You are on page 1of 23

Neuron

Review

The Pathobiology of Vascular Dementia


Costantino Iadecola1,*
1Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10021, USA

*Correspondence: coi2001@med.cornell.edu
http://dx.doi.org/10.1016/j.neuron.2013.10.008

Vascular cognitive impairment defines alterations in cognition, ranging from subtle deficits to full-blown de-
mentia, attributable to cerebrovascular causes. Often coexisting with Alzheimers disease, mixed vascular
and neurodegenerative dementia has emerged as the leading cause of age-related cognitive impairment.
Central to the disease mechanism is the crucial role that cerebral blood vessels play in brain health, not
only for the delivery of oxygen and nutrients, but also for the trophic signaling that inextricably links the
well-being of neurons and glia to that of cerebrovascular cells. This review will examine how vascular damage
disrupts these vital homeostatic interactions, focusing on the hemispheric white matter, a region at height-
ened risk for vascular damage, and on the interplay between vascular factors and Alzheimers disease.
Finally, preventative and therapeutic prospects will be examined, highlighting the importance of midlife
vascular risk factor control in the prevention of late-life dementia.

Introduction cognitive health. To this end, the major cerebrovascular causes


Age-related dementia, an irreversible condition resulting in pro- of cognitive dysfunction will be briefly reviewed, focusing on
gressive cognitive decline, has emerged as one of the leading neuropathology, emerging mechanisms, and overlap with neu-
health problems of our time. Advances in prevention and rodegeneration.
healthcare have increased life expectancy and produced a shift
in the burden of disease worldwide. Thus, noncommunicable Dementia through the Ages
diseases, including dementia, have been recognized for the first In Alois Alzheimers time (1900s), dementia was thought to be
time as the major threat to the world population (World Health caused predominantly by hardening of the arteries (arterio-
Organization, 2012). The World Health Organization estimates sclerotic dementia) (Bowler, 2007; Jellinger, 2006). Vascular
that 35.6 million people live with dementia, a number that is factors were considered a major player in dementia well into
anticipated to triple by 2050 (World Health Organization, 2012). the 20th century, until, in the 1980s, the Ab peptide was identified
Every year 7.7 million new cases of dementia are diagnosed, as the main component of parenchymal (amyloid plaque) and
imposing a tremendous burden on families and the primary care- vascular (amyloid angiopathy) amyloid deposits, pathological
givers, as well as a financial cost to society. Although recent data hallmarks of AD (Glenner and Wong, 1984; Kang et al., 1987).
suggest a decline in prevalence (Matthews et al., 2013), demen- Shortly after, mutations in the amyloid precursor protein (APP)
tia remains a devastating and costly disease. In the US such cost gene were identified in familial forms of AD (Bertram and Tanzi,
has already surpassed that of cancer and heart diseases (Hurd 2008). Since then, the emphasis shifted from vascular dementia
et al., 2013). The realization of its paramount public health impact to AD, a process defined as the Alzheimerization of dementia
has led nations, including the US, to develop national plans (Figure 1) (Bowler, 2007). However, an increasing appreciation
to cope with dementia and attempt to reduce its devastating of the impact of cerebrovascular lesions on AD brought to the
effects (National Alzheimers Project Act; Public Law 111-375). forefront the importance of cerebrovascular health in cognitive
Vascular dementia, a heterogeneous group of brain disorders function (Esiri et al., 1999; Gold et al., 2007; Snowdon et al.,
in which cognitive impairment is attributable to cerebrovascular 1997). Furthermore, community-based clinical-pathological
pathologies, is responsible for at least 20% of cases of dementia, studies revealed that the largest proportion of dementia cases
being second only to Alzheimers disease (AD) (Gorelick et al., have mixed pathology, comprising features of AD (amyloid
2011). Recent clinical-pathological studies have highlighted the plaques and neurofibrillary tangles) as well as ischemic lesions
role of cerebrovascular disease, not only as a primary cause of (Launer et al., 2008; Schneider et al., 2009). These developments
cognitive impairment, but also as an adjuvant to the expression have promoted an interest to gain a better understanding of
of dementia caused by other factors, including AD and other how vascular brain lesions affect cognition and how vascular pa-
neurodegenerative pathologies (Gorelick et al., 2011; Schneider thology and neurodegeneration interact to amplify their respec-
et al., 2007a; Toledo et al., 2013). At the same time, new exper- tive pathogenic contribution.
imental findings have revealed a previously unrecognized func-
tional and pathogenic synergy between neurons, glia, and Defining Dementia on Vascular Bases:
vascular cells (Iadecola, 2010; Quaegebeur et al., 2011; Zlo- From Arteriosclerotic Dementia to Vascular
kovic, 2011), providing a new framework to reevaluate how Cognitive Impairment
alterations in cerebral blood vessels could contribute to the The concept of dementia caused by cerebrovascular pathology
neuronal dysfunction underlying cognitive impairment. These has evolved considerably over the years (Figure 2). For many
advances call for a reappraisal of the role of vascular factors in decades vascular dementia was attributed to sclerosis of

844 Neuron 80, November 20, 2013 2013 Elsevier Inc.


Neuron

Review

Figure 2. Evolution of the Concept of Cognitive Impairment on


Vascular Bases
Hardening of the arteries was considered the main cause in the early 1900s.
The concept of multi-infarct dementia introduced the possibility of preventing
dementia by controlling vascular risk factors. The introduction of brain imaging
modalities (computer tomography, then magnetic resonance imaging) led to
the realization that white matter disease, termed leukoaraiosis, was a major
cause of cognitive impairment. In the 1990s the term VCI was introduced
to broaden the spectrum of cognitive deficits caused by vascular factors. At
this time, genetic causes of vascular damage causing dementia were also
discovered, CADASIL being the first monogenic cause of vascular cognitive
Figure 1. Changing Views about Dementia through the Years impairment, identified by M.-G. Bousser and colleagues.
In the early 1900s vascular factors were thought to be the main cause of
dementia. Over the next several decades Alzheimers disease was felt to be
the main cause. Clinical-pathological studies have revealed that mixed
dementia, combining features of vascular dementia and AD, is currently the affecting at least one cognitive domain (Gorelick et al., 2011),
most common cause of cognitive impairment in the aged. vascular dementia being the most severe form of VCI.

The Neurovascular Unit: Blood Flow Regulation


cerebral arteries leading to diffuse ischemic injury and brain and Beyond
atrophy (Jellinger, 2006). The first significant departure from The fundamental role that cerebral blood vessels play in the
this concept, inspired by the work of Tomlinson and colleagues broad spectrum of pathologies underlying cognitive impairment
(Tomlinson et al., 1970), was proposed by Hachinski and highlights the importance of vascular structure and function in
colleagues (Hachinski et al., 1974), who suggested that demen- brain health. Owing to its high energy needs and lack of fuel
tia on vascular bases was caused by multiple and discrete reserves, the brain requires a continuous and well-regulated
ischemic lesions in patients with vascular risk factors, such as supply of blood (Iadecola, 2004). Most energy is used by neurons
hypertension (multi-infarct dementia) (Figures 2 and 3). The to fuel ionic pumps to maintain and restore the ionic gradients
construct of multi-infarct dementia, by attributing cognitive dissipated by synaptic activity (Harris et al., 2012). Due to fewer
impairment to multiple strokes, raised the possibility that pre- synapses, white matter energy usage, and consequently blood
venting cerebrovascular diseases could also prevent dementia flow, is 1/3 lower of that of the gray matter (Harris and Attwell,
(Hachinski et al., 1974). The introduction of brain imaging led 2012). The brain vasculature has an intimate developmental,
to the realization that diffuse white matter lesions, termed leu- structural, and functional relationship with the brain tissue,
koaraiosis (Hachinski et al., 1987), were a frequent correlate of their cellular elements forming a functional domain termed the
cognitive impairment, much more common than multiple in- neurovascular unit (Iadecola, 2004). Due to their intimate asso-
farcts, which turned out to be a rare cause of dementia (Hulette ciation with brain cells, cerebral blood vessels have unique
et al., 1997) (Figures 2 and 3). Genetic causes of white matter characteristics that set them apart from vessels in other organs
lesions were discovered, the prototypical one being the Cerebral (Abboud, 1981; Bevan, 1979; Quaegebeur et al., 2011). The
Autosomal Dominant Arteriopathy with Subcortical Infarcts and salient structural and functional features of the cerebral circula-
Leukoencephalopathy (CADASIL) (Chabriat et al., 2009). In the tion are briefly examined next.
early 1990s the criteria for diagnosis of vascular dementia
were largely based on those used for AD, which emphasized The Brain Vascular Network and Neurovascular Unit
memory impairment, irreversibility of the deficits, and impaired The brain is supplied by arteries arising from the circle of Willis,
activities of daily living (Roman et al., 1993). This definition a polygon of interconnected vessels at the base of the brain
was felt to be restrictive since it did not take into due consider- formed by the confluence of the internal carotid arteries and
ation cognitive deficits more commonly associated with cere- the basilar artery (Figure 4). The main vessels arising from the
brovascular lesions, such as executive dysfunction and psycho- circle of Willisthe anterior middle and posterior cerebral
motor slowing (Table S1). Therefore, the term vascular cognitive arteries, and their branchesgive rise to a rich anastomotic
impairment (VCI) was introduced to better reflect the full range of network on the brain surface (pial arteries and arterioles). Pial
cognitive alterations resulting from vascular factors (Hachinski vessels are endowed with a smooth muscle cell coat, which sur-
and Bowler, 1993) (Figure 2). By doing so, it was hoped that rounds a monolayer of endothelial cells (Figure 4). Pial arterial
the vascular nature of the cognitive deficit could be recognized branches dive into the brain substance, giving rise to smaller
early, providing the opportunity to slow down disease progres- arterioles still surrounded by an extension of the subarachnoid
sion by controlling vascular risk factors (Hachinski and Bowler, space filled with cerebrospinal fluid (perivascular space or
1993). The concept of VCI has gained wide acceptance and is Virchow-Robin space) (Figure 4). Delimited by the vascular
currently defined as a syndrome with evidence of clinical stroke basement membrane and the basement membrane of the glia
or subclinical vascular brain injury and cognitive impairment limitans (Figure 4) (Dyrna et al., 2013), the perivascular space

Neuron 80, November 20, 2013 2013 Elsevier Inc. 845


Neuron

Review
Figure 3. Brain Lesions Responsible for
Vascular Cognitive Impairment
All MRI sequences are diffusion-weighted imag-
ing, except for the white matter lesions, which is a
fluid attenuated inversion recovery sequence.
Images are courtesy of Dr. Hooman Kamel.

of its cellular constituents. Consequently,


neural activity, which uses most of the
brains energy budget, is the major deter-
minant of the dynamic regulation of CBF.
The increases in blood flow induced by
activation depend on the concerted ac-
tion of neurons, astrocytes, and vascular
cells through a wide variety of molecular
signals including ions, arachidonic acid
has emerged as critically important for the disposal of unwanted metabolites, nitric oxide (NO), adenosine, neurotransmitters,
proteins and peptides, e.g., Ab (Carare et al., 2013; Iliff et al., and neuropeptides (Drake and Iadecola, 2007). The hemody-
2013; Laman and Weller, 2013). As intracerebral arterioles reach namic changes underlying the increases in blood flow are
deeper into the brain parenchyma and become smaller (diameter mediated by vasoactive agents with opposing vascular actions
< 100 mm), the perivascular space disappears and the vessels (vasodilatation or vasoconstriction), generated by synaptic
basement membrane enters in direct contact with the glial activity, astrocytes, interneurons, and afferent projections
basement membrane enveloping the end-feet of astrocytes from the basal forebrain and brainstem (Cauli and Hamel,
(Figure 4). In capillaries, smooth muscle cells are replaced by 2010; Drake and Iadecola, 2007; Kleinfeld et al., 2011). These
pericytes, contractile cells that are particularly abundant in highly coordinated signals converge on specific sites of the
brain vessels and are involved in the development and mainte- cerebrovascular network to shape the hemodynamic response
nance of the BBB (Armulik et al., 2010; Bell et al., 2010; Quaege- to neural activation with a remarkable degree of spatial and
beur et al., 2011). temporal precision (Iadecola, 2004). Thus, the regional hemo-
The outside in vascularization pattern of the brain differs dynamic changes induced by activation are widely used to
from that of other major organs, like the kidney and liver that localize neuronal activity in the living brain using functional
are vascularized from the inside out, and places key vessels imaging (Attwell and Iadecola, 2002).
regulating intracerebral blood flow, the pial arterioles, outside Like in other organs, endothelial cells regulate vascular tone
the brain parenchyma. Consequently, elaborate vascular by releasing vasoactive factors in response to chemical signals,
signaling mechanisms coordinate changes in vascular diameter e.g., transmitters (Andresen et al., 2006), or mechanical forces,
of pial arterioles on the brains surface with those of the intrace- e.g., shear stress (Ando and Yamamoto, 2013). Unlike other
rebral microvasculature (Bagher and Segal, 2011; Iadecola, organs, cerebral endothelial cells in most brain regions are
2004). Another consequence of this vascular arrangement is adjoined by intricate junctional complexes formed by occludins
that occlusion of penetrating arterioles or venules, unlike pial and claudins (tight junctions) that prevent the bidirectional
vessels, cannot be effectively compensated by anastomotic exchange of hydrophilic substances between blood the brain,
branches (Blinder et al., 2013), and results in reductions in flow a key feature of the BBB (Dyrna et al., 2013). Rather, specialized
sufficient to produce small ischemic lesions akin to microinfarcts transport proteins on the endothelial cell membrane control the
(Nguyen et al., 2011; Nishimura et al., 2010; Shih et al., 2013). In traffic of solutes in and out of the brain. For example, GLUT1
addition, regions of the deep white matter are supplied by long and aminoacid transporters regulate the transfer of glucose
penetrating arterioles arising from the pial cortical network at and aminoacids into the brain, whereas efflux transporters,
the border between nonoverlapping vascular territories of the such as LRP-1, ABC transporters, and others, remove drugs
anterior and middle cerebral arteries, and as such are more and metabolic by-products from the brain, including Ab and
vulnerable to reductions in blood flow (Brown and Thore, 2011; lactate (Neuwelt et al., 2011).
De Reuck, 1971) (Figure 4). On the other hand, the basal ganglia Vascular smooth muscle cells, owing to their ability to
and brainstem are supplied by penetrating arterioles arising constrict when intravascular pressure increases (myogenic
directly from the circle of Willis and its proximal branches tone), adjust vascular tone in response to changes in arterial
(Figure 4), rendering these vessels more susceptible to the me- pressure to maintain CBF relatively constant within a range of
chanical stresses imposed by chronic hypertension or stiffening pressures (cerebrovascular autoregulation) (Cipolla, 2009).
of large arteries (Scuteri et al., 2011; Soros et al., 2013). Autoregulation protects cerebral blood vessels from the wide
swings in arterial pressure associated with the activities of
Neurovascular Control Mechanisms daily living and provides a stabile CBF baseline on which the
The brain is endowed with vasoregulatory mechanisms that dynamic changes induced by neurovascular coupling and
assure that it receives enough blood to support the energy needs endothelium are superimposed. These neurovascular control

846 Neuron 80, November 20, 2013 2013 Elsevier Inc.


Neuron

Review
Figure 4. Anatomy of the Cerebral Blood
Supply
(A) Circle of Willis.
(B) The arterial supply of the deep white matter
arises from branches of the ACA and the MCA. The
supply of the basal ganglia white matter is pro-
vided by arteries arising directly from the circle of
Willis and its proximal branches. Abbreviations:
ACA: anterior cerebral artery; ICA: internal carotid
artery; MCA: middle cerebral artery; PCA: poste-
rior cerebral artery.
(C) Anatomy of the wall of arteries, arterioles, and
capillaries.

bolism in astrocytes through the produc-


tion of NO (Brix et al., 2012). Therefore,
neurovascular cells are trophically and
metabolically interdependent, such that
damage to one cell type removes a vital
source of support to the whole unit and
has deleterious consequences also for
the other cell types.

Immune Trafficking and Regulation


The cells of the neurovascular unit are
involved in the initiation and expression
mechanisms work in concert to assure that the brain receives of adaptive and innate immune responses of the brain.
sufficient blood flow to meet the metabolic needs of its active Pericytes and perivascular macrophages have the potential
cellular constituents. for antigen presentation, the first step in adaptive immunity,
whereas endothelial cells and microglia are richly endowed
Trophic Coupling in the Neurovascular Unit with innate immunity receptors including CD36, toll-like recep-
Neurons, astrocytes, oligodendrocytes, and vascular and peri- tors (TLR), and the receptor for advances glycation end-
vascular cells are in state of close trophic and metabolic products (RAGE) (Lampron et al., 2013; Park et al., 2011).
codependence that plays a defining role in brain development, The perivascular space, which drains into the subarachnoid
function, and reaction to injury. In the developing nervous sys- space and then into cervical lymphnodes (Laman and Weller,
tem, prototypical neural guidance signals, ephrins, netrins, slit 2013), is the afferent arm through which brain antigens reach
glycoproteins, and semaphorins also contribute to endothelial the systemic immune system (Galea et al., 2007). The cells of
tip cell guidance (Carmeliet and Jain, 2011). In turn, classical the neurovascular unit also regulate the efferent arm of the
angiogenic molecules, such as VEGF, participate in neurogene- immune system, which relies on the transfer of effector immune
sis (neurovascular niche), neuronal cell migration, axon guidance, cells into the brain. In conditions of hypoxia-ischemia, endo-
dendritogenesis, and oligodendrocyte precursor migration (But- thelial cells express adhesion receptors, such as P-selectin,
ler et al., 2010; Carmeliet and Ruiz de Almodovar, 2013; Quaege- E-selectin, ICAM, and VCAM, instrumental for the transfer of
beur et al., 2011). In the adult nervous system, neuroblasts circulating leukocytes into the perivascular space (Iadecola
migrate along blood vessels, a process dependent on BDNF and Anrather, 2011). In turn, perivascular macrophages,
secretion by endothelial cells (Snapyan et al., 2009). Endothelial located in contact with the vascular basement membrane, are
cells have the potential to stimulate the proliferation of neuronal required for inflammatory cells, such as lymphocytes, to cross
precursors and to stir their differentiation toward the neuronal the glial basement membrane and move from the perivascular
lineage (Shen et al., 2004). Furthermore, through BDNF, insulin space into the brain parenchyma (Tran et al., 1998). Astrocytes
growth factor 2, chemokine (C-X-C motif) ligand 12, and pleiotro- express death ligands (CD95L) on their perivascular end feet
phin, endothelial cells support neuronal survival and protect them and control immune trafficking by triggering apoptosis of
from injury (Dugas et al., 2008; Guo et al., 2008). Endothelial cells CD95+ lymphocytes attempting to enter the brain (Bechmann
can also promote the proliferation and survival of oligodendro- et al., 1999). Therefore, the neurovascular unit is an important
cytes (oligovascular niche) by activating the Akt/PI3 kinase checkpoint regulating the afferent and efferent arms of the
pathway through BDNF and FGF (Arai and Lo, 2009). In addition immune system and shaping the immune responses of the
to their well-established interactions with neurons, astrocytes are brain. Vital to vascular homeostasis are circulating endothelial
also needed for the development and maintenance of BBB char- progenitor cells (EPC), hematopoietic stem cells involved in
acteristics in endothelial cells (Wolburg et al., 2009) and for the the maintenance and repair of endothelial cells (Hill et al.,
reorganization of vascular networks after brain injury (Hayakawa 2003). EPC development and function is controlled by CD31+
et al., 2012). In turn, endothelial cells regulate glycolytic meta- T cells (angiogenic T cells) through the release proangiogenic

Neuron 80, November 20, 2013 2013 Elsevier Inc. 847


Neuron

Review
Figure 5. Vascular Lesions Leading to VCI
and Their Effects on the Brain
See text for details. CAA: cerebral amyloid angi-
opathy; ATS: atherosclerosis.

On the other hand, if the reduction in


CBF is sustained and severe, ischemic
stroke ensues (Moskowitz et al., 2010).
Stroke doubles the risk for dementia
(poststroke dementia), and approxi-
mately 30% of stroke patients go on to
develop cognitive dysfunction within 3
years (Allan et al., 2011; Leys et al.,
2005; Pendlebury and Rothwell, 2009).
The association between stroke and de-
mentia is also observed in patients
younger than 50 years, up to 50% of
whom exhibit cognitive deficits after a
decade (Schaapsmeerders et al., 2013).
As mentioned, multiple infarcts, caused
by multiple arterial occlusions over time,
are well known to impair cognition
(multi-infarct dementia), as can a single
infarct located in a brain regions critical
for cognitive function, such as frontal
lobe or thalamus (Table 1) (strategic
infarct dementia) (Figure 3). However,
cytokines (Hur et al., 2007; Kushner et al., 2010). Thus, immune ischemic strokes are often associated with many of the vascular
cells are also involved in the maintenance of vascular homeo- pathologies described below, which also contribute to the total
stasis. vascular burden.

