated adaptive
of systemic abnormalities can exacerbate the progres- phagocytosis58,59. Future studies will help to elucidate
sion of AD (FIG. 2). In turn, the downstream effects of the crosstalk between innate immunity and adaptive

NATURE REVIEWS | NEUROLOGY VOLUME 13 | OCTOBER 2017 | 615


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Disorders of systemic immunity


• Phagocytic function of mononuclear
phagocytes ↓ Blood abnormalities
• Altered profiles of autoreactive antibodies • APP expression on platelets ↑
• AD risk variants in immunity-associated • Altered APP isoform ratio and APP
genes: CD33, CR1, ABCA7, MS4A6A, processing pattern in platelets
MS4A4E, TREM2 • Quantity and Aβ-binding ability of RBC ↓
• Albumin ↓, Aβ-degrading enzymes ↓

Cardiovascular disease Respiratory and sleep disorders


• Myocardial function ↓ • OSA and COPD are associated with
• Ejection fractions ↓ increased plasma Aβ levels, which are
• Cerebral blood flow velocities ↓ negatively correlated with pulmonary
• PI and RI in BA, LTICA, RTICA ↑ function
• Intramyocardial deposits of Aβ
Renal dysfunction
Hepatic dysfunction • Serum Aβ levels are positively correlated
• Plasma Aβ levels are positively correlated with impaired renal function
with impaired hepatic function
• Aβ in liver tissue ↓
Microbiota disturbance and infection
• Disturbance of microbiome
Metabolic disorders • Plasma LPS and antibodies to
• Glucose metabolism ↓ anaerobes ↑
• Deficits in insulin and IGF signalling
• LDL-C ↑, HDL-C ↓
• Membrane fluidity of platelets and Systemic inflammation
leukocytes ↑ • Plasma proinflammatory cytokines ↑,
including TNF, IL-1β, IL-6, CRP
• AD risk variants in lipid-metabolism- • Infectious burden ↑
associated genes: APOE, BIN1, APOJ,
SORL1, PICALM, PLD3

Figure 2 | Systemic abnormalities in AD. Various systemic abnormalities have been found in patients with Alzheimer
Nature Reviews | Neurology
disease (AD). Red boxes highlight AD risk variants in genes related to innate immunity, phagocytosis of amyoid-β (Aβ)
by immune cells, and lipid metabolism in periphery and brain, respectively, which were identified in genome-wide
association studies and candidate-gene studies of sporadic AD. APP, amyloid precursor protein; BA, basilar artery; COPD,
chronic obstructive pulmonary disease; CRP, C‑reactive protein; HDL-C, HDL-cholesterol; IGF, insulin-like growth factor;
LDL-C, LDL-cholesterol; LPS, lipopolysaccharide; LTICA, left terminal internal carotid artery; OSA, obstructive sleep
apnoea; PI, pulsatility index; RBC, red blood cells; RI, resistance index; RTICA, right terminal internal carotid artery;
TNF, tumour necrosis factor.

immunity, and to discover how the interaction of identification of these protective components could be
these two immune systems might synergistically affect of importance in understanding the pathogenesis of AD
AD pathogenesis. and in developing systemic rejuvenation therapies68–71.

Blood abnormalities Metabolic disorders


Besides monocytes and leukocytes, other blood com- Diabetes mellitus. How diabetes mellitus affects Aβ
ponents are also involved in Aβ metabolism. Increased catabo­lism and AD risk is not yet well understood. Patients
expression of APP, altered APP isoform ratios and pro- with diabetes mellitus are estimated to be 1.4–2.0‑fold
cessing patterns, and enhanced β‑secretase activity are more likely than healthy individuals to develop AD72,73,
observed in platelets from patients with AD60–62, and although these claims need to be verified in patients with
these changes could result in overproduction of Aβ biomarker-­confirmed AD. In patients with diabetes mel-
in the periphery. Blood levels of albumin, an Aβ car- litus, insulin resistance substantially compromises the
rier, are reduced in patients with AD63. The quantity positive effects of insulin on both cognition and hepatic
and function of erythrocytes (another Aβ carrier) are clearance of circulating Aβ74,75, resulting in AD‑like alter-
also altered in such patients, and their erythrocytes ations in the brain. Moreover, excess insulin can competi-
show compromised binding of Aβ64,65. In addition, the tively inhibit IDE-mediated Aβ degradation76. Some other
activity of Aβ‑degrading enzymes in serum is thought pathological features of diabetes mellitus — including
to be decreased in AD28. These changes impede Aβ oxidative stress, BBB disruption and reduced cell energy
­transportation and clearance in the periphery. supply — can also affect Aβ generation and clearance77,78.
Some anti-ageing molecules, such as growth and dif- In addition, amylin (a misfolded protein deposited in the
ferentiation factor 11, granulocyte–macrophage colony pancreas in patients with type 2 diabetes mellitus) can
stimulating factor, and metalloproteinase inhibitor 2, enter the brain, where it accelerates and exacerbates the
have been identified in blood from young mice and misfolding and aggregation of Aβ79. However, atheroscle-
in human umbilical cord plasma66,67. Whether levels rosis and small vessel disease (discussed in more detail
of these anti-ageing molecules are reduced in patients below) can be important causes of cognitive dysfunction in
with AD, and whether they are pathophysiologically patients with diabetes mellitus80, and should be ­considered
relevant to this disease, remain unknown. However, in the differential diagnosis of AD in this setting.

616 | OCTOBER 2017 | VOLUME 13 www.nature.com/nrneurol


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 2 | Potential mechanisms of systemic abnormalities that affect Aβ metabolism


Abnormality Potential mechanisms Refs
Disorders Reduced expression of Aβ phagocytic receptors and Aβ‑degrading enzymes, and decreased phagocytic function in 41–45
of systemic mononuclear macrophages and neutrophils, might impede Aβ degradation and clearance
immunity
Increased levels of pathogenic autoreactive antibodies and decreased levels of protective antibodies could influence 52–54
Aβ generation, clearance and deposition
Lymphocytes produce immunoglobulins that bind to Aβ and form immune complexes, such as Aβ–IgG and Aβ–IgM, 58,59
which undergo clearance via phagocytosis
Blood Increased APP expression, altered APP isoform ratios and APP processing patterns, and enhanced β‑secretase activity 60–62
abnormalities in platelets could promote overproduction of Aβ in the periphery
Altered quantity and function of erythrocytes, and compromised adherence of Aβ to erythrocytes, might hinder Aβ 64,65
transportation and clearance
Reduced levels of albumin (an Aβ carrier) in blood might hinder Aβ transportation and clearance 63
Decreased activity of Aβ‑degrading enzymes could impede Aβ degradation and clearance 28
Metabolic Excess insulin in diabetes mellitus competitively inhibits insulin-degrading enzyme-mediated Aβ degradation 76
disorders
Insulin resistance compromises intracellular translocation of LRP1 to the plasma membrane in hepatocytes, potentially 74
hindering hepatic clearance of circulating Aβ
Increased oxidative metabolism of glucose might cause BBB disruption, further impeding Aβ clearance 77
Changes in mitochondrial function and mitochondria-derived free radicals could affect Aβ generation and degradation 78
Amylin interacts with Aβ by cross-seeding, which could accelerate or exacerbate the misfolding and aggregation of Aβ 79
in the brain
ApoE is an Aβ chaperone; the ApoE–Aβ complex facilitates Aβ clearance via autophagy, BBB transportation, enzymatic 83,84
degradation and glymphatic pathways
Low cholesterol levels could shift APP processing out of the lipid raft region, where α‑cleavage is preferred; high 85,86
cholesterol levels might disrupt the BBB through inflammation or other pathways
Abnormal membrane lipid components might affect membrane fluidity, which is involved in membrane APP 87–90
processing, cellular phagocytosis and endocytosis, which all affect Aβ generation and clearance
Cardiovascular The association between cardiovascular diseases and AD might be attributable to common risk factors or concomitant 96,97
diseases diseases, such as diabetes mellitus, hypertension, hypercholesterolemia and stroke
Cardiac systolic function drives blood flow, which is responsible for the transportation of Aβ and its chaperones; 98
cardiac systolic dysfunction, reflected by reduced ejection fractions, could impair Aβ clearance
Reduced cerebral blood flow could cause hypoxia, promoting Aβ generation via upregulating BACE1 100,101,
174,175
Reduced cerebral blood flow and hypoxia could lead to an energy deficiency in cells of the neurovascular unit, 102
breaking down the BBB and impairing clearance of Aβ
Hepatic Hepatic dysfunction might cause reduced hepatocyte uptake and degradation of Aβ directly from the blood 108
dysfunction
Hepatic dysfunction might result in dysregulation of Aβ‑related protein and lipid metabolism, thereby impairing Aβ 11,108
transportation and clearance
Hepatic dysfunction could lead to reduced Aβ excretion in bile 26
Renal Renal dysfunction could result in reduced Aβ excretion in urine 112,113
dysfunction
Respiratory Hypoxia in respiratory diseases might induce Aβ overproduction via upregulation of BACE1 174,175
and sleep
disorders Sleep fragmentation in patients with sleep-disordered breathing or obstructive sleep apnoea might cause increased 121,125,
neuronal activity, leading to elevated Aβ production and aggregation; increased wakefulness might also increase 126
sympathetic output, suppressing glymphatic clearance of Aβ and tau
Inflammation and immune responses in respiratory diseases might affect Aβ generation and clearance 122
Disturbance of Some micro-organisms or their products can enter the brain and cause local damage, either via direct pathogenic 128,131
microbiota and action or through neuroinflammation and oxidative effects
infection
Some microbiota or pathogens could inhibit autophagy, leading to impaired degradation of Aβ and phosphorylated tau 131
Some microbiota, pathogens and their products could damage the BBB and thereby impede Aβ clearance 131
Systemic Proinflammatory cytokines in blood could enter the CNS (via the BBB or neural afferent pathways such as the vagus 130,134,
inflammation nerve) and participate in AD pathogenesis either directly in the CNS or via inducing immune responses 135
Acute local inflammatory responses might promote recruitment of monocytes and macrophages into the brain, 47
facilitating clearance of cerebral Aβ
Aβ, amyloid‑β; APP, amyloid precursor protein; AD, Alzheimer disease; ApoE, apolipoprotein E; BBB, blood–brain barrier; LRP1, LDL receptor-related protein‑1.

