Você está na página 1de 15

Brain, Behavior, and Immunity xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

Brain, Behavior, and Immunity


journal homepage: www.elsevier.com/locate/ybrbi

Microglial dysfunction connects depression and Alzheimer’s disease


Luís Eduardo Santos a, Danielle Beckman b, Sergio T. Ferreira a,b,⇑
a
Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
b
Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil

a r t i c l e i n f o a b s t r a c t

Article history: Alzheimer’s disease (AD) and major depressive disorder (MDD) are highly prevalent neuropsychiatric
Received 5 July 2015 conditions with intriguing epidemiological overlaps. Depressed patients are at increased risk of develop-
Received in revised form 19 November 2015 ing late-onset AD, and around one in four AD patients are co-diagnosed with MDD. Microglia are the main
Accepted 19 November 2015
cellular effectors of innate immunity in the brain, and their activation is central to neuroinflammation – a
Available online xxxx
ubiquitous process in brain pathology, thought to be a causal factor of both AD and MDD. Microglia serve
several physiological functions, including roles in synaptic plasticity and neurogenesis, which may be dis-
Keywords:
rupted in neuroinflammation. Following early work on the ‘sickness behavior’ of humans and other ani-
Alzheimer’s disease
Microglia
mals, microglia-derived inflammatory cytokines have been shown to produce depressive-like symptoms
Major depressive disorder when administered exogenously or released in response to infection. MDD patients consistently show
Neuroinflammation increased circulating levels of pro-inflammatory cytokines, and anti-inflammatory drugs show promise
for treating depression. Activated microglia are abundant in the AD brain, and concentrate around senile
plaques, hallmark lesions composed of aggregated amyloid-b peptide (Ab). The Ab burden in affected
brains is regulated largely by microglial clearance, and the complex activation state of microglia may
be crucial for AD progression. Intriguingly, recent reports have linked soluble Ab oligomers, toxins that
accumulate in AD brains and are thought to cause memory impairment, to increased brain cytokine pro-
duction and depressive-like behavior in mice. Here, we review recent findings supporting the inflamma-
tory hypotheses of AD and MDD, focusing on microglia as a common player and therapeutic target linking
these devastating disorders.
Ó 2015 Published by Elsevier Inc.

1. Introduction In the following sections, we examine epidemiological data


linking depression and AD, and review mechanistic hypotheses
Although Alzheimer’s disease (AD) is often regarded solely as a that may explain this connection. We focus on neuroinflammation
disease of memory, nearly all patients display behavioral and psy- and microglial activation, common denominators to a number of
chological symptoms that accompany the characteristic memory/ brain-related pathologies thought to be causal factors in both AD
cognitive impairment (Lyketsos et al., 2011, 2002). Those symp- and MDD.
toms have significant clinical and societal relevance – they increase
the burden on patients and caregivers, precipitate institutionaliza- 1.1. Epidemiological link between AD and depression
tion, and ramp up the costs of managing AD (Allegri et al., 2006;
Hurt et al., 2008; Kales et al., 2015). Early assessments of the prevalence of depression in AD
Depression is consistently reported as one of the most prevalent patients date from the 1980s, and showed widely variable results,
co-morbidities in AD (Lyketsos and Olin, 2002; Steinberg et al., ranging from zero to 86% (Knesevich et al., 1983; Merriam et al.,
2008). Its incidence scales with AD progression (Steinberg et al., 1988). But after 1990, more consistent epidemiological data began
2008), and a diagnosis of major depressive disorder (MDD) predicts to emerge. Burns and colleagues published a notable case-register
a poorer cognitive outcome in later life, with increased risk of study of AD patients in the UK, finding that 20% of them were
developing AD (Ownby et al., 2006). aggressive, 41% apathetic and 24% depressed (Burns et al., 1990a,
b).
A decade later, cross-sectional analysis of data from the much
⇑ Corresponding author at: Institute of Biophysics Carlos Chagas Filho, Federal larger, US-based ‘Cache County Study on Memory, Health and
University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil. Aging’ found similar results. This longitudinal study enrolled
E-mail address: ferreira@bioqmed.ufrj.br (S.T. Ferreira). 5092 community-dwellers 65 and older, and found that 20% of

http://dx.doi.org/10.1016/j.bbi.2015.11.011
0889-1591/Ó 2015 Published by Elsevier Inc.

Please cite this article in press as: Santos, L.E., et al. Microglial dysfunction connects depression and Alzheimer’s disease. Brain Behav. Immun. (2015),
http://dx.doi.org/10.1016/j.bbi.2015.11.011
2 L.E. Santos et al. / Brain, Behavior, and Immunity xxx (2015) xxx–xxx

those diagnosed with AD also had depressive symptoms. Non- sample was limited, they further noted that all MCI patients meet-
demented elderly in the same cohort showed a third of this preva- ing DSM-IV criteria for MDD converted to AD within the 3-year
lence (Lyketsos et al., 2000). Working on a subset of data from the follow-up period (Modrego and Ferrández, 2004).
same study (408 dementia cases, 63% AD) Steinberg and colleagues
later concluded that the cumulative prevalence of depression in
1.3. Depression: Risk factor or prodromal symptom of AD?
dementia, over a 5-year period, is 77% (Steinberg et al., 2008). Sev-
eral different cohort studies have since helped establish depression
Even when it manifests before MCI or other cognitive symp-
as one the most frequent behavioral symptoms in AD (Benoit et al.,
toms, depression appears to predict the development of AD. In
2012; Porta-Etessam et al., 2011).
2003, Green and colleagues reported an analysis of data from the
Despite general consensus on the positive correlation between
Multi-institutional Research in Alzheimer’s Genetic Epidemiology
dementia and depression, it should be noted that results from dif-
(MIRAGE) study, a cross-sectional case-control study of 4046 sub-
ferent studies have varied considerably, and higher point preva-
jects—about half probable AD cases, half non-affected family mem-
lence values, of up to 50%, were recently reported (Chi et al.,
bers. That study showed a significant association between
2014; Lee and Lyketsos, 2003; Starkstein et al., 2005; Zubenko
depressive symptoms and the risk for development of AD later in
et al., 2003). Although differences among sampled populations
life. The association was stronger when symptoms appeared up
likely account for a significant part of this variability (Lee and
to 1 year before the onset of AD, but was significant even when
Lyketsos, 2003), the intrinsic difficulty of diagnosing depression
they first occurred more than 25 years before cognitive deficits
in old age may also be an important contributing factor, especially
(Green et al., 2003). These results were later included in a large
when dementia is comorbid (Nelson, 2001). Confounding cognitive
meta-analysis by Ownby and colleagues, which corroborated the
deficits, reduced perception of symptoms and impaired verbal
notion that a history of depression increases the risk of developing
communication complicate diagnosis in older patients (Fiske
AD (Ownby et al., 2006).
et al., 2009). Depression in AD tends to present differently from
Other studies, however, point to depression not as a risk factor,
depression in non-demented elderly, which in turn differs from
but as an early symptom of the neurodegenerative process of AD,
that of younger patients (Chemerinski et al., 2001; Fiske et al.,
advocating an overlap between the neuropathological events caus-
2009; Janzing et al., 2002). To address such differences, authors
ing cognitive deficits and those causing depressive symptoms in
must rely on one of numerous published diagnostic criteria, and
dementia (Kales et al., 2015; Panza et al., 2010; Richard et al.,
their choice of threshold largely defines reported results (Teng
2013). Considerable structural and functional imaging data sup-
et al., 2008).
port this hypothesis, showing that neuropathological lesions in
The 1982 Geriatric Depression Scale (GDS; Yesavage et al.,
AD brains correlate with the development of depressive behavior,
1982) was the first criterion to account for the particularities of
and may underlie them. These pathologies include reduced gray-
late-life depression. The widely used Neuropsychiatric Inventory
matter volume in frontal and temporal lobes (Son et al., 2013),
(NPI; Cummings et al., 1994) was not available until 1994, and only
lesioned subcortical regions (Palmqvist et al., 2011), and reduced
much later, in 2001, the National Institute of Mental Health devel-
glucose metabolism/brain perfusion in the prefrontal cortex
oped its specific diagnostic criteria for depression in AD (PDC-dAD;
(Holthoff et al., 2005; Kang et al., 2012; Kataoka et al., 2010;
Olin et al., 2002). Notably, implementation of PDC-dAD criteria to
Levy-Cooperman et al., 2008; Oshima et al., 2014; Terada et al.,
cohort studies revealed a larger prevalence of comorbid depression
2014).
in AD patients than could be estimated with more generic criteria
Despite the use of distinct diagnostic criteria, follow-up inter-
(Teng et al., 2008; Vilalta-Franch et al., 2006). A shift towards
vals, and sample characteristics, the general consensus emerging
specific, more sensitive diagnostic criteria could thus be related
from epidemiological studies leaves little doubt that depression
to the increasing reported prevalence of depression in AD, as illus-
and AD are tightly related. However, whether depression is a pro-
trated in a recent meta-analysis by Chi and colleagues (Chi et al.,
dromal symptom or a risk factor for AD remains to be determined.
2015).
Interestingly, some have argued that these hypotheses may be
simultaneously true, and that multiple pathways connect depres-
1.2. Depression in mild cognitive impairment
sion and AD (Butters et al., 2008; Green et al., 2003).
Given the long time scale of the association between depression
The prevalence of depression is also disproportionally high
and dementia (e.g. Green et al., 2003), neuroinflammation and
among patients diagnosed with mild cognitive impairment (MCI),
microglial activation appear as potential causal factors to explain
which presents as a relatively mild form of cognitive deficit
the AD-depression link.
between normal aging and dementia. In a systematic review of lit-
erature published before 2007, Apostolova and Cummings found
the prevalence of neuropsychiatric symptoms among MCI patients 2. Microglia function in the normal and diseased brain
to be 35–75%. Depression was listed as one of the most frequent,
with a majority of authors reporting prevalences of 20–50%, Microglia are resident immune cells of the central nervous sys-
depending on sampling and diagnostic criteria (Apostolova and tem (CNS), and account for approximately 10% of the total number
Cummings, 2008). of cells in a healthy mammalian brain (Prinz and Priller, 2014).
Although MCI affects memory to a certain extent, it does not While other major cell populations in the CNS share a neuroepithe-
represent a major impact on daily life activities (Gauthier et al., lial origin, microglia derive from myeloid progenitors, being more
2006). However, while some patients remain stable or even closely related to peripheral macrophages than to neighboring
recover normal cognition, 20–40% of MCI patients progress to AD astrocytes, oligodendrocytes or neurons (Ginhoux et al., 2010).
or other dementias within a few years, a much higher proportion It is now established that microglial progenitors originating in
than in cognitively normal elderly individuals (Roberts and the yolk sac migrate and colonize the CNS during embryonic devel-
Knopman, 2013). Notably, coexistence of depression and MCI opment, before the blood–brain barrier is formed. These progeni-
accelerates cognitive decline, and the risk of developing AD is tors subsequently differentiate and become confined to the CNS,
increased. In a prospective cohort study, Modrego and Ferrández where they function as mononuclear phagocytes, recognizing and
reported that depressed MCI patients had a twofold higher risk scavenging dead cells, pathogens and other targets. Microglia
of developing AD than non-depressed MCI subjects. Although their appear capable of local self-renewal, not depending on recruitment

Please cite this article in press as: Santos, L.E., et al. Microglial dysfunction connects depression and Alzheimer’s disease. Brain Behav. Immun. (2015),
http://dx.doi.org/10.1016/j.bbi.2015.11.011
L.E. Santos et al. / Brain, Behavior, and Immunity xxx (2015) xxx–xxx 3

of circulating progenitors to maintain its population (Ajami et al., anti-inflammatory (reviewed in Casano and Peri, 2015; Hu et al.,
2007; Casano and Peri, 2015; Kierdorf et al., 2013). 2014).
Microglia are well known for their role as cellular effectors of Historically, reports have focused on the detrimental conse-
innate immunity; together with astrocytes, they drive the neuroin- quences of microglia-mediated neuroinflammation, often shown
flammatory process, through phagocytosis and cytokine release. to exacerbate neuronal damage and hinder functional recovery
However, this comprises only part of their function in the CNS. after injury. However, it is now clear that a more complex scenario
Under physiological conditions and in the absence of inflammatory exists, in which microglial activation may also protect the lesioned
stimuli, microglia are found in what is often called a ‘resting state’, brain, depending on context and timeframe (Hellwig et al., 2013).
morphologically defined by extended and ramified processes. Far This matter has been surrounded by controversy in the literature,
from being dormant or inactive, however, these so-called resting largely due to the M1–M2 dichotomy; a persisting notion now
microglia are highly dynamic. Retracting and expanding their increasingly regarded as overly simplistic, and not necessarily fit
processes in response to environmental cues, they interact with to encompass all microglial phenotypes (Chen and Trapp, 2015;
blood vessels, neurons and astroglia, in constant surveillance of Mittelbronn, 2014). Although some authors may be inclined to
the environment (Nimmerjahn et al., 2005). In fact, many authors describe all M2-like microglia as neuroprotective (and, conversely,
have become critical of the word ‘resting’, preferring instead to M1-like microglia as potentially detrimental), relevant details are
use an alternative term, ‘surveillant microglia’ (Chen and Trapp, lost under this classification, which must be used with caution
2015). and in context. It may be possible for microglia to display M1
Recent work has highlighted the importance of microglial and M2 phenotypic characteristics simultaneously (Fenn et al.,
surveillance to CNS function. In early postnatal development, the 2014), and an M2-like phenotype does not always result in neuro-
elimination of excess synapses – a process known as synaptic prun- protection (Cherry et al., 2014).
ing – was shown to be dependent on microglia. Paolicelli et al.
(2011) reported that mice lacking the fractalkine receptor (CX3CR1
3. Neuroinflammation and the role of microglia in Alzheimer’s
KO), which suffer a transient reduction in brain microglia, had con-
disease
current synaptic pruning deficits, resulting in an abnormally high
number of dendritic spines. Work on CX3CR1 KO mice also helped
Alzheimer’s disease (AD), the most common form of dementia
establish a role of microglia in functional maturation of synapses
of the elderly, affects more than 35 million people worldwide (Alz-
(Paolicelli et al., 2011; Paolicelli and Gross, 2011). Most notably,
heimer’s Association, 2015) and is clinically characterized by cog-
pruning deficits found in this model of microglial dysfunction were
nitive deficits and memory loss. Although recent advances
implicated in behavioral abnormalities, including impaired social
towards diagnostic procedures have been made (Clark et al.,
interaction and autistic-like behavior (Zhan et al., 2014).
2011; Curtis et al., 2015; Doecke et al., 2012; Klunk et al., 2004;
In addition to its role in circuit refinement, physiologically
Mapstone et al., 2014; Sabri et al., 2015; Viola et al., 2014), defini-
active microglia modulate neuronal activity in the adult brain
tive diagnosis of AD still relies on histopathology. Plaques largely
through direct contact with neurons (Li et al., 2012). Interestingly,
composed of fibrillar aggregates of the amyloid-b peptide (Ab)
a novel type of microglia–neuron interaction was recently
and neurofibrillary tangles made up from hyperphosphorylated
described in the adult mouse brain, with a subpopulation of micro-
tau protein are the defining lesions found in AD-affected brains,
glia shown to specifically bind to the axon initial segment of corti-
and they are always accompanied by diffuse neuroinflammation.
cal neurons. While the importance of this particular interaction to
There is now little doubt that this inflammatory component is
neuronal function is not yet clear, it is of note that it is lost after
an important player in the pathogenesis of AD (reviewed in
brain injury (Baalman et al., 2015). This observation reinforces
Wilcock, 2012). However, it is still not clear whether brain inflam-
the role of surveillance microglia in brain physiology, and the
mation is a consequence of disease progression, a compensatory or
notion that a ‘loss-of-function’ effect takes place in pathology.
beneficial response, or a causal factor. Evidence supporting the lat-
Indeed, physiological functions depending on microglial shape,
ter possibility was recently summarized by Krstic and Knuesel
local interactions or secretory profile may become impaired as
(2013) in what they called the ‘inflammation hypothesis of AD’.
microglia undergo changes associated with inflammation and
This hypothesis incorporates correlations between late-onset AD
pathology, briefly described below.
and polymorphisms in genes of the innate immune system, as well
as animal and human studies linking inflammatory mediators to
cognitive decline.
2.1. The M1–M2 dichotomy

In both acute CNS lesions and chronic neurological diseases, 3.1. Inflammatory links
microglia become activated, gradually changing morphology and
function. The relevance of microglial activation and subsequent Traumatic brain injury (TBI) is a common primary cause of neu-
proliferation in many pathological contexts has been studied roinflammation (Ziebell and Morganti-Kossmann, 2010), known to
extensively in the past decades, and is discussed in a number of increase the risk of developing AD (Green et al., 2003; Lee et al.,
excellent recent reviews (e.g.: Heneka et al., 2014; Hu et al., 2013). Nonetheless, the possibility that cognitive deterioration fol-
2014; Lynch, 2014; Perry and Holmes, 2014; Von Bernhardi lowing brain trauma could be caused by direct neuronal loss
et al., 2010; Yirmiya et al., 2015). (related, for example, to localized ischemia or excitotoxicity)
Activated microglia acquire an amoeboid shape, with a larger makes it difficult to determine the specific role of TBI-induced
cell body and retracted processes. Functionally, this phenotypic inflammation in the functional outcomes of patients. Interestingly,
change was initially classified according to the same paradigm however, clinical and preclinical studies suggest that systemic
used for peripheral macrophages. According to this view, an initial inflammation, brought about by conditions not involving the
stimulus elicits an ‘M1’ response, centered around the production CNS, can also contribute to neurodegeneration and influence the
and release of mostly pro-inflammatory cytokines, such as TNF-a incidence and progression of AD (Ferreira et al., 2014; Holmes
and IL-6. With persisting stimulation, microglia may move towards et al., 2009; Perry et al., 2007;). For example, using data from over
an alternative activation pattern, the so-called ‘M2’ state, which 6000 community-dwelling older adults enrolled in the Rotterdam
involves active phagocytosis and release of factors regarded as Study, Ott and colleagues showed that AD prevalence is signifi-