Cerebrovascular Pathologies Underlying Cognitive Small Vessel Disease, Leukoaraiosis,


Impairment Are Diverse and Lacunar Infarcts
Considering the vital importance of the cerebral blood supply By far, the most prevalent vascular lesions associated with VCI
for the structural and functional integrity of the brain, it is not are related to alterations in small vessels in the hemispheric
surprising that alterations in cerebral blood vessels have a white matter (Jellinger, 2013). These microvascular alterations
profound impact on cognitive function. The vascular alterations result in different neuropathological lesions, which can occur
that cause cognitive impairment are diverse, and include in isolation but, more typically, coexist in the same brain (Table 1).
systemic conditions affecting global cerebral perfusion or Confluent white matter lesions, the imaging correlate of which
alterations involving cerebral blood vessels, most commonly is termed leukoaraiosis (Figure 3), and lacunes, small (<1.5 cm)
small size arterioles or venules (Figure 5). Table 1 describes white matter infarcts typically in the basal ganglia, are common
some of the most common conditions, their vascular bases, occurrence in VCI and are strongly associated with cardiovascu-
and neuropathological correlates (see Jellinger [2013] for a lar risk factors, especially hypertension, diabetes, hyperlipid-
more complete list). emia, and smoking (Gorelick et al., 2011; Wardlaw et al.,
2013a, 2013b). The vascular pathologies underlying these le-
Reduced Cerebral Perfusion and Poststroke Dementia sions consist of atherosclerotic plaques affecting small cerebral
Reduction in global cerebral perfusion caused by cardiac vessels, deposition of a hyaline substance in the vascular wall
arrest, arrhythmias, cardiac failure, or hypotension can produce (lipohyalinosis), fibrotic changes in the vessel wall resulting in
brain dysfunction and impair cognition transiently or perma- stiffening and microvascular distortion (arteriolosclerosis), and
nently (Table 1) (Alosco et al., 2013; Justin et al., 2013; total loss of integrity of the vascular wall (fibrinoid necrosis)
Marshall, 2012; Stefansdottir et al., 2013). High-grade stenosis (Figure 5) (Thal et al., 2012). Arterioles become tortuous,
or occlusion of the internal carotid arteries is associated with have thickened basement membranes, and are surrounded
chronic ischemia and can lead to cognitive impairment even by enlarged perivascular spaces (Brown and Thore, 2011).
in the absence of ischemic lesions (Balestrini et al., 2013; Capillaries are reduced in number and string vessels, nonfunc-
Cheng et al., 2012; Johnston et al., 2004; Marshall, 2012) tional capillaries that have lost endothelial cells and have only a
(Figure 5). basement membrane, are observed (Brown and Thore, 2011).

848 Neuron 80, November 20, 2013 2013 Elsevier Inc.


Neuron

Review

Table 1. Selected Causes of Cognitive Impairment Related to Vascular Factors


Predominant Target Vessel and Resulting Brain
Condition Association/Cause Vascular Pathology Lesions References
Hypoperfusion d Cardiac arrest/failure d Large vessel ATS d Watershed infarcts Jellinger, 2013;
dementia d Hypotension d Vascular stiffening d Cortical laminar necrosis Johnston et al., 2004;
d Carotid occlusion d Incomplete white Marshall et al., 2012
matter infarcts
Strategic infarct d Arterial occlusion d Large-medium size arteries d Infarct in regions involved Jellinger, 2013
dementia in cognition, e.g.,
frontal lobe, thalamus, etc.
Multiinfarct dementia d Multiple arterial d Large-medium size d Multiple large infarcts Thal et al., 2012
occlusions arteries and arterioles d Lacunar infarcts
(embolic thrombotic) d Microinfarcts
White matter lesions d Vascular risk factors d Arterioles (<300 mm ) d Axonal damage Black et al., 2009;
(Leukoaraiosis) d CADASIL and other d Small vessel ATS d Demyelination Brown and Thore, 2011;
and lacunes genetic causes d Arteriolosclerosis d Lacunar infarcts Thal et al., 2012
d Lipohyalinosis d Microinfarcts
d Venous collagenosis d Microbleeds
Microinfarcts d CADASIL d Arterioles (<300 mm ) d Infarcts not visible Smith et al., 2012
d CAA, AD d Small vessel ATS by naked eye
d Vascular risk factors d Arteriolosclerosis
Microbleeds and d CAA, AD d Arterioles (<300 mm ) d Small hemorrhage in Charidimou et al., 2012;
hemorrhages d CADASIL d Vascular rupture perivascular space Henskens et al., 2008
d Vascular risk factors d Lobar or basal ganglia
hemorrhage
CADASIL d Notch 3 mutations d Arterioles (<300 mm ) d White matter lesions Chabriat et al., 2009;
d Thickened wall d Lacunar infarcts Federico et al., 2012;
d Smooth muscle cell GOM d Microinfarcts Schmidt et al., 2012
d Pericyte loss d Microbleeds
d Brain atrophy
Cerebral amyloid d Hereditary d Amyloid deposits in arteries d Hemorrhage Attems et al., 2011;
angiopathy d Sporadic (<2 mm ), arterioles, capillaries d White matter lesions Charidimou et al., 2012
d AD d Smooth muscle degeneration d Microinfarcts
d Vascular rupture d Microbleeds
Poststroke dementia d Ischemic stroke of d Large-medium size arteries d Silent infarcts Leys et al., 2005;
any cause d Perivascular immune cells (?) d White matter lesions Iadecola and Anrather,
d Lacunar infarcts 2011
d Neuronal loss
d Brain atrophy
Mixed AD vascular d Sporadic d Large-medium size arteries d AD pathology Jellinger, 2013;
dementia d Vascular risk factors and arterioles d White matter lesions Thal et al., 2012
d Lacunar infarcts
d Microinfarcts
d Microbleeds
Large infarct: > 1 cm ; Lacunar infarct: 515 mm ; microinfarct: < 1 mm ; microbleeds: < 5 mm ; ATS: atherosclerosis; GOM: graular osmophilic
material; vascular risk factors: hypertension, diabetes, smoking, etc.

Collagen deposits are observed in venules (venous collagenosis) with the evolution of the cognitive impairment (Maillard et al.,
(Black et al., 2009; Brown and Thore, 2011). The white matter 2012), new lacunes causing a steeper decline, especially in
damage resulting from these lesions consists of vacuolation, motor speed and executive functions (Jokinen et al., 2011).
demyelination, axonal loss, and lacunar infarcts. The white mat- White matter lesions and lacunar infarcts are also present in
ter lesions generally correspond to hyperintensities observed uncommon genetic conditions resulting in VCI and vascular
on MRI, which, however, can also reflect other pathological dementia (Federico et al., 2012; Schmidt et al., 2012). The
substrates (Gouw et al., 2011). The white matter lesions evolve better studied of these, CADASIL, is associated with extensive
over time by expansion of existing lesions, rather than formation leukoaraiosis and lacunar infarcts (Chabriat et al., 2009).
of new foci (Maillard et al., 2012), resembling the patterns of CADASIL vascular lesions are related to accumulation of gran-
progression of amyloid angiopathy (Alonzo et al., 1998; Robbins ular osmiofilic material (GOM) in vascular and perivascular cells,
et al., 2006). The expansion of the white matter lesions correlates which include the Notch 3 ectodomain (Yamamoto et al., 2013).

Neuron 80, November 20, 2013 2013 Elsevier Inc. 849


Neuron

Review

Microinfarcts and Microhemorrhages How Do Vascular Factors Cause Cognitive Impairment?


Microscopic infarcts (microinfarcts) and hemorrhages (micro- As reviewed above, VCI can stem from a wide variety of cardio-
bleeds) (Figure 5) are independent predictors of cognitive vascular and cerebrovascular pathologies, but it has been diffi-
dysfunction, but are commonly associated with other vascular cult to pin down the contribution of each condition to cognitive
pathologies, such as leukoaraiosis, lacunar infarcts, large dysfunction because of the coexistence of the different lesions
infarcts, and hemorrhage (Smith et al., 2012; van Norden and overlap with neurodegenerative pathology (Gorelick et al.,
et al., 2013), as well as CADASIL and AD (Table 1). Microin- 2011). Reductions in global cerebral perfusion, such as those
farcts are sharply delineated lesions consisting of pallor, necro- caused by heart diseases or carotid artery stenosis/occlusion,
sis, cavitation, and inflammation (astrocytosis, microgliosis, if below a critical threshold, can impair cognition independently
and macrophage infiltration) (Thal et al., 2012), caused by the of brain lesions (Marshall et al., 2012). Reductions in CBF by
small vessel pathology described above (Table 1). Microbleeds 40%50% are associated with suppression of brain activity
are microscopic areas of blood extravasation from leaky and cognitive dysfunction, which are reversible upon re-estab-
arterioles, which are restricted to the perivascular space and lishing normal CBF levels (Marshall et al., 1999; Marshall, 2012;
do not disrupt the brain parenchyma (De Reuck, 2012). Tatemichi et al., 1995). As for the other pathologies underlying
Observed in 17% of demented patients (Cordonnier and van VCI, there is a general correlation between the total burden of
der Flier, 2011), cortical microbleeds are frequently associated vascular pathology and cognitive deficits (Gelber et al., 2012;
with cerebral amyloid angiopathy (CAA), whereas microbleeds Gorelick et al., 2011; Inzitari et al., 2009). A caveat is that, due
in deep regions tend to be associated with white matter dis- to confounding factors, such as overlap with AD, differences in
ease secondary to vascular risk factors (De Reuck, 2012; educational level (see below), and microscopic pathology not
Park et al., 2013a). seen by in vivo imaging, the exact parameters of the relationship
have been hard to define (Black et al., 2009; Brickman et al.,
Cerebral amyloid angiopathy 2011). However, there is general consensus that cognitive
It is well known that deposits of Ab in cerebral blood vessels impairment results from the brain dysfunction caused by cumu-
or CAA are associated with vascular cognitive impairment. lative tissue damage (Gorelick et al., 2011), as originally pro-
Although inherited forms of CAA have been described, CAA posed by Tomlinson et al. for large cerebral infarcts (Tomlinson
is most prevalent in AD, being present in over 90% of cases et al., 1970).
(Attems et al., 2011; Charidimou et al., 2012). CAA is also In addition to gray matter damage, disruption of the white
observed in demented (50%60%) and nondemented matter can have profound effects on the precision and fidelity
(20%40%) elderly people (Attems et al., 2011; Charidimou of the information transfer underlying brain function and cogni-
et al., 2012). The major risk factor for CAA is advanced tive health (Nave, 2010a). Fast-conducting myelinated white
age, and cardiovascular risk factors seem to play a lesser matter tracts are responsible for long-range connectivity, inter-
role (Charidimou et al., 2012). CAA is a major cause of micro- hemispheric synchronization, and neurotrophic effects through
bleeds and large hemorrhages, typically located in the cortex spike-timing-dependent plasticity and axonal transport (Dan
(lobar hemorrhages) (Auriel and Greenberg, 2012). The amyloid and Poo, 2004; Nave, 2010a; Stone and Tesche, 2013). Indeed,
accumulation occurs in the media and the adventitia of white matter lesions affect brain structure and function broadly
cerebral vessels, leading to degeneration of smooth muscle and are associated with reductions in frontal lobe glucose utiliza-
cells and pericytes (Thal et al., 2012). In extreme cases, the tion (DeCarli et al., 1995; Haight et al., 2013; Tullberg et al., 2004),
vascular wall develops fibrinoid necrosis and the vessels global reduction in cortical blood flow (Appelman et al., 2008;
assume a characteristic double barrel appearance (Thal et al., Chen et al., 2013a; ten Dam et al., 2007; Kobari et al., 1990),
2012). disruption of brain connectivity (Lawrence et al., 2013; Sun
et al., 2011), and cerebral atrophy (Appelman et al., 2009). In
Mixed lesions addition, since myelination of previously naked fibers partici-
Overlap of AD neuropathology (amyloid plaques and neuro- pates in neuroplasticity and skilled motor learning (Fields,
fibrillary tangles) with cerebrovascular lesions is observed in up 2010; Richardson et al., 2011), myelin damage could also
to 50% of cases of dementia (Jellinger, 2013). These lesions compromise these important functions and contribute to cogni-
include atherosclerosis of the circle of Willis and its branches, tive impairment.
leukoaraiosis, and lacunar infarcts, microbleeds, microinfarcts,
and CAA (Benedictus et al., 2013; Charidimou et al., 2012; Risk Factors for Vascular Cognitive Impairments
Honig et al., 2005; Jellinger, 2013; Richardson et al., 2012; Ascertaining the genetic and modifiable risk factors of VCI is
Roher et al., 2004; Toledo et al., 2013; Yarchoan et al., 2012). problematic due to the multiplicity of underlying pathologies,
Ischemic lesions in regions between arterial territories coexistence with cardiovascular diseases, and the frequent
(watershed infarcts) have also been reported in AD, implicating overlap with AD and other neurodegenerative diseases (Gorelick
hypoperfusion and CAA in their mechanisms (Miklossy, 2003; et al., 2011). Therefore, the risk attributable to specific factors
Suter et al., 2002). Vascular lesions are also present in remains unclear, although the recent development of bio-
other age-related neurodegenerative diseases, such as synu- markers for in vivo AD diagnosis (Hampel et al., 2012) promises
cleinopathies, hippocampal sclerosis, and frontotemporal lobar to alleviate this problem.
degeneration linked to tau or TDP-43, but the coexistence with Advanced age is a powerful risk factor for VCI, and the
AD is the most frequent (Toledo et al., 2013). prevalence and incidence of cognitive impairment increases

850 Neuron 80, November 20, 2013 2013 Elsevier Inc.


Neuron

Review
Figure 6. Potential Mechanisms of the
Blood Vessel Damage Induced by Vascular
Risk Factors
Endothelial dysfunction, impairment of autor-
egulation, and dysfunction of neurovascular
coupling, partly mediated by oxidative stress and
NO deficit, reduce CBF resulting in hypoperfusion
and tissue hypoxia. In addition to hypoperfusion,
a critical consequence of endothelial dysfunction
is increased BBB permeability, which leads to
extravasation of plasma proteins, including fibrin-
ogen, into the brain. Fibrinogen activates CD11b
and TLR leading to production of ROS, proin-
flammatory cytokines and MMPs from activated
microglia, reactive astrocytes and OPCs. Inflam-
mation, in turn, aggravates the BBB breakdown
and induces expression of adhesion molecules in
endothelial cells, contributing to leukocyte and
platelet adhesion and microvascular occlusion.

phy caused by frameshift deletions in the


exonuclease TREX1, and mutations of
the COL4A1 gene encoding the type IV
exponentially after age 65 (Gorelick et al., 2011). The level of collagen alpha 1 chain (Federico et al., 2012; Gorelick et al.,
education, a surrogate marker of cognitive reserves (Stern, 2011; Lanfranconi and Markus, 2010). The ApoE 4 allele is a
2012), is an important determinant of the expression of VCI, well-established susceptibility gene for increased cardiovascu-
such that for a given level of neuropathology higher education lar risk and Alzheimer disease (Verghese et al., 2011). The
is associated with less cognitive deficits (Zieren et al., 2013). ApoE4 allele is associated with increased risk of CAA, whereas
However, the education level does not influence the rate of both ApoE2 and 4 increase the risk of lobar hemorrhages
progression of VCI and no longer has an impact in advanced dis- (Charidimou et al., 2012). Nevertheless, a strong link between
ease (Elbaz et al., 2013; Zieren et al., 2013). Although education ApoE and sporadic VCI has not been established (Lee and
could account for individual differences in the susceptibility Kim, 2013; Yu et al., 2013). Studies of candidate genes have
to cognitive impairment given comparable burdens of disease, revealed weak associations with genes involved in the renin-
socioeconomic status, coexisting chronic diseases, ethnicity, angiotensin system, endothelial nitric oxide synthase, oxidative
and premorbid intellectual capacity are important confounders stress, lipid metabolism and inflammation, but have not been
(Gorelick et al., 2011). replicated (Fornage et al., 2011; Lee and Kim, 2013; Markus,
Vascular risk factors, including hypertension, diabetes, hyper- 2008). GWAS of vascular dementia have shown small effect of
lipidemia, smoking, atrial fibrillation, and hyperhomocystinemia, SNPs in the androgen receptor gene locus (Schrijvers et al.,
increase the risk of dementia independently of the associated 2012), a finding not observed in all ethnic groups (Lee and Kim,
increase in stroke risk (Sahathevan et al., 2012). Furthermore, 2013). The diversity of pathologies underlying VCI and the
the metabolic syndrome, including insulin resistance, hyperten- overlap with AD complicate the interpretation of these studies.
sion, and dyslipidemia, has been associated with lower cognitive Linkage studies in patients with white matter lesions on MRI
performance (Yates et al., 2012). However, recurrent stroke is have discovered several loci (Schmidt et al., 2012), but no
one of the strongest predictors of dementia onset (Pendlebury specific gene has been identified and the findings await replica-
and Rothwell, 2009). Remarkably, in VCI as in AD, the increase tion and validation (Lee and Kim, 2013; Markus, 2008).
in risk afforded by vascular risk factors is observed decades
later, a finding that may explain why some studies did not find Pathogenic Mechanisms Responsible for White
a cognitive improvement with risk factor control later in life Matter Injury
(Sahathevan et al., 2012). Although as described in the previous section severe ischemia
A host of rare genetic mutations are associated with VCI resulting from arterial occlusion can lead to brain damage and
(Federico et al., 2012; Schmidt et al., 2012). The most common VCI, e.g., multi-infarct dementia, cognitive dysfunction is most
of these is the CADASIL syndrome caused by a frame shift often associated with more subtle vascular alterations targeting
mutation of Notch-3 that either creates or eliminates a cystein predominantly the deep hemispheric white matter (Figure 5).
residue (Chabriat et al., 2009). Other hereditary cerebral vas- Here we examine the major pathogenic mechanisms leading
culopathies include familial CAAs caused by mutations or to white matter damage, inferred either from brain imaging and
duplications of APP (Auriel and Greenberg, 2012; Rannikmae postmortem studies in humans, or animal models (Figure 6).
et al., 2013), the cerebral autosomal recessive arteriopathy
with subcortical infarcts and leukoencephalopathy (CARASIL) Hypoperfusion and Hypoxia
caused by mutation of the TGFb repressor HTRA1, the auto- Owing to their location at the distal border between different
somal dominant retinal vasculopathy with cerebral leukodystro- vascular territories (De Reuck, 1971) (Figure 4) and to the