NATURE REVIEWS | NEUROLOGY VOLUME 13 | OCTOBER 2017 | 617


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Lipid and lipoprotein risk factors. Some evidence Hepatic dysfunction


suggests that abnormal lipid metabolism is associ- The liver is the major organ responsible for system-wide
ated with an increased risk of AD81. Several potential metabolic regulation, protein synthesis and metabolic
AD risk genes (including APOE, BIN1, CLU, SORL1, detoxification. Circulating Aβ is predominantly cleared
PICALM and PLD3) encode proteins linked to lipid by either degradation in hepatocytes or direct excretion
metabolism82. Among these, ApoE is known to partici- in bile; several peptide clearance experiments have sug-
pate in Aβ production, aggregation, and clearance in an gested that soluble Aβ has a short half-life of 2.5 min to
­isoform-dependent manner 83,84. 2.5 h in the circulation26,107. LRP1 is thought to mediate
Cholesterol levels in the brain can affect Aβ syn- the uptake of Aβ by hepatocytes25. The liver might also
thesis, clearance and neurotoxicity. High serum chol­ indirectly influence Aβ clearance by regulating albu-
esterol levels are associated with an increased cerebral min levels and Aβ‑related lipid metabolism. Plasma Aβ
burden of Aβ85,86. Abnormal cholesterol levels could levels inversely correlate with liver function, suggesting
reflect unmeasured genetic factors or dietary patterns that hepatic dysfunction attenuates peripheral Aβ clear-
that might affect the pathogenesis of AD85,86. Cell mem- ance108. Liver tissue from patients with AD contains less
brane fluidity is strongly affected by its lipid composi- Aβ than that from healthy individuals, which implies
tion, and increased membrane fluidity of platelets and that the Aβ‑clearance function of liver is compromised
leukocytes has been reported in patients with AD, as in patients with AD5.
well as in individuals with Down syndrome (who have Whether liver dysfunction also increases the Aβ load
a greatly increased risk of developing dementia and in the brain remains unknown; however, treatments that
AD)87–90. Cell membrane fluidity also influences the enhance LRP1‑mediated Aβ uptake by the liver alleviate
processing of APP and cellular phagocytosis, both of both the burden of Aβ in the brain and cognitive impair-
which might affect Aβ generation and clearance. A high ment74,109. These observations suggest that improving the
dietary intake of polyunsaturated fatty acids such as Aβ clearance capacity of the liver is a potential systemic
docosahexaenoic acid, which maintain membrane flu- therapeutic approach for AD.
idity, have a beneficial effect on cognition in patients
with AD91,92. The study of membrane fluidity in AD Renal dysfunction
might provide insights into the alteration of mem- Soluble Aβ is a normal component of human urine27.
brane-dependent biological functions related to Aβ, In addition, animal experiments have shown that, after
such as phagocytosis, endocytosis, m ­ acropinocytosis intracranial or intravenous infusion of 125I-labelled Aβ,
and autophagy. radioactivity is subsequently detected in the kidney and
urine23,110. These findings indicate that the kidney might
Cardiovascular disease participate in physiological clearance of Aβ by filtering
Emerging evidence indicates that cardiovascular dis- Aβ from blood to urine. Conversely, renal dysfunction
ease (CVD) is a major comorbidity in patients with probably leads to impaired peripheral Aβ clearance.
sporadic AD. A low cardiac index and heart failure are In support of this notion, serum Aβ levels inversely cor-
both associ­ated with dementia, and perhaps also with relate with measures of renal function (estimated glo-
AD93–95. However, as CVD and AD are both complex merular filtration rate and creatinine levels) in patients
and multifactorial age-related diseases, the association with chronic kidney disease111,112. Moreover, human
between them might be attributed partly to shared kidney donors have decreased estimated glomerular
risk factors, such as diabetes mellitus, hypertension, filtration rates and increased circulating levels of Aβ113,
­hypercholesterolemia and stroke96,97. suggesting that the reduction in renal function reserve
The presence of compromised myocardial func- associated with having a single kidney also attenuates
tion and intramyocardial deposits of Aβ in patients peripheral Aβ clearance.
with AD suggests that peripheral Aβ accumulation Whether renal dysfunction increases Aβ burden in
could affect heart function in patients with AD15,98. the brain or facilitates AD processes remains unknown.
In addition, cardiac systolic dysfunction could affect Renal dysfunction increases the risk of both cognitive
Aβ generation and clearance in the brain as a result impairment and dementia114, and this association could
of reduced cerebral blood flow 99–102. Regional cerebral involve AD pathogenetic pathways. However, kidney
blood flow and glucose uptake or metabolism are con- transplantation can reduce plasma Aβ levels113, and
sistently decreased in Aβ‑positive patients with AD103, haemodialysis alleviates Aβ deposition in the brain of
and correlate inversely with AD severity 104. Indeed, patients with chronic kidney disease115. These observa-
some degree of cerebral small vessel disease almost tions suggest that improvement of renal function is a
always accompanies AD. Increased stiffness of small promising approach to AD prevention and treatment.
vessel walls might attenuate Aβ clearance via the BBB,
interstitial fluid bulk flow and glymphatic pathways, Respiratory and sleep disorders
thereby accelerating AD105,106. However, a clear under- Patients with AD have an increased incidence of respira-
standing of the interaction between CVD and AD is tory disorders, such as bronchopneumonia, obstructive
lacking. Most of the evidence points to CVD being an sleep apnoea (OSA) and sleep-disordered breathing 116,117.
independent risk factor for cognitive impairment, and In addition, sleep-disordered breathing is associated
having an additive rather than synergistic effect on the with an increased risk of mild cognitive impairment or
AD neurodegenerative process. dementia and with earlier onset of AD118–120.