Please cite this article in press as: Santos, L.E., et al. Microglial dysfunction connects depression and Alzheimer’s disease. Brain Behav. Immun. (2015),
http://dx.doi.org/10.1016/j.bbi.2015.11.011
4 L.E. Santos et al. / Brain, Behavior, and Immunity xxx (2015) xxx–xxx

cantly higher among subjects with type 2 diabetes (T2D), a condi- phagocytosis of apoptotic neurons (Colonna, 2003; Hsieh et al.,
tion characterized by long-lasting, low grade peripheral inflamma- 2009). Although TREM2 was not shown to bind Ab directly, it inter-
tion, and that incidence of AD is almost doubled when T2D is acts with myelin phospholipids (Cannon et al., 2012), which asso-
pre-existing (Ott et al., 1999, 1996). ciate with Ab (Rivest, 2015). Recently, genome-wide association
In chronically inflamed patients, such as those suffering from studies (GWASs) showed that a polymorphism in TREM2 correlates
obesity and T2D, mechanisms triggered by inflammation may be with increased risk of developing sporadic AD, to an extent similar
shared by the brain and the periphery, and have been proposed to what is seen with the APOEe4 allele (Guerreiro et al., 2013;
as likely links between AD and peripheral metabolic deregulation Jonsson et al., 2013; Karch et al., 2014), the most relevant genetic
(reviewed in De Felice and Ferreira, 2014; Ferreira et al., 2014; risk factor known to date for sporadic AD (Hauser and Ryan,
Heneka et al., 2015b; Morales et al., 2014). 2013; Saunders et al., 1993).
A few years before TREM2 polymorphisms were first described
3.2. Microglial priming in AD patients, Frank and co-workers (2008) showed that its
expression is increased in microglia associated with amyloid pla-
The notion that systemic inflammation influences brain disease ques in a transgenic mouse model of AD. In similar models, others
is, to a good extent, based on microglial priming. Chronic low grade had shown that one of the consequences of TREM2 upregulation in
peripheral inflammation results in increased levels of inflamma- microglia is reduced production of pro-inflammatory cytokines
tory mediators, notably TNF-a and IL-1b, sufficient to transiently (Takahashi et al., 2005; Turnbull et al., 2006). Notably, experiments
activate microglia into a primed state (Cunningham, 2013). As using TREM2 knockout mice also carrying AD-related APP/PS1
mentioned above, activated microglia go through morphological mutations showed that deletion of even a single copy of the Trem2
changes and express certain types of cell surface antigens and cyto- gene is sufficient to reduce plaque-associated microglia (Ulrich
kines. Primed microglia do not actively release cytokines, but et al., 2014). Similar transgenic mice were used to demonstrate
retain several markers of activation (Perry and Holmes, 2014). that expression of TREM2 is required for the survival of Ab-
When presented with a secondary stimulus, primed microglia are activated microglia, and that its deficiency impacts microgliosis
hypersensitive, producing an amplified – and more often neuro- and efficient Ab clearance (Wang et al., 2015). Interestingly, how-
toxic – response, compared to naïve microglia (Norden et al., ever, in another recent paper, Jay and colleagues (2015) reported
2015; Perry and Holmes, 2014). Even in the absence of trauma or opposite results in TREM2-deficient AD mice, which exhibited
pathology, microglia priming may occur as a consequence of nor- reduced inflammation and milder amyloid and tau pathologies.
mal aging (Luo et al., 2010). A modest but persistent increase in Although these two studies used different AD animal models lack-
plasma levels of pro-inflammatory cytokines is associated with ing TREM2, such markedly different results appear difficult to rec-
aging, and could partially account for the increased susceptibility oncile. Experiments involving behavioral and cognitive analyses
to AD and other neurodegenerative disorders in older subjects. may help resolve these discrepancies (Rivest, 2015).
Age is, indeed, the strongest risk factor known for AD (Michaud Currently available data thus point to a dual role of microglia
et al., 2013; Ye and Johnson, 1999). and inflammation in AD. Decreased microglia-mediated Ab clear-
ance was shown to be detrimental in a number of models. It causes
3.3. Microglia in Ab clearance early mortality in a transgenic mouse model of AD (El Khoury et al.,
2007), and is likely linked to disease onset in humans, as suggested
Surface receptors found in activated microglia include many by data on TREM2 polymorphisms (Jonsson et al., 2013). This
proteins capable of binding soluble Ab, such as the scavenger notion is further corroborated by recent work showing that IL-10,
receptor SR-AI, toll-like (TLR2, TLR4, TLR6 and TLR9) and comple- the prototypical anti-inflammatory cytokine, may in fact nega-
ment receptors. Binding of Ab appears to be often followed by tively impact AD brains, precisely due to its effects on microglia.
phagocytosis, proteolysis and Ab clearance, events considered ben- Complementary results from two independent groups showed, in
eficial in AD (Doens and Fernández, 2014; Heneka et al., 2015a). In mouse models of AD, that IL-10 overexpression impairs microglial
addition, bound microglia may prevent the expansion of amyloid clearance of Ab (Chakrabarty et al., 2015), while knocking-out IL-10
plaques and protofibrils, by forming a barrier that alters their com- has the opposite effect, markedly reducing amyloidosis (Guillot-
position and toxicity (Condello et al., 2015). Nonetheless, the Sestier et al., 2015). In both reports, significant behavioral and bio-
increase in microglia recruitment and proliferation that takes place chemical consequences were found to accompany IL-10 modula-
in AD seems insufficient to halt brain accumulation of Ab. In fact, tion of microglia. Nonetheless, beneficial functions of microglia in
Ab clearance is reduced in late-onset AD patients, when compared Ab clearance seem to wane in late AD, and continued production
to age-matched controls, while Ab production is similar in both of TNF-a and other pro-inflammatory cytokines is ultimately neu-
groups (Mawuenyega et al., 2010). Recent studies further indicate rotoxic. This dual role may be one of the reasons why, to date, clin-
that direct transport across the blood–brain barrier accounts for ical trials of several anti-inflammatory therapies have failed to
25% of the clearance of Ab from the brain, with an additional slow AD progression (Perry and Holmes, 2014).
25% resulting from transport into the cerebrospinal fluid and sub- A successful approach to AD-related inflammation should pro-
sequent reabsorption into the venous circulation (Roberts et al., mote microglial clearance of Ab, while also mitigating the effects
2014), implying that 50% of total Ab clearance depends on intrac- of pro-inflammatory cytokines such as TNF-a. Pharmacological
erebral degradation. Studies in transgenic mouse models suggest strategies targeting TREM2 or its downstream effectors may prove
that microglia become dysfunctional in late-stage AD, with a pro- promising in this regard.
gressive reduction in the expression of Ab-binding receptors and
degrading enzymes, while maintaining production of pro-
inflammatory cytokines. These cytokines may, in turn, downregu- 4. Microglia deregulation in depression
late microglial enzymes, further impairing Ab clearance
(Hickman et al., 2008; Orre et al., 2014). Some of the earliest associations between inflammation and
The triggering receptor expressed on myeloid cells 2 (TREM2) is depressive symptoms were described in the late 1980s. In clinical
one of the receptors more recently implicated in Ab clearance. It is trials aimed at hepatitis and metastatic cancer, immunomodula-
a cell-surface protein of the immunoglobulin superfamily, highly tion strategies based on administering the cytokines IFN-a
expressed in microglia, and known to be involved in the (Renault, 1987) and IL-2 (Denicoff et al., 1987) caused unexpected

Please cite this article in press as: Santos, L.E., et al. Microglial dysfunction connects depression and Alzheimer’s disease. Brain Behav. Immun. (2015),
http://dx.doi.org/10.1016/j.bbi.2015.11.011
L.E. Santos et al. / Brain, Behavior, and Immunity xxx (2015) xxx–xxx 5

psychiatric outcomes, including mood alterations and depressive are largely unknown (Glebov et al., 2015; Kolodziejczak et al.,
behavior, in a significant subset of patients. In 1991, Smith was 2015; Krabbe et al., 2012). Nevertheless, these observations sug-
the first to formally propose that inflammatory cytokines could gest an interesting connection between the modulation of micro-
be the natural cause of MDD, in what he termed ‘The macrophage glial inflammatory responses by 5-HT and the anti-depressant
theory of depression’. effects of SSRIs.
Around the same time, the term ‘sickness behavior’ was being
coined, to describe behavioral alterations in animals stricken by 4.2. Microglial regulation of neurogenesis
viral or bacterial infections. Hart (1988) aggregated observations
that anorexia, sleepiness, reduced grooming and an overall The presence of neurotransmitter receptors in microglia illus-
depressive-like state were common traits of sick and feverish trates their functional connection to neurons, which is greater than
mammals. Based on his work and that of others, Hart suggested initially thought (Pocock and Kettenmann, 2007). Another relevant
such behavior was linked to cytokine release, and that it could be aspect of this connection, which could further causally link inflam-
evolutionarily advantageous, perhaps a deterrent to debilitated mation to MDD, is neurogenesis. Adult hippocampal neurogenesis
animals against certain dangerous activities. Evidence that sick- was first reported in the human brain in 1998 (Eriksson et al.,
ness behavior was indeed mediated by inflammatory cytokines 1998), and its inhibition was soon proposed as a mechanism for
came a few years later (Kent et al., 1992). MDD (Jacobs et al., 2000). Findings supporting this notion at the
A clearer association between sickness behavior and MDD was time included the positive regulation of neurogenesis by 5-HT
established in 1996, when Yirmiya used LPS-injected rats to show and its negative regulation by stressful events, likely due to ele-
both phenomena share a common mechanism, involving the vated glucocorticoid levels (Kempermann, 2002; Kempermann
release of inflammatory cytokines. In 2001, Reichenberg and co- and Kronenberg, 2003). Recent data have confirmed and extended
workers extended those results and conclusions to human sub- these observations by showing that MDD patients have smaller
jects. Since then, several lines of evidence have corroborated this hippocampi, and a reduced number of granule cells in the dentate
connection (reviewed in Dantzer et al., 2008; Irwin and Miller, gyrus (Bremner et al., 2000; Videbech, 2004). Further, it is increas-
2007; Walker, 2013), leading to the current notion that microglia ingly clear that SSRIs promote adult neurogenesis in mice and
are intimately connected to mechanisms implicated in depression humans, and that this has a role in their antidepressant effects
(Yirmiya et al., 2015). (Boldrini et al., 2012, 2013).
The involvement of microglia in this scenario was assessed
4.1. Inflammatory links early on (Ekdahl et al., 2003). Experiments were likely motivated
not only by the responsiveness of microglia to 5-HT or cytokine
Several central and peripheral conditions are correlated with levels, but also by their influence on neural precursor cells. At
increased incidence of MDD, including TBI, stroke, T2D and multi- the time, in vitro use of conditioned media had begun to show that
ple sclerosis (Anisman and Hayley, 2012; Jorge et al., 2004; Naranjo factors secreted by microglia guide the differentiation of adult and
et al., 2011; Pascoe et al., 2011; Sadovnick et al., 1997). Inflamma- embryonic neural precursors towards a neuronal fate (Aarum et al.,
tory mediators are common to all these pathologies, and have been 2003), an observation reproduced and expanded by several others
proposed as causal links to MDD (Anisman and Hayley, 2012; since then (Morgan et al., 2004; Ribeiro Xavier et al., 2015; Walton
Dowlati et al., 2010; Felger and Lotrich, 2013). In line with this pos- et al., 2006).
sibility, depressed patients present significantly increased levels of In line with these early data, it is now known that, under certain
pro-inflammatory cytokines, notably IL-6 and TNF-a, in both conditions, microglia may adopt a pro-neurogenic phenotype,
plasma and CSF (e.g. Alesci et al., 2005; Grassi-Oliveira et al., which involves the expression of neurotrophins and anti-
2009; reviewed in Raison et al., 2006). As suggested by this cyto- inflammatory cytokines, such as insulin-like growth factor 1
kine profile, microglial activation and neuroinflammation are also (IGF-1), brain derived neurotrophic factor (BDNF), and IL-4 (Chen
increased in the brains of patients undergoing major depressive and Trapp, 2015; Parkhurst et al., 2013; Ribeiro Xavier et al.,
episodes. This was recently confirmed in a neuroimaging study 2015). However, in pro-inflammatory activation states, microglia
involving depressed subjects with no concurrent psychiatric condi- are usually described as inhibitory to neurogenesis (Monje et al.,
tions, and who had been medication-free for at least 6 weeks 2003; Nakanishi et al., 2007: Kreisel et al., 2014). The implications
(Setiawan et al., 2015). Such correlative data, however, are still of this dual role may be interesting. While, in most cases, reduced
insufficient to determine whether microglial activation is a con- neurogenesis offers a link between inflammatory microglia,
tributing cause or a consequence of other aspects of MDD peripheral disorders and depressive symptoms, certain microglial
pathology. activation states could in fact lead to mood improvement. Favoring
Notably, and unlike what has been found for AD, anti- these pro-neurogenic states pharmacologically could thus be a
inflammatory drugs have shown positive results as adjunct thera- novel road to MDD therapy (Aarum et al., 2003; Gemma and
pies for MDD (Akhondzadeh et al., 2009; Köhler et al., 2014; Bachstetter, 2013; Morgan et al., 2004; Ziv et al., 2006; Ziv and
Müller, 2010). Further, selective 5-HT reuptake inhibitors (SSRIs), Schwartz, 2008).
widely prescribed as antidepressants, were found to have potent
anti-inflammatory effects in pre-clinical and clinical studies 4.3. The IDO hypothesis
(Powell et al., 2013; Sacre et al., 2010; Taraz et al., 2013). Initially
described in peripheral inflammation (Mårtensson and Nässberger, An additional mechanism that may link microglial activation to
1993; Roumestan et al., 2007), the anti-inflammatory actions of MDD has been suggested by several authors (e.g. Myint and Kim,
SSRIs are now known to extend to microglial function, inhibiting 2003; Wichers and Maes, 2004), and more recently revisited by
TNF-a and nitric oxide production (Su et al., 2015; Tynan et al., Dantzer and co-workers (2008). This hypothesis is based on the
2012; Zhang et al., 2012). Moreover, Macchi and co-workers observation that increased levels of pro-inflammatory cytokines
(2013) found that animals with partial deletions in the serotonin induce microglial expression of indolamine-2,3-dioxygenase
transporter (SERT) gene have increased levels of serum pro- (IDO), the enzyme responsible for converting tryptophan to
inflammatory cytokines and increased microglial activation, kynurenine. Tryptophan is the precursor amino acid for serotonin
whether or not under inflammatory stress. Microglial 5-HT recep- (5-HT) biosynthesis, and its reduced availability due to kynurenine
tors have only recently began to be described, and their functions production could impair 5-HT synthesis, leading to depressive

Please cite this article in press as: Santos, L.E., et al. Microglial dysfunction connects depression and Alzheimer’s disease. Brain Behav. Immun. (2015),
http://dx.doi.org/10.1016/j.bbi.2015.11.011
6 L.E. Santos et al. / Brain, Behavior, and Immunity xxx (2015) xxx–xxx

behavior. This is supported by the fact that 5-HT depletion pro- trophic factor for maintaining neuronal survival in the cortex dur-
duces depressive-like symptoms in animal models (O’Connor ing development (Ueno et al., 2013) or near sites of lesions
et al., 2009) and mood alterations in humans (Ruhé et al., 2007), (Lalancette-Hébert et al., 2007). Microglia-derived IGF-1 has sev-
at least in subjects with a family history of MDD, or in remission. eral important functions, but chronic neuroinflammation and
Alternatively, MDD could be linked to metabolic products of pro-inflammatory cytokines are capable of reducing its production,
excessive kynurenine, which include 3-hydroxykynurenine, a free as well as production of other factors secreted by microglia. Such a
radical donor, and quinolinic acid, a specific NMDAR agonist. In line suppression is currently regarded a likely facet of the neurodegen-
with this hypothesis, increased levels of microglial quinolinic acid erative process of AD (Álvarez et al., 2007).
(Steiner et al., 2011) and oxidative products (Ng et al., 2008) have Obesity and peripheral insulin resistance appear linked to lower
been reported in MDD patients. Despite considerable amount of IGF-1 levels (Dunger et al., 2003). Interestingly, brain insulin resis-
work in this regard (reviewed in Dantzer and Walker, 2014; tance has emerged as an early finding in AD, which is notably
Müller and Schwarz, 2007), the exact involvement of excitotoxicity accompanied by changes in the IGF-1 pathway and increased
and the glutamatergic system in this context is not fully under- expression of IGF-1 receptor (IGF-1R) in areas surrounding amyloid
stood, and would be beyond the scope of the current review. How- plaques (Bomfim et al., 2012; De Felice, 2013; De Felice et al., 2014;
ever, these results illustrate that the role of IDO and microglia in Moloney et al., 2010; Talbot et al., 2012). Circulating levels of IGF-1
MDD may involve multiple neurotransmitter systems. are reduced in AD patients (Carro and Torres-Aleman, 2004;
Watanabe et al., 2005), and recent epidemiological data suggest
that low IGF-1 levels are associated with increased risk of AD
5. Connecting neuroinflammation and depression in
(Westwood et al., 2014). High IGF-1 levels, on the other hand, pre-
Alzheimer’s disease
serve cognitive function and total brain volume in elderly subjects
(Okereke et al., 2006; Westwood et al., 2014).
Microglia are a major source, as well as a target, of inflamma-
In animal models of AD, increasing circulating levels of IGF-1
tory cytokines in the CNS. These cells play a pivotal role in neuroin-
accelerates Ab clearance, an effect countered by TNF-a (Carro
flammation, and several lines of evidence now implicate them in
et al., 2002). In addition, a recent study, using mice injected into
the incidence and progression of AD (Lynch, 2014) and MDD
the lateral ventricle with the Ab peptide, showed that reducing
(Yirmiya et al., 2015).
neuroinflammation by deleting the prostaglandin receptor Ep2
A vast literature encompassing clinical studies and basic
enhances IGF-1R signaling (Johansson et al., 2014). IGF-1R is highly
research has established that increased levels of microglia-
expressed in the hippocampus and other areas relevant for cogni-
derived cytokines correlate with AD and depression. For example,
tion and mood, where it regulates neuronal plasticity, neurogene-
two recent meta-analyses have highlighted the fact that increased
sis and oxidative balance (Sonntag et al., 2013; Sun, 2006; Yuan
TNF-a and IL-6 levels in peripheral blood correlate robustly with
et al., 2015). Chronic systemic or hippocampal IGF-1 deficiency
MDD (Dowlati et al., 2010), and that increased levels of IL-6,
produces depressive symptoms in mice (Mitschelen et al., 2011),
TNF-a, IL-1b, TGF-b, IL-12 and IL-18 correlate with AD
while administering IGF-1 exerts antidepressant effects in animal
(Swardfager et al., 2010). Further, polymorphisms in genes coding
models of depressive behavior (Malberg et al., 2007; Park et al.,
IL-1b and TNF-a have been linked to increased susceptibility to
2011; Paslakis et al., 2012). Further, studies in mice suggest that
both AD (Laws et al., 2005; Sciacca et al., 2003; Wang, 2015) and
treatment with IGF-1 reduces sickness behavior caused by LPS or
depression (Bufalino et al., 2013). In addition, IL-6 polymorphisms,
TNF-a injections (Bluthé et al., 2006; Dantzer et al., 1999), possibly
also risk factors for AD (Chen et al., 2012; Dai et al., 2012), were
through increased serotoninergic transmission and neurogenesis
linked to impaired therapeutic response in depressed patients
(Hoshaw et al., 2008; Yuan et al., 2015).
(Bufalino et al., 2013).
Notably, several authors have recently pointed out that
More recently, similar observations were extended to AD
increased IGF-1 levels may also counter depressive symptoms in
patients with comorbid depression. In a study of 55 AD cases —
humans (Cassilhas et al., 2010; Kopczak et al., 2015; Szczêsny
31 of whom were depressed — and 37 age-matched controls,
et al., 2013). In a recent cross-sectional study, direct association
Khemka and co-workers found that depressed AD patients had
between depressive symptoms and cognition was related to low
the highest levels of circulating IL-6 and TNF-a, followed by non-
IGF-1 levels, suggesting that IGF-1 could also mediate the connec-
depressed AD patients and by non-demented controls. Levels of
tion between memory deficits and depression (Lin et al., 2014).
IL-1b were significantly increased in all AD patients, regardless of
depression status. Further, AD patients with depression exhibited
a positive correlation between cognitive deficits and cytokine
5.2. BDNF
levels (Khemka et al., 2014).
Likewise, type 2 diabetes (T2D), a disorder known for its ties to
Brain-derived neurotrophic factor (BDNF), the most widely dis-
systemic inflammation (De Felice and Ferreira, 2014), significantly
tributed neurotrophin in the CNS, is critically involved with struc-
increases the risk of AD in depression. In an epidemiological study
ture and function of neural circuits throughout life. During
involving data collected from all Danish citizens over 50 years of
development, it regulates progenitor survival, neurogenesis and
age (nearly 14 million person-years), Katon and colleagues recently
synaptogenesis (Barnabé-Heider and Miller, 2003; Martínez et al.,
showed that the incidence of AD is higher in patients with comor-
1998), and its trophic roles, e.g. in the promotion of growth and
bid T2D and depression than in those with either condition alone
stabilization of dendritic spines, extend into adult life (Choi et al.,
(Katon et al., 2015). Nonetheless, T2D, depression and AD are com-
2009; Rauskolb et al., 2010). Although it is mostly released by neu-
plex disorders, and their connections may also involve other mech-
rons, BDNF may also be produced by microglia (Dougherty et al.,
anisms in addition to inflammation, as outlined below.
2000) and astrocytes (Parpura and Zorec, 2010). A recent study
showed an interesting role of microglial BDNF in microglia–neuron
5.1. IGF-1 signaling, involving activation of neuronal tropomyosin receptor
kinase B (TrkB) receptors. This interaction promotes learning-
Insulin-like growth factor 1 (IGF-1) is a hormone similar to related synapse formation, suggesting that BDNF secreted by
insulin in structure and biological actions. In the CNS, proliferative microglia modulates synapses and their plasticity, particularly dur-
microglia are an important source of IGF-1, which serves as a ing learning tasks (Parkhurst et al., 2013).

