Você está na página 1de 14

Arch. Pharm. Res.

DOI 10.1007/s12272-015-0648-x

REVIEW

Involvement of inflammation in Alzheimer’s disease pathogenesis


and therapeutic potential of anti-inflammatory agents
Sina Shadfar1 • Chul Ju Hwang2 • Mi-Sun Lim1 • Dong-Young Choi1 •

Jin Tae Hong2

Received: 4 May 2015 / Accepted: 8 August 2015


 The Pharmaceutical Society of Korea 2015

Abstract Alzheimer’s disease (AD) is the most common microglia, T cells and microRNA participate in AD and the
form of dementia. It is characterized by beta-amyloid (Ab) neuroprotective mechanisms of anti-inflammatory drugs.
peptide fibrils, which are extracellular depositions of a
specific protein, and is accompanied by extensive neu- Keywords Alzheimer’s disease  Amyloidogenesis 
roinflammation. Various studies have demonstrated risk Anti-inflammatory  Beta-amyloid  Neuroinflammation
factors that can affect AD pathogenesis, and they include
accumulation of Ab, hyperphosphorylation of tau protein,
and neuroinflammation. Among these detrimental factors, Introduction
neuroinflammation has been highlighted by epidemiologic
studies suggesting that use of anti-inflammatory drugs Alzheimer’s disease (AD) is the most prevalent neurode-
could significantly reduce the incidence of AD. Evidence generative disease and the most common form of dementia.
suggests that astrocytes, microglia, and infiltrating immune However, specific molecular mechanisms by which neu-
cells from periphery might contribute to or modify the rons degenerate in AD brains remain unclear, and there is
process of neuroinflammation and neurodegeneration in no known way to halt or cure the neurodegeneration in AD.
AD brains. In addition, recent data indicate that micro- It is estimated that about 10% of the population older than
RNAs may affect neuroinflammatory responses in the 65 years of age suffers from the disease. The number of
brain. This article focuses on supportive evidence that AD patients is predicted to reach 14 million in the United
neuroinflammation plays a critical role in AD development. States by 2050, with incidence of a million cases per year
In addition, we depict putative therapeutic capacity of anti- (Choi et al. 2012c). This situation underscores the urgency
inflammatory drugs for AD prevention or treatment. We for the development of therapeutic or preventative inter-
also discuss pathogenic mechanisms by which astrocytes, ventions of AD.
The pathologic hallmarks of AD are senile plaques and
neurofibrillary tangles. Senile plaques are extracellular
deposits of amyloid plaques, which are mainly composed
Sina Shadfar and Chul Ju Hwang have contributed equally.
of b-amyloid (Ab) 40 or 42. Ab is produced by sequential
& Dong-Young Choi proteolytic cleavage of amyloid precursor protein (APP) by
neochallenger@yahoo.co.kr beta and gamma secretases. Mutations in the genes of
& Jin Tae Hong presenilin 1, presenilin 2 and amyloid precursor protein are
jinthong@chungbuk.ac.kr associated with the pathogenesis of the familial form of
1
AD, suggesting that dysregulations of APP processing and
College of Pharmacy, Yeungnam University, 280 Daehak-ro,
Ab production are critical pathways of neurodegeneration
Gyeongsan, Gyeongbuk 712-749, Republic of Korea
2
leading to dementia in AD (Leuner et al. 2007). Neu-
College of Pharmacy and Medical Research Center,
rofibrillary tangles are intracellular protein depositions of
Chungbuk National University, 52 Naesudong-ro,
Heungdeok-gu, Cheongju, Chungbuk 361-763, hyperphosphorylated tau protein. Tau filaments accumulate
Republic of Korea in dystrophic neurites as fine neuropil threads or as bundles