618 | OCTOBER 2017 | VOLUME 13 www.nature.com/nrneurol


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Compared with healthy control individuals, patients associated with a reduced risk of AD136. These findings
with OSA or chronic obstructive pulmonary disease suggest that chronic systemic inflammation promotes
exhibit higher blood levels of Aβ, which negatively the AD process.
correlate with pulmonary function121,122. OSA is also By contrast, acute systemic inflammatory responses
associated with altered levels of AD biomarkers in CSF, seem to protect against AD — at least in animal ­models
including decreased levels of Aβ42 and elevated levels — by recruiting monocyte-derived macrophages into
of phosphorylated tau123. OSA and chronic obstructive the brain, where they clear cerebral Aβ47. However,
pulmonary disease could contribute to AD processes most studies in this area have not been repeated, and
via hypoxia, inflammation, or sleep disruption124. Sleep their results have not yet been validated in patients with
disruption has been suggested to increase Aβ produc- biomarker-confirmed AD.
tion and aggregation, suppress glymphatic clearance
of AD pathogenic proteins (tau as well as Aβ) and A systemic approach to understanding AD
aggravate oxidative stress, inflammation and synaptic The close interaction between the brain and the
damage125,126. periphery, in terms of Aβ metabolism, provides novel
insights into the pathogenesis of AD, and could lead to
Gut microbiota disturbance and infection new approaches to the diagnosis and treatment of AD,
The establishment of the gut–brain axis revealed a clear based on systems biology and systems neurophysiology
association between the gastrointestinal microbiota and paradigms.
cognition127. Gram-negative bacterial species (such as
Escherichia coli K99) are the predominant sources of Pathogenesis
bacteria-­derived factors in normal human brains, and Our current understanding of the role of Aβ in AD
­levels of these molecules are increased in AD brains, along focuses on its contribution to brain pathology and symp-
with levels of the bacterial cell wall component lipopoly- toms. However, as already discussed, this view might
saccharide128. Lipopolysaccharide colocalizes with Aβ in not be the whole story. First, although Aβ peptides are
plaques in AD brains128, suggesting that Gram-negative generated in the brain, a considerable amount of Aβ is
bacteria are associated with AD pathogenesis. Probiotic also generated in peripheral systems. Second, Aβ can be
supplementation is associated with improved cognition cleared — from peripheral organs or tissues as well as
in patients with AD129, which further supports a role for from the brain — by professional phagocytes, which can
the gut microbiota in AD development. transmigrate through the BBB. Third, Aβ deposits have
In addition to its relationship with normal microbial been detected in the periphery — although this claim
flora, emerging evidence indicates that AD is associ­ has not yet been replicated and its pathophysiological
ated with exposure to an ever-increasing number of relevance remains unknown. Last, a series of systemic
pathogens130,131. Moreover, a high infectious burden is abnormalities are both driven by and contribute to AD
associated with increased serum levels of Aβ and pro­ progression. On the basis of these findings, we propose
inflammatory cytokines in patients with AD and in that AD might not be solely a brain disorder, in the sense
healthy controls130. However, the underlying mechanisms that systemic factors might interact with the brain to
through which the microbiota or pathogens influence modify the AD process.
AD remain to be determined. Whether the microbiota As discussed, the central and peripheral Aβ pools
or pathogens contribute to AD development, or whether interact with and influence each other. For exam-
an increased infectious burden is a c­ onsequence of AD, ple, the rate of peripheral catabolism of Aβ seems to
also remains unknown. affect the rate of Aβ efflux from the brain, and periph-
erally derived Aβ can enter the brain and accelerate the
Systemic inflammation progression of cerebral AD pathology 34,35. Therefore, we
Chronic reactive gliosis and microgliosis are neuro­ hypothesize that the peripheral pool of Aβ is not simply
inflammatory responses that are important contributors associated with AD, but is causally linked to this disease.
to AD pathology. These processes might participate in Indeed, interactions between the brain and the periphery
a positive feedback loop of Aβ deposition, neurofibril- might have a crucial role in the natural history of AD,
lary tangle formation, and damage to synapses and and elucidation of the effects of peripheral processes on
neurons. Several studies have shown that other condi- AD development could lead to improved understand-
tions involving chronic systemic inflammation, such as ing of its pathogenesis. The crucial questions to answer
rheumatoid arthritis and periodontitis, are associated would be precisely how the brain and periphery interact
with an increased risk of AD132,133. These conditions are with each other to affect AD progression, and whether
also associated with elevated levels of C‑reactive pro- interventions that target systemic factors can modulate
tein and proinflammatory cytokines, such as tumour the pathogenesis or development of AD.
necrosis factor, IL‑6 and IL‑1β. These proinflammatory
molecules could participate in AD pathogenesis either Diagnosis
directly, by affecting brain Aβ metabolism (via entry to Several PET radiotracers can be used to detect Aβ in the
the CNS through the BBB or neural afferent pathways brain, and a few biomarkers for AD have been validated
such as the vagus nerve)134,135 or indirectly, by affect- for diagnostic use, including CSF levels of Aβ42, total tau
ing Aβ metabolism in the periphery. In this regard, the and phosphorylated tau. However, these approaches are
results of observational studies show that NSAID use is either invasive or expensive, and are impractical for the

NATURE REVIEWS | NEUROLOGY VOLUME 13 | OCTOBER 2017 | 619


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 1 | Novel peripheral biomarkers for AD Technical advances by the AD Neuroimaging


Initiative have enabled the use of microarrays to detect
Potential biomarkers related to Alzheimer disease (AD) are continually being serum anti‑Aβ autoantibodies with 100% accuracy 55;
discovered, and might enable prediction of mild cognitive impairment and progression however, this promising technique requires validation in
to AD, especially when combined with APOE genotyping and assessment for systemic large independent cohorts. This approach is particularly
dysfunctions linked to AD.
interesting in the light of the promising results obtained
Exosome cargo proteins with aducanumab, the first human anti‑Aβ autoantibody
Exosomes are cell-derived vesicles that are highly enriched in biological fluids. They to be developed for clinical trials57. In addition to Aβ
carry amyloid precursor protein (APP), amyloid-β (Aβ) and tau, and might facilitate and tau, β‑secretase 1 (encoded by BACE1, an enzyme
intercellular communication and propagation of Aβ and tau pathologies. Plasma
involved in Aβ production) has been suggested as a
exosome cargoes could have prognostic value in AD158; astrocyte-derived exosomes
contain higher levels of soluble Aβ42 and proteins of the Aβ42-generating pathway (such
potential biomarker for AD142. In general, the identifi-
as β‑secretase 1, γ‑secretase, soluble APPβ and soluble APPα) and phosphorylated tau cation of novel peripheral biomarkers for AD diagno-
than do neuron-derived exosomes. Levels of β‑secretase 1 and soluble APPβ in sis and prognosis represents a promising and rapidly
astrocyte-derived exosomes are higher in patients with AD than in healthy controls159. expanding research direction. (BOX 1)
Blood proteins
A new high-throughput multiplex protein biochip can identify patients with AD with an Treatment
accuracy of 92% when used to screen platelet lysates160. Another study found that a Currently, effective agents for AD prevention or treat-
panel of serum proteins has diagnostic value161, although the sensitivity and specificity ment are lacking. The traditional concept ‘one target,
of this test need improvement. Individual blood proteins, including biomarkers of one treatment’ inevitably ignores the complexity of AD
neurodegeneration (such as neurofilament light protein, neuron-specific enolase and pathogenetic mechanisms143. After the failure of over
heart fatty acid binding protein) and glial activation biomarkers (such as YKL‑40 100 clinical trials of monotherapies targeting Aβ, multi­
and monocyte chemotactic protein 1) also exhibit potential for AD diagnosis and targeted therapies that address various aspects of AD
prognostication162–165. Leukocyte surface protein phenotyping and functional pathogenesis at different disease stages are needed144. We
phenotyping might also prove useful; leukocyte surface expression of P2X7 is
argue that a comprehensive strategy targeting both brain
decreased in patients with AD and is linked to altered phagocytic function
of monocytes45.
and peripheral (systemic) abnormalities might be more
effective than strategies that target CNS abnormalities
Autoantibodies alone. As discussed, many comorbidities of (and risk
Autoantibodies have emerged as effective and noninvasive biomarkers for early
factors for) AD — such as diabetes mellitus, metabolic
diagnosis and staging of AD. A panel of blood antibodies can differentiate patients
disorders, cardiovascular diseases, and hypertension
with mild cognitive impairment from controls with 100% accuracy55.
— are systemic disorders.
Blood RNA profiles Many attempts have been made to prevent AD via
Circulating RNA profiles have diagnostic potential in AD. An exosomal microRNA peripheral interventions, and some have been associ-
signature exhibits high sensitivity and specificity for the detection of early AD, and
ated with beneficial outcomes. Improvements in overall
shows concordance with neuropsychological and neuroimaging data166; TRPC6 mRNA
levels in blood cells were reduced in patients with AD and mild cognitive impairment, population health have led to a decreased incidence and
and were related to brain Aβ burden and Aβ42 levels in cerebrospinal fluid167. prevalence of dementia over the past 10–30 years145–148,
perhaps through improved management of cardiovas-
cular risk factors145–148. For example, administration
early diagnosis of patients without obvious cognitive of statins (which reduce peripheral blood cholesterol
complaints. The search for peripheral blood or plasma ­levels) to healthy middle-aged individuals was associ-
biomarkers for AD that reflect AD‑related processes in ated with a reduced dementia risk in one large-scale
the brain has, therefore, received considerable attention. prospective cohort study 149; statins have also decreased
However, owing to the complexity of blood compo- the brain burden of Aβ in experimental models of
nents, the accurate measurement of plasma levels of Aβ AD150, although the results of most studies of statin
or tau is very challenging. A study published in 2017 did treatment in patients with AD have been disappoint-
not find a statistically significant difference in plasma ing 151. Furthermore, treatment with continuous positive
levels of free Aβ between patients with AD and age- airway pressure for sleep-disordered breathing or OSA
matched controls137, and similarly negative results have might delay the onset of mild cognitive impairment
been published for plasma tau levels138. However, these and slow or even improve cognitive decline in patients
results do not indicate the end of the road for AD bio- with AD120,152,153. These observations support the view
marker studies139. With the development of advanced that systemic management of an individual’s known
and highly sensitive techniques that are able to promote comorbidities or risk factors, with the aim of maintain-
efflux of Aβ and tau from the brain, and accurately ing bodily homeostasis, might help to prevent or slow
measure their levels even when bound to other serum the progression of AD.
proteins, researchers might eventually find a method Active removal of excess peripheral Aβ seems to be
to monitor cerebral Aβ accumulation. In addition, a particularly promising therapeutic strategy for AD23.
misfolded oligomeric species of Aβ and tau proteins Plasma albumin exchange both improves cognition
have been detected in CSF and suggested as potential and decreases the Aβ burden in patients with AD154.
biomarkers for AD140,141. If these misfolded protein spe- Peritoneal dialysis reduces blood Aβ levels in humans
cies also prove to be present in blood, they might be and also attenuates AD pathology in an AD mouse
useful for AD diagnosis, although no ­published reports model155; patients who have undergone haemodialysis
yet exist. exhibit a reduction in Aβ deposition in the brain115.