Please cite this article in press as: Santos, L.E., et al. Microglial dysfunction connects depression and Alzheimer’s disease. Brain Behav. Immun. (2015),
http://dx.doi.org/10.1016/j.bbi.2015.11.011
L.E. Santos et al. / Brain, Behavior, and Immunity xxx (2015) xxx–xxx 7

BDNF has emerged as a central molecule in memory formation, that changes in microglial kynurenine production may also be pre-
and a potential therapeutic target for cognitive deficits. Its expres- sent in neurodegenerative disorders (Maddison and Giorgini, 2015;
sion is reduced in a number of pathological conditions (Lu et al., Vécsei et al., 2013). AD patients were found to have reduced levels
2013). Reduced levels of BDNF, pro-BDNF and their corresponding of plasma tryptophan and increased quinolinic acid (Gulaj et al.,
mRNAs are well documented in AD, even at early stages, in brain 2010), as well as increased IDO immunoreactivity in hippocampal
regions such as the hippocampus and cerebral cortex (Holsinger microglia (Bonda et al., 2010). Targeting the kynurenine pathway
et al., 2000; Peng et al., 2005). In peripheral blood, BDNF concentra- using inhibitors of IDO or kynurenine hydroxylase could be a
tion is lowered in AD and other neurological disorders (Laske et al., potential strategy for simultaneously treating MDD and AD, a pro-
2007; Ventriglia et al., 2013; Yasutake et al., 2006). posal that already has some support from animal studies
Reduced levels of serum BDNF have been reported consistently (O’Connor et al., 2009; Yu et al., 2015).
in MDD patients (Bocchio-Chiavetto et al., 2010; Karege et al., Yet another way by which 5-HT may be involved in MDD and
2002; Molendijk et al., 2014), and the role of BDNF in MDD is well AD is by modulating hippocampal neurogenesis. In addition to its
established (Yu and Chen, 2011), particularly in aging (Dwivedi, role in MDD, detailed in a previous section, hippocampal neuroge-
2013). Notably, successful treatment with antidepressants restores nesis may be stimulated by SSRIs (Chang et al., 2012; Kim et al.,
BDNF levels in MDD patients (Wolkowitz et al., 2011). 2013). In AD, although its relevance to disease progression is not
Neuroinflammation has a known impact on BDNF expression, established, neurogenesis may have compensatory roles in mem-
first reported in animal experiments utilizing administration of ory loss, as it may counter neuronal damage to some extent
exogenous cytokines (Lapchak et al., 1993), and now regarded as (Demars et al., 2010; Jin et al., 2004; Lazarov and Marr, 2013; Mu
a potential factor in the pathophysiology of BDNF-related disorders and Gage, 2011). IGF-1, BDNF (two factors that can be induced
(Mondelli et al., 2011). The exact role of microglia-derived BDNF is by 5-HT) and 5-HT itself have all been shown to stimulate hip-
not yet established in the context of inflammation. However, pocampal neurogenesis (Ziv and Schwartz, 2008; Ziv et al., 2006).
down-regulation of BDNF levels in the brain may be a downstream The relationship between these three molecules has been proposed
event to inflammatory processes mediated by microglia, and a as central to the aging process (Mattson et al., 2004) and, given
common underlying contributor to AD and MDD. their ties to neuroinflammation, could also underlie the links
between depression and dementia.
5.3. Serotonin and neurogenesis In this context, an interesting non-pharmacological approach
that holds promise in terms of warding off depression and demen-
Involvement of the monoaminergic system in MDD was initially tia through immunomodulation is physical exercise. Anti-
proposed by Schildkraut, in 1965 (Schildkraut, 1965), who at the inflammatory effects of exercise were first described in the periph-
time focused entirely on catecholamines. Shortly after, Coppen ery, being credited to altered levels of circulating cytokines, macro-
highlighted the role of the serotonergic system (Coppen, 1967), phage TLRs and reduced visceral fat (Gleeson et al., 2011). But
suggesting that 5-HT deficit is the major cause of depression, and recent work suggests that physical exercise may modulate age-
that effective pharmacological treatment for depression could be related changes in microglia, reducing expression of pro-
attributed to increased 5-HT at the synaptic cleft. Since then, sero- inflammatory mediators and stimulating neurogenesis (Kohman
tonergic deficits have been associated with various aspects of MDD et al., 2013; Vukovic et al., 2012). Exercise was shown to favor a
pathophysiology. However, this simplistic hypothesis has been neuroprotective microglial profile, with positive consequences in
challenged by a number of conflicting results, paving the way for animal models of aging and of depressive-like behavior (Kohman
criticism (Holtzheimer and Mayberg, 2011; Lacasse and Leo, et al., 2012; Eyre et al., 2013), as well as decreased Ab accumula-
2005). As a consequence, the 5-HT hypothesis of depression has tion in a mouse model of AD (Adlard et al., 2005).
been refined in an attempt to account for its inconsistencies, which Interestingly, using a transgenic mouse that lacks brain 5-HT,
include the fact that two thirds of patients remain symptomatic Klempin et al. (2013) showed that serotonergic transmission is
after treatment with 5-HT-related drugs (Rush et al., 2006; strictly required for exercise-induced stimulation of hippocampal
Trivedi et al., 2006). neurogenesis. Those authors also noted that microglia were signif-
Current evidence links depressive episodes to several other icantly more abundant in the hippocampi of 5-HT knockout mice
alterations, including hyper-activation of the HPA axis, increased after exercise, which may be related to a lack of anti-
glucocorticoid release (Pariante and Lightman, 2008), and systemic inflammatory 5-HT input, and may be partially responsible for
inflammation, with increased pro-inflammatory cytokine produc- the lack of neurogenesis (Beckman and Santos, 2013). Indeed,
tion. These and other findings support what has been called the pro-inflammatory microglia are associated with a reduced number
‘new 5-HT hypothesis of depression’ (Maes et al., 2011), which sug- of new neurons in the hippocampus, and IFN-a was shown to
gests new pathways in the search for antidepressants (Berton and directly suppress neural stem cell proliferation (Zheng et al., 2014).
Nestler, 2006). Given its involvement in MDD and its links to mem- Notably, a number of clinical trials have addressed the effective-
ory and neuroinflammation, the serotonergic system has been ness of facilitating 5-HT-mediated synaptic communication, by
studied extensively as a target for therapeutic approaches that means of SSRIs, to treat depression in patients with dementia,
encompass AD and behavioral/psychological symptoms of demen- but so far they have produced conflicting results (Banerjee et al.,
tia (Geldenhuys et al., 2011; Rodríguez et al., 2012). 2011; Lyketsos et al., 2003; Nelson and Devanand, 2011; Sepehry
One of several mechanisms by which 5-HT may directly influ- et al., 2012). Fewer studies, however, have focused on cognitive
ence AD pathogenesis is by up-regulating a-secretase activity, via performance in AD patients treated with SSRIs. Additional trials
5-HT4 receptors (Bockaert et al., 2008). Interestingly, Cirrito et al. targeting SSRIs as potential disease modifying agents appear war-
(2011) showed that SSRIs reduce Ab production, accumulation in ranted to determine their potential usefulness in AD therapeutics
interstitial fluid and plaque deposition in a mouse model of AD (Chow et al., 2007).
as well as in cognitively normal humans (Cirrito et al., 2011;
Sheline et al., 2014), presumably through modulation of a- 5.4. Ab oligomers
secretase activity.
As noted above, reduced 5-HT production may follow microglial Ab oligomers (AbOs) are soluble forms of the Ab peptide that
activation through increased IDO levels and activity, leading to accumulate in AD brains, have been shown to cause synaptic defi-
depressive-like symptoms. Interestingly, recent evidence suggests cits and memory impairment in multiple animal models, and are

Please cite this article in press as: Santos, L.E., et al. Microglial dysfunction connects depression and Alzheimer’s disease. Brain Behav. Immun. (2015),
http://dx.doi.org/10.1016/j.bbi.2015.11.011
8 L.E. Santos et al. / Brain, Behavior, and Immunity xxx (2015) xxx–xxx

Fig. 1. Alternative microglial activation states as potential therapeutic targets in AD and MDD. Exogenous and endogenous factors such as aging, infections, stress, brain
trauma and systemic inflammation/metabolic deregulation can induce microglial activation and contribute to CNS pathologies, including AD and MDD. Inflammatory (often
called ‘‘M1-like”) microglia show increased production and secretion of cytokines such as IL-1b, IL-6 and TNF-a, with negative consequences to cognition and increased risk of
AD (see also Heneka et al., 2015a). Inflammatory microglia also exhibit upregulated indolamine-2,3-dioxygenase (IDO), reducing serotonin availability and possibly
contributing to depression. Pharmacological or non-pharmacological approaches favoring a neuroprotective microglial phenotype (classically referred to as ‘‘M2-like”) may
promote neurogenesis, improve Ab clearance, and contribute to an overall lower level of neuroinflammation and release of neurotrophins, potentially beneficial in both AD
and MDD.

increasingly recognized as proximal neurotoxins in AD (Ferreira use in AD are not disease modifying, and are useful only for symp-
et al., 2007; Ferreira and Klein, 2011). Remarkably, a single injec- tomatic management. Likewise, the mechanisms underlying MDD
tion of AbOs (10 pmol) into the lateral cerebral ventricle causes are still poorly understood, and available pharmacological
rapid and persistent memory impairment in mice (Figueiredo approaches are not always effective or reliable (Rush et al.,
et al., 2013). Intriguingly, we recently found that Ab oligomers 2006). In the past decade, considerable evidence has established
(AbOs) trigger depressive-like behavior in mice, accompanied by microglia-mediated inflammation as an important component of
brain inflammation comprising microglial/astrocyte activation both AD and MDD (Cunningham, 2013; Yirmiya et al., 2015).
and release of the pro-inflammatory cytokines TNF-a and IL-1b, Increased levels of pro-inflammatory cytokines and a shift in tryp-
and (Ledo et al., 2013). These findings suggest AbOs may provide tophan use towards the microglial kynurenine pathway have been
a mechanistic link between AD and MDD. Pre-treatment of mice reported in AD and MDD, and have been proposed as causal factors
with fluoxetine, a commonly used SSRI, prevented both in both disorders (Dantzer et al., 2008; Gong et al., 2011). While a
depressive-like behavior and memory loss induced by AbO infu- more aggressive, pro-inflammatory microglial phenotype may be
sion, and reduced microglia density and brain levels of pro- detrimental to neurons and lead to memory impairment and mood
inflammatory cytokines (Ledo et al., 2013). These results suggest disturbances, alternative, more benign phenotypes may actually be
that fluoxetine itself and/or the increased availability of 5-HT could protective and increase Ab clearance/detoxification. Rebalancing
have a relevant effect on AbO-activated microglia, simultaneously microglial responses may thus be beneficial, especially so for
contributing to ameliorating mood and memory in AD. comorbid AD and MDD (Fig. 1). As argued above, if the inflamma-
tion hypothesis linking AD and MDD proves valid, modulating
microglial reactivity could be more than a palliative approach.
6. Conclusion
Finally, understanding microglial priming, activation states and
age-related dysfunction may reveal relevant therapeutic targets
In line with a role of microglia as a link between AD and depres-
against dementia and virtually all brain related disorders, even
sion, an anti-inflammatory drug, minocycline, is currently going
those not causally linked to inflammation.
through phase two clinical trials for both MDD (Dean et al.,
2014) and AD (The MADE trial – ISRCTN16105064). In the context
of MDD, numerous animal studies underlie the potential of Acknowledgments
minocycline (reviewed by Carvalho et al., 2014; Dean et al.,
2012; Soczynska et al., 2012), including recent work showing it sig- Work in the authors’ laboratory is supported by grants from the
nificantly suppresses depressive-like behavior and inhibition of National Institute for Translational Neuroscience/Brazil, Conselho
neurogenesis in IFN-a-treated mice (Zheng et al., 2015). In AD, Nacional de Desenvolvimento Científico e Tecnológico (CNPq)
although anti-inflammatory approaches have so far failed to block and Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado
disease progression, minocycline appears promising due to its do Rio de Janeiro (FAPERJ). L.E.S. and D.B. are pre-doctoral fellows
selective effect on microglial activation, which seems to favor supported by CNPq.
lower TNF-a and IL-1b production (Kobayashi et al., 2013) while
preserving phagocytic behavior and, possibly, Ab clearance.
References
AD is a complex neurodegenerative disorder, and our limited
knowledge of its pathophysiology remains an obstacle for the Aarum, J., Sandberg, K., Haeberlein, S.L.B., Persson, M.A.A., 2003. Migration and
development of effective therapies. Drugs currently approved for differentiation of neural precursor cells can be directed by microglia. Proc. Natl.

Please cite this article in press as: Santos, L.E., et al. Microglial dysfunction connects depression and Alzheimer’s disease. Brain Behav. Immun. (2015),
http://dx.doi.org/10.1016/j.bbi.2015.11.011
L.E. Santos et al. / Brain, Behavior, and Immunity xxx (2015) xxx–xxx 9