Neuron 80, November 20, 2013 2013 Elsevier Inc. 851


Neuron

Review

susceptibility of their vasculature to risk factors (Brown and Increased Blood-Brain Barrier Permeability
Thore, 2011), deep white matter tracts are particularly vulnerable Reflecting another aspect of endothelial dysfunction, alterations
to vascular insufficiency. Even in healthy individuals, hypercap- in BBB permeability are also associated with leukoaraiosis and
nia, a potent vasodilator, does not increase, but reduces, CBF lacunar stroke (Wardlaw et al., 2013b; Yang and Rosenberg,
in the periventricular white matter, suggesting that vasodilatation 2011). Several lines of evidence indicate that the BBB is
of upstream vessels diverts blood flow to other regions (intrace- disrupted in the course of the disease. First, the plasma protein
rebral steal) (Mandell et al., 2008). This finding highlights the albumin is increased in the CSF of patient with VCI, reflecting
hemodynamic precariousness of the periventricular white mat- BBB breakdown (Candelario-Jalil et al., 2011). Second, plasma
ter, even in the absence of vascular damage. proteins, including complement, fibrinogen, albumin, and immu-
Increasing evidence suggests that the white matter cerebral noglobulins are detected in astrocytes in white matter lesions
blood supply is compromised in VCI (Figure 6). Resting flow is (Akiguchi et al., 1998; Alafuzoff et al., 1985; Simpson et al.,
reduced in areas of leukoaraiosis and vascular reactivity attenu- 2007; Tomimoto et al., 1996). Third, the permeability to MRI
ated (Kobari et al., 1990; Makedonov et al., 2013; Markus et al., tracers is increased in white matter lesions (Hanyu et al., 2002;
2000; 1994; Marstrand et al., 2002; OSullivan et al., 2002; Yao Taheri et al., 2011; Wardlaw et al., 2009) and in normal appearing
et al., 1992). In patients with VCI risk factors, like hypertension white matter (Topakian et al., 2010). The latter finding suggests
and diabetes, the ability of neural activity to increase blood that the BBB disruption could precede white matter injury and
flow in brain or retina is compromised (Delles et al., 2004; contribute to its development. BBB leakiness in white matter
Jennings et al., 2005; Sorond et al., 2011). Cerebrovascular was found in lacunar strokes, but not cortical strokes (Wardlaw
autoregulation is impaired, increasing the susceptibility of the et al., 2008), raising the possibility of a specific association
white matter to damage during fluctuation in blood pressure with small vessel disease of the deep white matter.
(Matsushita et al., 1994). Interestingly, CBF alterations have Several factors could contribute to the BBB disruption
also been described in normal appearing white matter (OSulli- (Rosenberg, 2012). Hypoxia-ischemia, which has been demon-
van et al., 2002), suggesting that the flow reduction precedes strated in white matter lesions, is well known to damage endo-
and, as such, may contribute to the white matter damage. thelial cells leading to increased BBB leakage in vitro (Al Ahmad
Indeed, in the general population, lower global CBF and lower et al., 2012). In vivo, hypoperfusion produced by bilateral carotid
cerebrovascular reactivity to hypercapnia is associated with a stenosis in rat increases BBB permeability (Ueno et al., 2002).
greater volume of white matter lesions (Bakker et al., 1999; In a similar model, the BBB alteration was found to be due to
Vernooij et al., 2008). The CBF reduction is observed prior MMP9 production by oligodendrocyte precursors, which are
to the onset of dementia (Ruitenberg et al., 2005). Due to their increased in ischemic white matter injury in rodent models
hemodynamic vulnerability, deep white matter regions are (Seo et al., 2013) and in patients with VCI (Candelario-Jalil
marginally perfused, and, in the presence of vascular risk fac- et al., 2011). In stroke prone spontaneously hypertensive rats,
tors, their vessels may be unable to adapt CBF to the metabolic which have a strong vascular risk factor profile, a high salt diet
needs of the tissue. Consistent with this hypothesis, postmortem induces fast-developing vasculopathy with BBB leakage that
studies have shown that areas of leukoaraiosis are chronically leads to ischemic injury in the absence of arterial occlusions
hypoxic, as indicated by the expression of hypoxia inducible (Schreiber et al., 2013). This finding indicates that chronic BBB
factors and related hypoxia-inducible genes (Fernando et al., disruption has the potential of induce ischemic damage. Indeed,
2006; Rosenberg et al., 2001). vascular risk factors, and the associated oxidative stress and
In addition to local factors affecting white matter microvessels, vascular inflammation also alter BBB permeability and could
broader-acting systemic factors are also involved. White matter play a role.
lesions and lacunar strokes are associated with increases in
circulating levels of the NO synthase inhibitor asymmetric Oxidative Stress and Inflammation
dimethylarginine (ADMA) (Notsu et al., 2009; Pikula et al., 2009; Pathological studies have shown markers of oxidative stress
Rufa et al., 2008). ADMA may contribute to the impairment of (isoprostanes) and inflammation (cytokines and adhesion
NO-dependent vasodilatation in peripheral and cerebral arteries molecules) in the damaged white matter associated with VCI
(Chen et al., 2006; Knottnerus et al., 2009; Pretnar-Oblak et al., (Back et al., 2011; Candelario-Jalil et al., 2011; Fernando et al.,
2006; Stevenson et al., 2010). Furthermore, stiffness of large 2006). Furthermore, microglial activation and reactive astrocytes
vessels and increased pulsatility are associated with reduced are also present in the lesions (Akiguchi et al., 1998; Simpson
white matter CBF and are strong predictors of leukoaraiosis et al., 2007; Tomimoto et al., 1996) (Figure 6). Markers of
and lacunes (Brisset et al., 2013; Tarumi et al., 2011; Webb endothelial activation, hemostasis, inflammation, and oxidative
et al., 2012), independently of vascular risk factors (Kearney- stress are also upregulated in blood, consistent with more
Schwartz et al., 2009). These findings implicate loss of large widespread effects in the systemic circulation (Gallacher et al.,
artery elasticity and increased pulsatile stress on microvessels, 2010; Knottnerus et al., 2010; Markus et al., 2005; Rouhl et al.,
especially those branching directly from the circle of Willis, in 2012a; Shibata et al., 2004; Xu et al., 2010) (Figure 6). The
the microvascular damage underlying white matter lesions mechanisms of these responses have not been fully elucidated,
(Scuteri et al., 2011). Similar microvascular changes occur also but several factors may play a role. Cerebral hypoperfusion
in other organs, suggesting that small vessel disease in brain is associated with white matter inflammation and oxidative
may be the manifestation of a systemic vasculopathy (Thompson stress in rodent models (Dong et al., 2011; Huang et al., 2010;
and Hakim, 2009). Ihara et al., 2001; Juma et al., 2011; Masumura et al., 2001;

852 Neuron 80, November 20, 2013 2013 Elsevier Inc.


Neuron

Review

Reimer et al., 2011; Yoshizaki et al., 2008), indicating that hypox- of colony forming units, a subset of EPC, to form vascular tubes
ia-ischemia is sufficient to trigger these responses. Vascular risk in a matrigel assay is impaired patients with large artery athero-
factors for VCI, such as hypertension, insulin resistance, and sclerosis or lacunar stroke (Chu et al., 2008). Interestingly, EPC
diabetes, lead to vascular oxidative stress and inflammation, colony forming units are also reduced in AD patients, in whom
both in animal models and in humans (Cohen and Tong, 2010; the magnitude of the reduction correlates with the degree of
Iadecola and Davisson, 2008; Yates et al., 2012), which, in cognitive impairment (Lee et al., 2009). Angiogenic T cells are
turn, impair the factors regulating the cerebral circulation (Faraci reduced in patients with vascular risk factors (Hur et al., 2007;
et al., 2011). Thus, functional hyperemia and endothelium Weil et al., 2011), and in hypertensive patients with small vessels
dependent responses are attenuated in models of aging, hyper- disease (Rouhl et al., 2012b). Furthermore, angiogenic T cells
tension, and diabetes (Ergul, 2011; Kazama et al., 2004; Park migration in vitro is positively correlated with preservation of
et al., 2007), whereas the ability of the vessels to adjust cerebral endothelium-dependent vasodilatation in patients with cardio-
perfusion in response to changes in blood pressure (autoregula- vascular risk factors (Weil et al., 2011), highlighting their protec-
tion) is blunted in patients with diabetes or hypertension (Kim tive role in vascular function. These findings, raise the possibility
et al., 2008b; Novak et al., 2003). Such neurovascular dysfunc- that vascular risk factors suppress the production of angiogenic
tion would aggravate the CBF reduction in critically perfused T cells, reduce the repair potential of EPC, and contribute to
deep white matter regions and contribute to the white matter the microvascular degeneration underlying leukoaraiosis and
damage. Accordingly, scavenging of free radicals or approaches lacunar stroke. Accordingly, capillary density is reduced not
to suppress inflammation counteract white matter damage and only at lesioned sites, but also in normal appearing white
behavioral deficits in rodent models of cerebral hypoperfusion matter in patients with VCI (Brown et al., 2007). Vessels devoid
(Dong et al., 2011; Kim et al., 2008a; Maki et al., 2011; Ueno of endothelium (string vessels) are often observed, reflecting a
et al., 2009; Wakita et al., 2008; Wang et al., 2010; Washida failure of endothelial repair, possibly due to EPC dysfunction or
et al., 2010; Zhang et al., 2011). NADPH oxidase, a multiunit loss of neuron and/or glial-derived growth factors.
enzyme particularly enriched in cerebral blood vessels (Miller Lesions of white matter tracts also lead to distant effects re-
et al., 2005), has emerges as an important source in vascular sulting from loss of trophic support at their site of termination.
oxidative stress in aging, hypertension, hyperlipidemia and dia- Leukoaraiosis is associated with focal cortical thinning espe-
betes (Faraci et al., 2011), and inhibition or genetic inactivation cially in frontal cortex, a finding correlated with executive
of this enzyme has been shown to ameliorate the vascular dysfunction (Seo et al., 2012). Focal cortical thinning was also
dysfunction (Drummond et al., 2011). Extravasation of plasma observed in a prospective study of patients with CADASIL
proteins, due to the BBB alterations, is also likely to play a subsequent to a subcortical infarct (Duering et al., 2012), indi-
role, since fibrinogen, immunoglobulins, and complement are cating a causal link between white matter lesions and cortical
potent activators of inflammation and free radical production atrophy. These processes are likely to play a role in the progres-
(Crehan et al., 2013; Davalos and Akassoglou, 2012; Yoshida sive cerebral atrophy observed in patients with leukoaraiosis,
et al., 2002). In particular, fibrinogen extravasation activates who experience a brain volume loss of 1% per year, twice that
inflammatory pathway through its interaction with integrin of age matches controls (Nitkunan et al., 2011). However, it
(CD11b/CD18) and non-integrin receptors (TLRs), leading to has not been established whether white matter lesions cause
activation of microglia and astrocytes (Davalos and Akassoglou, the atrophy independently of age and other risk factors (Appel-
2012; Davalos et al., 2012) (Figure 6). As discussed next, inflam- man et al., 2009; 2010). Trophic interactions are also critically
mation and oxidative stress have also deleterious effects on the involved in the demyelination and remyelination associated
trophic interaction among the cells of the neurovascular unit. with leukoaraiosis, which are examined next.

Trophic uncoupling Demyelination and Remyelination


ROS and inflammation suppress the prosurvival action of endo- One of the consequences of the oxidative and proinflammatory
thelial cells on neurons by reducing BDNF levels, an effect medi- environment induced by hypoperfusion and BBB breakdown
ated by impairing integrin linked kinase signaling (Guo et al., is damage to the myelin sheet and demyelination. Myelination
2008). In models of diabetes, advanced glycation end-products allows axons to conduct 100 times faster, and reduce energy
lead to MMP9 secretion by endothelial cells and cleavage of the expenditures by restricting the depolarization of the axonal
ectodomain of the BDNF receptor TRKB on neurons, reducing membrane to the Na+ channel rich Ranvier nodes (Nave,
neurotrophic signaling (Navaratna et al., 2013). Owing to their 2010b). Some of the energy saving afforded by myelination is
trophic support of vascular cells, dysfunction and damage to offset by the cost of maintaining the resting potential of oligo-
neurons and glia is associated with endothelial cell atrophy dendrocytes, which is estimated to be high (Harris and Attwell,
and microvascular rarefaction (Brown and Thore, 2011). Sys- 2012). Loss of myelin has important consequences for the white
temic factor also play a role in the mechanisms of trophic uncou- matter tracts. In addition to the brain dysfunction caused by
pling. EPC are reduced by stroke risk factors (Hill et al., 2003) and slowing down the transmission of axon potentials, demyelination
predict cardiovascular morbidity and mortality (Werner et al., threatens the integrity of the axons and leads to axonal loss
2005). EPC are reduced in age associated white matter lesions, (Franklin and Ffrench-Constant, 2008; Matute and Ransom,
the reduction correlating with lesion burden (Jickling et al., 2009). 2012). Several factors contribute to the demise of the axons.
In addition, EPC may be less functionally competent in patients Oligodendrocytes release growth factors, such as IGF-1 and
with vascular risk factors and stroke. For example, the ability glial cell-derived neurotrophic factor that support the survival

Neuron 80, November 20, 2013 2013 Elsevier Inc. 853


Neuron

Review

diseases, including leukoaraiosis, axons fail to fully remyelinate


(Franklin and Ffrench-Constant, 2008). Several factors are
thought to be responsible (Figure 7). First, OPC in the late stage
of development are particularly susceptible to injury in condi-
tions of chronic hypoxia and oxidative stress existing in the
ischemic white matter (Back et al., 2011; 2002; Fernando et al.,
2006; French et al., 2009). Oligodendrocytes are also suscepti-
ble to damage caused by extracellular ATP, which increases in
hypoxia-ischemia, through activation of the P2X7 purinergic re-
ceptors (Domercq et al., 2010). Second, withdrawal of trophic
support from damaged endothelial cells and astrocytes could
reduce the vitality of the OPC pool and contribute to their demise
in the hypoxic environment of the vulnerable white matter (Arai
and Lo, 2010). Third, failure to remyelinate could be related to
an arrest in OPC maturation. OPC are abundant in areas of
leukoaraiosis, which are enriched with hyluronan (HA), a high
molecular weight glycosaminoglycan produced by reactive as-
trocytes and other cells (Back et al., 2011). HA is a component
of the matrix and is involved in neurodevelopment promoting
neuronal migration (Sherman and Back, 2008). In white matter
lesions, HA is degraded by the hyaluronidase PH20 and its
cleavage products inhibit the maturation of OPC into oligoden-
drocytes capable of myelination (Preston et al., 2013) (Figure 7).
Dysregulation of the Wnt signaling pathway could also play a role
Figure 7. Potential Mechanisms of Failure to Remyelinate the
Damaged White Matter
in the OPC developmental arrest (Fancy et al., 2011b). In addi-
Inflammation-, oxidative-stress-, and hypoxia-induced demyelination. OPC tion, OPC produce MMP9, which, as seen in the previous
proliferate to attempt remyelination. High molecular weight hyaluronic acid sections, promotes BBB breakdown perpetuating the cytotoxic
(HMW-HA) produced by reactive astrocytes is cleaved by the hyaluronidase
milieu underlying demyelination (Seo et al., 2013).
PH20 generating digestion products that inhibit OPC maturation through
mechanisms involving TLR2 and 4 and GSK3b. The resulting OPC maturation
arrest prevents efficient remyelination. Putting It All Together
The evidence reviewed above suggests a convergence of
pathogenic factors on cerebral blood vessels, which in turn
of axons (Wilkins et al., 2003). Thus, loss of myelin deprives the leads to white matter damage (Figures 6 and 7). Oxidative
axons of trophic support and increases their vulnerability. In stress-induced endothelial dysfunction caused by risk factors
addition, demyelination exposes the axons to the deleterious is most likely an early event leading to white matter damage.
effects of cytokine and free radicals in the hypoxic white matter, Endothelial dysfunction has two major pathogenic conse-
which may impair axonal energy production leading to failure of quences: reductions in resting CBF in the marginally perfused
the Na+/K+ ATPase. The resulting accumulation of intracellular white matter, and alterations in the permeability of the BBB.
Na+ reverses the operation of the Na+/ Ca2+ exchanger, resulting In turn, hypoperfusion and BBB disruption lead to additional
in intracellular Ca2+ accumulation (Matute and Ransom, 2012; oxidative stress by inducing tissue hypoxia and by extravasa-
Stys et al., 1992). Furthermore, the adaptive upregulation of tion of plasma proteins, such as fibrinogen. Tissue edema re-
voltage-dependent Na+ channels (VNa+) in the denuded inter- sulting from increased BBB permeability may exacerbate these
nodal axoplasm, attempting to preserve impulse propagation alterations by compressing blood vessels and reducing CBF
in demyelinated axons, leads to Na+ entry and aggravates the further. Tissue hypoxia and oxidative stress activate inflamma-
energy deficit and Ca2+ overload. Upregulation of VNa+1.2 chan- tory pathways through NFkb-dependent transcription, leading
nels increases the activity of the Na+/K+ ATPase, stressing to production of cytokines and adhesion molecules in vascular
further the energy budget of the marginally perfused white matter cells, reactive astrocytes and activated microglia. Hypoxia,
(Trapp and Stys, 2009). In turn, excess intracellular Ca2+ acti- inflammation and oxidative stress damage oligodendrocytes
vates protease dependent processes that lead to microtubule and leads to trophic uncoupling in the neurovascular unit,
fragmentation and perturbation of axonal flow (Franklin and which, in turn, contribute to the damage to vascular cells and
Ffrench-Constant, 2008; Matute and Ransom, 2012). oligodendrocytes. Oligodendrocyte damage, oxidative stress
Attempts to remyelinate are present in the damaged white and inflammation lead to demyelination and attempted remye-
matter in leukoaraiosis (Jonsson et al., 2012). Oligodendrocytes lination through OPC proliferation. Developmental arrest of
are responsible for the formation and maintenance of the myelin OPC, due to HA degradation products, leads to accumulation
sheet. A large pool of oligodendrocyte progenitor cells (OPC) of these cells which secrete MMP9 and worsen the BBB
is present in the brain, which goes through several stages of impairment (Figure 7). Once demyelination occurs, the
development before becoming mature and competent to lay increased energy requirement of the denuded axons aggra-
down myelin (Fancy et al., 2011a). However, in demyelinating vates the hypoxic stress of the tissue, leading to a vicious circle

854 Neuron 80, November 20, 2013 2013 Elsevier Inc.


Neuron

Review

that perpetuates these pathogenic processes and exacerbates observations highlight the importance of neurovascular factors
the tissue damage. in maintaining white matter health.

Is Hypoperfusion Also Involved in Inherited and Overlap between Vascular and Neurodegenerative
Autoimmune White Matter Diseases? Dementia
There is emerging evidence that reduced cerebral perfusion The realization that most cases of dementia have mixed patho-
may contribute to other diseases characterized by white matter logical features has raised the intriguing possibility that vascular
damage. Multiple sclerosis (MS) is the prototypical neuroinflam- factors play role in AD and other neurodegenerative diseases.
matory disease in which demyelination is thought to be related As discussed in the section on Mixed lesions, AD brains
to a T cell mediated autoimmune attack on myelin (McFarland have a wide variety of vascular lesions, suggesting a potential
and Martin, 2007). However, in MS patients CBF is reduced in pathogenic interaction between vascular factors and AD.
the normal appearing white matter (Law et al., 2004), as well However, since cerebrovascular diseases and AD are common
as in the gray matter (Dhaeseleer et al., 2011). In contrast, in in the aged, the coexistence of the two pathologies could
active lesions displaying BBB disruption CBF is increased, simply be coincidental (Hachinski, 2011). The overall effect on
consistent with vasodilatation caused by inflammation (Dhae- cognition would results from the combined burden of vascular
seleer et al., 2011). The reduction in CBF in the normal white and neurodegenerative pathology, according to an additive
matter could be caused by a primary vascular dysfunction path- model. Alternatively, vascular disease could promote AD and
ogenically linked to the disease process, or could be secondary vice-versa, resulting in a reciprocal interaction amplifying their
to loss of white matter elsewhere, due to distal Wallerian degen- pathogenic effects. The cognitive impact of vascular and AD
eration, or reduced synaptic activity (De Keyser et al., 2008). neuropathology depends on the severity of the AD pathology
Studies in which CBF measurements in the normal appearing and location of the vascular lesions (Gold et al., 2007). In
white matter were coupled to diffusion tensor imaging, revealed advanced cases of AD, vascular lesions do not seem to have a
that the reductions in CBF are associated with restricted diffu- major influence on the progression of the cognitive deficits,
sion and not with increased fractional anisotropy, as anticipated suggesting the AD pathology is the major driver of the cognitive
if the CBF changes were secondary to Wallerian degeneration dysfunction (Chui et al., 2006; Jellinger, 2001). On the other
(Saindane et al., 2007). Although the possibility that the reduc- hand, in older individuals with moderate AD pathology subcor-
tion in CBF is secondary to reduced local synaptic activity tical vascular lesions are a major determinant of the expression
has not been ruled out, the fact that the hypoperfusion is of the dementia (Esiri et al., 1999; Schneider et al., 2007b;
normalized by an endothelin receptor antagonist suggest a pri- Snowdon et al., 1997).
mary vascular cause (Dhaeseleer et al., 2013). Consistent with
the hypoperfusion hypothesis, HIF-1a and dependent genes Cerebrovascular Factors and AD
are upregulated in normal appearing white matter (Graumann Cerebrovascular function is reduced in patients with early AD or
et al., 2003). at risk for AD (Claassen et al., 2009; Gao et al., 2013; Luckhaus
Reductions in white matter CBF has also been found X-linked et al., 2008; Mentis et al., 1996; Niedermeyer, 2006; Ruitenberg
adrenoleukodystrophy (ALD), a disease caused by mutations in et al., 2005; Sabayan et al., 2012; Tanaka et al., 2002), impli-
ABCD1, which encodes a peroxisomal membrane transporter cating reduced cerebral perfusion in the pathobiology of the
protein, leading to accumulation of very long chain fatty acids disease (Iadecola, 2004). Conversely, some studies (Jendroska
in brain, spinal cord and adrenal glands (Moser et al., 2000). In et al., 1995; Ly et al., 2012), but not others (Aho et al., 2006; Mas-
its infantile form, the disease starts between 4 and 8 years taglia et al., 2003), have reported increased amyloid deposition
of age and is characterized by a progressive cognitive decline in stroke patients, implicating that ischemia promotes AD pathol-
associated with rampant inflammatory demyelination of the ogy. Furthermore, AD and cerebrovascular diseases may have
white matter (Moser et al., 2000). BBB alterations predict disease common risk factors, such as hypertension, insulin resistance,
progression (Melhem et al., 2000). Cerebral blood volume, diabetes, obesity, hyperhomocystinemia, hyperlipidemia, etc.
assessed by susceptibility contrast MRI (Musolino et al., 2012), (Craft, 2009; Fillit et al., 2008; Honjo et al., 2012; Purnell et al.,
or CBF, assessed by single photon emission tomography 2009). However, the correlation was most evident when the
(al Suhaili et al., 1994), is reduced in the normal appearing and risk factors were considered together and not individually
abnormal white matter. The mechanisms of the white matter (Chui et al., 2012). Furthermore, the correlation was strongest
hypoperfusion remain to be defined. Reductions in CBF prior for vascular dementia and weakest for AD, suggesting that
to white matter damage were also observed in a patient with vascular risk factors may independently increase the likelihood
Alexander disease, a rare childhood disease caused by a domi- of dementia without exacerbating AD pathology (Chui et al.,
nant mutation of the GFAP gene (Ito et al., 2009). 2012). In contrast, studies that have prospectively evaluated
It is noteworthy that, despite fundamental differences in their representative patients cohorts with confirmation of the clinical
pathogenesis, inherited and autoimmune diseases of the white diagnosis at autopsy failed to establish a link between the
matter exhibit cerebrovascular alterations before pathology burden of AD pathology and vascular risk factors (Chui et al.,
develops, just like in white matter disease caused by vascular 2012). It is, therefore, conceivable that in cases in which AD
factors. Thus, hypoperfusion and BBB disruption seem neces- was diagnosed clinically there might have been a component
sary correlates of the process leading to white matter damage of vascular pathology. New imaging and CSF biomarkers for
independently of the primary disease cause. Collectively, these the in vivo diagnosis of AD may provide additional insights into