620 | OCTOBER 2017 | VOLUME 13 www.nature.com/nrneurol


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Approaches to improve peripheral Aβ clearance via treatment should have a corresponding focus not only on
enhancing phagocytosis45, proteolytic degradation and pathological changes in the brain but also on peripheral
excretion155,156, and identification of rejuvenation factors abnormalities, which vary among individuals. Identifying
in blood68, are other promising systemic therapeutic these peripheral abnormalities might offer new oppor-
strategies for AD157. tunities for diagnosis of early AD and lead to the design
of specific treatment strategies for individuals with pre-
Conclusions clinical, prodromal or frank AD. In conclusion, the sys-
AD might be not only a brain disorder, but also a systemic temic view of AD proposed in this Review provides a
disease with widespread abnormalities beyond the brain. novel perspective for understanding the pathogenesis of
Thus, systemic factors might interact with brain-related this disease, and fosters new opportunities for its early
factors to modify the AD process. AD diagnosis and diagnosis and successful management.

1. Mangialasche, F., Solomon, A., Winblad, B., peptide oligomerization and fibrillogenesis. J. Biol. 34. Eisele, Y. S. et al. Peripherally applied Aβ‑containing
Mecocci, P. & Kivipelto, M. Alzheimer’s disease: Chem. 278, 11612–11622 (2003). inoculates induce cerebral β‑amyloidosis. Science 330,
clinical trials and drug development. Lancet Neurol. 9, 17. Murray, M. et al. Amyloid β protein: Aβ40 inhibits Aβ42 980–982 (2010).
702–716 (2010). oligomerization. J. Am. Chem. Soc. 131, 6316–6317 This study suggests that peripherally derived Aβ
2. Berk, C., Paul, G. & Sabbagh, M. Investigational drugs (2009). might enter the brain and participate in AD
in Alzheimer’s disease: current progress. Expert Opin. 18. Tarasoff-Conway, J. M. et al. Clearance systems in the pathogenesis.
Investig. Drugs 23, 837–846 (2014). brain — implications for Alzheimer disease. Nat. Rev. 35. Eisele, Y. S. et al. Multiple factors contribute to the
3. Selkoe, D. J. & Hardy, J. The amyloid hypothesis of Neurol. 11, 457–470 (2015). peripheral induction of cerebral β‑amyloidosis.
Alzheimer’s disease at 25 years. EMBO Mol. Med. 8, This review summarizes the clearance systems of J. Neurosci. 34, 10264–10273 (2014).
595–608 (2016). Aβ and tau in the brain. 36. Ritchie, D. L. et al. Amyloid‑β accumulation in the CNS
This article reviews new evidence supporting the 19. Yao, X. Q. et al. p75NTR ectodomain is a physiological in human growth hormone recipients in the UK.
concept that an imbalance between production neuroprotective molecule against amyloid‑β toxicity in Acta Neuropathol. 134, 221–240 (2017).
and clearance of Aβ is a very early, often initiating the brain of Alzheimer’s disease. Mol. Psychiatry 20, 37. Jaunmuktane, Z. et al. Evidence for human
factor, in AD — a widely debated issue. 1301–1310 (2015). transmission of amyloid‑β pathology and cerebral
4. Yankner, B. A. & Mesulam, M. M. Seminars in 20. Liao, M. C. et al. N‑Terminal domain of myelin basic amyloid angiopathy. Nature 525, 247–250 (2015).
medicine of the Beth Israel Hospital, Boston. protein inhibits amyloid β‑protein fibril assembly. 38. Deane, R. et al. RAGE mediates amyloid‑β peptide
β‑Amyloid and the pathogenesis of Alzheimer’s J. Biol. Chem. 285, 35590–35598 (2010). transport across the blood–brain barrier and
disease. N. Engl. J. Med. 325, 1849–1857 (1991). 21. Qosa, H. et al. Differences in amyloid‑β clearance accumulation in brain. Nat. Med. 9, 907–913 (2003).
5. Roher, A. E. et al. Amyloid β peptides in human across mouse and human blood–brain barrier models: 39. Donahue, J. E. et al. RAGE, LRP‑1, and amyloid‑β
plasma and tissues and their significance for kinetic analysis and mechanistic modeling. protein in Alzheimer’s disease. Acta Neuropathol. 112,
Alzheimer’s disease. Alzheimers Dement. 5, 18–29 Neuropharmacology 79, 668–678 (2014). 405–415 (2006).
(2009). 22. Yuede, C. M. et al. Rapid in vivo measurement of 40. Yan, S. D. et al. RAGE and amyloid‑β peptide
This study evaluates Aβ levels in brain, peripheral β‑amyloid reveals biphasic clearance kinetics in an neurotoxicity in Alzheimer’s disease. Nature 382,
organs and tissues, suggesting that brain as well as Alzheimer’s mouse model. J. Exp. Med. 213, 685–691 (1996).
plasma Aβ levels are the consequence of intricate 677–685 (2016). 41. Zenaro, E. et al. Neutrophils promote Alzheimer’s
relationships between central and peripehral 23. Xiang, Y. et al. Physiological amyloid‑β clearance in the disease-like pathology and cognitive decline via LFA‑1
sources. periphery and its therapeutic potential for Alzheimer’s integrin. Nat. Med. 21, 880–886 (2015).
6. Li, Q. X., Fuller, S. J., Beyreuther, K. & Masters, C. L. disease. Acta Neuropathol. 130, 487–499 (2015). 42. Frenkel, D. et al. Scara1 deficiency impairs clearance
The amyloid precursor protein of Alzheimer disease in This article demonstrates that peripheral clearance of soluble amyloid‑β by mononuclear phagocytes and
human brain and blood. J. Leukoc. Biol. 66, 567–574 systems are potent in clearing brain Aβ and accelerates Alzheimer’s‑like disease progression.
(1999). preventing AD. Nat. Commun. 4, 2030 (2013).
7. Toledo, J. et al. Factors affecting Aβ plasma levels and 24. Bradshaw, E. M. et al. CD33 Alzheimer’s disease 43. Krabbe, G. et al. Functional impairment of microglia
their utility as biomarkers in ADNI. Acta Neuropathol. locus: altered monocyte function and amyloid biology. coincides with β‑amyloid deposition in mice with
122, 401–413 (2011). Nat. Neurosci. 16, 848–850 (2013). Alzheimer-like pathology. PLoS ONE 8, e60921
8. Mehta, P. D., Pirttila, T., Patrick, B. A., Barshatzky, M. 25. Kanekiyo, T. & Bu, G. The low-density lipoprotein (2013).
& Mehta, S. P. Amyloid β protein 1–40 and 1–42 receptor-related protein 1 and amyloid‑β clearance 44. Zaghi, J. et al. Alzheimer disease macrophages shuttle
levels in matched cerebrospinal fluid and plasma from in Alzheimer’s disease. Front. Aging Neurosci. 6, 93 amyloid‑β from neurons to vessels, contributing to
patients with Alzheimer disease. Neurosci. Lett. 304, (2014). amyloid angiopathy. Acta Neuropathol. 117, 111–124
102–106 (2001). 26. Ghiso, J. et al. Systemic catabolism of Alzheimer’s (2009).
9. Delvaux, E., Bentley, K., Stubbs, V., Sabbagh, M. & Aβ40 and Aβ42. J. Biol. Chem. 279, 45897–45908 45. Gu, B. J. et al. Innate phagocytosis by peripheral
Coleman, P. Differential processing of amyloid (2004). blood monocytes is altered in Alzheimer’s disease.
precursor protein in brain and in peripheral blood This article demonstrates that the liver is the Acta Neuropathol. 132, 377–389 (2016).
leukocytes. Neurobiol. Aging 34, 1680–1686 major organ responsible for uptake and This human study demonstrates that innate
(2013). degradation of circulating Aβ42 and Aβ40, followed immunity is compromised in patients with AD.
10. Evin, G., Zhu, A., Holsinger, R. M., Masters, C. & by the kidney. 46. Darlington, D. et al. Human umbilical cord blood-
Li, Q.‑X. Proteolytic processing of the Alzheimer’s 27. Ghiso, J. et al. Alzheimer’s soluble amyloid β is a derived monocytes improve cognitive deficits and
disease amyloid precursor protein in brain and normal component of human urine. FEBS Lett. 408, reduce amyloid‑β pathology in PSAPP mice. Cell
platelets. J. Neurosci. Res. 74, 386–392 (2003). 105–108 (1997). Transplant. 24, 2237–22350 (2015).
11. Biere, A. L. et al. Amyloid β‑peptide is transported 28. Liu, Z. et al. Characterization of insulin degrading 47. Baruch, K. et al. PD‑1 immune checkpoint blockade
on lipoproteins and albumin in human plasma. enzyme and other amyloid‑β degrading proteases reduces pathology and improves memory in mouse
J. Biol. Chem. 271, 32916–32922 (1996). in human serum: a role in Alzheimer’s disease? models of Alzheimer’s disease. Nat. Med. 22,
12. Kuo, Y. M. et al. Amyloid‑β peptides interact with J. Alzheimers Dis. 29, 329–340 (2012). 135–137 (2016).
plasma proteins and erythrocytes: implications for 29. Mackic, J. B. et al. Human blood–brain barrier 48. Prokop, S. et al. Impact of peripheral myeloid cells on
their quantitation in plasma. Biochem. Biophys. Res. receptors for Alzheimer’s amyloid-β1–40. Asymmetrical amyloid‑β pathology in Alzheimer’s disease-like mice.
Commun. 268, 750–756 (2000). binding, endocytosis, and transcytosis at the apical J. Exp. Med. 212, 1811–1818 (2015).
13. Joachim, C. L., Mori, H. & Selkoe, D. J. Amyloid side of brain microvascular endothelial cell monolayer. 49. Hollingworth, P. et al. Common variants at ABCA7.
β‑protein deposition in tissues other than brain in J. Clin. Invest. 102, 734–743 (1998). MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are
Alzheimer’s disease. Nature 341, 226–230 (1989). 30. Silverberg, G. D., Mayo, M., Saul, T., Rubenstein, E. & associated with Alzheimer’s disease. Nat. Genet. 43,
14. Koronyo, Y., Salumbides, B., Black, K. & Koronyo McGuire, D. Alzheimer’s disease, normal-pressure 429–435 (2011).
Hamaoui, M. Alzheimer’s disease in the retina: hydrocephalus, and senescent changes in CSF 50. Jonsson, T. et al. Variant of TREM2 associated with
imaging retinal Aβ plaques for early diagnosis and circulatory physiology: a hypothesis. Lancet Neurol. 2, the risk of Alzheimer’s disease. N. Engl. J. Med. 368,
therapy assessment. Neurodegener. Dis. 10, 506–511 (2003). 107–116 (2013).
285–293 (2012). 31. Aspelund, A. et al. A dural lymphatic vascular system 51. Jay, T. R. et al. TREM2 deficiency eliminates TREM2+
15. Troncone, L. et al. Aβ amyloid pathology affects the that drains brain interstitial fluid and macromolecules. inflammatory macrophages and ameliorates pathology
hearts of patients with Alzheimer’s disease: mind the J. Exp. Med. 212, 991–999 (2015). in Alzheimer’s disease mouse models. J. Exp. Med.
heart. J. Am. Coll. Cardiol. 68, 2395–2407 (2016). 32. Louveau, A. et al. Structural and functional features 212, 287–295 (2015).
This article was the first to describe the presence of central nervous system lymphatic vessels. Nature 52. Bartos, A., Fialova, L., Svarcova, J. & Ripova, D.
of compromised myocardial function and 523, 337–341 (2015). Patients with Alzheimer disease have elevated
intramyocardial deposits of Aβ in patients with AD. 33. Iliff, J. J., Goldman, S. A. & Nedergaard, M. intrathecal synthesis of antibodies against tau protein
16. Stine, W. B., Dahlgren, K., Krafft, G. & LaDu, M. Implications of the discovery of brain lymphatic and heavy neurofilament. J. Neuroimmunol. 252,
In vitro characterization of conditions for amyloid‑β pathways. Lancet Neurol. 14, 977–979 (2015). 100–105 (2012).