Acad. Sci. U.S.A. 100, 15983–15988. http://dx.doi.org/10.1073/ found in the neuropathology of Alzheimer’s disease. Redox Rep. 15, 161–168.
pnas.2237050100. http://dx.doi.org/10.1179/174329210X12650506623645.
Adlard, P.A., Perreau, V.M., Pop, V., Cotman, C.W., 2005. Voluntary exercise Bremner, J.D., Narayan, M., Anderson, E.R., Staib, L.H., Miller, H.L., Charney, D.S.,
decreases amyloid load in a transgenic model of Alzheimer’s disease. J. 2000. Hippocampal volume reduction in major depression. Am. J. Psychiatry
Neurosci. 25, 4217–4221. http://dx.doi.org/10.1523/JNEUROSCI.0496-05.2005. 157, 115–118. http://dx.doi.org/10.1176/appi.ajp.157.1.115.
Ajami, B., Bennett, J.L., Krieger, C., Tetzlaff, W., Rossi, F.M.V., 2007. Local self-renewal Bufalino, C., Hepgul, N., Aguglia, E., Pariante, C.M., 2013. The role of immune genes
can sustain CNS microglia maintenance and function throughout adult life. Nat. in the association between depression and inflammation: a review of recent
Neurosci. 10, 1538–1543. http://dx.doi.org/10.1038/nn2014. clinical studies. Brain Behav. Immun. 31, 31–47. http://dx.doi.org/10.1016/j.
Akhondzadeh, S., Jafari, S., Raisi, F., Nasehi, A.A., Ghoreishi, A., Salehi, B., Mohebbi- bbi.2012.04.009.
Rasa, S., Raznahan, M., Kamalipour, A., 2009. Clinical trial of adjunctive Burns, A., Jacoby, R., Levy, R., 1990a. Psychiatric phenomena in Alzheimer’s disease.
celecoxib treatment in patients with major depression: a double blind and III. Disorders of mood. Br. J. Psychiatry 157, 81–86. http://dx.doi.org/10.1192/
placebo controlled trial. Depress. Anxiety 26, 607–611. http://dx.doi.org/ bjp.157.1.81.
10.1002/da.20589. Burns, A., Jacoby, R., Levy, R., 1990b. Psychiatric phenomena in Alzheimer’s disease.
Alesci, S., Martinez, P.E., Kelkar, S., Ilias, I., Ronsaville, D.S., Listwak, S.J., Ayala, A.R., IV. Disorders of behaviour. Br. J. Psychiatry 157, 86–94. http://dx.doi.org/
Licinio, J., Gold, H.K., Kling, M.A., Chrousos, G.P., Gold, P.W., 2005. Major 10.1192/bjp.157.1.86.
depression is associated with significant diurnal elevations in plasma Butters, M.A., Young, J.B., Lopez, O., Aizenstein, H.J., Mulsant, B.H., Reynolds, C.F.,
interleukin-6 levels, a shift of its circadian rhythm, and loss of physiological DeKosky, S.T., Becker, J.T., 2008. Pathways linking late-life depression to
complexity in its secretion: clinical implications. J. Clin. Endocrinol. Metab. 90, persistent cognitive impairment and dementia. Dialogues Clin. Neurosci. 10,
2522–2530. http://dx.doi.org/10.1210/jc.2004-1667. 345–357. http://dx.doi.org/10.1016/j.bbi.2008.05.010.
Allegri, R.F., Sarasola, D., Serrano, C.M., Taragano, F.E., Arizaga, R.L., Butman, J., Loñ, Cannon, J.P., O’Driscoll, M., Litman, G.W., 2012. Specific lipid recognition is a general
L., 2006. Neuropsychiatric symptoms as a predictor of caregiver burden in feature of CD300 and TREM molecules. Immunogenetics 64, 39–47. http://dx.
Alzheimer’s disease. Neuropsychiatr. Dis. Treat. 2, 105–110. doi.org/10.1007/s00251-011-0562-4.
Álvarez, A., Cacabelos, R., Sanpedro, C., García-Fantini, M., Aleixandre, M., 2007. Carro, E., Torres-Aleman, I., 2004. Insulin-like growth factor I and Alzheimer’s
Serum TNF-alpha levels are increased and correlate negatively with free IGF-I in disease: therapeutic prospects? Expert Rev. Neurother. 4, 79–86. http://dx.doi.
Alzheimer disease. Neurobiol. Aging 28, 533–536. http://dx.doi.org/10.1016/j. org/10.1586/14737175.4.1.79.
neurobiolaging.2006.02.012. Carro, E., Trejo, J.L., Gomez-Isla, T., LeRoith, D., Torres-Aleman, I., 2002. Serum
Anisman, H., Hayley, S., 2012. Inflammatory factors contribute to depression and its insulin-like growth factor I regulates brain amyloid-beta levels. Nat. Med. 8,
comorbid conditions. Sci. Signal. 5. http://dx.doi.org/10.1126/ 1390–1397. http://dx.doi.org/10.1038/nm793.
scisignal.2003579, pe45–pe45. Carvalho, A.F., Miskowiak, K.K., Hyphantis, T.N., Köhler, C.A., Alves, G.S., Bortolato,
Apostolova, L.G., Cummings, J.L., 2008. Neuropsychiatric manifestations in mild B., Sales, P.M.G., Machado-vieira, R., Berk, M., Mcintyre, R.S., 2014. Cognitive
cognitive impairment: a systematic review of the literature. Dement. Geriatr. dysfunction in depression – pathophysiology and novel targets. CNS Neurol.
Cogn. Disord. 25, 115–126, 000112509 [pii] 10.1159/000112509. Disord.: Drug Targets 13, 1819–1835.
Baalman, K., Marin, M.A., Ho, T.S.-Y., Godoy, M., Cherian, L., Robertson, C., Rasband, Casano, A.M., Peri, F., 2015. Microglia: multitasking specialists of the brain. Dev. Cell
M.N., 2015. Axon initial segment-associated microglia. J. Neurosci. 35, 2283– 32, 469–477. http://dx.doi.org/10.1016/j.devcel.2015.01.018.
2292. http://dx.doi.org/10.1523/JNEUROSCI.3751-14.2015. Cassilhas, R.C., Antunes, H.K.M., Tufik, S., de Mello, M.T., 2010. Mood, anxiety, and
Banerjee, S., Hellier, J., Dewey, M., Romeo, R., Ballard, C., Baldwin, R., Bentham, P., serum IGF-1 in elderly men given 24 weeks of high resistance exercise. Percept.
Fox, C., Holmes, C., Katona, C., Knapp, M., Lawton, C., Lindesay, J., Livingston, G., Mot. Skills 110, 265–276. http://dx.doi.org/10.2466/PMS.110.1.265-276.
McCrae, N., Moniz-Cook, E., Murray, J., Nurock, S., Orrell, M., O’Brien, J., Poppe, Chakrabarty, P., Li, A., Das, P., Golde, T.E., Chakrabarty, P., Li, A., Ceballos-diaz, C.,
M., Thomas, A., Walwyn, R., Wilson, K., Burns, A., 2011. Sertraline or mirtazapine Eddy, J.A., Funk, C.C., Moore, B., Dinunno, N., Rosario, A.M., Cruz, P.E., Verbeeck,
for depression in dementia (HTA-SADD): a randomised, multicentre, double- C., Sacino, A., Nix, S., 2015. IL-10 alters immunoproteostasis in APP mice,
blind, placebo-controlled trial. Lancet 378, 403–411. http://dx.doi.org/10.1016/ increasing plaque burden and worsening cognitive article IL-10 alters
S0140-6736(11)60830-1. immunoproteostasis in APP Mice, increasing plaque burden and worsening
Barnabé-Heider, F., Miller, F.D., 2003. Endogenously produced neurotrophins cognitive behavior. Neuron 85, 519–533. http://dx.doi.org/10.1016/j.
regulate survival and differentiation of cortical progenitors via distinct neuron.2014.11.020.
signaling pathways. J. Neurosci. 23, 5149–5160. Chang, K.A., Kim, J.A., Kim, S.S., Joo, Y., Shin, K.Y., Kim, S.S., Kim, H.S., Suh, Y.H., 2012.
Beckman, D., Santos, L.E., 2013. The importance of serotonin in exercise-induced Therapeutic potentials of neural stem cells treated with fluoxetine in
adult neurogenesis: new evidence from Tph2/ mice. J. Neurosci. 33, 14283– Alzheimer’s disease. Neurochem. Int. 61, 885–891. http://dx.doi.org/10.1016/j.
14284. http://dx.doi.org/10.1523/JNEUROSCI.2911-13.2013. neuint.2012.03.017.
Benoit, M., Berrut, G., Doussaint, J., Bakchine, S., Bonin-Guillaume, S., Frémont, P., Chemerinski, E., Petracca, G., Sabe, L., Kremer, J., Starkstein, S.E., 2001. The
Gallarda, T., Krolak-Salmon, P., Marquet, T., Mékiès, C., Sellal, F., Schuck, S., specificity of depressive symptoms in patients with Alzheimer’s disease. Am.
David, R., Robert, P., 2012. Apathy and depression in mild Alzheimer’s disease: a J. Psychiatry 158, 68–72. http://dx.doi.org/10.1176/appi.ajp.158.1.68.
cross-sectional study using diagnostic criteria. J. Alzheimers. Dis. 31, 325–334. Chen, Z., Trapp, B.D., 2015. Microglia and neuroprotection. J. Neurochem. 1–8.
http://dx.doi.org/10.3233/JAD-2012-112003. http://dx.doi.org/10.1111/jnc.13062.
Berton, O., Nestler, E.J., 2006. New approaches to antidepressant drug discovery: Chen, S.Y., Chen, T.F., Lai, L.C., Chen, J.H., Sun, Y., Wen, L.L., Yip, P.K., Chu, Y.M., Chen,
beyond monoamines. Nat. Rev. Neurosci. 7, 137–151. http://dx.doi.org/10.1038/ Y.C., 2012. Sequence variants of interleukin 6 (IL-6) are significantly associated
nrn1846. with a decreased risk of late-onset Alzheimer’s disease. J. Neuroinflamm. 9, 21.
Bluthé, R.-M., Kelley, K.W., Dantzer, R., 2006. Effects of insulin-like growth factor-I http://dx.doi.org/10.1186/1742-2094-9-21.
on cytokine-induced sickness behavior in mice. Brain Behav. Immun. 20, 57–63. Cherry, J.D., Olschowka, J.A., O’Banion, M.K., 2014. Neuroinflammation and M2
http://dx.doi.org/10.1016/j.bbi.2005.02.003. microglia: the good, the bad, and the inflamed. J. Neuroinflamm. 11, 98. http://
Bocchio-Chiavetto, L., Bagnardi, V., Zanardini, R., Molteni, R., Nielsen, M.G., dx.doi.org/10.1186/1742-2094-11-98.
Placentino, A., Giovannini, C., Rillosi, L., Ventriglia, M., Riva, M.A., Gennarelli, Chi, S., Yu, J.-T., Tan, M.-S., Tan, L., 2014. Depression in Alzheimer’s disease:
M., 2010. Serum and plasma BDNF levels in major depression: a replication epidemiology, mechanisms, and management. J. Alzheimers. Dis. 42, 739–755.
study and meta-analyses. World J. Biol. Psychiatry 11, 763–773. http://dx.doi. http://dx.doi.org/10.3233/JAD-140324.
org/10.3109/15622971003611319. Chi, S., Wang, C., Jiang, T., Zhu, X.-C., Yu, J.-T., Tan, L., 2015. The prevalence of
Bockaert, J., Claeysen, S., Compan, V., Dumuis, A., 2008. 5-HT4 receptors: history, depression in Alzheimer’s disease: a systematic review and meta-analysis. Curr.
molecular pharmacology and brain functions. Neuropharmacology 55, 922– Alzheimer Res. 12, 189–198.
931. http://dx.doi.org/10.1016/j.neuropharm.2008.05.013. Choi, S., Li, Y., Parada, L.F., Sisodia, S.S., 2009. Regulation of hippocampal progenitor
Boldrini, M., Hen, R., Underwood, M.D., Rosoklija, G.B., Dwork, A.J., Mann, J.J., cell survival, proliferation and dendritic development by BDNF. Mol.
Arango, V., 2012. Hippocampal angiogenesis and progenitor cell proliferation Neurodegener. 4, 52. http://dx.doi.org/10.1186/1750-1326-4-52.
are increased with antidepressant use in major depression. Biol. Psychiatry 72, Chow, T.W., Pollock, B.G., Milgram, N.W., 2007. Potential cognitive enhancing and
562–571. http://dx.doi.org/10.1016/j.biopsych.2012.04.024. disease modification effects of SSRIs for Alzheimer’s disease. Neuropsychiatr.
Boldrini, M., Santiago, A.N., Hen, R., Dwork, A.J., Rosoklija, G.B., Tamir, H., Arango, V., Dis. Treat. 3, 627–636.
John Mann, J., 2013. Hippocampal granule neuron number and dentate gyrus Cirrito, J.R., Disabato, B.M., Restivo, J.L., Verges, D.K., Goebel, W.D., Sathyan, A.,
volume in antidepressant-treated and untreated major depression. Hayreh, D., D’Angelo, G., Benzinger, T., Yoon, H., Kim, J., Morris, J.C., Mintun, M.
Neuropsychopharmacology 38, 1068–1077. http://dx.doi.org/10.1038/ A., Sheline, Y.I., 2011. Serotonin signaling is associated with lower amyloid-
npp.2013.5. {beta} levels and plaques in transgenic mice and humans. Proc. Natl. Acad. Sci.
Bomfim, T.R., Forny-Germano, L., Sathler, L.B., Brito-Moreira, J., Houzel, J.C., Decker, U.S.A. 108, 14968–14973. http://dx.doi.org/10.1073/pnas.1107411108.
H., Silverman, M.A., Kazi, H., Melo, H.M., McClean, P.L., Holscher, C., Arnold, S.E., Clark, C.M., Schneider, J.A., Bedell, B.J., Beach, T.G., Bilker, W.B., Mintun, M.A.,
Talbot, K., Klein, W.L., Munoz, D.P., Ferreira, S.T., De Felice, F.G., 2012. An anti- Pontecorvo, M.J., Hefti, F., Carpenter, A.P., Flitter, M.L., Krautkramer, M.J., Kung,
diabetes agent protects the mouse brain from defective insulin signaling caused H.F., Coleman, R.E., Doraiswamy, P.M., Fleisher, A.S., Sabbagh, M.N., Sadowsky,
by Alzheimer’s disease-associated Ab oligomers. J. Clin. Invest. 122, 1339–1353. C.H., Reiman, E.P., Zehntner, S.P., Skovronsky, D.M., 2011. Use of florbetapir-PET
http://dx.doi.org/10.1172/JCI57256. for imaging beta-amyloid pathology. JAMA 305, 275–283. http://dx.doi.org/
Bonda, D.J., Mailankot, M., Stone, J.G., Garrett, M.R., Staniszewska, M., Castellani, R.J., 10.1001/jama.2010.2008.
Siedlak, S.L., Zhu, X., Lee, H., Perry, G., Nagaraj, R.H., Smith, M.A., 2010. Colonna, M., 2003. TREMs in the immune system and beyond. Nat. Rev. Immunol. 3,
Indoleamine 2,3-dioxygenase and 3-hydroxykynurenine modifications are 445–453. http://dx.doi.org/10.1038/nri1106.

Please cite this article in press as: Santos, L.E., et al. Microglial dysfunction connects depression and Alzheimer’s disease. Brain Behav. Immun. (2015),
http://dx.doi.org/10.1016/j.bbi.2015.11.011
10 L.E. Santos et al. / Brain, Behavior, and Immunity xxx (2015) xxx–xxx