Neuron 80, November 20, 2013 2013 Elsevier Inc. 855


Neuron

Review

whether vascular factors are pathogenically linked to AD (Chui tion through a proinflammatory pathway involving cyclophilin A
et al., 2012; Haight et al., 2013; Purnell et al., 2009). in pericytes (Bell et al., 2012). Activation of this pathway causes
MMP-9-mediated degradation of endothelial tight junctions
Vascular Effects of Ab and basement membrane proteins, as shown in human
Mounting evidence that Ab has powerful vascular effects also ApoE4 targeted replacement mice (Bell et al., 2012). ApoE4
suggests a link between AD and vascular disease. Ab1 40 positive individuals may develop a similar age-dependent
constrict isolated cerebral and systemic blood vessels (Niwa BBB breakdown prior to cognitive decline (Halliday et al.,
et al., 2001; Paris et al., 2003; Thomas et al., 1996), whereas 2013). In patients with vascular risk factors, such as hyperten-
application of Ab1 40 to the exposed cerebral cortex of mice sion, sedentary life style, or ApoE4 genotype, there is a greater
reduces CBF and impairs the increase in CBF induced by tendency for amyloid accumulation (Head et al., 2012; Rodrigue
endothelium-dependent vasodilators and functional hyperemia et al., 2013), whereas amyloid accumulation is reduced in pa-
(Niwa et al., 2000a; 2000b). Similarly, functional hyperemia, tients who exercise regularly (Liang et al., 2010). Experimental
endothelium-dependent responses and autoregulation are pro- studies indicate that this clearance mechanism is altered in
foundly impaired in young mice overexpressing mutated forms the presence of vascular dysfunction and damage, contributing
of APP, in which brain Ab is elevated, but there are no plaques, to parenchymal and vascular Ab accumulation (Deane et al.,
behavioral alterations, or reductions in resting glucose utilization 2004; Park et al., 2013b). In particular, suppression of LRP1
(Niwa et al., 2000b; 2002; Tong et al., 2012). These data suggest in vascular smooth muscle cells due to upregulation of serum
that the cerebrovascular effects of Ab are not attributable to response factor and myocardin, is a key factor in the clearance
CAA or amyloid plaques, and are not a consequence of impairment (Bell et al., 2009). Collectively, these observations
neuronal energy hypometabolism. APP-overexpressing mice suggest a link between cerebrovascular health and brain Ab
have increased brain damage following occlusion of the middle clearance.
cerebral artery (Koistinaho et al., 2002; Zhang et al., 1997), an These lines of evidence suggest that AD is frequently associ-
effect in part related to poor collateral circulation due to vascular ated with cerebral macro- and micro-vascular pathology, which
dysregulation (Zhang et al., 1997). The vascular alterations can contribute to the expression of the dementia. Vascular risk
induced by Ab are abrogated by overexpression of the ROS factors can increase amyloid accumulation and the risk of
scavenging enzyme superoxide dismutase or deficiency of the clinically defined AD. The vasoactivity of Ab and the influence
NADPH oxidase subunit NOX2 (Iadecola et al., 1999; Park of cerebral perfusion on APP processing and Ab clearance
et al., 2005; 2008), implicating ROS produced by the enzyme suggest that cerebral blood vessels can have a role the accu-
NADPH oxidase in the vascular dysfunction. The mechanisms mulation of Ab in the brain parenchyma and cerebral blood
of NADPH oxidase activation involve the Ab-binding scavenger vessels. Preliminary evidence suggests that control of vascular
receptor CD36 (Park et al., 2011). Aged APP mice deficient in risk factors reduces vascular lesions in AD (Richard et al.,
CD36 are protected from cerebrovascular alterations and 2010), and may delay disease progression (Deschaintre
behavioral deficits, effects associated with reduced CAA et al., 2009), at least early in the disease course (Richard et al.,
compared to controls, but no reduction of amyloid plaques 2010). Although replication in representative cohorts in which
(Park et al., 2013b). Thus, CD36, which is located in vascular AD is confirmed pathologically or with biomarkers is needed,
and perivascular cells, may contribute to the accumulation of these observations provide initial evidence that improving
Ab in cerebral blood vessels. vascular health may also help in AD.

Ab Production and Clearance Prospects for Prevention and Therapy


Hypoperfusion and hypoxia caused by vascular insufficiency The development of treatments for VCI has been hampered by
may also facilitate Ab production by activating the APP cleav- the lack of a suitable animal model recapitulating the multifac-
age enzyme b-secretase (Kitaguchi et al., 2009; Sun et al., eted features of the disease (Gorelick et al., 2011). Although
2006; Tesco et al., 2007; Wen et al., 2004a). Cerebral ischemia several animal models have been developed (Hainsworth et al.,
promotes amyloid plaque formation (Garcia-Alloza et al., 2011; 2012; Lee et al., 2012), the most widely used has been white
Kitaguchi et al., 2009; Okamoto et al., 2012), and tau phos- matter damage produced by chronic forebrain ischemia (Ihara
phorylation (Koike et al., 2010; Wen et al., 2007; 2004b). The and Tomimoto, 2011). These models have demonstrated
vascular effects of Ab may also impair the clearance of the pep- that counteracting some of the pathogenic factors, including
tide, a key factor in brain Ab accumulation in sporadic AD chronic ischemia, inflammation, and oxidative stress, reduce
(Mawuenyega et al., 2010). The vascular pathway is estimated white matter damage and/or behavioral deficits (Dong et al.,
to be a major route of removal of Ab from the brain (Castellano 2011; Ihara and Tomimoto, 2011; Maki et al., 2011). Other
et al., 2012; Shibata et al., 2000). Brain Ab is transported along approaches have attempted to promote remyelination by
the perivascular pathway draining into the cervical lymphnodes stimulating the survival and differentiation of OPC (Miyamoto
(Carare et al., 2013; Iliff et al., 2013). In addition, Ab is cleared et al., 2010). Despite these positive results in models of hypo-
from the brain through a transvascular transport system perfusion-induced white matter damage, there are no FDA
involving LRP-1 (Shibata et al., 2000), a protein that acts in con- approved treatments for VCI and vascular dementia (Butler
cert with P-glycoprotein, ApoE, ApoJ, and a2-macroglobulin to and Radhakrishnan, 2012). Treatment with antioxidants, anti-
regulate brain Ab homeostasis (Zlokovic, 2008). Interestingly, inflammatory agents or agents increasing cerebral perfusion
ApoE4, a major genetic risk factor for AD, leads to BBB disrup- have not led to consistent results (Butler and Radhakrishnan,

856 Neuron 80, November 20, 2013 2013 Elsevier Inc.


Neuron

Review

2012). Some agents, like the neurotrophic factor cerebrolysin, topology and precarious blood supply are likely to play a role, the
showed a modest cognitive improvement, but the evidence is cellular and molecular bases determining the characteristic
not sufficiently strong to justify clinical use (Chen et al., 2013b). vascular lesions remain to be defined. In particular, how aging
Clinical trials are currently exploring other agents, including and vascular risk factors interact with the vascular wall to induce
cholinergic stimulants (choline alphoscerate), vasodilators vascular lesions preferentially in the white matter remains un-
(udenafil), inhibitor of platelet aggregation (cilostazol) and clear. The relative contribution of hypoperfusion, BBB damage,
delta-9-tetrahydrocannabinol (a complete list can be found at and oxidative stress to vascular and parenchymal damage
www.clinicaltrials.gov). remain to be defined. Furthermore, what determines the
On the other hand, increasing evidence indicates that the pathological diversity, e.g., lacunes, microinfarcts, microhe-
risk of VCI and vascular dementia can be reduced by preven- morrhages, etc., and spatial localization of the brain lesions
tive measures. A study in the UK population suggests that resulting from similar vascular pathology remain unexplained.
the prevalence of dementia may be decreasing, a finding A better understanding of ischemic demyelination and abortive
interpreted to reflect the beneficial effects of controlling blood remyelination is needed. Fundamental questions concerning
pressure and other risk factors (Matthews et al., 2013). Indeed, the interaction of AD pathology with vascular pathology also
rigorous blood pressure control reduces white matter damage remain unanswered. Studies elucidating the vascular biology of
and staves off cognitive decline (Sharp et al., 2011; Soros the white matter and the interaction with risk factors and
et al., 2013). Physical and mental activity, social engagement, AD pathology would be needed to shed light on some of
and a diet rich in antioxidants or polyunsaturated fatty acids these issues and provide better insight into potential therapeutic
reduce dementia risk (Aarsland et al., 2010; Akinyemi et al., targets. These mechanistic studies can benefit from the increas-
2013; Middleton and Yaffe, 2009; Verdelho et al., 2012). There- ing availability of cell-specific conditional genetic models,
fore, controlling vascular risk factors and promoting a healthy viral-based gene delivery methods, and novel approaches for
diet, exercise, and mental activity are promising strategies targeted cell replacement/modification in the brain, e.g., (Gold-
to reduce VCI. This hypothesis is supported by a study indi- man et al., 2012).
cating that weight control, a healthy diet, nonsmoking, physical Developing treatments for VCI remains a challenge. In addition
activity, and keeping total cholesterol, blood pressure, and to the lack of predictive animal models to guide target selection,
fasting glucose at goal levels are associated with better the heterogeneity of the underlying pathology represents a ther-
cognitive performance later in life (Reis et al., 2013). However, apeutic challenge. The role of hypoperfusion, BBB disruption,
most of the evidence is based on observational studies, which oxidative stress, and inflammation is well established in animal
have not been confirmed by randomized clinical trials of risk models of white matter damage, but therapies based on these
factor modification, stressing the need for further large-scale pathogenic mechanisms have not been successful. Although it
studies (Dichgans and Zietemann, 2012; Middleton and Yaffe, has been difficult to prove that these approaches achieved the
2009). intended effect on cerebral perfusion, ROS production, and
inflammation in the white matter at risk, other considerations
Conclusions make the development of treatments particularly challenging.
VCI and vascular dementia are major contributors to age-related For example, the long preclinical phase of dementia is problem-
dementing illnesses and comprise a heterogeneous group of atic, since, in VCI as in AD, initiating therapy when patients
cognitive disorders attributable to vascular causes. Vascular become symptomatic may be too late. Furthermore, due to
pathology is also an integral part of AD and other late-life neuro- frequent overlap with AD, the diagnosis of VCI can be chal-
degenerative conditions associated with dementia and plays a lenging, complicating the choice of the best therapeutic
defining role in the expression of the cognitive dysfunction. approach (Wang et al., 2012). Novel imaging modalities,
Despite the diversity of the underlying brain pathology, the including amyloid and tau imaging, as well as high-resolution
vascular alterations have a similar pathogenic basis, resulting MRI, will go a long way in addressing some of these challenges
from hypoperfusion, oxidative stress and inflammation, which and will make it possible to characterize the pathology in vivo
in turn lead to endothelial damage, BBB breakdown, activation with an unprecedented spatial, temporal, and morphological
of innate immunity, and disruption of trophic coupling between accuracy. At the same time, these approaches offer the prospect
vascular and brain cells. The hemispheric white matter, which of developing new biomarkers that will be critical for identifying
is particularly susceptible to the deleterious effects of vascular patients at risk, staging the progression of the disease, and
risk factors, is a major target of these vascular alterations. The assessing therapeutic efficacy.
resulting demyelination and axonal loss play a role in the broad Considering that mixed dementia is the most common
functional brain changes underlying cognitive impairment and cause of dementia in the elderly, it has become increasingly
in the associated cerebral atrophy. This chain of events high- important to harmonize basic science, translational, and clinical
lights the critical role that vascular cells play in the maintenance approaches in AD and vascular dementia. Thus, the impact
of the health of neurons, glia, and myelin. of both pathologies should be considered, independently of
However, several fundamental questions remain to be whether their contribution is additive or synergistic. In the
addressed. The predilection of the vascular pathology for the absence of effective therapies, promoting and maintaining
deep hemispheric white matter, a remarkable constant in condi- vascular health seems critical to prevent both the vascular and
tions as diverse as CADASIL and sporadic white matter disease, neurodegenerative components of the disease and is probably
remains incompletely understood. Although its peculiar vascular the best possible course of action at the present.

Neuron 80, November 20, 2013 2013 Elsevier Inc. 857


Neuron

Review
SUPPLEMENTAL INFORMATION Arai, K., and Lo, E.H. (2010). Astrocytes protect oligodendrocyte precursor
cells via MEK/ERK and PI3K/Akt signaling. J. Neurosci. Res. 88, 758763.
Supplemental Information includes one table and can be found with this article
Armulik, A., Genove, G., Mae, M., Nisancioglu, M.H., Wallgard, E., Niaudet, C.,
online at http://dx.doi.org/10.1016/j.neuron.2013.10.008.
He, L., Norlin, J., Lindblom, P., Strittmatter, K., et al. (2010). Pericytes regulate
the blood-brain barrier. Nature 468, 557561.
ACKNOWLEDGMENTS
Attems, J., Jellinger, K., Thal, D.R., and Van Nostrand, W. (2011). Review: spo-
radic cerebral amyloid angiopathy. Neuropathol. Appl. Neurobiol. 37, 7593.
We gratefully acknowledge the support from the NIH (NINDS: NS37853,
NHLBI: HL96571), the Alzheimers Association (ZEN-11-202707), and the Attwell, D., and Iadecola, C. (2002). The neural basis of functional brain
Feil Family Foundation. Dr. Giuseppe Faraco provided invaluable help with imaging signals. Trends Neurosci. 25, 621625.
the figures.
Auriel, E., and Greenberg, S.M. (2012). The pathophysiology and clinical pre-
REFERENCES sentation of cerebral amyloid angiopathy. Curr. Atheroscler. Rep. 14, 343350.

Back, S.A., Han, B.H., Luo, N.L., Chricton, C.A., Xanthoudakis, S., Tam, J.,
Aarsland, D., Sardahaee, F.S., Anderssen, S., and Ballard, C.; Alzheimers Arvin, K.L., and Holtzman, D.M. (2002). Selective vulnerability of late oligoden-
Society Systematic Review group (2010). Is physical activity a potential drocyte progenitors to hypoxia-ischemia. J. Neurosci. 22, 455463.
preventive factor for vascular dementia? A systematic review. Aging Ment.
Health 14, 386395. Back, S.A., Kroenke, C.D., Sherman, L.S., Lawrence, G., Gong, X., Taber,
E.N., Sonnen, J.A., Larson, E.B., and Montine, T.J. (2011). White matter lesions
Abboud, F.M. (1981). Special characteristics of the cerebral circulation. Fed. defined by diffusion tensor imaging in older adults. Ann. Neurol. 70, 465476.
Proc. 40, 22962300.
Bagher, P., and Segal, S.S. (2011). Regulation of blood flow in the microcircu-
Aho, L., Jolkkonen, J., and Alafuzoff, I. (2006). Beta-amyloid aggregation in lation: role of conducted vasodilation. Acta Physiol. (Oxf.) 202, 271284.
human brains with cerebrovascular lesions. Stroke 37, 29402945.
Bakker, S.L., de Leeuw, F.E., de Groot, J.C., Hofman, A., Koudstaal, P.J., and
Akiguchi, I., Tomimoto, H., Suenaga, T., Wakita, H., and Budka, H. (1998). Breteler, M.M. (1999). Cerebral vasomotor reactivity and cerebral white matter
Blood-brain barrier dysfunction in Binswangers disease; an immunohisto- lesions in the elderly. Neurology 52, 578583.
chemical study. Acta Neuropathol. 95, 7884.
Balestrini, S., Perozzi, C., Altamura, C., Vernieri, F., Luzzi, S., Bartolini, M.,
Akinyemi, R.O., Mukaetova-Ladinska, E.B., Attems, J., Ihara, M., and Kalaria, Provinciali, L., and Silvestrini, M. (2013). Severe carotid stenosis and impaired
R.N. (2013). Vascular risk factors and neurodegeneration in ageing related cerebral hemodynamics can influence cognitive deterioration. Neurology 80,
dementias: Alzheimers disease and vascular dementia. Curr. Alzheimer 21452150.
Res. 10, 642653.
Bechmann, I., Mor, G., Nilsen, J., Eliza, M., Nitsch, R., and Naftolin, F. (1999).
Al Ahmad, A., Gassmann, M., and Ogunshola, O.O. (2012). Involvement FasL (CD95L, Apo1L) is expressed in the normal rat and human brain: evi-
of oxidative stress in hypoxia-induced blood-brain barrier breakdown. dence for the existence of an immunological brain barrier. Glia 27, 6274.
Microvasc. Res. 84, 222225.
Bell, R.D., Deane, R., Chow, N., Long, X., Sagare, A., Singh, I., Streb, J.W.,
Alafuzoff, I., Adolfsson, R., Grundke-Iqbal, I., and Winblad, B. (1985). Perivas- Guo, H., Rubio, A., Van Nostrand, W., et al. (2009). SRF and myocardin regu-
cular deposits of serum proteins in cerebral cortex in vascular dementia. Acta late LRP-mediated amyloid-beta clearance in brain vascular cells. Nat. Cell
Neuropathol. 66, 292298. Biol. 11, 143153.

Allan, L.M., Rowan, E.N., Firbank, M.J., Thomas, A.J., Parry, S.W., Polvikoski, Bell, R.D., Winkler, E.A., Sagare, A.P., Singh, I., LaRue, B., Deane, R., and
T.M., OBrien, J.T., and Kalaria, R.N. (2011). Long term incidence of dementia, Zlokovic, B.V. (2010). Pericytes control key neurovascular functions and
predictors of mortality and pathological diagnosis in older stroke survivors. neuronal phenotype in the adult brain and during brain aging. Neuron 68,
Brain 134, 37163727. 409427.

Alonzo, N.C., Hyman, B.T., Rebeck, G.W., and Greenberg, S.M. (1998). Bell, R.D., Winkler, E.A., Singh, I., Sagare, A.P., Deane, R., Wu, Z., Holtzman,
Progression of cerebral amyloid angiopathy: accumulation of amyloid- D.M., Betsholtz, C., Armulik, A., Sallstrom, J., et al. (2012). Apolipoprotein E
beta40 in affected vessels. J. Neuropathol. Exp. Neurol. 57, 353359. controls cerebrovascular integrity via cyclophilin A. Nature 485, 512516.

Alosco, M.L., Brickman, A.M., Spitznagel, M.B., Garcia, S.L., Narkhede, A., Benedictus, M.R., Goos, J.D.C., Binnewijzend, M.A.A., Muller, M., Barkhof, F.,
Griffith, E.Y., Raz, N., Cohen, R., Sweet, L.H., Colbert, L.H., et al. (2013). Scheltens, P., Prins, N.D., and van der Flier, W.M. (2013). Specific risk factors
Cerebral perfusion is associated with white matter hyperintensities in older for microbleeds and white matter hyperintensities in Alzheimers disease.
adults with heart failure. Congest. Heart Fail. 19, E29E34. Neurobiol. Aging 34, 24882494.