NATURE REVIEWS | NEUROLOGY VOLUME 13 | OCTOBER 2017 | 621


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

53. Liu, Y. H. et al. Immunity and Alzheimer’s disease: 77. Salameh, T. S., Shah, G. N., Price, T. O., Hayden, M. R. 100. Zetterberg, H. et al. Hypoxia due to cardiac arrest
immunological perspectives on the development of & Banks, W. A. Blood–brain barrier disruption and induces a time-dependent increase in serum amyloid β
novel therapies. Drug Discov. Today 18, 1212–1220 neurovascular unit dysfunction in diabetic mice: levels in humans. PLoS ONE 6, e28263 (2011).
(2013). protection with the mitochondrial carbonic anhydrase 101. Wang, L. et al. Chronic cerebral hypoperfusion induces
54. Wang, T. et al. Naturally occurring autoantibodies inhibitor topiramate. J. Pharmacol. Exp. Ther. 359, memory deficits and facilitates Aβ generation in
against Aβ oligomers exhibited more beneficial effects 452–459 (2016). C57BL/6J mice. Exp. Neurol. 283, 353–364 (2016).
in the treatment of mouse model of Alzheimer’s 78. Leuner, K. et al. Mitochondrion-derived reactive 102. Cermakova, P. et al. Heart failure and Alzheimer’s
disease than intravenous immunoglobulin. oxygen species lead to enhanced amyloid β formation. disease. J. Intern. Med. 277, 406–425 (2015).
Neuropharmacology 105, 561–576 (2016). Antioxid. Redox Signal. 16, 1421–1433 (2012). 103. Mattsson, N. et al. Association of brain amyloid‑β
55. DeMarshall, C. A. et al. Detection of Alzheimer’s 79. Moreno-Gonzalez, I. et al. Molecular interaction with cerebral perfusion and structure in Alzheimer’s
disease at mild cognitive impairment and disease between type 2 diabetes and Alzheimer’s disease disease and mild cognitive impairment. Brain 137,
progression using autoantibodies as blood-based through cross-seeding of protein misfolding. 1550–1561 (2014).
biomarkers. Alzheimers Dement. (Amst.) 3, 51–62 Mol. Psychiatry 22, 1327–1334 (2017). 104. Leeuwis, A. E. et al. Lower cerebral blood flow is
(2016). This study proposes a new molecular interaction associated with impairment in multiple cognitive
56. Monning, U. E. A. in Alzheimer’s Disease: Basic between AD and diabetes mellitus: misfolded domains in Alzheimer’s disease. Alzheimers Dement.
Mechanisms, Diagnosis and Therapeutic Strategies amylin (generated in the pancreas in type 2 13, 531–540 (2017).
(eds Iqbal, K. et al.) 557–563 (Wiley–Blackwell, diabetes mellitus) and Aβ accelerate or exacerbate 105. Marnane, M. & Hsiung, G. Y. Could better
1991). the misfolding and aggregation of each other by phenotyping small vessel disease provide new insights
57. Sevigny, J. et al. The antibody aducanumab reduces cross-seeding. into Alzheimer disease and improve clinical trial
Aβ plaques in Alzheimer’s disease. Nature 537, 80. Biessels, G. J. & Reijmer, Y. D. Brain changes outcomes? Curr. Alzheimer Res. 13, 750–763
50–56 (2016). underlying cognitive dysfunction in diabetes: what can (2016).
58. Marsh, S. E. et al. The adaptive immune system we learn from MRI? Diabetes 63, 2244–2252 (2014). 106. Kester, M. I. et al. Associations between cerebral
restrains Alzheimer’s disease pathogenesis by 81. Lesser, G. T. Association of Alzheimer disease small-vessel disease and Alzheimer disease pathology
modulating microglial function. Proc. Natl Acad. Sci. pathology with abnormal lipid metabolism: the as measured by cerebrospinal fluid biomarkers.
USA 113, E1316–E1325 (2016). Hisayama study. Neurology 78, 1280 (2012). JAMA Neurol. 71, 855–862 (2014).
59. Baruch, K. et al. Breaking immune tolerance by 82. Sato, N. & Morishita, R. The roles of lipid and glucose 107. Mackic, J. B. et al. Cerebrovascular accumulation and
targeting Foxp3+ regulatory T cells mitigates metabolism in modulation of β‑amyloid, tau, and increased blood-brain barrier permeability to
Alzheimer’s disease pathology. Nat. Commun. 6, 7967 neurodegeneration in the pathogenesis of Alzheimer circulating Alzheimer’s amyloid-β peptide in aged
(2015). disease. Front. Aging Neurosci. 7, 199 (2015). squirrel monkey with cerebral amyloid angiopathy.
60. Rosenberg, R. N. et al. Altered amyloid protein 83. Yu, J. T., Tan, L. & Hardy, J. Apolipoprotein E in J. Neurochem. 70, 210–215 (1998).
processing in platelets of patients with Alzheimer Alzheimer’s disease: an update. Annu. Rev. Neurosci. 108. Wang, Y. R. et al. Associations between hepatic
disease. Arch. Neurol. 54, 139–144 (1997). 37, 79–100 (2014). functions and plasma amyloid-β levels.— implications
61. Di Luca, M. et al. Abnormal pattern of platelet APP 84. Lee, C. Y., Tse, W., Smith, J. D. & Landreth, G. E. for the capacity of liver in peripheral amyloid-β
isoforms in Alzheimer disease and Down syndrome. Apolipoprotein E promotes β‑amyloid trafficking and clearance. Mol. Neurobiol. 54, 2338–2344 (2017).
Arch. Neurol. 53, 1162–1166 (1996). degradation by modulating microglial cholesterol 109. Sehgal, N. et al. Withania somnifera reverses
62. Srisawat, C. et al. The platelet amyloid precursor levels. J. Biol. Chem. 287, 2032–2044 (2012). Alzheimer’s disease pathology by enhancing low-
protein ratio as a diagnostic marker for Alzheimer’s 85. Zissimopoulos, J. M., Barthold, D., Brinton, R. D. & density lipoprotein receptor-related protein in liver.
disease in Thai patients. J. Clin. Neurosci. 20, Joyce, G. Sex and race differences in the association Proc. Natl Acad. Sci. USA 109, 3510–3515 (2012).
644–648 (2013). between statin use and the incidence of Alzheimer 110. Ghersi-Egea, J. F. et al. Fate of cerebrospinal fluid-
63. Doecke, J. D. et al. Blood-based protein biomarkers disease. JAMA Neurol. 74, 225–232 (2016). borne amyloid β‑peptide: rapid clearance into blood
for diagnosis of Alzheimer disease. Arch. Neurol. 69, 86. Reed, B. et al. Associations between serum cholesterol and appreciable accumulation by cerebral arteries.
1318–1325 (2012). levels and cerebral amyloidosis. JAMA Neurol. 71, J. Neurochem. 67, 880–883 (1996).
64. Rogers, J. et al. Peripheral clearance of amyloid β 195–200 (2014). 111. Arvanitakis, Z., Lucas, J. A., Younkin, L. H.,
peptide by complement C3‑dependent adherence to 87. Zubenko, G. S. et al. Platelet membrane fluidity in Younkin, S. G. & Graff-Radford, N. R. Serum creatinine
erythrocytes. Neurobiol. Aging 27, 1733–1739 Alzheimer’s disease and major depression. levels correlate with plasma amyloid β protein.