Condello, C., Yuan, P., Schain, A., Grutzendler, J., 2015. Microglia constitute a barrier Eyre, H.A., Papps, E., Baune, B.T., 2013. Treating Depression and Depression-Like
that prevents neurotoxic protofibrillar Ab42 hotspots around plaques. Nat. Behavior with Physical Activity: An Immune Perspective. Front. Psychiatry 4, 1–
Commun. 6, 6176. http://dx.doi.org/10.1038/ncomms7176. 27. http://dx.doi.org/10.3389/fpsyt.2013.00003.
Coppen, A., 1967. The biochemistry of affective disorders. Br. J. Psychiatry 113, Felger, J.C., Lotrich, F.E., 2013. Inflammatory cytokines in depression:
1237–1264. http://dx.doi.org/10.1192/bjp.113.504.1237. neurobiological mechanisms and therapeutic implications. Neuroscience 246,
Cummings, J.L., Mega, M., Gray, K., Rosenberg-Thompson, S., Carusi, D.A., Gornbein, 199–229. http://dx.doi.org/10.1016/j.neuroscience.2013.04.060.
J., 1994. The neuropsychiatric inventory: comprehensive assessment of Fenn, A.M., Hall, J.C.E., Gensel, J.C., Popovich, P.G., Godbout, J.P., 2014. IL-4 signaling
psychopathology in dementia. Neurology 44, 2308–2314. http://dx.doi.org/ drives a unique arginase+/IL-1+ microglia phenotype and recruits macrophages
10.1212/WNL.44.12.2308. to the inflammatory CNS: consequences of age-related deficits in IL-4R after
Cunningham, C., 2013. Microglia and neurodegeneration: the role of systemic traumatic spinal cord injury. J. Neurosci. 34, 8904–8917. http://dx.doi.org/
inflammation. Glia 61, 71–90. http://dx.doi.org/10.1002/glia.22350. 10.1523/JNEUROSCI.1146-14.2014.
Curtis, C., Gamez, J.E., Singh, U., Al, E., 2015. Phase 3 trial of flutemetamol labeled Ferreira, S.T., Klein, W.L., 2011. The Ab oligomer hypothesis for synapse failure and
with radioactive fluorine 18 imaging and neuritic plaque density. JAMA Neurol. memory loss in Alzheimer’s disease. Neurobiol. Learn. Mem. 96, 529–543.
72, 287–294. http://dx.doi.org/10.1016/j.nlm.2011.08.003.
Dai, L., Liu, D., Guo, H., Wang, Y., Bai, Y., 2012. Association between polymorphism in Ferreira, S.T., Vieira, M.N.N., De Felice, F.G., 2007. Soluble protein oligomers as
the promoter region of Interleukin 6 (-174 G/C) and risk of Alzheimer’s disease: emerging toxins in Alzheimer’s and other amyloid diseases. IUBMB Life 59,
a meta-analysis. J. Neurol. 259, 414–419. http://dx.doi.org/10.1007/s00415- 332–345. http://dx.doi.org/10.1080/15216540701283882.
011-6164-0. Ferreira, S.T., Clarke, J.R., Bomfim, T.R., De Felice, F.G., 2014. Inflammation,
Dantzer, R., Walker, A.K., 2014. Is there a role for glutamate-mediated excitotoxicity defective insulin signaling, and neuronal dysfunction in Alzheimer’s
in inflammation-induced depression? J. Neural Transm. 925–932. http://dx.doi. disease. Alzheimer’s Dement. 10, S76–S83. http://dx.doi.org/10.1016/
org/10.1007/s00702-014-1187-1. j.jalz.2013.12.010.
Dantzer, R., Gheusi, G., Johnson, R.W., Kelley, K.W., 1999. Central administration of Figueiredo, C.P., Clarke, J.R., Ledo, J.H., Ribeiro, F.C., Costa, C.V., Melo, H.M., Mota-
insulin-like growth factor-1 inhibits lipopolysaccharide-induced sickness Sales, A.P., Saraiva, L.M., Klein, W.L., Sebollela, A., De Felice, F.G., Ferreira, S.T.,
behavior in mice. NeuroReport 10, 289–292. 2013. Memantine rescues transient cognitive impairment caused by high-
Dantzer, R., O’Connor, J.C., Freund, G.G., Johnson, R.W., Kelley, K.W., 2008. From molecular-weight ab oligomers but not the persistent impairment induced by
inflammation to sickness and depression: when the immune system subjugates low-molecular-weight oligomers. J. Neurosci. 33, 9626–9634. http://dx.doi.org/
the brain. Nat. Rev. Neurosci. 9, 46–56. http://dx.doi.org/10.1038/nrn2297. 10.1523/JNEUROSCI.0482-13.2013.
De Felice, F.G., 2013. Alzheimer’s disease and insulin resistance: translating basic Fiske, A., Wetherell, J.L., Gatz, M., 2009. Depression in older adults. Annu. Rev. Clin.
science into clinical applications. J. Clin. Invest. 123, 531–539. http://dx.doi.org/ Psychol. 5, 363–389. http://dx.doi.org/10.1146/
10.1172/JCI64595. annurev.clinpsy.032408.153621.
De Felice, F.G., Ferreira, S.T., 2014. Inflammation, defective insulin signaling, and Frank, S., Burbach, G.J., Bonin, M., Walter, M., Streit, W., Bechmann, I., Deller, T.,
mitochondrial dysfunction as common molecular denominators connecting 2008. TREM2 is upregulated in amyloid plaque-associated microglia in aged
type 2 diabetes to Alzheimer disease. Diabetes 63, 2262–2272. http://dx.doi. APP23 transgenic mice. Glia 56, 1438–1447. http://dx.doi.org/10.1002/
org/10.2337/db13-1954. glia.20710.
De Felice, F.G., Lourenco, M.V., Ferreira, S.T., 2014. How does brain insulin resistance Gauthier, S., Reisberg, B., Zaudig, M., Petersen, R.C., Ritchie, K., Broich, K., Belleville,
develop in Alzheimer’s disease? Alzheimer’s Dement. 10, S26–S32. http://dx. S., Brodaty, H., Bennett, D., Chertkow, H., Cummings, J.L., de Leon, M., Feldman,
doi.org/10.1016/j.jalz.2013.12.004. H., Ganguli, M., Hampel, H., Scheltens, P., Tierney, M.C., Whitehouse, P.,
Dean, O.M., Data-Franco, J., Giorlando, F., Berk, M., 2012. Minocycline: therapeutic Winblad, B., 2006. Mild cognitive impairment. Lancet 367, 1262–1270. http://
potential in psychiatry. CNS Drugs 26, 391–401. http://dx.doi.org/10.2165/ dx.doi.org/10.1016/S0140-6736(06)68542-5.
11632000-000000000-00000. Geldenhuys, W.J., der Schyf, C.J., Van Der Schyf, C.J., der Schyf, C.J., 2011. Role of
Dean, O.M., Maes, M., Ashton, M., Berk, L., Kanchanatawan, B., Sughondhabirom, A., serotonin in Alzheimer’s disease: a new therapeutic target? CNS Drugs 25, 765–
Tangwongchai, S., Ng, C., Dowling, N., Malhi, G.S., Berk, Mi., 2014. Protocol and 781. http://dx.doi.org/10.2165/11590190-000000000-00000.
rationale-the efficacy of minocycline as an adjunctive treatment for major Gemma, C., Bachstetter, A.D., 2013. The role of microglia in adult hippocampal
depressive disorder: a double blind, randomised, placebo controlled trial. Clin. neurogenesis. Front. Cell. Neurosci. 7, 229. http://dx.doi.org/10.3389/
Psychopharmacol. Neurosci. 12, 180–188. http://dx.doi.org/10.9758/ fncel.2013.00229.
cpn.2014.12.3.180. Ginhoux, F., Greter, M., Leboeuf, M., Nandi, S., See, P., Gokhan, S., Mehler, M.F.,
Demars, M., Hu, Y.-S., Gadadhar, A., Lazarov, O., 2010. Impaired neurogenesis is an Conway, S.J., Ng, L.G., Stanley, E.R., Samokhvalov, I.M., Merad, M., 2010. Fate
early event in the etiology of familial Alzheimer’s disease in transgenic mice. J. mapping analysis reveals that adult microglia derive from primitive
Neurosci. Res. 88, 2103–2117. http://dx.doi.org/10.1002/jnr.22387. macrophages. Science 330, 841–845. http://dx.doi.org/
Denicoff, K.D., Rubinow, D.R., Papa, M.Z., Simpson, C., Seipp, C.A., Lotze, M.T., Chang, 10.1126/science.1194637.
A.E., Rosenstein, D., Rosenberg, S.A., 1987. The neuropsychiatric effects of Glebov, K., Löchner, M., Jabs, R., Lau, T., Merkel, O., Schloss, P., Steinhäuser, C.,
treatment with interleukin-2 and lymphokine-activated killer cells. Ann. Intern. Walter, J., 2015. Serotonin stimulates secretion of exosomes from microglia
Med. 107, 293–300. cells. Glia 63, 626–634. http://dx.doi.org/10.1002/glia.22772.
Doecke, J.D., Laws, S.M., Faux, N.G., Wilson, W., Burnham, S.C., Lam, C.-P., Mondal, A., Gleeson, M., Bishop, N.C., Stensel, D.J., Lindley, M.R., Mastana, S.S., Nimmo, M.A.,
Bedo, J., Bush, A.I., Brown, B., De Ruyck, K., Ellis, K.A., Fowler, C., Gupta, V.B., 2011. The anti-inflammatory effects of exercise: mechanisms and implications
Head, R., Macaulay, S.L., Pertile, K., Rowe, C.C., Rembach, A., Rodrigues, M., for the prevention and treatment of disease. Nat. Rev. Immunol. 11, 607–615.
Rumble, R., Szoeke, C., Taddei, K., Taddei, T., Trounson, B., Ames, D., Masters, C.L., http://dx.doi.org/10.1038/nri3041.
Martins, R.N., 2012. Blood-based protein biomarkers for diagnosis of Alzheimer Gong, C.Y., Li, Z., Wang, H.M., Liu, J., Chen, L., Zhang, H.W., Wang, X., Yang, J., 2011.
disease. Arch. Neurol. 69, 1318–1325. http://dx.doi.org/10.1001/ Targeting the kynurenine pathway as a potential strategy to prevent and treat
archneurol.2012.1282. Alzheimer’s disease. Med. Hypotheses 77, 383–385. http://dx.doi.org/10.1016/j.
Doens, D., Fernández, P.L., 2014. Microglia receptors and their implications in the mehy.2011.05.022.
response to amyloid b for Alzheimer’s disease pathogenesis. J. Neuroinflamm. Grassi-Oliveira, R., Brietzke, E., Pezzi, J.C., Lopes, R.P., Teixeira, A.L., Bauer, M.E., 2009.
11, 48. http://dx.doi.org/10.1186/1742-2094-11-48. Increased soluble tumor necrosis factor-alpha receptors in patients with major
Dougherty, K.D., Dreyfus, C.F., Black, I.B., 2000. Brain-derived neurotrophic factor in depressive disorder. Psychiatry Clin. Neurosci. 63, 202–208. http://dx.doi.org/
astrocytes, oligodendrocytes, and microglia/macrophages after spinal cord 10.1111/j.1440-1819.2008.01918.x.
injury. Neurobiol. Dis. 7, 574–585. http://dx.doi.org/10.1006/nbdi.2000.0318. Green, R.C., Cupples, L.A., Kurz, A., Auerbach, S., Go, R., Sadovnick, D., Duara, R.,
Dowlati, Y., Herrmann, N., Swardfager, W., Liu, H., Sham, L., Reim, E.K., Lanctôt, K.L., Kukull, W.A., Chui, H., Edeki, T., Griffith, P.A., Friedland, R.P., Bachman, D., Farrer,
2010. A meta-analysis of cytokines in major depression. Biol. Psychiatry 67, L., 2003. Depression as a risk factor for Alzheimer disease: the MIRAGE Study.
446–457. http://dx.doi.org/10.1016/j.biopsych.2009.09.033. Arch. Neurol. 60, 753–759. http://dx.doi.org/10.1001/archneur.60.5.753.
Dunger, D.B., Ong, K.K.L., Sandhu, M.S., 2003. Serum insulin-like growth factor-I Guerreiro, R., Wojtas, A., Bras, J., Carrasquillo, M., Rogaeva, E., Majounie, E.,
levels and potential risk of type 2 diabetes. Horm. Res. 60 (Suppl. 3), 131–135. Cruchaga, C., Sassi, C., Kauwe, J.S.K., Younkin, S., Hazrati, L., Collinge, J., Pocock, J.,
http://dx.doi.org/10.1159/000074514. Lashley, T., Williams, J., Lambert, J.-C., Amouyel, P., Goate, A., Rademakers, R.,
Dwivedi, Y., 2013. Involvement of brain-derived neurotrophic factor in late-life Morgan, K., Powell, J., St George-Hyslop, P., Singleton, A., Hardy, J., 2013. TREM2
depression. Am. J. Geriatr. Psychiatry 21, 433–449. http://dx.doi.org/10.1016/ variants in Alzheimer’s disease. N. Engl. J. Med. 368, 117–127. http://dx.doi.org/
j.jagp.2012.10.026. 10.1056/NEJMoa1211851.
Ekdahl, C.T., Claasen, J.-H., Bonde, S., Kokaia, Z., Lindvall, O., 2003. Inflammation is Guillot-Sestier, M.V., Doty, K.R., Gate, D., Rodriguez, J., Leung, B.P., Rezai-Zadeh, K.,
detrimental for neurogenesis in adult brain. Proc. Natl. Acad. Sci. U.S.A. 100, Town, T., 2015. Il10 deficiency rebalances innate immunity to mitigate
13632–13637. http://dx.doi.org/10.1073/pnas.2234031100. Alzheimer-like pathology article Il10 deficiency rebalances innate immunity
El Khoury, J., Toft, M., Hickman, S.E., Means, T.K., Terada, K., Geula, C., Luster, A.D., to mitigate Alzheimer-like pathology. Neuron 85, 1–15. http://dx.doi.org/
2007. Ccr2 deficiency impairs microglial accumulation and accelerates 10.1016/j.neuron.2014.12.068.
progression of Alzheimer-like disease. Nat. Med. 13, 432–438. http://dx.doi. Gulaj, E., Pawlak, K., Bien, B., Pawlak, D., 2010. Kynurenine and its metabolites in
org/10.1038/nm1555. Alzheimer’s disease patients. Adv. Med. Sci. 55, 204–211. http://dx.doi.org/
Eriksson, P.S., Perfilieva, E., Björk-Eriksson, T., Alborn, A.M., Nordborg, C., Peterson, 10.2478/v10039-010-0023-6.
D.A., Gage, F.H., 1998. Neurogenesis in the adult human hippocampus. Nat. Hart, B., 1988. Biological basis of the behavior of sick animals. Neurosci. Biobehav.
Med. 4, 1313–1317. http://dx.doi.org/10.1038/3305. Rev. 12, 123–137.

Please cite this article in press as: Santos, L.E., et al. Microglial dysfunction connects depression and Alzheimer’s disease. Brain Behav. Immun. (2015),
http://dx.doi.org/10.1016/j.bbi.2015.11.011
L.E. Santos et al. / Brain, Behavior, and Immunity xxx (2015) xxx–xxx 11

Hauser, P.S., Ryan, R.O., 2013. Impact of apolipoprotein E on Alzheimer’s disease. Jorge, R.E., Robinson, R.G., Moser, D., Tateno, A., Crespo-Facorro, B., Arndt, S., 2004.
Curr. Alzheimer Res. 10, 809–817. http://dx.doi.org/10.2174/ Major depression following traumatic brain injury. Arch. Gen. Psychiatry 61,
15672050113109990156. 42–50. http://dx.doi.org/10.1001/archpsyc.61.1.42.
Hellwig, S., Heinrich, A., Biber, K., 2013. The brain’s best friend: microglial Kales, H.C., Gitlin, L.N., Lyketsos, C.G., 2015. Assessment and management of
neurotoxicity revisited. Front. Cell. Neurosci. 7, 71. http://dx.doi.org/10.3389/ behavioral and psychological symptoms of dementia. BMJ 350, h369. http://dx.
fncel.2013.00071. doi.org/10.1136/bmj.h369.
Heneka, M.T., Kummer, M.P., Latz, E., 2014. Innate immune activation in Kang, J.Y., Lee, J.S., Kang, H., Lee, H.-W., Kim, Y.K., Jeon, H.J., Chung, J.-K., Lee, M.C.,
neurodegenerative disease. Nat. Rev. Immunol. 14, 463–477. http://dx.doi.org/ Cho, M.J., Lee, D.S., 2012. Regional cerebral blood flow abnormalities associated
10.1038/nri3705. with apathy and depression in Alzheimer disease. Alzheimer Dis. Assoc. Disord.
Heneka, M.T., Carson, M.J., Khoury, J. .El., Landreth, G.E., Brosseron, F., Feinstein, D.L., 26, 217–224. http://dx.doi.org/10.1097/WAD.0b013e318231e5fc.
Jacobs, A.H., Wyss-Coray, T., Vitorica, J., Ransohoff, R.M., Herrup, K., Frautschy, S. Karch, C.M., Cruchaga, C., Goate, A.M., 2014. Alzheimer’s disease genetics: from the
A., Finsen, B., Brown, G.C., Verkhratsky, A., Yamanaka, K., Koistinaho, J., Latz, E., bench to the clinic. Neuron 83, 11–26. http://dx.doi.org/10.1016/j.
Halle, A., Petzold, G.C., Town, T., Morgan, D., Shinohara, M.L., Perry, V.H., neuron.2014.05.041.
Holmes, C., Bazan, N.G., Brooks, D.J., Hunot, S., Joseph, B., Deigendesch, N., Karege, F., Perret, G., Bondolfi, G., Schwald, M., Bertschy, G., Aubry, J.M., 2002.
Garaschuk, O., Boddeke, E., Dinarello, C.A., Breitner, J.C., Cole, G.M., Golenbock, Decreased serum brain-derived neurotrophic factor levels in major depressed
D.T., Kummer, M.P., 2015a. Neuroinflammation in Alzheimer’s disease. Lancet patients. Psychiatry Res. 109, 143–148. http://dx.doi.org/10.1016/S0165-1781
Neurol. 14, 388–405. http://dx.doi.org/10.1016/S1474-4422(15)70016-5. (02)00005-7.
Heneka, M.T., Golenbock, D.T., Latz, E., 2015b. Innate immunity in Alzheimer’s Kataoka, K., Hashimoto, H., Kawabe, J., Higashiyama, S., Akiyama, H., Shimada, A.,
disease. Nat. Immunol. 16, 229–236. http://dx.doi.org/10.1038/ni.3102. Kai, T., Inoue, K., Shiomi, S., Kiriike, N., 2010. Frontal hypoperfusion in depressed
Hickman, S.E., Allison, E.K., El Khoury, J., 2008. Microglial dysfunction and defective patients with dementia of Alzheimer type demonstrated on 3DSRT. Psychiatry
beta-amyloid clearance pathways in aging Alzheimer’s disease mice. J. Clin. Neurosci. 64, 293–298. http://dx.doi.org/10.1111/j.1440-
Neurosci. 28, 8354–8360. http://dx.doi.org/10.1523/JNEUROSCI.0616-08.2008. 1819.2010.02083.x.
Holmes, C., Cunningham, C., Zotova, E., Woolford, J., Dean, C., Kerr, S., Culliford, D., Katon, W., Pedersen, H.S., Ribe, A.R., Fenger-Grøn, M., Davydow, D., Waldorff, F.B.,
Perry, V.H., 2009. Systemic inflammation and disease progression in Alzheimer Vestergaard, M., 2015. Effect of depression and diabetes mellitus on the risk for
disease. Neurology 73, 768–774. http://dx.doi.org/10.1212/ dementia. JAMA Psychiatry 72, 612. http://dx.doi.org/
WNL.0b013e3181b6bb95. 10.1001/jamapsychiatry.2015.0082.
Holsinger, R.M., Schnarr, J., Henry, P., Castelo, V.T., Fahnestock, M., 2000. Kempermann, G., 2002. Regulation of adult hippocampal neurogenesis –
Quantitation of BDNF mRNA in human parietal cortex by competitive reverse implications for novel theories of major depression. Bipolar Disord. 4, 17–33.
transcription-polymerase chain reaction: decreased levels in Alzheimer’s http://dx.doi.org/10.1034/j.1399-5618.2002.40101.x.
disease. Brain Res. Mol. Brain Res. 76, 347–354. Kempermann, G., Kronenberg, G., 2003. Depressed new neurons – adult
Holthoff, V.A., Beuthien-Baumann, B., Kalbe, E., Lüdecke, S., Lenz, O., Zündorf, G., hippocampal neurogenesis and a cellular plasticity hypothesis of major
Spirling, S., Schierz, K., Winiecki, P., Sorbi, S., Herholz, K., 2005. Regional cerebral depression. Biol. Psychiatry 54, 499–503. http://dx.doi.org/10.1016/S0006-
metabolism in early Alzheimer’s disease with clinically significant apathy or 3223(03)00319-6.
depression. Biol. Psychiatry 57, 412–421. http://dx.doi.org/10.1016/j. Kent, S., Bluthé, R.M., Kelley, K.W., Dantzer, R., 1992. Sickness behavior as a new
biopsych.2004.11.035. target for drug development. Trends Pharmacol. Sci. 13, 24–28. http://dx.doi.
Holtzheimer, P.E., Mayberg, H.S., 2011. Stuck in a rut: rethinking depression and its org/10.1016/0165-6147(92)90012-U.
treatment. Trends Neurosci. 34, 1–9. http://dx.doi.org/10.1016/j. Khemka, V.K., Ganguly, A., Bagchi, D., Ghosh, A., Bir, A., Biswas, A., Chattopadhyay, S.,
tins.2010.10.004. Chakrabarti, S., 2014. Raised serum proinflammatory cytokines in Alzheimer’s
Hoshaw, B.A., Hill, T.I., Crowley, J.J., Malberg, J.E., Khawaja, X., Rosenzweig-Lipson, S., disease with depression. Aging Dis. 5, 170–176. http://dx.doi.org/10.14336/
Schechter, L.E., Lucki, I., 2008. Antidepressant-like behavioral effects of IGF-I AD.2014.0500170.
produced by enhanced serotonin transmission. Eur. J. Pharmacol. 594, 109–116. Kierdorf, K., Erny, D., Goldmann, T., Sander, V., Schulz, C., Perdiguero, E.G.,
http://dx.doi.org/10.1016/j.ejphar.2008.07.023. Wieghofer, P., Heinrich, A., Riemke, P., Hölscher, C., Müller, D.N., Luckow, B.,
Hsieh, C.L., Koike, M., Spusta, S.C., Niemi, E.C., Yenari, M., Nakamura, M.C., Seaman, Brocker, T., Debowski, K., Fritz, G., Opdenakker, G., Diefenbach, A., Biber, K.,
W.E., 2009. A role for TREM2 ligands in the phagocytosis of apoptotic neuronal Heikenwalder, M., Geissmann, F., Rosenbauer, F., Prinz, M., 2013. Microglia
cells by microglia. J. Neurochem. 109, 1144–1156. http://dx.doi.org/10.1111/ emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent
j.1471-4159.2009.06042.x. pathways. Nat. Neurosci. 16, 273–280. http://dx.doi.org/10.1038/nn.3318.
Hu, X., Leak, R.K., Shi, Y., Suenaga, J., Gao, Y., Zheng, P., Chen, J., 2014. Microglial and Kim, H.J., Kim, W., Kong, S.Y., 2013. Antidepressants for neuro-regeneration: from
macrophage polarization—new prospects for brain repair. Nat. Rev. Neurol. 11, depression to Alzheimer’s disease. Arch. Pharm. Res. 36, 1279–1290. http://dx.
56–64. http://dx.doi.org/10.1038/nrneurol.2014.207. doi.org/10.1007/s12272-013-0238-8.
Hurt, C., Bhattacharyya, S., Burns, A., Camus, V., Liperoti, R., Marriott, A., Nobili, F., Klempin, F., Beis, D., Mosienko, V., Kempermann, G., Bader, M., Alenina, N., 2013.
Robert, P., Tsolaki, M., Vellas, B., Verhey, F., Byrne, E.J., 2008. Patient and Serotonin is required for exercise-induced adult hippocampal neurogenesis. J.
caregiver perspectives of quality of life in dementia: an investigation of the Neurosci. 33, 8270–8275. http://dx.doi.org/10.1523/JNEUROSCI.5855-12.2013.
relationship to behavioural and psychological symptoms in dementia. Dement. Klunk, W.E., Engler, H., Nordberg, A., Wang, Y., Blomqvist, G., Holt, D.P., Bergström,
Geriatr. Cogn. Disord. 26, 138–146. http://dx.doi.org/10.1159/000149584. M., Savitcheva, I., Huang, G.F., Estrada, S., Ausén, B., Debnath, M.L., Barletta, J.,
Irwin, M.R., Miller, A.H., 2007. Depressive disorders and immunity: 20 years of Price, J.C., Sandell, J., Lopresti, B.J., Wall, A., Koivisto, P., Antoni, G., Mathis, C.A.,
progress and discovery. Brain Behav. Immun. 21, 374–383. http://dx.doi.org/ Långström, B., 2004. Imaging Brain Amyloid in Alzheimer’s Disease with
10.1016/j.bbi.2007.01.010. Pittsburgh Compound-B. Ann. Neurol. 55, 306–319. http://dx.doi.org/10.1002/
Jacobs, B.L., van Praag, H., Gage, F.H., 2000. Adult brain neurogenesis and psychiatry: ana.20009.
a novel theory of depression. Mol. Psychiatry 5, 262–269. http://dx.doi.org/ Knesevich, J.W., Martin, R.L., Berg, L., Danziger, W., 1983. Preliminary report on
10.1038/sj.mp.4000712. affective symptoms in the early stages of senile dementia of the Alzheimer type.
Janzing, J.G.E., Hooijer, C., Van ’T Hof, M.A., Zitman, F.G., 2002. Depression in Am. J. Psychiatry 140, 233–235.
subjects with and without dementia: a comparison using GMS-AGECAT. Int. J. Kobayashi, K., Imagama, S., Ohgomori, T., Hirano, K., Uchimura, K., Sakamoto, K.,
Geriatr. Psychiatry 17, 1–5. http://dx.doi.org/10.1002/gps.526. Hirakawa, A., Takeuchi, H., Suzumura, A., Ishiguro, N., Kadomatsu, K., 2013.
Jay, T.R., Miller, C.M., Cheng, P.J., Graham, L.C., Bemiller, S., Broihier, M.L., Xu, G., Minocycline selectively inhibits M1 polarization of microglia. Cell Death Dis. 4,
Margevicius, D., Karlo, J.C., Sousa, G.L., Cotleur, A.C., Butovsky, O., Bekris, L., e525. http://dx.doi.org/10.1038/cddis.2013.54.
Staugaitis, S.M., Leverenz, J.B., Pimplikar, S.W., Landreth, G.E., Howell, G.R., Köhler, O., Benros, M.E., Nordentoft, M., Farkouh, M.E., Iyengar, R.L., Mors, O., Krogh,
Ransohoff, R.M., Lamb, B.T., 2015. TREM2 deficiency eliminates TREM2+ J., 2014. Effect of anti-inflammatory treatment on depression, depressive
inflammatory macrophages and ameliorates pathology in Alzheimer’s disease symptoms, and adverse effects. JAMA Psychiatry 71, 1381. http://dx.doi.org/
mouse models. J. Exp. Med. 212, 287–295. http://dx.doi.org/ 10.1001/jamapsychiatry.2014.1611.
10.1084/jem.20142322. Kohman, R.A., DeYoung, E.K., Bhattacharya, T.K., Peterson, L.N., Rhodes, J.S., 2012.
Jin, K., Peel, A.L., Mao, X.O., Xie, L., Cottrell, B.A., Henshall, D.C., Greenberg, D.A., Wheel running attenuates microglia proliferation and increases expression of a
2004. Increased hippocampal neurogenesis in Alzheimer’s disease. Proc. Natl. proneurogenic phenotype in the hippocampus of aged mice. Brain Behav.
Acad. Sci. U.S.A. 101, 343–347. http://dx.doi.org/10.1073/pnas.2634794100. Immun. 26, 803–810. http://dx.doi.org/10.1016/j.bbi.2011.10.006.
Johansson, J.U., Woodling, N.S., Wang, Q., Panchal, M., Liang, X., Trueba-Saiz, A., Kohman, R.A., Bhattacharya, T.K., Wojcik, E., Rhodes, J.S., 2013. Exercise reduces
Brown, H.D., Mhatre, S.D., Loui, T., Andreasson, K.I., 2014. Prostaglandin activation of microglia isolated from hippocampus and brain of aged mice. J.
signaling suppresses beneficial microglial function in Alzheimer’s Neuroinflamm. 10, 114. http://dx.doi.org/10.1186/1742-2094-10-114.
disease models. J. Clin. Invest. 125, 350–364. http://dx.doi.org/10.1172/ Kolodziejczak, M., Béchade, C., Gervasi, N., Irinopoulou, T., Banas, S.M., Cordier, C.,
JCI77487. Rebsam, A., Roumier, A., Maroteaux, L., 2015. Serotonin modulates
Jonsson, T., Stefansson, H., Steinberg, S., Jonsdottir, I., Jonsson, P.V., Snaedal, J., developmental microglia via 5-HT 2B receptors: potential implication during
Bjornsson, S., Huttenlocher, J., Levey, A.I., Lah, J.J., Rujescu, D., Hampel, H., synaptic refinement of retinogeniculate projections. ACS Chem. Neurosci. 6,
Giegling, I., Andreassen, O.A., Engedal, K., Ulstein, I., Djurovic, S., Ibrahim- 1219–1230. http://dx.doi.org/10.1021/cn5003489.
Verbaas, C., Hofman, A., Ikram, M.A., van Duijn, C.M., Thorsteinsdottir, U., Kong, Kopczak, A., Stalla, G.K., Uhr, M., Lucae, S., Hennings, J., Ising, M., Holsboer, F.,
A., Stefansson, K., 2013. Variant of TREM2 associated with the risk of Kloiber, S., 2015. IGF-I in major depression and antidepressant treatment
Alzheimer’s disease. N. Engl. J. Med. 368, 107–116. http://dx.doi.org/10.1056/ response. Eur. Neuropsychopharmacol. 25, 864–872. http://dx.doi.org/10.1016/
NEJMoa1211103. j.euroneuro.2014.12.013.