Ando, J., and Yamamoto, K. (2013). Flow detection and calcium signalling in Bertram, L., and Tanzi, R.E. (2008). Thirty years of Alzheimers disease
vascular endothelial cells. Cardiovasc. Res. 99, 260268. genetics: the implications of systematic meta-analyses. Nat. Rev. Neurosci.
9, 768778.
Andresen, J., Shafi, N.I., and Bryan, R.M., Jr. (2006). Endothelial influences on
cerebrovascular tone. J. Appl. Physiol. 100, 318327. Bevan, J.A. (1979). Sites of transition between functional systemic and
cerebral arteries of rabbits occur at embryological junctional sites. Science
Appelman, A.P.A., van der Graaf, Y., Vincken, K.L., Tiehuis, A.M., Witkamp, 204, 635637.
T.D., Mali, W.P.T.M., and Geerlings, M.I.; SMART Study Group (2008). Total
cerebral blood flow, white matter lesions and brain atrophy: the SMART-MR Black, S., Gao, F., and Bilbao, J. (2009). Understanding white matter dis-
study. J. Cereb. Blood Flow Metab. 28, 633639. ease: imaging-pathological correlations in vascular cognitive impairment.
Stroke 40 (Suppl ), S48S52.
Appelman, A.P.A., Exalto, L.G., van der Graaf, Y., Biessels, G.J., Mali, W.P.T.M.,
and Geerlings, M.I. (2009). White matter lesions and brain atrophy: more than Blinder, P., Tsai, P.S., Kaufhold, J.P., Knutsen, P.M., Suhl, H., and Kleinfeld, D.
shared risk factors? A systematic review. Cerebrovasc. Dis. 28, 227242. (2013). The cortical angiome: an interconnected vascular network with nonco-
lumnar patterns of blood flow. Nat. Neurosci. 16, 889897.
Appelman, A.P.A., Vincken, K.L., van der Graaf, Y., Vlek, A.L.M., Witkamp,
T.D., Mali, W.P.T.M., and Geerlings, M.I.; SMART Study Group (2010). White Bowler, J.V. (2007). Modern concept of vascular cognitive impairment.
matter lesions and lacunar infarcts are independently and differently associ- Br. Med. Bull. 83, 291305.
ated with brain atrophy: the SMART-MR study. Cerebrovasc. Dis. 29, 2835.
Brickman, A.M., Siedlecki, K.L., Muraskin, J., Manly, J.J., Luchsinger, J.A.,
Arai, K., and Lo, E.H. (2009). An oligovascular niche: cerebral endothelial cells Yeung, L.-K., Brown, T.R., DeCarli, C., and Stern, Y. (2011). White matter
promote the survival and proliferation of oligodendrocyte precursor cells. hyperintensities and cognition: testing the reserve hypothesis. Neurobiol.
J. Neurosci. 29, 43514355. Aging 32, 15881598.

858 Neuron 80, November 20, 2013 2013 Elsevier Inc.


Neuron

Review
Brisset, M., Boutouyrie, P., Pico, F., Zhu, Y., Zureik, M., Schilling, S., Dufouil, impact of subcortical vascular and Alzheimers disease pathology. Ann. Neu-
C., Mazoyer, B., Laurent, S., Tzourio, C., and Debette, S. (2013). Large-vessel rol. 60, 677687.
correlates of cerebral small-vessel disease. Neurology 80, 662669.
Chui, H.C., Zheng, L., Reed, B.R., Vinters, H.V., and Mack, W.J. (2012).
Brix, B., Mesters, J.R., Pellerin, L., and Johren, O. (2012). Endothelial cell- Vascular risk factors and Alzheimers disease: are these risk factors for
derived nitric oxide enhances aerobic glycolysis in astrocytes via HIF- plaques and tangles or for concomitant vascular pathology that increases
1a-mediated target gene activation. J. Neurosci. 32, 97279735. the likelihood of dementia? An evidence-based review. Alzheimers Res
Ther 4, 1.
Brown, W.R., and Thore, C.R. (2011). Review: cerebral microvascular pathology
in ageing and neurodegeneration. Neuropathol. Appl. Neurobiol. 37, 5674. Cipolla, M.J. (2009). The Cerebral Circulation. Colloquium Series on Integrated
Systems Physiology: From Molecule to Function. doi: http://dx.doi.org/
Brown, W.R., Moody, D.M., Thore, C.R., Challa, V.R., and Anstrom, J.A. 10.4199/C00005ED1V01Y200912ISP002).
(2007). Vascular dementia in leukoaraiosis may be a consequence of capillary
loss not only in the lesions, but in normal-appearing white matter and cortex Claassen, J.A.H.R., Diaz-Arrastia, R., Martin-Cook, K., Levine, B.D., and
as well. J. Neurol. Sci. 257, 6266. Zhang, R. (2009). Altered cerebral hemodynamics in early Alzheimer disease:
a pilot study using transcranial Doppler. J. Alzheimers Dis. 17, 621629.
Butler, R., and Radhakrishnan, R. (2012). Dementia. Clin. Evid. (Online),
Sep 10, 2012, 127. Cohen, R.A., and Tong, X. (2010). Vascular oxidative stress: the common link
in hypertensive and diabetic vascular disease. J. Cardiovasc. Pharmacol. 55,
Butler, J.M., Kobayashi, H., and Rafii, S. (2010). Instructive role of the vascular 308316.
niche in promoting tumour growth and tissue repair by angiocrine factors. Nat.
Rev. Cancer 10, 138146. Cordonnier, C., and van der Flier, W.M. (2011). Brain microbleeds and
Alzheimers disease: innocent observation or key player? Brain 134, 335344.
Candelario-Jalil, E., Thompson, J., Taheri, S., Grossetete, M., Adair, J.C.,
Edmonds, E., Prestopnik, J., Wills, J., and Rosenberg, G.A. (2011). Matrix Craft, S. (2009). The role of metabolic disorders in Alzheimer disease and
metalloproteinases are associated with increased blood-brain barrier opening vascular dementia: two roads converged. Arch. Neurol. 66, 300305.
in vascular cognitive impairment. Stroke 42, 13451350.
Crehan, H., Hardy, J., and Pocock, J. (2013). Blockage of CR1 prevents
Carare, R.O., Hawkes, C.A., Jeffrey, M., Kalaria, R.N., and Weller, R.O. (2013). activation of rodent microglia. Neurobiol. Dis. 54, 139149.
Review: cerebral amyloid angiopathy, prion angiopathy, CADASIL and the
spectrum of protein elimination failure angiopathies (PEFA) in neurodegene- Dhaeseleer, M., Cambron, M., Vanopdenbosch, L., and De Keyser, J. (2011).
rative disease with a focus on therapy. Neuropathol. Appl. Neurobiol. 39, Vascular aspects of multiple sclerosis. Lancet Neurol. 10, 657666.
593611.
Dhaeseleer, M., Beelen, R., Fierens, Y., Cambron, M., Vanbinst, A.-M.,
Carmeliet, P., and Jain, R.K. (2011). Molecular mechanisms and clinical Verborgh, C., Demey, J., and De Keyser, J. (2013). Cerebral hypoperfusion
applications of angiogenesis. Nature 473, 298307. in multiple sclerosis is reversible and mediated by endothelin-1. Proc. Natl.
Acad. Sci. USA 110, 56545658.
Carmeliet, P., and Ruiz de Almodovar, C. (2013). VEGF ligands and receptors:
implications in neurodevelopment and neurodegeneration. Cell. Mol. Life Sci. ten Dam, V.H., van den Heuvel, D.M.J., de Craen, A.J.M., Bollen, E.L.E.M.,
70, 17631778. Murray, H.M., Westendorp, R.G.J., Blauw, G.J., and van Buchem, M.A.
(2007). Decline in total cerebral blood flow is linked with increase in periventric-
Castellano, J.M., Deane, R., Gottesdiener, A.J., Verghese, P.B., Stewart, F.R., ular but not deep white matter hyperintensities. Radiology 243, 198203.
West, T., Paoletti, A.C., Kasper, T.R., DeMattos, R.B., Zlokovic, B.V., and
Holtzman, D.M. (2012). Low-density lipoprotein receptor overexpression Dan, Y., and Poo, M.-M. (2004). Spike timing-dependent plasticity of neural
enhances the rate of brain-to-blood Ab clearance in a mouse model of circuits. Neuron 44, 2330.
b-amyloidosis. Proc. Natl. Acad. Sci. USA 109, 1550215507.
Davalos, D., and Akassoglou, K. (2012). Fibrinogen as a key regulator of
Cauli, B., and Hamel, E. (2010). Revisiting the role of neurons in neurovascular inflammation in disease. Semin. Immunopathol. 34, 4362.
coupling. Front Neuroenergetics 2, 9.
Davalos, D., Ryu, J.K., Merlini, M., Baeten, K.M., Le Moan, N., Petersen, M.A.,
Chabriat, H., Joutel, A., Dichgans, M., Tournier-Lasserve, E., and Bousser, Deerinck, T.J., Smirnoff, D.S., Bedard, C., Hakozaki, H., et al. (2012).
M.-G. (2009). Cadasil. Lancet Neurol. 8, 643653. Fibrinogen-induced perivascular microglial clustering is required for the devel-
opment of axonal damage in neuroinflammation. Nat. Commun. 3, 1227.
Charidimou, A., Gang, Q., and Werring, D.J. (2012). Sporadic cerebral amyloid
angiopathy revisited: recent insights into pathophysiology and clinical spec- De Keyser, J., Steen, C., Mostert, J.P., and Koch, M.W. (2008). Hypoperfusion
trum. J. Neurol. Neurosurg. Psychiatry 83, 124137. of the cerebral white matter in multiple sclerosis: possible mechanisms and
pathophysiological significance. J. Cereb. Blood Flow Metab. 28, 16451651.
Chen, P.L., Wang, P.Y., Sheu, W.H., Chen, Y.T., Ho, Y.P., Hu, H.H., and Hsu,
H.Y. (2006). Changes of brachial flow-mediated vasodilation in different De Reuck, J. (1971). The human periventricular arterial blood supply and the
ischemic stroke subtypes. Neurology 67, 10561058. anatomy of cerebral infarctions. Eur. Neurol. 5, 321334.

Chen, J.J., Rosas, H.D., and Salat, D.H. (2013a). The relationship between De Reuck, J.L. (2012). Histopathological stainings and definitions of vascular
cortical blood flow and sub-cortical white-matter health across the adult age disruptions in the elderly brain. Exp. Gerontol. 47, 834837.
span. PLoS ONE 8, e56733.
Deane, R., Wu, Z., Sagare, A., Davis, J., Du Yan, S., Hamm, K., Xu, F., Parisi,
Chen, N., Yang, M., Guo, J., Zhou, M., Zhu, C., and He, L. (2013b). Cerebroly- M., LaRue, B., Hu, H.W., et al. (2004). LRP/amyloid beta-peptide interaction
sin for vascular dementia. Cochrane Database Syst. Rev. 1, CD008900, http:// mediates differential brain efflux of Abeta isoforms. Neuron 43, 333344.
dx.doi.org/10.1002/14651858.CD008900.pub2.
DeCarli, C., Murphy, D.G., Tranh, M., Grady, C.L., Haxby, J.V., Gillette, J.A.,
Cheng, H.-L., Lin, C.-J., Soong, B.-W., Wang, P.-N., Chang, F.-C., Wu, Y.-T., Salerno, J.A., Gonzales-Aviles, A., Horwitz, B., Rapoport, S.I., et al. (1995).
Chou, K.-H., Lin, C.-P., Tu, P.-C., and Lee, I.-H. (2012). Impairments in cogni- The effect of white matter hyperintensity volume on brain structure, cognitive
tive function and brain connectivity in severe asymptomatic carotid stenosis. performance, and cerebral metabolism of glucose in 51 healthy adults.
Stroke 43, 25672573. Neurology 45, 20772084.

Chu, K., Jung, K.-H., Lee, S.-T., Park, H.-K., Sinn, D.-I., Kim, J.-M., Kim, D.-H., Delles, C., Michelson, G., Harazny, J., Oehmer, S., Hilgers, K.F., and
Kim, J.-H., Kim, S.-J., Song, E.-C., et al. (2008). Circulating endothelial progen- Schmieder, R.E. (2004). Impaired endothelial function of the retinal vasculature
itor cells as a new marker of endothelial dysfunction or repair in acute stroke. in hypertensive patients. Stroke 35, 12891293.
Stroke 39, 14411447.
Deschaintre, Y., Richard, F., Leys, D., and Pasquier, F. (2009). Treatment of
Chui, H.C., Zarow, C., Mack, W.J., Ellis, W.G., Zheng, L., Jagust, W.J., vascular risk factors is associated with slower decline in Alzheimer disease.
Mungas, D., Reed, B.R., Kramer, J.H., Decarli, C.C., et al. (2006). Cognitive Neurology 73, 674680.

Neuron 80, November 20, 2013 2013 Elsevier Inc. 859


Neuron

Review
Dichgans, M., and Zietemann, V. (2012). Prevention of vascular cognitive French, H.M., Reid, M., Mamontov, P., Simmons, R.A., and Grinspan, J.B.
impairment. Stroke 43, 31373146. (2009). Oxidative stress disrupts oligodendrocyte maturation. J. Neurosci.
Res. 87, 30763087.
Domercq, M., Perez-Samartin, A., Aparicio, D., Alberdi, E., Pampliega, O., and
Matute, C. (2010). P2X7 receptors mediate ischemic damage to oligodendro- Galea, I., Bechmann, I., and Perry, V.H. (2007). What is immune privilege (not)?
cytes. Glia 58, 730740. Trends Immunol. 28, 1218.

Dong, Y.-F., Kataoka, K., Toyama, K., Sueta, D., Koibuchi, N., Yamamoto, E., Gallacher, J., Bayer, A., Lowe, G., Fish, M., Pickering, J., Pedro, S., Dunstan,
Yata, K., Tomimoto, H., Ogawa, H., and Kim-Mitsuyama, S. (2011). Attenuation F., White, J., Yarnell, J., and Ben-Shlomo, Y. (2010). Is sticky blood bad for the
of brain damage and cognitive impairment by direct renin inhibition in mice brain?: Hemostatic and inflammatory systems and dementia in the Caerphilly
with chronic cerebral hypoperfusion. Hypertension 58, 635642. Prospective Study. Arterioscler. Thromb. Vasc. Biol. 30, 599604.

Drake, C.T., and Iadecola, C. (2007). The role of neuronal signaling in control- Gao, Y.-Z., Zhang, J.-J., Liu, H., Wu, G.-Y., Xiong, L., and Shu, M. (2013).
ling cerebral blood flow. Brain Lang. 102, 141152. Regional cerebral blood flow and cerebrovascular reactivity in Alzheimers
disease and vascular dementia assessed by arterial spinlabeling magnetic
Drummond, G.R., Selemidis, S., Griendling, K.K., and Sobey, C.G. (2011). resonance imaging. Curr. Neurovasc. Res. 10, 4953.
Combating oxidative stress in vascular disease: NADPH oxidases as thera-
peutic targets. Nat. Rev. Drug Discov. 10, 453471. Garcia-Alloza, M., Gregory, J., Kuchibhotla, K.V., Fine, S., Wei, Y., Ayata, C.,
Frosch, M.P., Greenberg, S.M., and Bacskai, B.J. (2011). Cerebrovascular
lesions induce transient b-amyloid deposition. Brain 134, 36973707.
Duering, M., Righart, R., Csanadi, E., Jouvent, E., Herve, D., Chabriat, H., and
Dichgans, M. (2012). Incident subcortical infarcts induce focal thinning in
Gelber, R.P., Launer, L.J., and White, L.R. (2012). The Honolulu-Asia Aging
connected cortical regions. Neurology 79, 20252028.
Study: epidemiologic and neuropathologic research on cognitive impairment.
Curr. Alzheimer Res. 9, 664672.
Dugas, J.C., Mandemakers, W., Rogers, M., Ibrahim, A., Daneman, R., and
Barres, B.A. (2008). A novel purification method for CNS projection neurons Glenner, G.G., and Wong, C.W. (1984). Alzheimers disease: initial report of the
leads to the identification of brain vascular cells as a source of trophic support purification and characterization of a novel cerebrovascular amyloid protein.
for corticospinal motor neurons. J. Neurosci. 28, 82948305. Biochem. Biophys. Res. Commun. 120, 885890.
Dyrna, F., Hanske, S., Krueger, M., and Bechmann, I. (2013). The blood-brain Gold, G., Giannakopoulos, P., Herrmann, F.R., Bouras, C., and Kovari, E.
barrier. J. Neuroimmune Pharmacol. 8, 763773. (2007). Identification of Alzheimer and vascular lesion thresholds for mixed
dementia. Brain 130, 28302836.
Elbaz, A., Vicente-Vytopilova, P., Tavernier, B., Sabia, S., Dumurgier, J.,
Mazoyer, B., Singh-Manoux, A., and Tzourio, C. (2013). Motor function in the Goldman, S.A., Nedergaard, M., and Windrem, M.S. (2012). Glial progenitor
elderly: evidence for the reserve hypothesis. Neurology 81, 417426. cell-based treatment and modeling of neurological disease. Science 338,
491495.
Ergul, A. (2011). Endothelin-1 and diabetic complications: focus on the
vasculature. Pharmacol. Res. 63, 477482. Gorelick, P.B., Scuteri, A., Black, S.E., Decarli, C., Greenberg, S.M., Iadecola,
C., Launer, L.J., Laurent, S., Lopez, O.L., Nyenhuis, D., et al.; American Heart
Esiri, M.M., Nagy, Z., Smith, M.Z., Barnetson, L., and Smith, A.D. (1999). Association Stroke Council, Council on Epidemiology and Prevention, Council
Cerebrovascular disease and threshold for dementia in the early stages of on Cardiovascular Nursing, Council on Cardiovascular Radiology and
Alzheimers disease. Lancet 354, 919920. Intervention, and Council on Cardiovascular Surgery and Anesthesia (2011).
Vascular contributions to cognitive impairment and dementia: a statement
Fancy, S.P.J., Chan, J.R., Baranzini, S.E., Franklin, R.J.M., and Rowitch, D.H. for healthcare professionals from the american heart association/american
(2011a). Myelin regeneration: a recapitulation of development? Annu. Rev. stroke association. Stroke 42, 26722713.
Neurosci. 34, 2143.
Gouw, A.A., Seewann, A., van der Flier, W.M., Barkhof, F., Rozemuller, A.M.,
Fancy, S.P.J., Harrington, E.P., Yuen, T.J., Silbereis, J.C., Zhao, C., Baranzini, Scheltens, P., and Geurts, J.J.G. (2011). Heterogeneity of small vessel
S.E., Bruce, C.C., Otero, J.J., Huang, E.J., Nusse, R., et al. (2011b). Axin2 as disease: a systematic review of MRI and histopathology correlations.
regulatory and therapeutic target in newborn brain injury and remyelination. J. Neurol. Neurosurg. Psychiatry 82, 126135.
Nat. Neurosci. 14, 10091016.
Graumann, U., Reynolds, R., Steck, A.J., and Schaeren-Wiemers, N. (2003).
Faraci, F.M., Harrington, E.P., Yuen, T.J., Silbereis, J.C., Zhao, C., Baranzini, Molecular changes in normal appearing white matter in multiple sclerosis
S.E., Bruce, C.C., Otero, J.J., Huang, E.J., Nusse, R., et al. (2011). Protecting are characteristic of neuroprotective mechanisms against hypoxic insult.
against vascular disease in brain. Am. J. Physiol. Heart Circ. Physiol. 300, Brain Pathol. 13, 554573.
H1566H1582.
Guo, S., Kim, W.J., Lok, J., Lee, S.R., Besancon, E., Luo, B.H., Stins, M.F.,
Federico, A., Di Donato, I., Bianchi, S., Di Palma, C., Taglia, I., and Dotti, M.T. Wang, X., Dedhar, S., and Lo, E.H. (2008). Neuroprotection via matrix-trophic
(2012). Hereditary cerebral small vessel diseases: a review. J. Neurol. Sci. 322, coupling between cerebral endothelial cells and neurons. Proc. Natl. Acad.
2530. Sci. USA 105, 75827587.