123
S. Shadfar et al.

of paired helical filaments in neuronal bodies, forming (IL)-1b, IL-6, tumor necrosis factor-a (TNF-a) and trans-
neurofibrillary tangles that become extracellular ghost forming growth factor-b (TGF-b). In addition, infiltration
tangles after the death of the neuron (Liu et al. 2005). of peripheral immune cells including T cells, macrophages
Deposition of tau protein in the brain causes loss of and others was detected in AD brains, and they may modify
synaptic function and finally neuronal death (Crespo-Biel the contribution of microglia and astrocytes to neuroin-
et al. 2012). The number and localization of neurofibrillary flammation. Recently, experimental results suggest that
tangles have been well correlated with the level of microRNAs may affect neuroinflammatory reactions and
dementia, while no such correlation has been demonstrated can be associated with AD pathogenesis (Thounaojam et al.
for senile plaques (Giannakopoulos et al. 2003). Tau 2013). Putative role of microglia, astrocytes and inflam-
pathology, therefore, appears to be cardinal to AD clinical matory stimulants such as IFN-c produced by T cells in AD
manifestations. pathogenesis is depicted in Fig. 1, and contribution of
Many therapeutic targets have been identified and microRNAs to neuroinflammatory responses is described
studied for modification of neurodegeneration in AD in Fig. 2.
brains. These include neuroinflammation, oxidative stress,
Ab accumulation, tau pathology, neuronal loss, and Astrocytes in AD brains
synaptic loss (Frautschy & Cole 2010). However, multiple
disease-modifying therapeutic attempts to date have failed. Astrocytes are the most abundant cells in the central ner-
For instance, passive or active immunization against Ab or vous system and constitute about 25% of the cerebral
tau protein was conducted to attenuate burden of the pep- volume (Morales et al. 2014). They have multiple physi-
tide, but this trial was not successful and even caused ological functions including synaptogenesis, formation and
severe adverse effects such as brain microhemorrhages in maintenance of blood-brain barrier, maintenance of ion
some AD patients (Panza et al. 2012). Neuroinflammation balance, maintenance of homeostasis, and turnover of
appears to play a critical role in AD pathophysiology and glutamate (Morales et al. 2014). However, astrocytes may
has been considered a promising target for AD treatment, play a significant role in AD pathogenesis. Reactive
although none of the anti-neuroinflammatory drugs is astrocytes are found in the post mortem brains of AD
clinically available for AD treatment. The role of neu- patients and they are manifested by hypertrophy and
roinflammation in AD pathogenesis was highlighted by increased expression of glial fibrillary acidic protein
multiple epidemiological and animal studies in which (Verkhratsky et al. 2010). Activated astrocytes are mostly
NSAIDs had substantial sparing effects on AD neurode- observed in areas with amyloid plaques (Nagele et al.
generation (Etminan et al. 2003; McGeer & McGeer 2013). 2004). Effects of astrogliosis on accumulation of Ab are
Furthermore, neuroprotective effects of other classes of still debated, but Ab deposition (Mucke et al. 2000; Nils-
anti-inflammatory drugs were demonstrated in models of son et al. 2001) and tau phosphorylation (Padmanabhan
AD (Heneka et al. 2011). et al. 2006) are enhanced by a1-antichymotrypsin, an
In this article, we describe the association of neuroin- acute-phase protein that is upregulated by activated astro-
flammation with the neurodegenerative process in AD and cytes. Critical evidence for the role of astrogliosis in AD
the putative roles of microglia, astrocytes and T cells in AD pathogenesis is the correlation between the extent of
pathogenesis. In addition, we briefly describe the dysreg- astrogliosis and cognitive impairment (Simpson et al.
ulation of microRNA expression in AD brains and its 2010). In normal brains, expression of BACE1 is exclu-
involvement in neuroinflammation and the process of AD. sively confined to neurons. However, astrocytes acquire the
Furthermore, we emphasize the therapeutic potential of ability to produce Ab and turn into Ab producers under
anti-inflammatory agents for treatment of AD and the conditions of AD or chronic stress (Rossner et al. 2005). In
molecular mechanisms of their neuroprotective effects. addition, expression of BACE1 in the astrocytes related to
Ab plaques is detected in several transgenic mouse AD
models (Hartlage-Rubsamen et al. 2003; Heneka et al.
Neuroinflammatory responses in AD brains 2005a; Rossner et al. 2001).
Reactive astrocytes seem to neglect their neurosup-
Accumulating evidence indicates that there is a close link portive functions, thus rendering neurons vulnerable to
between AD pathogenesis and neuroinflammatory cascades excitotoxicity and oxidative stress (Steele & Robinson
in the brain. Neuroinflammation in AD brains might be 2012). Furthermore, fragments of Ab promote inflamma-
exerted by damaged neurons, highly insoluble Ab deposits, tory response in the brain via calcineurin and NF-jB (Lim
and neurofibrillary tangles (Wenk 2003). Neuroinflamma- et al. 2013). When astrocytes are stimulated by aggregated
tion manifests as astrogliosis, microglial activation, and an Ab, they produce inflammatory cytokines (IL-1b and
increase in inflammatory cytokines such as interleukin TNFa) and reactive oxygen species. These cytokines could