Please cite this article in press as: Santos, L.E., et al. Microglial dysfunction connects depression and Alzheimer’s disease. Brain Behav. Immun. (2015),
http://dx.doi.org/10.1016/j.bbi.2015.11.011
12 L.E. Santos et al. / Brain, Behavior, and Immunity xxx (2015) xxx–xxx

Krabbe, G., Matyash, V., Pannasch, U., Mamer, L., Boddeke, H.W.G.M., Kettenmann, Maddison, D.C., Giorgini, F., 2015. The kynurenine pathway and neurodegenerative
H., 2012. Activation of serotonin receptors promotes microglial injury-induced disease. Semin. Cell Dev. Biol. 44. http://dx.doi.org/10.1016/j.
motility but attenuates phagocytic activity. Brain Behav. Immun. 26, 419–428. semcdb.2015.03.002.
http://dx.doi.org/10.1016/j.bbi.2011.12.002. Maes, M., Leonard, B.E., Myint, A.M., Kubera, M., Verkerk, R., 2011. The new ‘‘5-HT”
Kreisel, T., Frank, M.G., Licht, T., Reshef, R., Ben-Menachem-Zidon, O., Baratta, M.V., hypothesis of depression. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 35,
Maier, S.F., Yirmiya, R., 2014. Dynamic microglial alterations underlie stress- 702–721. http://dx.doi.org/10.1016/j.pnpbp.2010.12.017.
induced depressive-like behavior and suppressed neurogenesis. Mol. Psychiatry Malberg, J.E., Platt, B., Rizzo, S.J.S., Ring, R.H., Lucki, I., Schechter, L.E., Rosenzweig-
19, 699–709. http://dx.doi.org/10.1038/mp.2013.155. Lipson, S., 2007. Increasing the levels of insulin-like growth factor-I by an IGF
Krstic, D., Knuesel, I., 2013. Deciphering the mechanism underlying late-onset binding protein inhibitor produces anxiolytic and antidepressant-like effects.
Alzheimer disease. Nat. Rev. Neurol. 9, 25–34. http://dx.doi.org/10.1038/ Neuropsychopharmacology 32, 2360–2368. http://dx.doi.org/10.1038/sj.
nrneurol.2012.236. npp.1301358.
Lacasse, J.R., Leo, J., 2005. Serotonin and depression: a disconnect between the Mapstone, M., Cheema, A.K., Fiandaca, M.S., Zhong, X., Mhyre, T.R., MacArthur, L.H.,
advertisements and the scientific literature. PLoS Med. http://dx.doi.org/ Hall, W.J., Fisher, S.G., Peterson, D.R., Haley, J.M., Nazar, M.D., Rich, S.A., Berlau,
10.1371/journal.pmed.0020392. D.J., Peltz, C.B., Tan, M.T., Kawas, C.H., Federoff, H.J., 2014. Plasma phospholipids
Lalancette-Hébert, M., Gowing, G., Simard, A., Weng, Y.C., Kriz, J., 2007. Selective identify antecedent memory impairment in older adults. Nat. Med. 20, 415–
ablation of proliferating microglial cells exacerbates ischemic injury in the 418. http://dx.doi.org/10.1038/nm.3466.
brain. J. Neurosci. 27, 2596–2605. http://dx.doi.org/10.1523/JNEUROSCI.5360- Mårtensson, U., Nässberger, L., 1993. Influence of antidepressants on mitogen
06.2007. stimulation of human lymphocytes. Toxicol. In Vitro 7, 241–245. http://dx.doi.
Lapchak, P.A., Araujo, D.M., Hefti, F., 1993. Systemic interleukin-1 beta decreases org/10.1016/0887-2333(93)90007-R.
brain-derived neurotrophic factor messenger RNA expression in the rat Martínez, A., Alcántara, S., Borrell, V., Del Río, J.A., Blasi, J., Otal, R., Campos, N.,
hippocampal formation. Neuroscience 53, 297–301, 0306-4522(93)90196-M Boronat, A., Barbacid, M., Silos-Santiago, I., Soriano, E., 1998. TrkB and TrkC
[pii]. signaling are required for maturation and synaptogenesis of hippocampal
Laske, C., Stransky, E., Leyhe, T., Eschweiler, G.W., Maetzler, W., Wittorf, A., connections. J. Neurosci. 18, 7336–7350.
Soekadar, S., Richartz, E., Koehler, N., Bartels, M., Buchkremer, G., Schott, K., Mattson, M.P., Maudsley, S., Martin, B., 2004. A neural signaling triumvirate that
2007. BDNF serum and CSF concentrations in Alzheimer’s disease, normal influences ageing and age-related disease: insulin/IGF-1, BDNF and serotonin.
pressure hydrocephalus and healthy controls. J. Psychiatr. Res. 41, 387–394. Ageing Res. Rev. 3, 445–464. http://dx.doi.org/10.1016/j.arr.2004.08.001.
http://dx.doi.org/10.1016/j.jpsychires.2006.01.014. Mawuenyega, K.G., Sigurdson, W., Ovod, V., Munsell, L., Kasten, T., Morris, J.C.,
Laws, S.M., Perneczky, R., Wagenpfeil, S., Müller, U., Förstl, H., Martins, R.N., Kurz, A., Yarasheski, K.E., Bateman, R.J., 2010. Decreased clearance of CNS beta-amyloid
Riemenschneider, M., 2005. TNF polymorphisms in Alzheimer disease and in Alzheimer’s disease. Science 330, 1774. http://dx.doi.org/
functional implications on CSF beta-amyloid levels. Hum. Mutat. 26, 29–35. 10.1126/science.1197623.
http://dx.doi.org/10.1002/humu.20180. Merriam, A.E., Aronson, M.K., Gaston, P., Wey, S.L., Katz, I., 1988. The psychiatric
Lazarov, O., Marr, R.A., 2013. Of mice and men: neurogenesis, cognition, and symptoms of Alzheimer’s disease. J. Am. Geriatr. Soc. 36, 7–12.
Alzheimer’s disease. Front. Aging Neurosci. 5, 1–8. http://dx.doi.org/10.3389/ Michaud, M., Balardy, L., Moulis, G., Gaudin, C., Peyrot, C., Vellas, B., Cesari, M.,
fnagi.2013.00043. Nourhashemi, F., 2013. Proinflammatory cytokines, aging, and age-related
Ledo, J.H., Azevedo, E.P., Clarke, J.R., Ribeiro, F.C., Figueiredo, C.P., Foguel, D., De diseases. J. Am. Med. Dir. Assoc. 14, 877–882. http://dx.doi.org/10.1016/
Felice, F.G., Ferreira, S.T., 2013. Amyloid-b oligomers link depressive-like j.jamda.2013.05.009.
behavior and cognitive deficits in mice. Mol. Psychiatry 18, 1053–1054. Mitschelen, M., Yan, H., Farley, J.A., Warrington, J.P., Han, S., Hereñú, C.B., Csiszar, A.,
http://dx.doi.org/10.1038/mp.2012.168. Ungvari, Z., Bailey-Downs, L.C., Bass, C.E., Sonntag, W.E., 2011. Long-term
Lee, H.B., Lyketsos, C.G., 2003. Depression in Alzheimer’s disease: heterogeneity and deficiency of circulating and hippocampal insulin-like growth factor I induces
related issues. Biol. Psychiatry 54, 353–362. http://dx.doi.org/10.1016/S0006- depressive behavior in adult mice: a potential model of geriatric depression.
3223(03)00543-2. Neuroscience 185, 50–60. http://dx.doi.org/10.1016/j.
Lee, Y.-K., Hou, S.-W., Lee, C.-C., Hsu, C.-Y., Huang, Y.-S., Su, Y.-C., 2013. Increased neuroscience.2011.04.032.
Risk of Dementia in Patients with Mild Traumatic Brain Injury: A Nationwide Mittelbronn, M., 2014. The M1/M2 immune polarization concept in microglia: a fair
Cohort Study. PLoS One 8, e62422. http://dx.doi.org/10.1371/journal. transfer? Neuroimmunol. Neuroinflamm. 1, 6. http://dx.doi.org/10.4103/2347-
pone.0062422. 8659.135567.
Levy-Cooperman, N., Burhan, A.M., Rafi-Tari, S., Kusano, M., Ramirez, J., Caldwell, C., Modrego, P.J., Ferrández, J., 2004. Depression in patients with mild cognitive
Black, S.E., 2008. Frontal lobe hypoperfusion and depressive symptoms in impairment increases the risk of developing dementia of Alzheimer type: a
Alzheimer disease. J. Psychiatry Neurosci. 33, 218–226. prospective cohort study. Arch. Neurol. 61, 1290–1293. http://dx.doi.org/
Li, Y., Du, X.F., Liu, C.S., Wen, Z.L., Du, J.L., 2012. Reciprocal regulation between 10.1001/archneur.61.8.1290.
resting microglial dynamics and neuronal activity in vivo. Dev. Cell 23, 1189– Molendijk, M.L., Spinhoven, P., Polak, M., Bus, B.A.A., Penninx, B.W.J.H., Elzinga, B.M.,
1202. http://dx.doi.org/10.1016/j.devcel.2012.10.027. 2014. Serum BDNF concentrations as peripheral manifestations of depression:
Lin, F., Suhr, J., Diebold, S., Heffner, K.L., 2014. Associations between depressive evidence from a systematic review and meta-analyses on 179 associations
symptoms and memory deficits vary as a function of insulin-like growth factor (N=9484). Mol. Psychiatry 19, 791–800. http://dx.doi.org/10.1038/
(IGF-1) levels in healthy older adults. Psychoneuroendocrinology 42, 118–123. mp.2013.105.
http://dx.doi.org/10.1016/j.psyneuen.2014.01.006. Moloney, A.M., Griffin, R.J., Timmons, S., O’Connor, R., Ravid, R., O’Neill, C., 2010.
Lu, B., Nagappan, G., Guan, X., Nathan, P.J., Wren, P., 2013. BDNF-based synaptic Defects in IGF-1 receptor, insulin receptor and IRS-1/2 in Alzheimer’s disease
repair as a disease-modifying strategy for neurodegenerative diseases. Nat. Rev. indicate possible resistance to IGF-1 and insulin signalling. Neurobiol. Aging 31,
Neurosci. 14, 401–416, nrn3505 [pii]nr10.1038/nrn3505. 224–243. http://dx.doi.org/10.1016/j.neurobiolaging.2008.04.002.
Luo, X.-G., Ding, J.-Q., Chen, S.-D., 2010. Microglia in the aging brain: relevance to Mondelli, V., Cattaneo, A., Murri, M.B., Forti, M., Di Handley, R., Hepgul, N., Miorelli,
neurodegeneration. Mol. Neurodegener. 5, 12. http://dx.doi.org/10.1186/1750- A., Navari, S., Papadopoulos, A.S., Aitchison, K.J., Morgan, C., Murray, R.M.,
1326-5-12. Dazzan, P., Pariante, C.M., 2011. Stress and inflammation reduce brain-derived
Lyketsos, C.G., Olin, J., 2002. Depression in Alzheimer’s disease: overview and neurotrophic factor expression in first-episode psychosis: a pathway to smaller
treatment. Biol. Psychiatry 52, 243–252. http://dx.doi.org/10.1016/S0006-3223 hippocampal volume. J. Clin. Psychiatry 72, 1677–1684. http://dx.doi.org/
(02)01348-3. 10.4088/JCP.10m06745.
Lyketsos, C.G., Steinberg, M., Tschanz, J.T., Norton, M.C., Steffens, D.C., Breitner, J.C.S., Monje, M.L., Toda, H., Palmer, T.D., 2003. Inflammatory blockade restores adult
2000. Mental and behavioral disturbance in dementia: findings from the Cache hippocampal neurogenesis. Science 302, 1760–1765. http://dx.doi.org/
County Study on Memory in Aging. Am. J. Psychiatry 157, 708–714. http://dx. 10.1126/science.1088417.
doi.org/10.1176/appi.ajp.157.5.708. Morales, I., Guzmán-Martínez, L., Cerda-Troncoso, C., Farías, G.A., Maccioni, R.B.,
Lyketsos, C.G., Lopez, O., Jones, B., Fitzpatrick, A.L., Breitner, J., DeKosky, S., 2002. 2014. Neuroinflammation in the pathogenesis of Alzheimer’s disease. A rational
Prevalence of neuropsychiatric symptoms in dementia and mild cognitive framework for the search of novel therapeutic approaches. Front. Cell. Neurosci.
impairment: results from the cardiovascular health study. JAMA 288, 1475– 8, 112. http://dx.doi.org/10.3389/fncel.2014.00112.
1483. http://dx.doi.org/10.1001/jama.288.12.1475. Morgan, S.C., Taylor, D.L., Pocock, J.M., 2004. Microglia release activators of
Lyketsos, C.G., DelCampo, L., Steinberg, M., Miles, Q., Steele, C.D., Munro, C., Baker, A. neuronal proliferation mediated by activation of mitogen-activated protein
S., Sheppard, J.-M.E., Frangakis, C., Brandt, J., Rabins, P.V., 2003. Treating kinase, phosphatidylinositol-3-kinase/Akt and delta-Notch signalling cascades.
depression in Alzheimer disease. Arch. Gen. Psychiatry 60, 737. http://dx.doi. J. Neurochem. 90, 89–101. http://dx.doi.org/10.1111/j.1471-4159.2004.
org/10.1001/archpsyc.60.7.737. 02461.x.
Lyketsos, C.G., Carrillo, M.C., Ryan, J.M., Khachaturian, A.S., Trzepacz, P., Amatniek, J., Mu, Y., Gage, F.H., 2011. Adult hippocampal neurogenesis and its role in Alzheimer’s
Cedarbaum, J., Brashear, R., Miller, D.S., 2011. Neuropsychiatric symptoms in disease. Mol. Neurodegener. 6, 85. http://dx.doi.org/10.1186/1750-1326-6-85.
Alzheimer’s disease. Alzheimer’s Dement. http://dx.doi.org/10.1016/ Müller, N., 2010. COX-2 inhibitors as antidepressants and antipsychotics: clinical
j.jalz.2011.05.2410. evidence. Curr. Opin. Investig. Drugs 11, 31–42.
Lynch, M.A., 2014. The impact of neuroimmune changes on development of amyloid Müller, N., Schwarz, M.J., 2007. The immune-mediated alteration of serotonin and
pathology; relevance to Alzheimer’s disease. Immunology 141, 292–301. http:// glutamate: towards an integrated view of depression. Mol. Psychiatry 12, 988–
dx.doi.org/10.1111/imm.12156. 1000. http://dx.doi.org/10.1038/sj.mp.4002006.
Macchi, F., Homberg, J.R., Calabrese, F., Zecchillo, C., Racagni, G., Riva, M.A., Molteni, Myint, A.M., Kim, Y.K., 2003. Cytokine-serotonin interaction through IDO: a
R., 2013. Altered inflammatory responsiveness in serotonin transporter mutant neurodegeneration hypothesis of depression. Med. Hypotheses 61, 519–525.
rats. J. Neuroinflamm. 10, 116. http://dx.doi.org/10.1186/1742-2094-10-116. http://dx.doi.org/10.1016/S0306-9877(03)00207-X.