(2006). Am. J. Psychiatry 144, 860–868 (1987). Alzheimer Dis. Assoc. Disord. 16, 187–190 (2002).
65. Chen, S. H. et al. Altered peripheral profile of blood 88. Collins, J. M., Scott, R. B., McClish, D. K., Taylor, J. R. 112. Liu, Y. H. et al. Association between serum amyloid‑β
cells in Alzheimer disease: a hospital-based case– & Grogan, W. M. Altered membrane anisotropy and renal functions: implications for roles of kidney in
control study. Medicine (Baltimore) 96, e6843 (2017). gradients of plasma membranes of living peripheral amyloid‑β clearance. Mol. Neurobiol. 52, 115–119
66. Loffredo, F. S. et al. Growth differentiation factor 11 is blood leukocytes in aging and Alzheimer’s disease. (2015).
a circulating factor that reverses age-related cardiac Mech. Ageing Dev. 59, 153–162 (1991). 113. Gronewold, J. et al. Factors responsible for plasma
hypertrophy. Cell 153, 828–839 (2013). 89. Zubenko, G. S. & Howland, R. Markedly increased β‑amyloid accumulation in chronic kidney disease.
67. Castellano, J. M. et al. Human umbilical cord plasma platelet membrane fluidity in Down syndrome with Mol. Neurobiol. 53, 3136–3145 (2016).
proteins revitalize hippocampal function in aged mice. a (14q, 21q) translocation. J. Geriatr. Psychiatry 114. Deckers, K. et al. Dementia risk in renal dysfunction:
Nature 544, 488–492 (2017). Neurol. 1, 218–219 (1988). a systematic review and meta-analysis of prospective
68. Villeda, S. A. et al. Young blood reverses age-related 90. Scott, R. B., Collins, J. M. & Hunt, P. A. Alzheimer’s studies. Neurology 88, 198–208 (2017).
impairments in cognitive function and synaptic disease and Down syndrome: leukocyte membrane 115. Sakai, K. et al. Patients that have undergone
plasticity in mice. Nat. Med. 20, 659–663 (2014). fluidity alterations. Mech. Ageing Dev. 75, 1–10 hemodialysis exhibit lower amyloid deposition in the
This article suggests that anti-ageing molecules (1994). brain: evidence supporting a therapeutic strategy for
exist in young blood. Identification of these 91. Yassine, H. N. et al. Association of serum Alzheimer’s disease by removal of blood amyloid.
protective components could be important in docosahexaenoic acid with cerebral amyloidosis. J. Alzheimers Dis. 51, 997–1002 (2016).
understanding the pathogenesis of AD and in JAMA Neurol. 73, 1208–1216 (2016). 116. Emamian, F. et al. The association between
developing systemic rejuvenation therapeutics. 92. Nishihira, J. et al. Associations between serum obstructive sleep apnea and Alzheimer’s disease:
69. Villeda, S. A. et al. The ageing systemic milieu omega‑3 fatty acid levels and cognitive functions a meta-analysis perspective. Front. Aging Neurosci. 8,
negatively regulates neurogenesis and cognitive among community-dwelling octogenarians in Okinawa, 78 (2016).
function. Nature 477, 90–94 (2011). Japan: the KOCOA study. J. Alzheimers Dis. 51, 117. Brunnstrom, H. R. & Englund, E. M. Cause of death in
70. Katsimpardi, L. et al. Vascular and neurogenic 857–866 (2016). patients with dementia disorders. Eur. J. Neurol. 16,
rejuvenation of the aging mouse brain by young 93. Rusanen, M. et al. Heart diseases and long-term risk 488–492 (2009).
systemic factors. Science 344, 630–634 (2014). of dementia and Alzheimer’s disease: a population- 118. Pan, W. & Kastin, A. J. Can sleep apnea cause
71. Middeldorp, J. et al. Preclinical assessment of young based CAIDE study. J. Alzheimers Dis. 42, 183–191 Alzheimer’s disease? Neurosci. Biobehav. Rev. 47,
blood plasma for Alzheimer disease. JAMA Neurol. (2014). 656–669 (2014).
73, 1325–1333 (2016). 94. Qiu, C. et al. Heart failure and risk of dementia and 119. Yaffe, K. et al. Sleep-disordered breathing, hypoxia,
72. Xu, W. et al. Meta-analysis of modifiable risk factors Alzheimer disease: a population-based cohort study. and risk of mild cognitive impairment and dementia
for Alzheimer’s disease. J. Neurol. Neurosurg. Arch. Intern. Med. 166, 1003–1008 (2006). in older women. JAMA 306, 613–619 (2011).
Psychiatry 86, 1299–1306 (2015). 95. Jefferson, A. L. et al. Low cardiac index is associated 120. Osorio, R. S. et al. Sleep-disordered breathing
73. Velayudhan, L. et al. Risk of developing dementia in with incident dementia and Alzheimer disease: the advances cognitive decline in the elderly. Neurology
people with diabetes and mild cognitive impairment. Framingham Heart Study. Circulation 131, 84, 1964–1971 (2015).
Br. J. Psychiatry 196, 36–40 (2010). 1333–1339 (2015). 121. Bu, X. L. et al. Serum amyloid‑β levels are increased
74. Tamaki, C., Ohtsuki, S. & Terasaki, T. Insulin facilitates 96. Luchsinger, J. A. et al. Aggregation of vascular risk in patients with obstructive sleep apnea syndrome.
the hepatic clearance of plasma amyloid β‑peptide factors and risk of incident Alzheimer disease. Sci. Rep. 5, 13917 (2015).
(1–40) by intracellular translocation of low-density Neurology 65, 545–551 (2005). 122. Bu, X. L. et al. Serum amyloid‑β levels are increased in
lipoprotein receptor-related protein 1 (LRP‑1) to the 97. Li, J. et al. Vascular risk factors promote conversion patients with chronic obstructive pulmonary disease.
plasma membrane in hepatocytes. Mol. Pharmacol. from mild cognitive impairment to Alzheimer disease. Neurotox. Res. 28, 346–351 (2015).
72, 850–855 (2007). Neurology 76, 1485–1491 (2011). 123. Osorio, R. S. et al. Interaction between sleep-
75. Kang, S., Lee, Y. H. & Lee, J. E. Metabolism-centric 98. Jin, W. S. et al. Reduced cardiovascular functions in disordered breathing and apolipoprotein E genotype
overview of the pathogenesis of Alzheimer’s disease. patients with Alzheimer’s disease. J. Alzheimers Dis. on cerebrospinal fluid biomarkers for Alzheimer’s
Yonsei Med. J. 58, 479–488 (2017). 58, 919–925 (2017). disease in cognitively normal elderly individuals.
76. Gasparini, L. et al. Stimulation of β‑amyloid precursor 99. Okamoto, Y. et al. Cerebral hypoperfusion accelerates Neurobiol. Aging 35, 1318–1324 (2014).
protein trafficking by insulin reduces intraneuronal cerebral amyloid angiopathy and promotes cortical 124. Rosenzweig, I. et al. Sleep apnoea and the brain:
β‑amyloid and requires mitogen-activated protein microinfarcts. Acta Neuropathol. 123, 381–394 a complex relationship. Lancet Respir. Med. 3,
kinase signaling. J. Neurosci. 21, 2561–2570 (2001). (2012). 404–414 (2015).