Hachinski, V. (2011). Stroke and Alzheimer disease: fellow travelers or partners


Fernando, M.S., Simpson, J.E., Matthews, F., Brayne, C., Lewis, C.E., Barber,
in crime? Arch. Neurol. 68, 797798.
R., Kalaria, R.N., Forster, G., Esteves, F., Wharton, S.B., et al.; MRC Cognitive
Function and Ageing Neuropathology Study Group (2006). White matter
Hachinski, V.C., and Bowler, J.V. (1993). Vascular dementia. Neurology 43,
lesions in an unselected cohort of the elderly: molecular pathology suggests
21592160, author reply 21602161.
origin from chronic hypoperfusion injury. Stroke 37, 13911398.
Hachinski, V.C., Lassen, N.A., and Marshall, J. (1974). Multi-infarct dementia.
Fields, R.D. (2010). Neuroscience. Change in the brains white matter. Science A cause of mental deterioration in the elderly. Lancet 2, 207210.
330, 768769.
Hachinski, V.C., Potter, P., and Merskey, H. (1987). Leuko-araiosis.
Fillit, H., Nash, D.T., Rundek, T., and Zuckerman, A. (2008). Cardiovascular risk Arch. Neurol. 44, 2123.
factors and dementia. Am. J. Geriatr. Pharmacother. 6, 100118.
Haight, T.J., Landau, S.M., Carmichael, O., Schwarz, C., DeCarli, C., and
Fornage, M., Debette, S., Bis, J.C., Schmidt, H., Ikram, M.A., Dufouil, C., Jagust, W.J.; Alzheimers Disease Neuroimaging Initiative (2013). Dissociable
Sigurdsson, S., Lumley, T., DeStefano, A.L., Fazekas, F., et al. (2011). effects of Alzheimer disease and white matter hyperintensities on brain meta-
Genome-wide association studies of cerebral white matter lesion burden: bolism. JAMA Neurol. 70, 10391045.
the CHARGE consortium. Ann. Neurol. 69, 928939.
Hainsworth, A.H., Brittain, J.F., and Khatun, H. (2012). Pre-clinical models of
Franklin, R.J.M., and Ffrench-Constant, C. (2008). Remyelination in the CNS: human cerebral small vessel disease: utility for clinical application. J. Neurol.
from biology to therapy. Nat. Rev. Neurosci. 9, 839855. Sci. 322, 237240.

860 Neuron 80, November 20, 2013 2013 Elsevier Inc.


Neuron

Review
Halliday, M.R., Pomara, N., Sagare, A.P., Mack, W.J., Frangione, B., and Ihara, M., and Tomimoto, H. (2011). Lessons from a mouse model character-
Zlokovic, B.V. (2013). Relationship between cyclophilin a levels and matrix izing features of vascular cognitive impairment with white matter changes.
metalloproteinase 9 activity in cerebrospinal fluid of cognitively normal apoli- J. Aging Res. 2011, 978761.
poprotein e4 carriers and blood-brain barrier breakdown. JAMA Neurol. 70,
11981200. Ihara, M., Tomimoto, H., Kinoshita, M., Oh, J., Noda, M., Wakita, H., Akiguchi,
I., and Shibasaki, H. (2001). Chronic cerebral hypoperfusion induces MMP-2
Hampel, H., Lista, S., and Khachaturian, Z.S. (2012). Development of but not MMP-9 expression in the microglia and vascular endothelium of white
biomarkers to chart all Alzheimers disease stages: the royal road to cutting matter. J. Cereb. Blood Flow Metab. 21, 828834.
the therapeutic Gordian Knot. Alzheimers Dement. 8, 312336.
Iliff, J.J., Lee, H., Yu, M., Feng, T., Logan, J., Nedergaard, M., and Benveniste,
Hanyu, H., Asano, T., Tanaka, Y., Iwamoto, T., Takasaki, M., and Abe, K. H. (2013). Brain-wide pathway for waste clearance captured by contrast-
(2002). Increased blood-brain barrier permeability in white matter lesions of enhanced MRI. J. Clin. Invest. 123, 12991309.
Binswangers disease evaluated by contrast-enhanced MRI. Dement. Geriatr.
Cogn. Disord. 14, 16. Inzitari, D., Pracucci, G., Poggesi, A., Carlucci, G., Barkhof, F., Chabriat, H.,
Erkinjuntti, T., Fazekas, F., Ferro, J.M., Hennerici, M., et al.; LADIS Study
Harris, J.J., and Attwell, D. (2012). The energetics of CNS white matter. Group (2009). Changes in white matter as determinant of global functional
J. Neurosci. 32, 356371. decline in older independent outpatients: three year follow-up of LADIS
(leukoaraiosis and disability) study cohort. BMJ 339, b2477.
Harris, J.J., Jolivet, R., and Attwell, D. (2012). Synaptic energy use and supply.
Neuron 75, 762777. Ito, K., Takeuchi, S., Hasuo, K., Morooka, M., Kubota, K., and Matsuda, H.
(2009). Cerebral blood flow on ECD SPECT in a patient with adult onset
Hayakawa, K., Pham, L.-D.D., Katusic, Z.S., Arai, K., and Lo, E.H. (2012).
Alexander disease. Clin. Nucl. Med. 34, 931933.
Astrocytic high-mobility group box 1 promotes endothelial progenitor
cell-mediated neurovascular remodeling during stroke recovery. Proc. Natl.
Jellinger, K.A. (2001). Small concomitant cerebrovascular lesions are not
Acad. Sci. USA 109, 75057510.
important for cognitive decline in severe Alzheimer disease. Arch. Neurol.
Head, D., Bugg, J.M., Goate, A.M., Fagan, A.M., Mintun, M.A., Benzinger, T., 58, 520521.
Holtzman, D.M., and Morris, J.C. (2012). Exercise Engagement as a Moderator
of the Effects of APOE Genotype on Amyloid Deposition. Arch. Neurol. 69, Jellinger, K.A. (2006). Alzheimer 100highlights in the history of Alzheimer
636643. research. J. Neural Transm. 113, 16031623.

Henskens, L.H.G., van Oostenbrugge, R.J., Kroon, A.A., de Leeuw, P.W., and Jellinger, K.A. (2013). Pathology and pathogenesis of vascular cognitive
Lodder, J. (2008). Brain microbleeds are associated with ambulatory blood impairment-a critical update. Front Aging Neurosci 5, 17.
pressure levels in a hypertensive population. Hypertension 51, 6268.
Jendroska, K., Poewe, W., Daniel, S.E., Pluess, J., Iwerssen-Schmidt, H.,
Hill, J.M., Zalos, G., Halcox, J.P.J., Schenke, W.H., Waclawiw, M.A., Quyyumi, Paulsen, J., Barthel, S., Schelosky, L., Cervos-Navarro, J., and DeArmond,
A.A., and Finkel, T. (2003). Circulating endothelial progenitor cells, vascular S.J. (1995). Ischemic stress induces deposition of amyloid beta immuno-
function, and cardiovascular risk. N. Engl. J. Med. 348, 593600. reactivity in human brain. Acta Neuropathol. 90, 461466.

Honig, L.S., Kukull, W., and Mayeux, R. (2005). Atherosclerosis and AD: Jennings, J.R., Muldoon, M.F., Ryan, C., Price, J.C., Greer, P., Sutton-Tyrrell,
analysis of data from the US National Alzheimers Coordinating Center. K., van der Veen, F.M., and Meltzer, C.C. (2005). Reduced cerebral blood flow
Neurology 64, 494500. response and compensation among patients with untreated hypertension.
Neurology 64, 13581365.
Honjo, K., Black, S.E., and Verhoeff, N.P.L.G. (2012). Alzheimers disease,
cerebrovascular disease, and the b-amyloid cascade. Can. J. Neurol. Sci. Jickling, G., Salam, A., Mohammad, A., Hussain, M.S., Scozzafava, J., Nasser,
39, 712728. A.M., Jeerakathil, T., Shuaib, A., and Camicioli, R. (2009). Circulating endo-
thelial progenitor cells and age-related white matter changes. Stroke 40,
Huang, Y., Zhang, W., Lin, L., Feng, J., Chen, F., Wei, W., Zhao, X., Guo, W., Li, 31913196.
J., Yin, W., and Li, L. (2010). Is endothelial dysfunction of cerebral small
vessel responsible for white matter lesions after chronic cerebral hypoperfu- Johnston, S.C., OMeara, E.S., Manolio, T.A., Lefkowitz, D., OLeary, D.H.,
sion in rats? J. Neurol. Sci. 299, 7280. Goldstein, S., Carlson, M.C., Fried, L.P., and Longstreth, W.T., Jr. (2004).
Cognitive impairment and decline are associated with carotid artery disease
Hulette, C., Nochlin, D., McKeel, D., Morris, J.C., Mirra, S.S., Sumi, S.M., and in patients without clinically evident cerebrovascular disease. Ann. Intern.
Heyman, A. (1997). Clinical-neuropathologic findings in multi-infarct dementia: Med. 140, 237247.
a report of six autopsied cases. Neurology 48, 668672.
Jokinen, H., Gouw, A.A., Madureira, S., Ylikoski, R., van Straaten, E.C.W., van
Hur, J., Yang, H.-M., Yoon, C.-H., Lee, C.-S., Park, K.-W., Kim, J.-H., Kim, der Flier, W.M., Barkhof, F., Scheltens, P., Fazekas, F., Schmidt, R., et al.;
T.-Y., Kim, J.-Y., Kang, H.-J., Chae, I.-H., et al. (2007). Identification of a novel LADIS Study Group (2011). Incident lacunes influence cognitive decline: the
role of T cells in postnatal vasculogenesis: characterization of endothelial LADIS study. Neurology 76, 18721878.
progenitor cell colonies. Circulation 116, 16711682.
Jonsson, M., Zetterberg, H., Rolstad, S., Edman, A., Gouw, A.A., Bjerke, M.,
Hurd, M.D., Martorell, P., Delavande, A., Mullen, K.J., and Langa, K.M. (2013).
Lind, K., Blennow, K., Pantoni, L., Inzitari, D., and Wallin, A. (2012). Low
Monetary costs of dementia in the United States. N. Engl. J. Med. 368, 1326
cerebrospinal fluid sulfatide predicts progression of white matter lesions:
1334.
The LADIS study. Dement. Geriatr. Cogn. Disord. 34, 6167.
Iadecola, C. (2004). Neurovascular regulation in the normal brain and in
Alzheimers disease. Nat. Rev. Neurosci. 5, 347360. Juma, W.M., Lira, A., Marzuk, A., Marzuk, Z., Hakim, A.M., and Thompson,
C.S. (2011). C-reactive protein expression in a rodent model of chronic
Iadecola, C. (2010). The overlap between neurodegenerative and vascular cerebral hypoperfusion. Brain Res. 1414, 8593.
factors in the pathogenesis of dementia. Acta Neuropathol. 120, 287296.
Justin, B.N., Turek, M., and Hakim, A.M. (2013). Heart disease as a risk factor
Iadecola, C., and Anrather, J. (2011). The immunology of stroke: from mecha- for dementia. Clin. Epidemiol. 5, 135145.
nisms to translation. Nat. Med. 17, 796808.
Kang, J., Lemaire, H.G., Unterbeck, A., Salbaum, J.M., Masters, C.L.,
Iadecola, C., and Davisson, R.L. (2008). Hypertension and cerebrovascular Grzeschik, K.H., Multhaup, G., Beyreuther, K., and Muller-Hill, B. (1987). The
dysfunction. Cell Metab. 7, 476484. precursor of Alzheimers disease amyloid A4 protein resembles a cell-surface
receptor. Nature 325, 733736.
Iadecola, C., Zhang, F., Niwa, K., Eckman, C., Turner, S.K., Fischer, E., Youn-
kin, S., Borchelt, D.R., Hsiao, K.K., and Carlson, G.A. (1999). SOD1 rescues Kazama, K., Anrather, J., Zhou, P., Girouard, H., Frys, K., Milner, T.A., and
cerebral endothelial dysfunction in mice overexpressing amyloid precursor Iadecola, C. (2004). Angiotensin II impairs neurovascular coupling in neocortex
protein. Nat. Neurosci. 2, 157161. through NADPH oxidase-derived radicals. Circ. Res. 95, 10191026.

Neuron 80, November 20, 2013 2013 Elsevier Inc. 861


Neuron

Review
Kearney-Schwartz, A., Rossignol, P., Bracard, S., Felblinger, J., Fay, R., Lee, S.-T., Chu, K., Jung, K.-H., Park, H.-K., Kim, D.-H., Bahn, J.-J., Kim, J.-H.,
Boivin, J.-M., Lecompte, T., Lacolley, P., Benetos, A., and Zannad, F. Oh, M.-J., Lee, S.K., Kim, M., and Roh, J.K. (2009). Reduced circulating
(2009). Vascular structure and function is correlated to cognitive performance angiogenic cells in Alzheimer disease. Neurology 72, 18581863.
and white matter hyperintensities in older hypertensive patients with subjective
memory complaints. Stroke 40, 12291236. Lee, J.H., Bacskai, B.J., and Ayata, C. (2012). Genetic animal models of
cerebral vasculopathies. Prog. Mol. Biol. Transl. Sci. 105, 2555.
Kim, J.-S., Yun, I., Choi, Y.B., Lee, K.-S., and Kim, Y.-I. (2008a). Ramipril
protects from free radical induced white matter damage in chronic hypo- Leys, D., Henon, H., Mackowiak-Cordoliani, M.-A., and Pasquier, F. (2005).
perfusion in the rat. J. Clin. Neurosci. 15, 174178. Poststroke dementia. Lancet Neurol. 4, 752759.

Kim, Y.-S., Immink, R.V., Stok, W.J., Karemaker, J.M., Secher, N.H., and Liang, K.Y., Mintun, M.A., Fagan, A.M., Goate, A.M., Bugg, J.M., Holtzman,
van Lieshout, J.J. (2008b). Dynamic cerebral autoregulatory capacity is D.M., Morris, J.C., and Head, D. (2010). Exercise and Alzheimers disease
affected early in Type 2 diabetes. Clin. Sci. 115, 255262. biomarkers in cognitively normal older adults. Ann. Neurol. 68, 311318.

Kitaguchi, H., Tomimoto, H., Ihara, M., Shibata, M., Uemura, K., Kalaria, R.N., Luckhaus, C., Flub, M.O., Wittsack, H.-J., Grass-Kapanke, B., Janner, M.,
Kihara, T., Asada-Utsugi, M., Kinoshita, A., and Takahashi, R. (2009). Chronic Khalili-Amiri, R., Friedrich, W., Supprian, T., Gaebel, W., Modder, U., and
cerebral hypoperfusion accelerates amyloid beta deposition in APPSwInd Cohnen, M. (2008). Detection of changed regional cerebral blood flow in
transgenic mice. Brain Res. 1294, 202210. mild cognitive impairment and early Alzheimers dementia by perfusion-
weighted magnetic resonance imaging. Neuroimage 40, 495503.
Kleinfeld, D., Blinder, P., Drew, P.J., Driscoll, J.D., Muller, A., Tsai, P.S., and
Shih, A.Y. (2011). A guide to delineate the logic of neurovascular signaling in Ly, J.V., Rowe, C.C., Villemagne, V.L., Zavala, J.A., Ma, H., Sahathevan, R.,
the brain. Front. Neuroenergetics 3, 19. OKeefe, G., Gong, S.J., Gunawan, R., Churilov, L., et al. (2012). Subacute
ischemic stroke is associated with focal 11C PiB positron emission tomogra-
Knottnerus, I.L.H., Ten Cate, H., Lodder, J., Kessels, F., and van Oosten- phy retention but not with global neocortical Ab deposition. Stroke 43, 1341
brugge, R.J. (2009). Endothelial dysfunction in lacunar stroke: a systematic 1346.
review. Cerebrovasc. Dis. 27, 519526.
Maillard, P., Carmichael, O., Fletcher, E., Reed, B., Mungas, D., and DeCarli, C.
Knottnerus, I.L.H., Govers-Riemslag, J.W.P., Hamulyak, K., Rouhl, R.P.W., (2012). Coevolution of white matter hyperintensities and cognition in the
Staals, J., Spronk, H.M.H., van Oerle, R., van Raak, E.P.M., Lodder, J., ten elderly. Neurology 79, 442448.
Cate, H., and van Oostenbrugge, R.J. (2010). Endothelial activation in
lacunar stroke subtypes. Stroke 41, 16171622. Makedonov, I., Black, S.E., and MacIntosh, B.J. (2013). Cerebral small vessel
disease in aging and Alzheimers disease: a comparative study using MRI and
Kobari, M., Meyer, J.S., Ichijo, M., and Oravez, W.T. (1990). Leukoaraiosis: SPECT. Eur. J. Neurol. 20, 243250.
correlation of MR and CT findings with blood flow, atrophy, and cognition.
AJNR Am. J. Neuroradiol. 11, 273281. Maki, T., Ihara, M., Fujita, Y., Nambu, T., Miyashita, K., Yamada, M., Washida,
K., Nishio, K., Ito, H., Harada, H., et al. (2011). Angiogenic and vasoprotective
effects of adrenomedullin on prevention of cognitive decline after chronic
Koike, M.A., Green, K.N., Blurton-Jones, M., and Laferla, F.M. (2010). Oligemic
cerebral hypoperfusion in mice. Stroke 42, 11221128.
hypoperfusion differentially affects tau and amyloid-beta. Am. J. Pathol. 177,
300310.
Mandell, D.M., Han, J.S., Poublanc, J., Crawley, A.P., Kassner, A., Fisher, J.A.,
and Mikulis, D.J. (2008). Selective reduction of blood flow to white matter
Koistinaho, M., Kettunen, M.I., Goldsteins, G., Keinanen, R., Salminen, A., Ort,
during hypercapnia corresponds with leukoaraiosis. Stroke 39, 19931998.
M., Bures, J., Liu, D., Kauppinen, R.A., Higgins, L.S., and Koistinaho, J. (2002).
Beta-amyloid precursor protein transgenic mice that harbor diffuse A beta
Markus, H.S. (2008). Genes, endothelial function and cerebral small vessel
deposits but do not form plaques show increased ischemic vulnerability:
disease in man. Exp. Physiol. 93, 121127.
role of inflammation. Proc. Natl. Acad. Sci. USA 99, 16101615.
Markus, H.S., Vallance, P., and Brown, M.M. (1994). Differential effect of
Kushner, E.J., Weil, B.R., MacEneaney, O.J., Morgan, R.G., Mestek, M.L., Van three cyclooxygenase inhibitors on human cerebral blood flow velocity and
Guilder, G.P., Diehl, K.J., Stauffer, B.L., and DeSouza, C.A. (2010). Human carbon dioxide reactivity. Stroke 25, 17601764.
aging and CD31+ T-cell number, migration, apoptotic susceptibility, and
telomere length. J. Appl. Physiol. 109, 17561761. Markus, H.S., Lythgoe, D.J., Ostegaard, L., OSullivan, M., and Williams, S.C.
(2000). Reduced cerebral blood flow in white matter in ischaemic leukoaraiosis
Laman, J.D., and Weller, R.O. (2013). Drainage of cells and soluble antigen from demonstrated using quantitative exogenous contrast based perfusion MRI.
the CNS to regional lymph nodes. J. Neuroimmune Pharmacol. 8, 840856. J. Neurol. Neurosurg. Psychiatry 69, 4853.
Lampron, A., Elali, A., and Rivest, S. (2013). Innate immunity in the CNS: Markus, H.S., Hunt, B., Palmer, K., Enzinger, C., Schmidt, H., and Schmidt, R.
redefining the relationship between the CNS and Its environment. Neuron (2005). Markers of endothelial and hemostatic activation and progression of
78, 214232. cerebral white matter hyperintensities: longitudinal results of the Austrian
Stroke Prevention Study. Stroke 36, 14101414.
Lanfranconi, S., and Markus, H.S. (2010). COL4A1 mutations as a monogenic
cause of cerebral small vessel disease: a systematic review. Stroke 41, e513 Marshall, R.S. (2012). Effects of altered cerebral hemodynamics on cognitive
e518. function. J. Alzheimers Dis. 32, 633642.

Launer, L.J., Petrovitch, H., Ross, G.W., Markesbery, W., and White, L.R. Marshall, R.S., Lazar, R.M., Mohr, J.P., Pile-Spellman, J., Hacein-Bey, L.,
(2008). AD brain pathology: vascular origins? Results from the HAAS autopsy Duong, D.H., Joshi, S., Chen, X., Levin, B., and Young, W.L. (1999). Higher
study. Neurobiol. Aging 29, 15871590. cerebral function and hemispheric blood flow during awake carotid artery
balloon test occlusions. J. Neurol. Neurosurg. Psychiatry 66, 734738.
Law, M., Saindane, A.M., Ge, Y., Babb, J.S., Johnson, G., Mannon, L.J.,
Herbert, J., and Grossman, R.I. (2004). Microvascular abnormality in relaps- Marshall, R.S., Festa, J.R., Cheung, Y.K., Chen, R., Pavol, M.A., Derdeyn,
ing-remitting multiple sclerosis: perfusion MR imaging findings in normal- C.P., Clarke, W.R., Videen, T.O., Grubb, R.L., Adams, H.P., et al. (2012).
appearing white matter. Radiology 231, 645652. Cerebral hemodynamics and cognitive impairment: baseline data from the
RECON trial. Neurology 78, 250255.
Lawrence, A.J., Patel, B., Morris, R.G., MacKinnon, A.D., Rich, P.M., Barrick,
T.R., and Markus, H.S. (2013). Mechanisms of cognitive impairment in Marstrand, J.R., Garde, E., Rostrup, E., Ring, P., Rosenbaum, S., Mortensen,
cerebral small vessel disease: multimodal MRI results from the St Georges E.L., and Larsson, H.B.W. (2002). Cerebral perfusion and cerebrovascular
cognition and neuroimaging in stroke (SCANS) study. PLoS ONE 8, e61014. reactivity are reduced in white matter hyperintensities. Stroke 33, 972976.