Please cite this article in press as: Santos, L.E., et al. Microglial dysfunction connects depression and Alzheimer’s disease. Brain Behav. Immun. (2015),
http://dx.doi.org/10.1016/j.bbi.2015.11.011
L.E. Santos et al. / Brain, Behavior, and Immunity xxx (2015) xxx–xxx 13

Nakanishi, M., Niidome, T., Matsuda, S., Akaike, A., Kihara, T., Sugimoto, H., 2007. Paslakis, G., Blum, W.F., Deuschle, M., 2012. Intranasal insulin-like growth factor I
Microglia-derived interleukin-6 and leukaemia inhibitory factor promote (IGF-I) as a plausible future treatment of depression. Med. Hypotheses 79, 222–
astrocytic differentiation of neural stem/progenitor cells. Eur. J. Neurosci. 25, 225. http://dx.doi.org/10.1016/j.mehy.2012.04.045.
649–658. http://dx.doi.org/10.1111/j.1460-9568.2007.05309.x. Peng, S., Wuu, J., Mufson, E.J., Fahnestock, M., 2005. Precursor form of brain-derived
Naranjo, D.M., Fisher, L., Arean, P.A., Hessler, D., Mullan, J., 2011. Patients with type neurotrophic factor and mature brain-derived neurotrophic factor are
2 diabetes at risk for major depressive disorder over time. Ann. Fam. Med. 9, decreased in the pre-clinical stages of Alzheimer’s disease. J. Neurochem. 93,
115–120. http://dx.doi.org/10.1370/afm.1212. 1412–1421. http://dx.doi.org/10.1111/j.1471-4159.2005.03135.x.
Nelson, J.C., 2001. Diagnosing and treating depression in the elderly. J. Clin. Perry, V.H., Holmes, C., 2014. Microglial priming in neurodegenerative disease. Nat.
Psychiatry 62, 18–22. Rev. Neurol. 10, 217–224. http://dx.doi.org/10.1038/nrneurol.2014.38.
Nelson, J.C., Devanand, D.P., 2011. A systematic review and meta-analysis of Perry, V.H., Cunningham, C., Holmes, C., 2007. Systemic infections and inflammation
placebo-controlled antidepressant studies in people with depression and affect chronic neurodegeneration. Nat. Rev. Immunol. 7, 161–167. http://dx.doi.
dementia. J. Am. Geriatr. Soc. 59, 577–585. http://dx.doi.org/10.1111/j.1532- org/10.1038/nri2015.
5415.2011.03355.x. Pocock, J.M., Kettenmann, H., 2007. Neurotransmitter receptors on microglia.
Ng, F., Berk, M., Dean, O., Bush, A.I., 2008. Oxidative stress in psychiatric disorders: Trends Neurosci. 30, 527–535. http://dx.doi.org/10.1016/j.tins.2007.07.007.
evidence base and therapeutic implications. Int. J. Neuropsychopharmacol. 11, Porta-Etessam, J., Tobaruela-González, J.L., Rabes-Berendes, C., 2011. Depression in
851–876. http://dx.doi.org/10.1017/S1461145707008401. patients with moderate Alzheimer disease. Alzheimer Dis. Assoc. Disord. 25,
Nimmerjahn, A., Kirchhoff, F., Helmchen, F., 2005. Resting microglial cells are highly 317–325. http://dx.doi.org/10.1097/WAD.0b013e31820e7c45.
dynamic surveillants of brain parenchyma in vivo. Neuroforum 11, 95–96. Powell, T.R., Schalkwyk, L.C., Heffernan, A.L., Breen, G., Lawrence, T., Price, T.,
http://dx.doi.org/10.1126/science.1110647. Farmer, A.E., Aitchison, K.J., Craig, I.W., Danese, A., Lewis, C., McGuffin, P., Uher,
Norden, D.M., Muccigrosso, M.M., Godbout, J.P., 2015. Microglial priming and R., Tansey, K.E., D’Souza, U.M., 2013. Tumor necrosis factor and its targets in the
enhanced reactivity to secondary insult in aging, and traumatic CNS injury, and inflammatory cytokine pathway are identified as putative transcriptomic
neurodegenerative disease. Neuropharmacology 96, 29–41. http://dx.doi.org/ biomarkers for escitalopram response. Eur. Neuropsychopharmacol. 23, 1105–
10.1016/j.neuropharm.2014.10.028. 1114. http://dx.doi.org/10.1016/j.euroneuro.2012.09.009.
O’Connor, J.C., Lawson, M.A., André, C., Moreau, M., Lestage, J., Castanon, N., Kelley, Prinz, M., Priller, J., 2014. Microglia and brain macrophages in the molecular age:
K.W., Dantzer, R., 2009. Lipopolysaccharide-induced depressive-like behavior is from origin to neuropsychiatric disease. Nat. Rev. Neurosci. 15, 300–312. http://
mediated by indoleamine 2,3-dioxygenase activation in mice. Mol. Psychiatry dx.doi.org/10.1038/nrn3722.
14, 511–522. http://dx.doi.org/10.1038/sj.mp.4002148. Raison, C.L., Capuron, L., Miller, A.H., 2006. Cytokines sing the blues: inflammation
Okereke, O.I., Kang, J.H., Ma, J., Gaziano, J.M., Grodstein, F., 2006. Midlife plasma and the pathogenesis of depression. Trends Immunol. 27, 24–31. http://dx.doi.
insulin-like growth factor I and cognitive function in older men. J. Clin. org/10.1016/j.it.2005.11.006.
Endocrinol. Metab. 91, 4306–4312. http://dx.doi.org/10.1210/jc.2006-1325. Rauskolb, S., Zagrebelsky, M., Dreznjak, A., Deogracias, R., Matsumoto, T., Wiese, S.,
Olin, J.T., Schneider, L.S., Katz, I.R., Meyers, B.S., Alexopoulos, G.S., Breitner, J.C., Erne, B., Sendtner, M., Schaeren-Wiemers, N., Korte, M., Barde, Y.-A., 2010.
Bruce, M.L., Caine, E.D., Cummings, J.L., Devanand, D.P., Krishnan, K.R.R., Global deprivation of brain-derived neurotrophic factor in the CNS reveals an
Lyketsos, C.G., Lyness, J.M., Rabins, P.V., Reynolds, C.F., Rovner, B.W., Steffens, area-specific requirement for dendritic growth. J. Neurosci. 30, 1739–1749.
D.C., Tariot, P.N., Lebowitz, B.D., 2002. Provisional diagnostic criteria for http://dx.doi.org/10.1523/JNEUROSCI.5100-09.2010.
depression of Alzheimer disease. Am. J. Geriatr. Psychiatry 10, 125–128. Reichenberg, A., Yirmiya, R., Schuld, A., Kraus, T., Haack, M., Morag, A., Pollmächer,
http://dx.doi.org/10.1097/00019442-200203000-00003. T., 2001. Cytokine-associated emotional and cognitive disturbances in humans.
Orre, M., Kamphuis, W., Osborn, L.M., Jansen, A.H.P., Kooijman, L., Bossers, K., Hol, E. Arch. Gen. Psychiatry 58, 445–452. http://dx.doi.org/10.1001/
M., 2014. Isolation of glia from Alzheimer’s mice reveals inflammation and archpsyc.58.5.445.
dysfunction. Neurobiol. Aging 35, 2746–2760. http://dx.doi.org/10.1016/j. Renault, P.F., 1987. Psychiatric complications of long-term interferon alfa therapy.
neurobiolaging.2014.06.004. Arch. Intern. Med. 147, 1577. http://dx.doi.org/10.1001/
Oshima, E., Terada, S., Sato, S., Ikeda, C., Oda, K., Inoue, S., Kawada, K., Yokota, O., archinte.1987.00370090055011.
Uchitomi, Y., 2014. Left frontal lobe hypoperfusion and depressive symptoms in Ribeiro Xavier, A.L., Kress, B.T., Goldman, S.A., Lacerda de Menezes, J.R., Nedergaard,
Alzheimer’s disease patients taking cholinesterase inhibitors. Psychiatry Res. M., 2015. A distinct population of microglia supports adult neurogenesis in the
Neuroimaging 224, 319–323. http://dx.doi.org/10.1016/j. subventricular zone. J. Neurosci. 35, 11848–11861. http://dx.doi.org/10.1523/
pscychresns.2014.10.008. JNEUROSCI.1217-15.2015.
Ott, A., Stolk, R.P., Hofman, A., Van Harskamp, F., Grobbee, D.E., Breteler, M.M.B., Richard, E., Reitz, C., Honig, L.H., Schupf, N., Tang, M.X., Manly, J.J., Mayeux, R.,
1996. Association of diabetes mellitus and dementia: the Rotterdam Study. Devanand, D., Luchsinger, J.A., 2013. Late-life depression, mild cognitive
Diabetologia 39, 1392–1397. http://dx.doi.org/10.1007/s001250050588. impairment, and dementia. JAMA Neurol. 70, 383. http://dx.doi.org/
Ott, A., Stolk, R.P., van Harskamp, F., Pols, H.A., Hofman, A., Breteler, M.M., 1999. 10.1001/jamaneurol.2013.603.
Diabetes mellitus and the risk of dementia: the Rotterdam Study. Neurology 53, Rivest, S., 2015. TREM2 enables amyloid b clearance by microglia. Cell Res. 25, 535–
1937–1942. http://dx.doi.org/10.1016/S0140-6736(97)07541-7. 536. http://dx.doi.org/10.1038/cr.2015.37.
Ownby, R.L., Crocco, E., Acevedo, A., John, V., Loewenstein, D., 2006. Depression and Roberts, R., Knopman, D.S., 2013. Classification and epidemiology of MCI. Clin.
risk for Alzheimer disease. Arch. Gen. Psychiatry 63, 530–538. Geriatr. Med. 29, 753–772. http://dx.doi.org/10.1016/j.cger.2013.07.003.
Palmqvist, S., Sarwari, A., Wattmo, C., Bronge, L., Zhang, Y., Wahlund, L.O., Nägga, K., Roberts, K.F., Elbert, D.L., Kasten, T.P., Patterson, B.W., Sigurdson, W.C., Connors, R.E.,
2011. Association between subcortical lesions and behavioral and psychological Ovod, V., Munsell, L.Y., Mawuenyega, K.G., Miller-Thomas, M.M., Moran, C.J.,
symptoms in patients with Alzheimer’s disease. Dement. Geriatr. Cogn. Disord. Cross, D.T., Derdeyn, C.P., Bateman, R.J., 2014. Amyloid-b efflux from the central
32, 417–423. http://dx.doi.org/10.1159/000335778. nervous system into the plasma. Ann. Neurol. 76, 837–844. http://dx.doi.org/
Panza, F., Frisardi, V., Capurso, C., D’Introno, A., Colacicco, A.M., Imbimbo, B.P., 10.1002/ana.24270.
Santamato, A., Vendemiale, G., Seripa, D., Pilotto, A., Capurso, A., Solfrizzi, V., Rodríguez, J.J., Noristani, H.N., Verkhratsky, A., 2012. The serotonergic system in
2010. Late-life depression, mild cognitive impairment, and dementia: possible ageing and Alzheimer’s disease. Prog. Neurobiol. 99, 15–41. http://dx.doi.org/
continuum? Am. J. Geriatr. Psychiatry 18, 98–116. http://dx.doi.org/10.1097/ 10.1016/j.pneurobio.2012.06.010.
JGP.0b013e3181b0fa13. Roumestan, C., Michel, A., Bichon, F., Portet, K., Detoc, M., Henriquet, C., Jaffuel, D.,
Paolicelli, R.C., Gross, C.T., 2011. Microglia in development: linking brain wiring to Mathieu, M., 2007. Anti-inflammatory properties of desipramine and fluoxetine.
brain environment. Neuron Glia Biol. 7, 77–83. http://dx.doi.org/10.1017/ Respir. Res. 8, 35. http://dx.doi.org/10.1186/1465-9921-8-35.
S1740925X12000105. Ruhé, H.G., Mason, N.S., Schene, A.H., 2007. Mood is indirectly related to serotonin,
Paolicelli, R.C., Bolasco, G., Pagani, F., Maggi, L., Scianni, M., Panzanelli, P., Giustetto, norepinephrine and dopamine levels in humans: a meta-analysis of
M., Ferreira, T.A., Guiducci, E., Dumas, L., Ragozzino, D., Gross, C.T., 2011. monoamine depletion studies. Mol. Psychiatry 12, 331–359. http://dx.doi.org/
Synaptic pruning by microglia is necessary for normal brain development. 10.1038/sj.mp.4001949.
Science 333, 1456–1458. http://dx.doi.org/10.1126/science.1202529. Rush, A.J., Trivedi, M.H., Wisniewski, S.R., Nierenberg, A.A., Stewart, J.W., Warden, D.,
Pariante, C.M., Lightman, S.L., 2008. The HPA axis in major depression: classical Niederehe, G., Thase, M.E., Lavori, P.W., Lebowitz, B.D., McGrath, P.J.,
theories and new developments. Trends Neurosci. 31, 464–468. http://dx.doi. Rosenbaum, J.F., Sackeim, H.A., Kupfer, D.J., Luther, J., Fava, M., 2006. Acute
org/10.1016/j.tins.2008.06.006. and longer-term outcomes in depressed outpatients requiring one or several
Park, S.-E., Dantzer, R., Kelley, K.W., McCusker, R.H., 2011. Central administration of treatment steps: a STAR*D report. Am. J. Psychiatry 163, 1905–1917. http://dx.
insulin-like growth factor-I decreases depressive-like behavior and brain doi.org/10.1176/appi.ajp.163.11.1905.
cytokine expression in mice. J. Neuroinflamm. 8, 12. http://dx.doi.org/ Sabri, O., Seibyl, J., Rowe, C., Barthel, H., 2015. Beta-amyloid imaging with
10.1186/1742-2094-8-12. florbetaben. Clin. Transl. Imaging 3, 13–26. http://dx.doi.org/10.1007/s40336-
Parkhurst, C.N., Yang, G., Ninan, I., Savas, J.N., Yates, J.R., Lafaille, J.J., Hempstead, B.L., 015-0102-6.
Littman, D.R., Gan, W.B., 2013. Microglia promote learning-dependent synapse Sacre, S., Medghalchi, M., Gregory, B., Brennan, F., Williams, R., 2010. Fluoxetine and
formation through brain-derived neurotrophic factor. Cell 155, 1596–1609. citalopram exhibit potent antiinflammatory activity in human and murine
http://dx.doi.org/10.1016/j.cell.2013.11.030. models of rheumatoid arthritis and inhibit toll-like receptors. Arthritis Rheum.
Parpura, V., Zorec, R., 2010. Gliotransmission: exocytotic release from astrocytes. 62, 683–693. http://dx.doi.org/10.1002/art.27304.
Brain Res. Rev. 63, 83–92. http://dx.doi.org/10.1016/j.brainresrev.2009.11.008. Sadovnick, A.D., Remick, R.A., Allen, J., Swartz, E., Yee, I.M., Eisen, K., Farquhar, R.,
Pascoe, M.C., Crewther, S.G., Carey, L.M., Crewther, D.P., 2011. Inflammation and Hashimoto, S.A., Hooge, J., Kastrukoff, L.F., Morrison, W., Nelson, J., Oger, J., Paty,
depression: why poststroke depression may be the norm and not the exception. D.W., 1997. Depression and multiple sclerosis. Eur. Psychiatry J. Assoc. Eur.
Int. J. Stroke 6, 128–135. http://dx.doi.org/10.1111/j.1747-4949.2010.00565.x. Psychiatry 46, 628–632. http://dx.doi.org/10.1212/WNL.46.3.628.