622 | OCTOBER 2017 | VOLUME 13 www.nature.com/nrneurol


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

125. Musiek, E. S. & Holtzman, D. M. Mechanisms linking 147. Satizabal, C. L. et al. Incidence of dementia over three 166. Kayano, M. et al. Plasma microRNA biomarker
circadian clocks, sleep, and neurodegeneration. decades in the Framingham heart study. N. Engl. detection for mild cognitive impairment using
Science 354, 1004–1008 (2016). J. Med. 374, 523–532 (2016). differential correlation analysis. Biomark. Res. 4, 22
126. Cedernaes, J. et al. Candidate mechanisms underlying This study finds that the incidence of dementia has (2016).
the association between sleep–wake disruptions and decreased over the past three decades, suggesting 167. Lu, R. et al. Reduced TRPC6 mRNA levels in the
Alzheimer’s disease. Sleep Med. Rev. 31, 102–111 that control of systemic comorbidities and risk blood cells of patients with Alzheimer’s disease
(2017). factors, as well as maintenance of body and mild cognitive impairment. Mol. Psychiatry
127. Gareau, M. Microbiota–gut–brain axis and cognitive homeostasis, could bring improved results for AD http://dx.doi.org/10.1038/mp.2017.136 (2017).
function. Adv. Exp. Med. Biol. 817, 357–371 (2014). prevention. 168. Roberts, B. R. et al. Biochemically-defined pools
128. Zhan, X. et al. Gram-negative bacterial molecules 148. Langa, K. M. et al. A comparison of the prevalence of of amyloid‑β in sporadic Alzheimer’s disease:
associate with Alzheimer disease pathology. dementia in the United States in 2000 and 2012. correlation with amyloid PET. Brain 140, 1486–1498
Neurology 87, 2324–2332 (2016). JAMA Intern. Med. 177, 51–58 (2017). (2017).
129. Akbari, E. et al. Effect of probiotic supplementation on 149. Haag, M. D., Hofman, A., Koudstaal, P. J., 169. Bush, A. I. et al. The amyloid precursor protein of
cognitive function and metabolic status in Alzheimer’s Stricker, B. H. & Breteler, M. M. Statins are associated Alzheimer’s disease is released by human platelets.
disease: a randomized, double-blind and controlled with a reduced risk of Alzheimer disease regardless of J. Biol. Chem. 265, 15977–15983 (1990).
trial. Front. Aging Neurosci. 8, 256 (2016). lipophilicity. The Rotterdam Study. J. Neurol. 170. Li, Q. X. et al. Secretion of Alzheimer’s disease Aβ
130. Bu, X. L. et al. A study on the association between Neurosurg. Psychiatry 80, 13–17 (2009). amyloid peptide by activated human platelets.
infectious burden and Alzheimer’s disease. 150. Papadopoulos, P., Tong, X. K. & Hamel, E. Selective Lab. Invest. 78, 461–469 (1998).
Eur. J. Neurol. 22, 1519–1525 (2015). benefits of simvastatin in bitransgenic APPSwe, Ind/ 171. Citron, M. et al. Excessive production of amyloid
This study offers the first evidence that an TGF‑β1 mice. Neurobiol. Aging 35, 203–212 (2014). β‑protein by peripheral cells of symptomatic and
increased infectious burden is associated with AD, 151. Richardson, K. et al. Statins and cognitive function: presymptomatic patients carrying the Swedish familial
supporting the role of systemic infection and/or a systematic review. Ann. Intern. Med. 159, 688–697 Alzheimer disease mutation. Proc. Natl Acad. Sci. USA
inflammation in the aetiopathogenesis of AD. (2013). 91, 11993–11997 (1994).
131. Harris, S. A. & Harris, E. A. Herpes simplex virus 152. Ancoli-Israel, S. et al. Cognitive effects of treating 172. Li, S., Liu, B., Zhang, L. & Rong, L. Amyloid β peptide
type 1 and other pathogens are key causative factors obstructive sleep apnea in Alzheimer’s disease: is elevated in osteoporotic bone tissues and enhances
in sporadic Alzheimer’s disease. J. Alzheimers Dis. 48, a randomized controlled study. J. Am. Geriatr. Soc. osteoclast function. Bone 61, 164–175 (2014).
319–353 (2015). 56, 2076–2081 (2008). 173. Kuo, Y. M. et al. Elevated Aβ42 in skeletal muscle of
132. Abbayya, K., Puthanakar, N. Y., Naduwinmani, S. & 153. Cooke, J. R. et al. Sustained use of CPAP slows Alzheimer disease patients suggests peripheral
Chidambar, Y. S. Association between periodontitis deterioration of cognition, sleep, and mood in patients alterations of AβPP metabolism. Am. J. Pathol. 156,
and Alzheimer’s disease. N. Am. J. Med. Sci. 7, with Alzheimer’s disease and obstructive sleep apnea: 797–805 (2000).
241–246 (2015). a preliminary study. J. Clin. Sleep Med. 5, 305–309 174. Zhang, X. et al. Hypoxia-inducible factor 1α (HIF‑1α)-
133. Wallin, K. et al. Midlife rheumatoid arthritis increases (2009). mediated hypoxia increases BACE1 expression and
the risk of cognitive impairment two decades later: 154. Boada, M. et al. Amyloid-targeted therapeutics in β‑amyloid generation. J. Biol. Chem. 282,
a population-based study. J. Alzheimers Dis. 31, Alzheimer’s disease: use of human albumin in plasma 10873–10880 (2007).
669–676 (2012). exchange as a novel approach for Aβ mobilization. 175. Sun, X. et al. Hypoxia facilitates Alzheimer’s disease
134. Rivest, S. Regulation of innate immune responses in Drug News Perspect. 22, 325–339 (2009). pathogenesis by up‑regulating BACE1 gene
the brain. Nat. Rev. Immunol. 9, 429–439 (2009). 155. Jin, W. S. et al. Peritoneal dialysis reduces amyloid‑β expression. Proc. Natl Acad. Sci. USA 103,
135. Gao, H. M. & Hong, J. S. Why neurodegenerative plasma levels in humans and attenuates Alzheimer- 18727–18732 (2006).
diseases are progressive: uncontrolled inflammation associated phenotypes in an APP/PS1 mouse model.