Lee, C., and Kim, Y. (2013). Complex genetic susceptibility to vascular Mastaglia, F.L., Byrnes, M.L., Johnsen, R.D., and Kakulas, B.A. (2003). Preva-
dementia and an evidence for its underlying genetic factors associated with lence of cerebral vascular amyloid-beta deposition and stroke in an aging
memory and associative learning. Gene 516, 152157. Australian population: a postmortem study. J. Clin. Neurosci. 10, 186189.

862 Neuron 80, November 20, 2013 2013 Elsevier Inc.


Neuron

Review
Masumura, M., Hata, R., Nagai, Y., and Sawada, T. (2001). Oligodendroglial Nguyen, J., Nishimura, N., Fetcho, R.N., Iadecola, C., and Schaffer, C.B.
cell death with DNA fragmentation in the white matter under chronic cerebral (2011). Occlusion of cortical ascending venules causes blood flow decreases,
hypoperfusion: comparison between normotensive and spontaneously reversals in flow direction, and vessel dilation in upstream capillaries. J. Cereb.
hypertensive rats. Neurosci. Res. 39, 401412. Blood Flow Metab. 31, 22432254.

Matsushita, K., Kuriyama, Y., Nagatsuka, K., Nakamura, M., Sawada, T., and Niedermeyer, E. (2006). Alzheimer disease: caused by primary deficiency of
Omae, T. (1994). Periventricular white matter lucency and cerebral blood the cerebral blood flow. Clin. EEG Neurosci. 37, 175177.
flow autoregulation in hypertensive patients. Hypertension 23, 565568.
Nishimura, N., Rosidi, N.L., Iadecola, C., and Schaffer, C.B. (2010). Limitations
Matthews, F.E., Arthur, A., Barnes, L.E., Bond, J., Jagger, C., Robinson, L., of collateral flow after occlusion of a single cortical penetrating arteriole.
and Brayne, C.; on behalf of the Medical Research Council Cognitive Function J. Cereb. Blood Flow Metab. 30, 19141927.
and Ageing Collaboration (2013). A two-decade comparison of prevalence
of dementia in individuals aged 65 years and older from three geographical Nitkunan, A., Lanfranconi, S., Charlton, R.A., Barrick, T.R., and Markus, H.S.
areas of England: results of the Cognitive Function and Ageing Study I and (2011). Brain atrophy and cerebral small vessel disease: a prospective
II. Lancet. http://dx.doi.org/10.1016/S0140-6736(13)61570-6. follow-up study. Stroke 42, 133138.

Matute, C., and Ransom, B.R. (2012). Roles of white matter in central nervous Niwa, K., Carlson, G.A., and Iadecola, C. (2000a). Exogenous A beta1-40
system pathophysiologies. ASN Neuro 4, 89101. reproduces cerebrovascular alterations resulting from amyloid precursor
protein overexpression in mice. J. Cereb. Blood Flow Metab. 20, 16591668.
Mawuenyega, K.G., Sigurdson, W., Ovod, V., Munsell, L., Kasten, T., Morris,
Niwa, K., Younkin, L., Ebeling, C., Turner, S.K., Westaway, D., Younkin, S.,
J.C., Yarasheski, K.E., and Bateman, R.J. (2010). Decreased clearance of
Ashe, K.H., Carlson, G.A., and Iadecola, C. (2000b). Abeta 1-40-related
CNS beta-amyloid in Alzheimers disease. Science 330, 1774.
reduction in functional hyperemia in mouse neocortex during somatosensory
activation. Proc. Natl. Acad. Sci. USA 97, 97359740.
McFarland, H.F., and Martin, R. (2007). Multiple sclerosis: a complicated
picture of autoimmunity. Nat. Immunol. 8, 913919. Niwa, K., Porter, V.A., Kazama, K., Cornfield, D., Carlson, G.A., and Iadecola,
C. (2001). A beta-peptides enhance vasoconstriction in cerebral circulation.
Melhem, E.R., Loes, D.J., Georgiades, C.S., Raymond, G.V., and Moser, H.W. Am. J. Physiol. Heart Circ. Physiol. 281, H2417H2424.
(2000). X-linked adrenoleukodystrophy: the role of contrast-enhanced MR
imaging in predicting disease progression. AJNR Am. J. Neuroradiol. 21, Niwa, K., Kazama, K., Younkin, S.G., Carlson, G.A., and Iadecola, C. (2002).
839844. Alterations in cerebral blood flow and glucose utilization in mice overexpress-
ing the amyloid precursor protein. Neurobiol. Dis. 9, 6168.
Mentis, M.J., Horwitz, B., Grady, C.L., Alexander, G.E., VanMeter, J.W.,
Maisog, J.M., Pietrini, P., Schapiro, M.B., and Rapoport, S.I. (1996). Visual Notsu, Y., Nabika, T., Bokura, H., Suyama, Y., Kobayashi, S., Yamaguchi, S.,
cortical dysfunction in Alzheimers disease evaluated with a temporally graded and Masuda, J. (2009). Evaluation of asymmetric dimethylarginine and homo-
stress test during PET. Am. J. Psychiatry 153, 3240. cysteine in microangiopathy-related cerebral damage. Am. J. Hypertens. 22,
257262.
Middleton, L.E., and Yaffe, K. (2009). Promising strategies for the prevention
of dementia. Arch. Neurol. 66, 12101215. Novak, V., Chowdhary, A., Farrar, B., Nagaraja, H., Braun, J., Kanard, R.,
Novak, P., and Slivka, A. (2003). Altered cerebral vasoregulation in hyperten-
Miklossy, J. (2003). Cerebral hypoperfusion induces cortical watershed sion and stroke. Neurology 60, 16571663.
microinfarcts which may further aggravate cognitive decline in Alzheimers
disease. Neurol. Res. 25, 605610. OSullivan, M., Lythgoe, D.J., Pereira, A.C., Summers, P.E., Jarosz, J.M.,
Williams, S.C.R., and Markus, H.S. (2002). Patterns of cerebral blood flow
Miller, A.A., Drummond, G.R., Schmidt, H.H., and Sobey, C.G. (2005). NADPH reduction in patients with ischemic leukoaraiosis. Neurology 59, 321326.
oxidase activity and function are profoundly greater in cerebral versus sys-
temic arteries. Circ. Res. 97, 10551062. Okamoto, Y., Yamamoto, T., Kalaria, R.N., Senzaki, H., Maki, T., Hase, Y.,
Kitamura, A., Washida, K., Yamada, M., Ito, H., et al. (2012). Cerebral hypo-
Miyamoto, N., Tanaka, R., Shimura, H., Watanabe, T., Mori, H., Onodera, M., perfusion accelerates cerebral amyloid angiopathy and promotes cortical
Mochizuki, H., Hattori, N., and Urabe, T. (2010). Phosphodiesterase III inhi- microinfarcts. Acta Neuropathol. 123, 381394.
bition promotes differentiation and survival of oligodendrocyte progenitors
and enhances regeneration of ischemic white matter lesions in the adult Paris, D., Humphrey, J., Quadros, A., Patel, N., Crescentini, R., Crawford, F.,
mammalian brain. J. Cereb. Blood Flow Metab. 30, 299310. and Mullan, M. (2003). Vasoactive effects of A beta in isolated human cerebro-
vessels and in a transgenic mouse model of Alzheimers disease: role of
Moser, H.W., Loes, D.J., Melhem, E.R., Raymond, G.V., Bezman, L., Cox, inflammation. Neurol. Res. 25, 642651.
C.S., and Lu, S.E. (2000). X-Linked adrenoleukodystrophy: overview and
Park, L., Anrather, J., Zhou, P., Frys, K., Pitstick, R., Younkin, S., Carlson, G.A.,
prognosis as a function of age and brain magnetic resonance imaging
and Iadecola, C. (2005). NADPH-oxidase-derived reactive oxygen species
abnormality. A study involving 372 patients. Neuropediatrics 31, 227239.
mediate the cerebrovascular dysfunction induced by the amyloid beta pep-
tide. J. Neurosci. 25, 17691777.
Moskowitz, M.A., Lo, E.H., and Iadecola, C. (2010). The science of stroke:
mechanisms in search of treatments. Neuron 67, 181198. Park, L., Anrather, J., Girouard, H., Zhou, P., and Iadecola, C. (2007).
Nox2-derived reactive oxygen species mediate neurovascular dysregulation
Musolino, P.L., Rapalino, O., Caruso, P., Caviness, V.S., and Eichler, F.S. in the aging mouse brain. J. Cereb. Blood Flow Metab. 27, 19081918.
(2012). Hypoperfusion predicts lesion progression in cerebral X-linked adreno-
leukodystrophy. Brain 135, 26762683. Park, L., Zhou, P., Pitstick, R., Capone, C., Anrather, J., Norris, E.H., Younkin,
L., Younkin, S., Carlson, G., McEwen, B.S., and Iadecola, C. (2008). Nox2-
Navaratna, D., Fan, X., Leung, W., Lok, J., Guo, S., Xing, C., Wang, X., and Lo, derived radicals contribute to neurovascular and behavioral dysfunction in
E.H. (2013). Cerebrovascular degradation of TRKB by MMP9 in the diabetic mice overexpressing the amyloid precursor protein. Proc. Natl. Acad. Sci.
brain. J. Clin. Invest. 123, 33733377. USA 105, 13471352.

Nave, K.-A. (2010a). Myelination and support of axonal integrity by glia. Nature Park, L., Wang, G., Zhou, P., Zhou, J., Pitstick, R., Previti, M.L., Younkin, L.,
468, 244252. Younkin, S.G., Van Nostrand, W.E., Cho, S., et al. (2011). Scavenger receptor
CD36 is essential for the cerebrovascular oxidative stress and neurovascular
Nave, K.-A. (2010b). Myelination and the trophic support of long axons. Nat. dysfunction induced by amyloid-beta. Proc. Natl. Acad. Sci. USA 108, 5063
Rev. Neurosci. 11, 275283. 5068.

Neuwelt, E.A., Bauer, B., Fahlke, C., Fricker, G., Iadecola, C., Janigro, D., Park, J.-H., Seo, S.W., Kim, C., Kim, G.H., Noh, H.J., Kim, S.T., Kwak, K.-C.,
Leybaert, L., Molnar, Z., ODonnell, M.E., Povlishock, J.T., et al. (2011). Yoon, U., Lee, J.M., Lee, J.W., et al. (2013a). Pathogenesis of cerebral micro-
Engaging neuroscience to advance translational research in brain barrier bleeds: In vivo imaging of amyloid and subcortical ischemic small vessel dis-
biology. Nat. Rev. Neurosci. 12, 169182. ease in 226 individuals with cognitive impairment. Ann. Neurol. 73, 584593.

Neuron 80, November 20, 2013 2013 Elsevier Inc. 863


Neuron

Review
Park, L., Zhou, J., Zhou, P., Pistick, R., El Jamal, S., Younkin, L., Pierce, J., Rosenberg, G.A., Sullivan, N., and Esiri, M.M. (2001). White matter damage is
Arreguin, A., Anrather, J., Younkin, S.G., et al. (2013b). Innate immunity recep- associated with matrix metalloproteinases in vascular dementia. Stroke 32,
tor CD36 promotes cerebral amyloid angiopathy. Proc. Natl. Acad. Sci. USA 11621168.
110, 30893094.
Rouhl, R.P.W., Damoiseaux, J.G.M.C., Lodder, J., Theunissen, R.O.M.F.I.H.,
Pendlebury, S.T., and Rothwell, P.M. (2009). Prevalence, incidence, and Knottnerus, I.L.H., Staals, J., Henskens, L.H.G., Kroon, A.A., de Leeuw,
factors associated with pre-stroke and post-stroke dementia: a systematic P.W., Tervaert, J.-W.C., and van Oostenbrugge, R.J. (2012a). Vascular inflam-
review and meta-analysis. Lancet Neurol. 8, 10061018. mation in cerebral small vessel disease. Neurobiol. Aging 33, 18001806.

Pikula, A., Boger, R.H., Beiser, A.S., Maas, R., DeCarli, C., Schwedhelm, E., Rouhl, R.P.W., Mertens, A.E.C.S., van Oostenbrugge, R.J., Damoiseaux,
Himali, J.J., Schulze, F., Au, R., Kelly-Hayes, M., et al. (2009). Association of J.G.M.C., Debrus-Palmans, L.L., Henskens, L.H.G., Kroon, A.A., de Leeuw,
plasma ADMA levels with MRI markers of vascular brain injury: Framingham P.W., Lodder, J., and Tervaert, J.-W.C. (2012b). Angiogenic T-cells and puta-
offspring study. Stroke 40, 29592964. tive endothelial progenitor cells in hypertension-related cerebral small vessel
disease. Stroke 43, 256258.
Preston, M., Gong, X., Su, W., Matsumoto, S.G., Banine, F., Winkler, C.,
Foster, S., Xing, R., Struve, J., Dean, J., et al. (2013). Digestion products of Rufa, A., Blardi, P., De Lalla, A., Cevenini, G., De Stefano, N., Zicari, E., Auteri,
the PH20 hyaluronidase inhibit remyelination. Ann. Neurol. 73, 266280. A., Federico, A., and Dotti, M.T. (2008). Plasma levels of asymmetric dimethy-
larginine in cerebral autosomal dominant arteriopathy with subcortical infarct
Pretnar-Oblak, J., Sabovic, M., Sebestjen, M., Pogacnik, T., and Zaletel, M. and leukoencephalopathy. Cerebrovasc. Dis. 26, 636640.
(2006). Influence of atorvastatin treatment on L-arginine cerebrovascular
reactivity and flow-mediated dilatation in patients with lacunar infarctions. Ruitenberg, A., den Heijer, T., Bakker, S.L., van Swieten, J.C., Koudstaal, P.J.,
Stroke 37, 25402545. Hofman, A., and Breteler, M.M. (2005). Cerebral hypoperfusion and clinical
onset of dementia: the Rotterdam Study. Ann. Neurol. 57, 789794.
Purnell, C., Gao, S., Callahan, C.M., and Hendrie, H.C. (2009). Cardiovascular
risk factors and incident Alzheimer disease: a systematic review of the litera- Sabayan, B., Jansen, S., Oleksik, A.M., van Osch, M.J.P., van Buchem, M.A.,
ture. Alzheimer Dis. Assoc. Disord. 23, 110. van Vliet, P., de Craen, A.J.M., and Westendorp, R.G.J. (2012). Cerebrovascu-
lar hemodynamics in Alzheimers disease and vascular dementia: a meta-
Quaegebeur, A., Lange, C., and Carmeliet, P. (2011). The neurovascular link in analysis of transcranial Doppler studies. Ageing Res. Rev. 11, 271277.
health and disease: molecular mechanisms and therapeutic implications.
Neuron 71, 406424. Sahathevan, R., Brodtmann, A., and Donnan, G.A. (2012). Dementia, stroke,
and vascular risk factors; a review. Int. J. Stroke 7, 6173.
Rannikmae, K., Samarasekera, N., Martnez-Gonzalez, N.A., Al-Shahi Salman,
R., and Sudlow, C.L.M. (2013). Genetics of cerebral amyloid angiopathy: Saindane, A.M., Law, M., Ge, Y., Johnson, G., Babb, J.S., and Grossman, R.I.
systematic review and meta-analysis. J. Neurol. Neurosurg. Psychiatry 84, (2007). Correlation of diffusion tensor and dynamic perfusion MR imaging
901908. metrics in normal-appearing corpus callosum: support for primary hypoperfu-
sion in multiple sclerosis. AJNR Am. J. Neuroradiol. 28, 767772.
Reimer, M.M., McQueen, J., Searcy, L., Scullion, G., Zonta, B., Desmazieres,
A., Holland, P.R., Smith, J., Gliddon, C., Wood, E.R., et al. (2011). Rapid Schaapsmeerders, P., Maaijwee, N.A.M., van Dijk, E.J., Rutten-Jacobs,
disruption of axon-glial integrity in response to mild cerebral hypoperfusion. L.C.A., Arntz, R.M., Schoonderwaldt, H.C., Dorresteijn, L.D.A., Kessels,
J. Neurosci. 31, 1818518194. R.P.C., and de Leeuw, F.-E. (2013). Long-term cognitive impairment after
first-ever ischemic stroke in young adults. Stroke 44, 16211628.
Reis, J.P., Loria, C.M., Launer, L.J., Sidney, S., Liu, K., Jacobs, D.R., Jr., Zhu,
N., Lloyd-Jones, D.M., He, K., and Yaffe, K. (2013). Cardiovascular health Schmidt, H., Freudenberger, P., Seiler, S., and Schmidt, R. (2012). Genetics of
through young adulthood and cognitive functioning in midlife. Ann. Neurol. subcortical vascular dementia. Exp. Gerontol. 47, 873877.
73, 170179.
Schneider, J.A., Arvanitakis, Z., Bang, W., and Bennett, D.A. (2007a). Mixed
Richard, E., Gouw, A.A., Scheltens, P., and van Gool, W.A. (2010). Vascular brain pathologies account for most dementia cases in community-dwelling
care in patients with Alzheimer disease with cerebrovascular lesions slows older persons. Neurology 69, 21972204.
progression of white matter lesions on MRI: the evaluation of vascular care
in Alzheimers disease (EVA) study. Stroke 41, 554556. Schneider, J.A., Boyle, P.A., Arvanitakis, Z., Bienias, J.L., and Bennett, D.A.
(2007b). Subcortical infarcts, Alzheimers disease pathology, and memory
Richardson, W.D., Young, K.M., Tripathi, R.B., and McKenzie, I. (2011). function in older persons. Ann. Neurol. 62, 5966.
NG2-glia as multipotent neural stem cells: fact or fantasy? Neuron 70, 661673.
Schneider, J.A., Arvanitakis, Z., Leurgans, S.E., and Bennett, D.A. (2009). The
Richardson, K., Stephan, B.C.M., Ince, P.G., Brayne, C., Matthews, F.E., and neuropathology of probable Alzheimer disease and mild cognitive impairment.
Esiri, M.M. (2012). The neuropathology of vascular disease in the Medical Ann. Neurol. 66, 200208.
Research Council Cognitive Function and Ageing Study (MRC CFAS). Curr.
Alzheimer Res. 9, 687696. Schreiber, S., Bueche, C.Z., Garz, C., and Braun, H. (2013). Blood brain
barrier breakdown as the starting point of cerebral small vessel disease? -
Robbins, E.M., Betensky, R.A., Domnitz, S.B., Purcell, S.M., Garcia-Alloza, M., New insights from a rat model. Exp. Transl. Stroke Med. 5, 4.
Greenberg, C., Rebeck, G.W., Hyman, B.T., Greenberg, S.M., Frosch, M.P.,
and Bacskai, B.J. (2006). Kinetics of cerebral amyloid angiopathy progression Schrijvers, E.M.C., Schurmann, B., Koudstaal, P.J., van den Bussche, H.,
in a transgenic mouse model of Alzheimer disease. J. Neurosci. 26, 365371. Van Duijn, C.M., Hentschel, F., Heun, R., Hofman, A., Jessen, F., Kolsch, H.,
et al. (2012). Genome-wide association study of vascular dementia. Stroke
Rodrigue, K.M., Rieck, J.R., Kennedy, K.M., Devous, M.D., Diaz-Arrastia, R., 43, 315319.
and Park, D.C. (2013). Risk Factors for b-Amyloid Deposition in Healthy Aging:
Vascular and Genetic Effects. JAMA Neurol. 70, 600606. Scuteri, A., Nilsson, P.M., Tzourio, C., Redon, J., and Laurent, S. (2011). Micro-
vascular brain damage with aging and hypertension: pathophysiological
Roher, A.E., Esh, C., Rahman, A., Kokjohn, T.A., and Beach, T.G. (2004). consideration and clinical implications. J. Hypertens. 29, 14691477.
Atherosclerosis of cerebral arteries in Alzheimer disease. Stroke 35 (Suppl 1 ),
26232627. Seo, S.W., Lee, J.M., Im, K., Park, J.-S., Kim, S.-H., Kim, S.T., Ahn, H.-J., Chin,
J., Cheong, H.-K., Weiner, M.W., and Na, D.L. (2012). Cortical thinning related
Roman, G.C., Tatemichi, T.K., Erkinjuntti, T., Cummings, J.L., Masdeu, J.C., to periventricular and deep white matter hyperintensities. Neurobiol. Aging 33,
Garcia, J.H., Amaducci, L., Orgogozo, J.M., Brun, A., Hofman, A., and Moody, 11561167.
D. (1993). Vascular dementia: diagnostic criteria for research studies. Report
of the NINDS-AIREN International Workshop. Neurology 43, 250260. Seo, J.H., Miyamoto, N., Hayakawa, K., Pham, L.-D.D., Maki, T., Ayata, C.,
Kim, K.-W., Lo, E.H., and Arai, K. (2013). Oligodendrocyte precursors induce
Rosenberg, G.A. (2012). Neurological diseases in relation to the blood-brain early blood-brain barrier opening after white matter injury. J. Clin. Invest.
barrier. J. Cereb. Blood Flow Metab. 32, 11391151. 123, 782786.