Please cite this article in press as: Santos, L.E., et al. Microglial dysfunction connects depression and Alzheimer’s disease. Brain Behav. Immun. (2015),
http://dx.doi.org/10.1016/j.bbi.2015.11.011
14 L.E. Santos et al. / Brain, Behavior, and Immunity xxx (2015) xxx–xxx

Saunders, A.M., Strittmatter, W.J., Schmechel, D., George-Hyslop, P.H., Pericak- Terada, S., Oshima, E., Sato, S., Ikeda, C., Nagao, S., Hayashi, S., Hayashibara, C.,
Vance, M.A., Joo, S.H., Rosi, B.L., Gusella, J.F., Crapper-MacLachlan, D.R., Alberts, Yokota, O., Uchitomi, Y., 2014. Depressive symptoms and regional cerebral
M.J., 1993. Association of apolipoprotein E allele epsilon 4 with late-onset blood flow in Alzheimer’s disease. Psychiatry Res. 221, 86–91. http://dx.doi.org/
familial and sporadic Alzheimer’s disease. Neurology 43, 1467–1472. http://dx. 10.1016/j.pscychresns.2013.11.002.
doi.org/10.1212/WNL.43.8.1467. Trivedi, M.H., Rush, A.J., Wisniewski, S.R., Nierenberg, A.A., Warden, D., Ritz, L.,
Schildkraut, J.J., 1965. The catecholamine hypothesis of affective disorders: a review Norquist, G., Howland, R.H., Lebowitz, B., McGrath, P.J., Shores-Wilson, K., Biggs,
of supporting evidence. Am. J. Psychiatry 122, 509–522. http://dx.doi.org/ M.M., Balasubramani, G.K., Fava, M., 2006. Evaluation of outcomes with
10.1176/appi.ajp.122.5.509. citalopram for depression using measurement-based care in STAR*D:
Sciacca, F., Ferri, C., Licastro, F., Veglia, F., Biunno, I., Gavazzi, A., Calabrese, E., implications for clinical practice. Am. J. Psychiatry 163, 28–40. http://dx.doi.
Martinelli Boneschi, F., Sorbi, S., Mariani, C., Franceschi, M., Grimaldi, L.M., 2003. org/10.1176/appi.ajp.163.1.28.
Interleukin-1B polymorphism is associated with age at onset of Alzheimer’s Turnbull, I.R., Gilfillan, S., Cella, M., Aoshi, T., Miller, M., Piccio, L., Hernandez, M.,
disease. Neurobiol. Aging 24, 927–931. http://dx.doi.org/10.1016/S0197-4580 Colonna, M., 2006. Cutting edge: TREM-2 attenuates macrophage activation. J.
(03)00011-3. Immunol. 177, 3520–3524. http://dx.doi.org/10.4049/jimmunol.177.6.3520.
Sepehry, A.A., Lee, P.E., Hsiung, G.Y.R., Beattie, B.L., Jacova, C., 2012. Effect of Tynan, R.J., Weidenhofer, J., Hinwood, M., Cairns, M.J., Day, T.A., Walker, F.R., 2012. A
selective serotonin reuptake inhibitors in Alzheimer’s disease with comorbid comparative examination of the anti-inflammatory effects of SSRI and SNRI
depression: a meta-analysis of depression and cognitive outcomes. Drugs Aging antidepressants on LPS stimulated microglia. Brain Behav. Immun. 26, 469–479.
29, 793–806. http://dx.doi.org/10.1007/s40266-012-0012-5. http://dx.doi.org/10.1016/j.bbi.2011.12.011.
Setiawan, E., Wilson, A.A., Mizrahi, R., Rusjan, P.M., Miler, L., Rajkowska, G., Suridjan, Ueno, M., Fujita, Y., Tanaka, T., Nakamura, Y., Kikuta, J., Ishii, M., Yamashita, T., 2013.
I., Kennedy, J.L., Rekkas, P.V., Houle, S., Meyer, J.H., 2015. Role of translocator Layer V cortical neurons require microglial support for survival during postnatal
protein density, a marker of neuroinflammation, in the brain during major development. Nat. Neurosci. 16, 543–551. http://dx.doi.org/10.1038/nn.3358.
depressive episodes. JAMA Psychiatry 72, 268. http://dx.doi.org/ Ulrich, J.D., Finn, M.B., Wang, Y., Shen, A., Mahan, T.E., Jiang, H., Stewart, F.R., Piccio,
10.1001/jamapsychiatry.2014.2427. L., Colonna, M., Holtzman, D.M., 2014. Altered microglial response to Abeta
Sheline, Y.I., West, T., Yarasheski, K., Swarm, R., Jasielec, M.S., Fisher, J.R., Ficker, W. plaques in APPPS1-21 mice heterozygous for TREM2. Mol. Neurodegener. 9, 20.
D., Yan, P., Xiong, C., Frederiksen, C., Grzelak, M.V., Chott, R., Bateman, R.J., http://dx.doi.org/10.1186/1750-1326-9-20.
Morris, J.C., Mintun, M.A., Lee, J.-M., Cirrito, J.R., 2014. An antidepressant Vécsei, L., Szalárdy, L., Fülöp, F., Toldi, J., 2013. Kynurenines in the CNS: recent
decreases CSF Ab production in healthy individuals and in transgenic AD mice. advances and new questions. Nat. Rev. Drug Discov. 12, 64–82. http://dx.doi.
Sci. Transl. Med. 6, 236re4. http://dx.doi.org/10.1126/scitranslmed.3008169. org/10.1038/nrd3793.
Smith, R.S., 1991. The macrophage theory of depression. Med. Hypotheses 35, 298– Ventriglia, M., Zanardini, R., Bonomini, C., Zanetti, O., Volpe, D., Pasqualetti, P.,
306. http://dx.doi.org/10.1016/0306-9877(91)90272-Z. Gennarelli, M., Bocchio-Chiavetto, L., 2013. Serum brain-derived neurotrophic
Soczynska, J.K., Mansur, R.B., Brietzke, E., Swardfager, W., Kennedy, S.H., factor levels in different neurological diseases. Biomed Res. Int. 2013, 1–7.
Woldeyohannes, H.O., Powell, A.M., Manierka, M.S., McIntyre, R.S., 2012. http://dx.doi.org/10.1155/2013/901082.
Novel therapeutic targets in depression: minocycline as a candidate Videbech, P., 2004. Hippocampal volume and depression: a meta-analysis of MRI
treatment. Behav. Brain Res. http://dx.doi.org/10.1016/j.bbr.2012.07.026. studies. Am. J. Psychiatry 161, 1957–1966. http://dx.doi.org/10.1176/appi.
Son, J.H., Han, D.H., Min, K.J., Kee, B.S., 2013. Correlation between gray matter ajp.161.11.1957.
volume in the temporal lobe and depressive symptoms in patients with Vilalta-Franch, J., Garre-Olmo, J., Lopez-Pousa, S., Turon-Estrada, A., Lozano-Gallego,
Alzheimer’s disease. Neurosci. Lett. 548, 15–20. http://dx.doi.org/10.1016/j. M., Hernandez-Ferrandiz, M., Pericot-Nierga, I., Feijoo-Lorza, R., 2006.
neulet.2013.05.021. Comparison of different clinical diagnostic criteria for depression in
Sonntag, W.E., Deak, F., Ashpole, N., Toth, P., Csiszar, A., Freeman, W., Ungvari, Z., Alzheimer disease. Am. J. Geriatr. Psychiatry 14, 589–597, doi: 10.1097/01.
2013. Insulin-like growth factor-1 in CNS and cerebrovascular aging. Front. JGP.0000209396.15788.9d.
Aging Neurosci. 5, 27. http://dx.doi.org/10.3389/fnagi.2013.00027. Viola, K.L., Sbarboro, J., Sureka, R., De, M., Bicca, M.A., Wang, J., Vasavada, S.,
Starkstein, S.E., Jorge, R., Mizrahi, R., Robinson, R.G., 2005. The construct of minor Satpathy, S., Wu, S., Joshi, H., Velasco, P.T., MacRenaris, K., Waters, E.A., Lu, C.,
and major depression in Alzheimer’s disease. Am. J. Psychiatry 162, 2086–2093. Phan, J., Lacor, P., Prasad, P., Dravid, V.P., Klein, W.L., 2014. Towards non-
http://dx.doi.org/10.1176/appi.ajp.162.11.2086. invasive diagnostic imaging of early-stage Alzheimer’s disease. Nat.
Steinberg, M., Shao, H., Zandi, P., Lyketsos, C.G., Welsh-bohmer, K.A., Norton, M.C., Nanotechnol. 10, 91–98. http://dx.doi.org/10.1038/nnano.2014.254.
Breitner, J.C.S., Steffens, D.C., Tschanz, J.T.Cache County Investigators, 2008. Von Bernhardi, R., Tichauer, J.E., Eugenín, J., 2010. Aging-dependent changes of
Point and 5-year period prevalence of neuropsychiatric symptoms in dementia: microglial cells and their relevance for neurodegenerative disorders. J.
the Cache County Study. Int. J. Geriatr. Psychiatry 23, 170–177. http://dx.doi. Neurochem. 112, 1099–1114. http://dx.doi.org/10.1111/j.1471-
org/10.1002/gps. 4159.2009.06537.x.
Steiner, J., Walter, M., Gos, T., Guillemin, G.J., Bernstein, H.-G., Sarnyai, Z., Mawrin, C., Vukovic, J., Colditz, M.J., Blackmore, D.G., Ruitenberg, M.J., Bartlett, P.F., 2012.
Brisch, R., Bielau, H., Zu Schwabedissen, L., Bogerts, B., Myint, A.-M., 2011. Microglia modulate hippocampal neural precursor activity in response to
Severe depression is associated with increased microglial quinolinic acid in exercise and aging. J. Neurosci. 32, 6435–6443. http://dx.doi.org/10.1523/
subregions of the anterior cingulate gyrus: evidence for an immune-modulated JNEUROSCI.5925-11.2012.
glutamatergic neurotransmission? J. Neuroinflamm. 8, 94. http://dx.doi.org/ Walker, F.R., 2013. A critical review of the mechanism of action for the selective
10.1186/1742-2094-8-94. serotonin reuptake inhibitors: do these drugs possess anti-inflammatory
Su, F., Yi, H., Xu, L., Zhang, Z., 2015. Fluoxetine and S-citalopram inhibit M1 properties and how relevant is this in the treatment of depression?
activation and promote M2 activation of microglia in vitro. Neuroscience 294, Neuropharmacology 67, 304–317. http://dx.doi.org/10.1016/j.
60–68. http://dx.doi.org/10.1016/j.neuroscience.2015.02.028. neuropharm.2012.10.002.
Sun, L.Y., 2006. Hippocampal IGF-1 expression, neurogenesis and slowed aging: Walton, N.M., Sutter, B.M., Laywell, E.D., Levkoff, L.H., Kearns, S.M., Marshall, G.P.,
clues to longevity from mutant mice. Age (Omaha) 28, 181–189. http://dx.doi. Scheffler, B., Steindler, D.A., 2006. Microglia instruct subventricular zone
org/10.1007/s11357-006-9009-5. neurogenesis. Glia 54, 815–825. http://dx.doi.org/10.1002/glia.20419.
Swardfager, W., Lanctt, K., Rothenburg, L., Wong, A., Cappell, J., Herrmann, N., 2010. Wang, T., 2015. TNF-alpha G308A polymorphism and the susceptibility to
A meta-analysis of cytokines in Alzheimer’s disease. Biol. Psychiatry 68, 930– Alzheimer’s Disease: an updated meta-analysis. Arch. Med. Res. 46 (24–30),
941. http://dx.doi.org/10.1016/j.biopsych.2010.06.012. e1. http://dx.doi.org/10.1016/j.arcmed.2014.12.006.
Szczêsny, E., OElusarczyk, J., Glombik, K., Budziszewska, B., Kubera, M., Lasoñz, W., Wang, Y., Cella, M., Mallinson, K., Ulrich, J.D., Young, K.L., Robinette, M.L., Gilfillan, S.,
Basta-Kaim, A., 2013. Possible contribution of IGF-1 to depressive disorder. Krishnan, G.M., Sudhakar, S., Zinselmeyer, B.H., Holtzman, D.M., Cirrito, J.R.,
Pharmacol. Rep. 65, 1622–1631. Colonna, M., 2015. TREM2 lipid sensing sustains the microglial response in an
Takahashi, K., Rochford, C.D.P., Neumann, H., 2005. Clearance of apoptotic neurons Alzheimer’s disease model. Cell 160, 1061–1071. http://dx.doi.org/10.1016/
without inflammation by microglial triggering receptor expressed on myeloid j.cell.2015.01.049.
cells-2. J. Exp. Med. 201, 647–657. http://dx.doi.org/10.1084/jem.20041611. Watanabe, T., Miyazaki, A., Katagiri, T., Yamamoto, H., Idei, T., Iguchi, T., 2005.
Talbot, K., Wang, H.-Y., Kazi, H., Han, L.-Y., Bakshi, K.P., Stucky, A., Fuino, R.L., Relationship between serum insulin-like growth factor-1 levels and Alzheimer’s
Kawaguchi, K.R., Samoyedny, A.J., Wilson, R.S., Arvanitakis, Z., Schneider, J.A., disease and vascular dementia. J. Am. Geriatr. Soc. 53, 1748–1753. http://dx.doi.
Wolf, B.A., Bennett, D.A., Trojanowski, J.Q., Arnold, S.E., 2012. Demonstrated org/10.1111/j.1532-5415.2005.53524.x.
brain insulin resistance in Alzheimer’s disease patients is associated with IGF-1 Westwood, A.J., Beiser, A., Decarli, C., Harris, T.B., Chen, T.C., He, X.-M., Roubenoff, R.,
resistance, IRS-1 dysregulation, and cognitive decline. J. Clin. Invest. 122, 1316– Pikula, A., Au, R., Braverman, L.E., Wolf, P.A., Vasan, R.S., Seshadri, S., 2014.
1338. http://dx.doi.org/10.1172/JCI59903. Insulin-like growth factor-1 and risk of Alzheimer dementia and brain atrophy.
Taraz, M., Khatami, M., Dashti-khavidaki, S., Akhonzadeh, S., 2013. Sertraline Neurology 82, 1613–1619. http://dx.doi.org/10.1212/WNL.0000000000000382.
decreases serum level of interleukin-6 (IL-6) in hemodialysis patients with Wichers, M.C., Maes, M., 2004. The role of indoleamine 2,3-dioxygenase (IDO) in the
depression: results of a randomized double-blind, placebo-controlled clinical pathophysiology of interferon-alpha-induced depression. J. Psychiatry Neurosci.
trial. Int. Immunopharmacol. 17, 917–923. http://dx.doi.org/10.1016/j. 29, 11–17.
intimp.2013.09.020. Wilcock, D.M., 2012. A changing perspective on the role of neuroinflammation in
Teng, E., Ringman, J.M., Ross, L.K., Mulnard, R.A., Dick, M.B., Bartzokis, G., Davies, H. Alzheimer’s disease. Int. J. Alzheimers. Dis. 2012, 1–7. http://dx.doi.org/
D., Galasko, D., Hewett, L., Mungas, D., Reed, B.R., Schneider, L.S., Segal-Gidan, F., 10.1155/2012/495243.
Yaffe, K., Cummings, J.L., 2008. Diagnosing depression in Alzheimer disease with Wolkowitz, O.M., Wolf, J., Shelly, W., Rosser, R., Burke, H.M., Lerner, G.K., Reus, V.I.,
the national institute of mental health provisional criteria. Am. J. Geriatr. Nelson, J.C., Epel, E.S., Mellon, S.H., 2011. Serum BDNF levels before treatment
Psychiatry 16, 469–477. http://dx.doi.org/10.1097/JGP.0b013e318165dbae. predict SSRI response in depression. Prog. Neuro-Psychopharmacol. Biol.
Psychiatry 35, 1623–1630. http://dx.doi.org/10.1016/j.pnpbp.2011.06.013.

Please cite this article in press as: Santos, L.E., et al. Microglial dysfunction connects depression and Alzheimer’s disease. Brain Behav. Immun. (2015),
http://dx.doi.org/10.1016/j.bbi.2015.11.011
L.E. Santos et al. / Brain, Behavior, and Immunity xxx (2015) xxx–xxx 15

Yasutake, C., Kuroda, K., Yanagawa, T., Okamura, T., Yoneda, H., 2006. Serum BDNF, Zhang, F., Zhou, H., Wilson, B.C., Shi, J.-S., Hong, J.-S., Gao, H.-M., 2012. Fluoxetine
TNF-alpha and IL-1beta levels in dementia patients: comparison between protects neurons against microglial activation-mediated neurotoxicity.
Alzheimer’s disease and vascular dementia. Eur. Arch. Psychiatry Clin. Neurosci. Parkinsonism Relat. Disord. 18, S213–S217. http://dx.doi.org/10.1016/S1353-
256, 402–406. http://dx.doi.org/10.1007/s00406-006-0652-8. 8020(11)70066-9.
Ye, S.M., Johnson, R.W., 1999. Increased interleukin-6 expression by microglia from Zheng, L.S., Hitoshi, S., Kaneko, N., Takao, K., Miyakawa, T., Tanaka, Y., Xia, H.,
brain of aged mice. J. Neuroimmunol. 93, 139–148. http://dx.doi.org/10.1016/ Kalinke, U., Kudo, K., Kanba, S., Ikenaka, K., Sawamoto, K., 2014. Mechanisms for
S0165-5728(98)00217-3. interferon-a-induced depression and neural stem cell dysfunction. Stem Cell
Yesavage, J.A., Brink, T.L., Rose, T.L., Lum, O., Huang, V., Adey, M., Leirer, V.O., 1982. Rep. 3, 73–84. http://dx.doi.org/10.1016/j.stemcr.2014.05.015.
Development and validation of a geriatric depression screening scale: a Zheng, L.-S., Kaneko, N., Sawamoto, K., 2015. Minocycline treatment ameliorates
preliminary report. J. Psychiatr. Res. 17, 37–49. http://dx.doi.org/10.1016/ interferon-alpha-induced neurogenic defects and depression-like behaviors in
0022-3956(82)90033-4. mice. Front. Cell. Neurosci. 9, 1–10. http://dx.doi.org/10.3389/fncel.2015.00005.
Yirmiya, R., 1996. Endotoxin produces a depressive-like episode in rats. Brain Res. Ziebell, J.M., Morganti-Kossmann, M.C., 2010. Involvement of pro- and anti-
711, 163–174. http://dx.doi.org/10.1016/0006-8993(95)01415-2. inflammatory cytokines and chemokines in the pathophysiology of traumatic
Yirmiya, R., Rimmerman, N., Reshef, R., 2015. Depression as a microglial disease. brain injury. Neurotherapeutics 7, 22–30. http://dx.doi.org/10.1016/j.
Trends Neurosci. 38, 637–658. http://dx.doi.org/10.1016/j.tins.2015.08.001. nurt.2009.10.016.
Yu, H., Chen, Z., 2011. The role of BDNF in depression on the basis of its location in Ziv, Y., Schwartz, M., 2008. Immune-based regulation of adult neurogenesis:
the neural circuitry. Acta Pharmacol. Sin. 32, 3–11. http://dx.doi.org/10.1038/ implications for learning and memory. Brain Behav. Immun. 22, 167–176.
aps.2010.184. http://dx.doi.org/10.1016/j.bbi.2007.08.006.
Yu, D., Tao, B.-B., Yang, Y.-Y., Du, L.-S., Yang, S.-S., He, X.-J., Zhu, Y.-W., Yan, J.-K., Ziv, Y., Ron, N., Butovsky, O., Landa, G., Sudai, E., Greenberg, N., Cohen, H., Kipnis, J.,
Yang, Q., 2015. The IDO inhibitor coptisine ameliorates cognitive impairment in Schwartz, M., 2006. Immune cells contribute to the maintenance of
a mouse model of Alzheimer’s disease. J. Alzheimers. Dis. 43, 291–302. http:// neurogenesis and spatial learning abilities in adulthood. Nat. Neurosci. 9,
dx.doi.org/10.3233/JAD-140414. 268–275. http://dx.doi.org/10.1038/nn1629.
Yuan, H., Chen, R., Wu, L., Chen, Q., Hu, A., Zhang, T., Wang, Z., Zhu, X., 2015. The Zubenko, G.S., Zubenko, W.N., McPherson, S., Spoor, E., Marin, D.B., Farlow, M.R.,
regulatory mechanism of neurogenesis by IGF-1 in adult mice. Mol. Neurobiol. Smith, G.E., Geda, Y.E., Cummings, J.L., Petersen, R.C., Sunderland, T., 2003. A
51, 512–522. http://dx.doi.org/10.1007/s12035-014-8717-6. collaborative study of the emergence and clinical features of the major
Zhan, Y., Paolicelli, R.C., Sforazzini, F., Weinhard, L., Bolasco, G., Pagani, F., Vyssotski, depressive syndrome of Alzheimer’s disease. Am. J. Psychiatry 160, 857–866.
A.L., Bifone, A., Gozzi, A., Ragozzino, D., Gross, C.T., 2014. Deficient neuron- http://dx.doi.org/10.1176/appi.ajp.160.5.857.
microglia signaling results in impaired functional brain connectivity and social
behavior. Nat. Neurosci. 17, 400–406. http://dx.doi.org/10.1038/nn.3641.

Please cite this article in press as: Santos, L.E., et al. Microglial dysfunction connects depression and Alzheimer’s disease. Brain Behav. Immun. (2015),
http://dx.doi.org/10.1016/j.bbi.2015.11.011

Você também pode gostar