drives disease progression. Trends Immunol. 29, Acta Neuropathol. 134, 207–220 (2017). Acknowledgements
357–365 (2008). 156. Liu, Y. et al. Expression of neprilysin in skeletal muscle The authors’ research work is supported by the National
136. Wang, J. et al. Anti-inflammatory drugs and risk of reduces amyloid burden in a transgenic mouse model Natural Science Foundation of China (grants 81471296 and
Alzheimer’s disease: an updated systematic review of Alzheimer disease. Mol. Ther. 17, 1381–1386 81625007 to Y.-J.W., and grant 81600949 to J.W.), and the
and meta-analysis. J. Alzheimers Dis. 44, 385–396 (2009). Chinese Ministry of Science and Technology (grant
(2015). 157. Liu, Y. H. et al. Clearance of amyloid‑β in Alzheimer’s 2016YFC1306401 to Y.-J.W.). The authors thank Dr J. Piña-
137. Lövheim, H. et al. Plasma concentrations of free disease: shifting the action site from center to Crespo and Dr H. Xu at Sanford Burnham Prebys Medical
amyloid‑β cannot predict the development of periphery. Mol. Neurobiol. 51, 1–7 (2015). Discovery Institute, USA, for critical reading of the paper.
Alzheimer’s disease. Alzheimers Dement. 13, 778–782 158. Winston, C. N. et al. Prediction of conversion from
(2017). mild cognitive impairment to dementia with neuronally Author contributions
138. Mattsson, N. et al. Plasma tau in Alzheimer disease. derived blood exosome protein profile. Alzheimers All authors contributed to researching data for the article,
Neurology 87, 1827–1835 (2016). Dement. (Amst.) 3, 63–72 (2016). discussion of the content, writing the article, and to review
139. Wood, H. Alzheimer disease: biomarkers of AD risk 159. Goetzl, E. J. et al. Cargo proteins of plasma astrocyte- and/or editing of the manuscript before submission.
— the end of the road for plasma amyloid‑β? derived exosomes in Alzheimer’s disease. FASEB J. 30,
Nat. Rev. Neurol. 12, 613 (2016). 3853–3859 (2016). Competing interests statement
140. Herskovits, A. Z., Locascio, J. J., Peskind, E. R., Li, G. 160. Veitinger, M. et al. A platelet protein biochip rapidly The authors declare no competing interests.
& Hyman, B. T. A. Luminex assay detects amyloid β detects an Alzheimer’s disease-specific phenotype.
oligomers in Alzheimer’s disease cerebrospinal fluid. Acta Neuropathol. 128, 665–677 (2014). Publisher’s note
PLoS ONE 8, e67898 (2013). This article demonstrates platelet changes in AD, Springer Nature remains neutral with regard to jurisdictional
141. Sengupta, U. et al. Tau oligomers in cerebrospinal fluid providing potential biomarkers for early diagnosis claims in published maps and institutional affiliations.
in Alzheimer’s disease. Ann. Clin. Transl Neurol. 4, of AD.
226–235 (2017). 161. Burnham, S. C. et al. A blood-based predictor for Review criteria
142. Shen, Y. et al. Increased plasma β‑secretase 1 may neocortical Aβ burden in Alzheimer’s disease: results Articles for inclusion in this Review were identified by
predict conversion to Alzheimer’s disease dementia from the AIBL study. Mol. Psychiatry 19, 519–526 searches of the PubMed database using the following terms:
in individuals with mild cognitive impairment. (2014). “Alzheimer disease”, “amyloid beta”, “peripheral clearance”,
Biol. Psychiatry http://dx.doi.org/10.1016/ 162. Mattsson, N., Andreasson, U., Zetterberg, H., “innate immunity”, “adaptive immunity”, “macrophage”,
j.biopsych.2017.02.007 (2017). Blennow, K. & Alzheimer’s Disease Neuroimaging “monocyte”, “phagocytosis”, “blood”, “circulation”, “plasma”,
143. Brooks, M. One target, one treatment? Not for Initiative. Association of plasma neurofilament light “platelet”, “erythrocyte”, “exosome”, “diabetes”, “insulin”,
Alzheimer’s disease. Medscape http://www.medscape. with neurodegeneration in patients with Alzheimer “amylin”, “apolipoprotein E”, “cholesterol”, “lipid”, “lipo­
com/viewarticle/848322 (2015). disease. JAMA Neurol. 74, 557–566 (2017). protein”, “cerebrovascular diseases”, “heart failure”, “cerebral
144. Wang, Y. J. Alzheimer disease: lessons from 163. Chaves, M. L. et al. Serum levels of S100B and NSE blood flow”, “hypoperfusion”, “hypoxia”, “liver”, “hepatic func‑
immunotherapy for Alzheimer disease. Nat. Rev. proteins in Alzheimer’s disease patients. tion”, “kidney”, “renal function”, “respiratory”, “sleep”, “micro‑
Neurol. 10, 188–189 (2014). J. Neuroinflammation 7, 6 (2010). biome”, “gastrointestinal microbiota”, “gut flora”, “infection”,
145. Larson, E. B., Yaffe, K. & Langa, K. M. New insights 164. Teunissen, C. E. et al. Brain-specific fatty acid-binding “inflammation”, “immune responses”, “GWAS”, “risk factor”,
into the dementia epidemic. N. Engl. J. Med. 369, protein is elevated in serum of patients with dementia- “biomarker”, “diagnosis”, “therapy”, “treatment”, “prevent”.
2275–2277 (2013). related diseases. Eur. J. Neurol. 18, 865–871 (2011). Only articles published in English were retrieved. Full-text
146. Wu, Y. T. et al. Dementia in western Europe: 165. Zhang, R. et al. Systemic immune system alterations in papers were available for most of the articles that were
epidemiological evidence and implications for policy early stages of Alzheimer’s disease. J. Neuroimmunol. ­chosen for review, and the reference lists of these articles
making. Lancet Neurol. 15, 116–124 (2016). 256, 38–42 (2013). were searched for further relevant material.

NATURE REVIEWS | NEUROLOGY VOLUME 13 | OCTOBER 2017 | 623


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like