864 Neuron 80, November 20, 2013 2013 Elsevier Inc.


Neuron

Review
Sharp, S.I., Aarsland, D., Day, S., Snnesyn, H., and Ballard, C.; Alzheimers Sun, Y.-W., Qin, L.-D., Zhou, Y., Xu, Q., Qian, L.-J., Tao, J., and Xu, J.-R.
Society Vascular Dementia Systematic Review Group (2011). Hypertension (2011). Abnormal functional connectivity in patients with vascular cognitive
is a potential risk factor for vascular dementia: systematic review. Int. J. Ger- impairment, no dementia: a resting-state functional magnetic resonance
iatr. Psychiatry 26, 661669. imaging study. Behav. Brain Res. 223, 388394.

Shen, Q., Goderie, S.K., Jin, L., Karanth, N., Sun, Y., Abramova, N., Vincent, P., Suter, O.-C., Sunthorn, T., Kraftsik, R., Straubel, J., Darekar, P., Khalili, K., and
Pumiglia, K., and Temple, S. (2004). Endothelial cells stimulate self-renewal Miklossy, J. (2002). Cerebral hypoperfusion generates cortical watershed
and expand neurogenesis of neural stem cells. Science 304, 13381340. microinfarcts in Alzheimer disease. Stroke 33, 19861992.

Sherman, L.S., and Back, S.A. (2008). A GAG reflex prevents repair of the Taheri, S., Gasparovic, C., Huisa, B.N., Adair, J.C., Edmonds, E., Prestopnik,
damaged CNS. Trends Neurosci. 31, 4452. J., Grossetete, M., Shah, N.J., Wills, J., Qualls, C., and Rosenberg, G.A. (2011).
Blood-brain barrier permeability abnormalities in vascular cognitive impair-
Shibata, M., Yamada, S., Kumar, S.R., Calero, M., Bading, J., Frangione, B., ment. Stroke 42, 21582163.
Holtzman, D.M., Miller, C.A., Strickland, D.K., Ghiso, J., and Zlokovic, B.V.
(2000). Clearance of Alzheimers amyloid-ss(1-40) peptide from brain by LDL Tanaka, M., Fukuyama, H., Yamauchi, H., Narita, M., Nabatame, H., Yokode,
receptor-related protein-1 at the blood-brain barrier. J. Clin. Invest. 106, M., Fujimoto, N., Kita, T., and Murakami, M. (2002). Regional cerebral blood
14891499. flow abnormalities in nondemented patients with memory impairment.
J. Neuroimaging 12, 112118.
Shibata, H., Nabika, T., Moriyama, H., Masuda, J., and Kobayashi, S. (2004).
Correlation of NO metabolites and 8-iso-prostaglandin F2a with periventricular Tarumi, T., Shah, F., Tanaka, H., and Haley, A.P. (2011). Association between
hyperintensity severity. Arterioscler. Thromb. Vasc. Biol. 24, 16591663. central elastic artery stiffness and cerebral perfusion in deep subcortical gray
and white matter. Am. J. Hypertens. 24, 11081113.
Shih, A.Y., Blinder, P., Tsai, P.S., Friedman, B., Stanley, G., Lyden, P.D., and
Kleinfeld, D. (2013). The smallest stroke: occlusion of one penetrating vessel Tatemichi, T.K., Desmond, D.W., Prohovnik, I., and Eidelberg, D. (1995).
leads to infarction and a cognitive deficit. Nat. Neurosci. 16, 5563. Dementia associated with bilateral carotid occlusions: neuropsychological
and haemodynamic course after extracranial to intracranial bypass surgery.
Simpson, J.E., Fernando, M.S., Clark, L., Ince, P.G., Matthews, F., Forster, G., J. Neurol. Neurosurg. Psychiatry 58, 633636.
OBrien, J.T., Barber, R., Kalaria, R.N., Brayne, C., et al.; MRC Cognitive Func-
tion and Ageing Neuropathology Study Group (2007). White matter lesions in Tesco, G., Koh, Y.H., Kang, E.L., Cameron, A.N., Das, S., Sena-Esteves, M., Hil-
an unselected cohort of the elderly: astrocytic, microglial and oligodendrocyte tunen, M., Yang, S.-H., Zhong, Z., Shen, Y., et al. (2007). Depletion of GGA3 sta-
precursor cell responses. Neuropathol. Appl. Neurobiol. 33, 410419. bilizes BACE and enhances beta-secretase activity. Neuron 54, 721737.

Smith, E.E., Schneider, J.A., Wardlaw, J.M., and Greenberg, S.M. (2012). Ce- Thal, D.R., Grinberg, L.T., and Attems, J. (2012). Vascular dementia: different
rebral microinfarcts: the invisible lesions. Lancet Neurol. 11, 272282. forms of vessel disorders contribute to the development of dementia in the
elderly brain. Exp. Gerontol. 47, 816824.
Snapyan, M., Lemasson, M., Brill, M.S., Blais, M., Massouh, M., Ninkovic, J.,
Thomas, T., Thomas, G., McLendon, C., Sutton, T., and Mullan, M. (1996).
Gravel, C., Berthod, F., Gotz, M., Barker, P.A., et al. (2009). Vasculature guides
b-Amyloid-mediated vasoactivity and vascular endothelial damage. Nature
migrating neuronal precursors in the adult mammalian forebrain via brain-
380, 168171.
derived neurotrophic factor signaling. J. Neurosci. 29, 41724188.
Thompson, C.S., and Hakim, A.M. (2009). Living beyond our physiological
Snowdon, D.A., Greiner, L.H., Mortimer, J.A., Riley, K.P., Greiner, P.A., and
means: small vessel disease of the brain is an expression of a systemic failure
Markesbery, W.R. (1997). Brain infarction and the clinical expression of
in arteriolar function: a unifying hypothesis. Stroke 40, e322e330.
Alzheimer disease. The Nun Study. JAMA 277, 813817.
Toledo, J.B., Arnold, S.E., Raible, K., Brettschneider, J., Xie, S.X., Grossman,
Sorond, F.A., Kiely, D.K., Galica, A., Moscufo, N., Serrador, J.M., Iloputaife, I.,
M., Monsell, S.E., Kukull, W.A., and Trojanowski, J.Q. (2013). Contribution of
Egorova, S., DellOglio, E., Meier, D.S., Newton, E., et al. (2011). Neurovascular
cerebrovascular disease in autopsy confirmed neurodegenerative disease
coupling is impaired in slow walkers: the MOBILIZE Boston Study. Ann.
cases in the National Alzheimers Coordinating Centre. Brain 136, 26972706.
Neurol. 70, 213220.
Tomimoto, H., Akiguchi, I., Suenaga, T., Nishimura, M., Wakita, H., Nakamura,
Soros, P., Whitehead, S., Spence, J.D., and Hachinski, V. (2013). Antihyper- S., and Kimura, J. (1996). Alterations of the blood-brain barrier and glial cells in
tensive treatment can prevent stroke and cognitive decline. Nat. Rev. Neurol. white-matter lesions in cerebrovascular and Alzheimers disease patients.
9, 174178. Stroke 27, 20692074.
Stefansdottir, H., Arnar, D.O., Aspelund, T., Sigurdsson, S., Jonsdottir, M.K., Tomlinson, B.E., Blessed, G., and Roth, M. (1970). Observations on the brains
Hjaltason, H., Launer, L.J., and Gudnason, V. (2013). Atrial fibrillation is of demented old people. J. Neurol. Sci. 11, 205242.
associated with reduced brain volume and cognitive function independent of
cerebral infarcts. Stroke 44, 10201025. Tong, X.-K., Lecrux, C., Rosa-Neto, P., and Hamel, E. (2012). Age-dependent
rescue by simvastatin of Alzheimers disease cerebrovascular and memory
Stern, Y. (2012). Cognitive reserve in ageing and Alzheimers disease. Lancet deficits. J. Neurosci. 32, 47054715.
Neurol. 11, 10061012.
Topakian, R., Barrick, T.R., Howe, F.A., and Markus, H.S. (2010). Blood-brain
Stevenson, S.F., Doubal, F.N., Shuler, K., and Wardlaw, J.M. (2010). A system- barrier permeability is increased in normal-appearing white matter in patients
atic review of dynamic cerebral and peripheral endothelial function in lacunar with lacunar stroke and leucoaraiosis. J. Neurol. Neurosurg. Psychiatry 81,
stroke versus controls. Stroke 41, e434e442. 192197.

Stone, D.B., and Tesche, C.D. (2013). Topological dynamics in spike-timing Tran, E.H., Hoekstra, K., van Rooijen, N., Dijkstra, C.D., and Owens, T. (1998).
dependent plastic model neural networks. Front. Neural Circuits 7, 70. Immune invasion of the central nervous system parenchyma and experimental
allergic encephalomyelitis, but not leukocyte extravasation from blood, are
Stys, P.K., Waxman, S.G., and Ransom, B.R. (1992). Ionic mechanisms of prevented in macrophage-depleted mice. J. Immunol. 161, 37673775.
anoxic injury in mammalian CNS white matter: role of Na+ channels and
Na(+)-Ca2+ exchanger. J. Neurosci. 12, 430439. Trapp, B.D., and Stys, P.K. (2009). Virtual hypoxia and chronic necrosis of
demyelinated axons in multiple sclerosis. Lancet Neurol. 8, 280291.
al Suhaili, A.R., Hertecant, J., and Sztriha, L. (1994). Adrenoleukodystrophy:
cortical hypoperfusion demonstrated with 99mTc-HMPAO SPECT. J. Child Tullberg, M., Fletcher, E., DeCarli, C., Mungas, D., Reed, B.R., Harvey, D.J.,
Neurol. 9, 284286. Weiner, M.W., Chui, H.C., and Jagust, W.J. (2004). White matter lesions impair
frontal lobe function regardless of their location. Neurology 63, 246253.
Sun, X., He, G., Qing, H., Zhou, W., Dobie, F., Cai, F., Staufenbiel, M., Huang,
L.E., and Song, W. (2006). Hypoxia facilitates Alzheimers disease patho- Ueno, M., Tomimoto, H., Akiguchi, I., Wakita, H., and Sakamoto, H. (2002).
genesis by up-regulating BACE1 gene expression. Proc. Natl. Acad. Sci. Blood-brain barrier disruption in white matter lesions in a rat model of chronic
USA 103, 1872718732. cerebral hypoperfusion. J. Cereb. Blood Flow Metab. 22, 97104.

Neuron 80, November 20, 2013 2013 Elsevier Inc. 865


Neuron

Review
Ueno, Y., Zhang, N., Miyamoto, N., Tanaka, R., Hattori, N., and Urabe, T. Wen, Y., Yang, S.-H., Liu, R., Perez, E.J., Brun-Zinkernagel, A.M., Koulen, P.,
(2009). Edaravone attenuates white matter lesions through endothelial protec- and Simpkins, J.W. (2007). Cdk5 is involved in NFT-like tauopathy induced
tion in a rat chronic hypoperfusion model. Neuroscience 162, 317327. by transient cerebral ischemia in female rats. Biochim. Biophys. Acta 1772,
473483.
van Norden, A.G.W., van Uden, I.W.M., de Laat, K.F., Gons, R.A.R., Kessels,
R.P.C., van Dijk, E.J., and de Leeuw, F.-E. (2013). Cerebral microbleeds are Werner, N., Kosiol, S., Schiegl, T., Ahlers, P., Walenta, K., Link, A., Bohm, M.,
related to subjective cognitive failures: the RUN DMC study. Neurobiol. Aging and Nickenig, G. (2005). Circulating endothelial progenitor cells and cardio-
34, 22252230. vascular outcomes. N. Engl. J. Med. 353, 9991007.

Verdelho, A., Madureira, S., Ferro, J.M., Baezner, H., Blahak, C., Poggesi, A., Wilkins, A., Majed, H., Layfield, R., Compston, A., and Chandran, S. (2003).
Hennerici, M., Pantoni, L., Fazekas, F., Scheltens, P., et al.; LADIS Study Oligodendrocytes promote neuronal survival and axonal length by distinct
(2012). Physical activity prevents progression for cognitive impairment and intracellular mechanisms: a novel role for oligodendrocyte-derived glial cell
vascular dementia: results from the LADIS (Leukoaraiosis and Disability) study. line-derived neurotrophic factor. J. Neurosci. 23, 49674974.
Stroke 43, 33313335.
Wolburg, H., Noell, S., Mack, A., Wolburg-Buchholz, K., and Fallier-Becker, P.
Verghese, P.B., Castellano, J.M., and Holtzman, D.M. (2011). Apolipoprotein E (2009). Brain endothelial cells and the glio-vascular complex. Cell Tissue Res.
in Alzheimers disease and other neurological disorders. Lancet Neurol. 10, 335, 7596.
241252.
World Health Organization (2012). Dementia A Public Health Priority.
Vernooij, M.W., van der Lugt, A., Ikram, M.A., Wielopolski, P.A., Vrooman, (World Health Organization).
H.A., Hofman, A., Krestin, G.P., and Breteler, M.M.B. (2008). Total cerebral
blood flow and total brain perfusion in the general population: the Rotterdam Xu, H., Stamova, B., Jickling, G., Tian, Y., Zhan, X., Ander, B.P., Liu, D., Turner,
Scan Study. J. Cereb. Blood Flow Metab. 28, 412419. R., Rosand, J., Goldstein, L.B., et al. (2010). Distinctive RNA expression pro-
files in blood associated with white matter hyperintensities in brain. Stroke
Wakita, H., Ruetzler, C., Illoh, K.O., Chen, Y., Takanohashi, A., Spatz, M., and 41, 27442749.
Hallenbeck, J.M. (2008). Mucosal tolerization to E-selectin protects against
memory dysfunction and white matter damage in a vascular cognitive impair- Yamamoto, Y., Craggs, L.J.L., Watanabe, A., Booth, T., Attems, J., Low,
ment model. J. Cereb. Blood Flow Metab. 28, 341353. R.W.C., Oakley, A.E., and Kalaria, R.N. (2013). Brain microvascular accumu-
lation and distribution of the NOTCH3 ectodomain and granular osmiophilic
Wang, J., Zhang, H.Y., and Tang, X.C. (2010). Huperzine a improves chronic material in CADASIL. J. Neuropathol. Exp. Neurol. 72, 416431.
inflammation and cognitive decline in rats with cerebral hypoperfusion.
J. Neurosci. Res. 88, 807815. Yang, Y., and Rosenberg, G.A. (2011). Blood-brain barrier breakdown in acute
and chronic cerebrovascular disease. Stroke 42, 33233328.
Wang, B.W., Lu, E., Mackenzie, I.R.A., Assaly, M., Jacova, C., Lee, P.E.,
Yao, H., Sadoshima, S., Ibayashi, S., Kuwabara, Y., Ichiya, Y., and Fujishima,
Beattie, B.L., and Hsiung, G.Y.R. (2012). Multiple pathologies are common in
M. (1992). Leukoaraiosis and dementia in hypertensive patients. Stroke 23,
Alzheimer patients in clinical trials. Can. J. Neurol. Sci. 39, 592599.
16731677.
Wardlaw, J.M., Farrall, A., Armitage, P.A., Carpenter, T., Chappell, F., Doubal,
Yarchoan, M., Xie, S.X., Kling, M.A., Toledo, J.B., Wolk, D.A., Lee, E.B.,
F., Chowdhury, D., Cvoro, V., and Dennis, M.S. (2008). Changes in background
Van Deerlin, V., Lee, V.M.-Y., Trojanowski, J.Q., and Arnold, S.E. (2012).
blood-brain barrier integrity between lacunar and cortical ischemic stroke
Cerebrovascular atherosclerosis correlates with Alzheimer pathology in
subtypes. Stroke 39, 13271332.
neurodegenerative dementias. Brain 135, 37493756.
Wardlaw, J.M., Doubal, F., Armitage, P., Chappell, F., Carpenter, T., Munoz
Yates, K.F., Sweat, V., Yau, P.L., Turchiano, M.M., and Convit, A. (2012).
Maniega, S., Farrall, A., Sudlow, C., Dennis, M., and Dhillon, B. (2009). Lacunar
Impact of metabolic syndrome on cognition and brain: a selected review of
stroke is associated with diffuse blood-brain barrier dysfunction. Ann. Neurol.
the literature. Arterioscler. Thromb. Vasc. Biol. 32, 20602067.
65, 194202.
Yoshida, T., Tanaka, M., and Okamoto, K. (2002). Immunoglobulin G induces
Wardlaw, J.M., Smith, E.E., Biessels, G.J., Cordonnier, C., Fazekas, F., microglial superoxide production. Neurol. Res. 24, 361364.
Frayne, R., Lindley, R.I., OBrien, J.T., Barkhof, F., Benavente, O.R., et al.;
STandards for ReportIng Vascular changes on nEuroimaging (STRIVE v1) Yoshizaki, K., Adachi, K., Kataoka, S., Watanabe, A., Tabira, T., Takahashi, K.,
(2013a). Neuroimaging standards for research into small vessel disease and and Wakita, H. (2008). Chronic cerebral hypoperfusion induced by right
its contribution to ageing and neurodegeneration. Lancet Neurol. 12, 822838. unilateral common carotid artery occlusion causes delayed white matter
lesions and cognitive impairment in adult mice. Exp. Neurol. 210, 585591.
Wardlaw, J.M., Smith, C., and Dichgans, M. (2013b). Mechanisms of sporadic
cerebral small vessel disease: insights from neuroimaging. Lancet Neurol. 12, Yu, L., Boyle, P., Schneider, J.A., Segawa, E., Wilson, R.S., Leurgans, S., and
483497. Bennett, D.A. (2013). APOE 4, Alzheimers Disease Pathology, Cerebrovascu-
lar Disease, and Cognitive Change Over the Years Prior to Death. Psychol.
Washida, K., Ihara, M., Nishio, K., Fujita, Y., Maki, T., Yamada, M., Takahashi, Aging. http://dx.doi.org/10.1037/a0031642.
J., Wu, X., Kihara, T., Ito, H., et al. (2010). Nonhypotensive dose of telmisartan
attenuates cognitive impairment partially due to peroxisome proliferator- Zhang, F., Eckman, C., Younkin, S., Hsiao, K.K., and Iadecola, C. (1997).
activated receptor-gamma activation in mice with chronic cerebral hypo- Increased susceptibility to ischemic brain damage in transgenic mice overex-
perfusion. Stroke 41, 17981806. pressing the amyloid precursor protein. J. Neurosci. 17, 76557661.

Webb, A.J.S., Simoni, M., Mazzucco, S., Kuker, W., Schulz, U., and Rothwell, Zhang, G., Zhao, Z., Gao, L., Deng, J., Wang, B., Xu, D., Liu, B., Qu, Y., Yu, J.,
P.M. (2012). Increased cerebral arterial pulsatility in patients with leukoaraio- Li, J., and Gao, G. (2011). Gypenoside attenuates white matter lesions induced
sis: arterial stiffness enhances transmission of aortic pulsatility. Stroke 43, by chronic cerebral hypoperfusion in rats. Pharmacol. Biochem. Behav. 99,
26312636. 4251.

Weil, B.R., Kushner, E.J., Diehl, K.J., Greiner, J.J., Stauffer, B.L., and Desouza, Zieren, N., Duering, M., Peters, N., Reyes, S., Jouvent, E., Herve, D.,
C.A. (2011). CD31+ T cells, endothelial function and cardiovascular risk. Heart Gschwendtner, A., Mewald, Y., Opherk, C., Chabriat, H., and Dichgans, M.
Lung Circ. 20, 659662. (2013). Education modifies the relation of vascular pathology to cognitive func-
tion: cognitive reserve in cerebral autosomal dominant arteriopathy with
Wen, Y., Onyewuchi, O., Yang, S., Liu, R., and Simpkins, J.W. (2004a). subcortical infarcts and leukoencephalopathy. Neurobiol. Aging 34, 400407.
Increased beta-secretase activity and expression in rats following transient
cerebral ischemia. Brain Res. 1009, 18. Zlokovic, B.V. (2008). The blood-brain barrier in health and chronic neurode-
generative disorders. Neuron 57, 178201.
Wen, Y., Yang, S., Liu, R., and Simpkins, J.W. (2004b). Transient cerebral
ischemia induces site-specific hyperphosphorylation of tau protein. Brain Zlokovic, B.V. (2011). Neurovascular pathways to neurodegeneration in Alz-
Res. 1022, 3038. heimers disease and other disorders. Nat. Rev. Neurosci. 12, 723738.

866 Neuron 80, November 20, 2013 2013 Elsevier Inc.

You might also like