123
Involvement of inflammation in Alzheimer’s disease pathogenesis and therapeutic potential…

Fig. 1 The plausible roles of neuroinflammatory responses in AD pathogenesis. Inflammatory stress including environmental and genetic factors
might cause microglial and astroglial activation. Activated glial cells can release determental mediators including cytotoxic cytokines, reactive
oxygen species, nitric oxide, glutamate and byproducts of COX-2. Further, activated glial cells can contribute to accumulation of Ab via
expression of BACE1 and decreased phagocytosis of Ab. Therefore, anti-inflammatory drugs such as NSAIDs have been suggested to prevent
from AD neurodegenertion or delay AD progression in spite of their failure in multiple clinical trials

Fig. 2 MicroRNAs involved in inflammation and AD. MicroRNAs including miRNA-9, -34a, -125b, -146a, and -155 can be upregulated by
proinfalmmatory cytokine and Ab. These miRNAs are sensitive to NF-jB which increases levels of the miRNA transcription in AD brains.
MicroRNA-34a appear to downregulate TREM2 expression resulting in elevation of immune response. MicroRNA-125b and 146a reduce
expression of CFH which suppress the neuroinflammaotry response in the brains

be causative factors in the cognitive dysfunction of AD. In and homeostasis of the CNS (Kettenmann et al. 2011). For
addition, a secondary inflammatory response to IL-1b and instance, they can secrete growth factors for the formation
TNFa leads to astrocyte activation, with the long-term of the CNS in the embryo and contribute to neurogenesis,
effect of these cytokines being detrimental to the survival neuroprotection and synaptic pruning when they are in the
of neurons (Morales et al. 2014). resting state (Morales et al. 2014).
In AD brains, microglia are prone to a pro-inflammatory
Microglia in AD brains rather than phagocytic status (Mandrekar-Colucci & Lan-
dreth 2010). Furthermore, a pro-inflammatory environment
Microglia are rich in the central nervous system (CNS), but impaired microglial phagocytosis of Ab in vitro, while
they are mostly concentrated in the hippocampus and anti-inflammatory factors enhanced Ab clearance (Koe-
substantia nigra (Kim et al. 2000). These cells constitute nigsknecht-Talboo & Landreth 2005). Consistent with this,
the major immune system in the CNS since they produce deletion of PGE2 E prostanoid subtype 2 receptor in
cytotoxic mediators, perform phagocytosis, and behave APPSwe-PS1DeltaE9 mice markedly attenuated Ab plaque
like antigen-presenting cells (Ousman & Kubes 2012; deposition and enhanced phagocytosis (Liang et al. 2005).
Ransohoff & Engelhardt 2012). In addition, physiological In addition, neuroinflammation could contribute to tau
functions of microglia are essential for the development protein-mediated neuronal loss as shown in an animal

123
S. Shadfar et al.

model for taupathy (Jaworski et al. 2011). Indeed, activated increased Ab deposition and reduced microglial recruit-
microglia accumulated around tau-positive neurons, and ment into the brain, particularly phagocytic macrophages
synapses were deleted before formation of neurofibrillary (El Khoury et al. 2007).
tangles in P301S tau transgenic mice (Yoshiyama et al. Microglial activation presents a first line of defense in
2007). Lipopolysaccharide (LPS)-induced peripheral the CNS, but it can contribute to neurodegeneration. Thus,
inflammation exacerbates tau pathology via CDK5-medi- understanding the underlying mechanisms by which
ated pathway in a transgenic model of AD (Kitazawa et al. microglia shift between these two opposite phenotypes
2005). Furthermore, a recent study showed that inflam- may allow us to minimize microglia-mediated neuronal
mation induced by infection increased GSK3 activity in the damage and find a way to treat neurodegenerative diseases
triple-transgenic mouse model of AD; this was associated such as AD.
with a shift of tau from the detergent-soluble to the
detergent-insoluble fraction (Sy et al. 2011). Another study T-cell-mediated immune response associated
demonstrated detrimental effects of activated microglia in with AD
AD (Qiao et al. 2001). In this study, lipopolysaccharide
(LPS) was chronically infused in the lateral ventricle of Strong Ab-reactive T cell responses are more common in
APPV717F mice with (apoE?/?) or without (apoE-/-) healthy elderly subjects and patients with AD than in
expression of apolipoprotein E. Chronic LPS led to amy- middle-aged subjects, suggesting that intrinsic T cell
loid plaque-localized microglial activation, but significant reactivity to the self-antigen Ab may be linked to AD
LPS-mediated acceleration of amyloid deposition was seen susceptibility (Monsonego et al. 2003). In addition, T-cell
only in APPV717F-apoE?/? mice, suggesting that experi- infiltration was detected in the brains of AD patients in
mental induction of microglial activation by chronic proximity with Ab deposition; this is also true in mice of
administration of LPS can accelerate amyloidosis in a the brains of AD model (Monsonego et al. 2006; Togo
transgenic mouse model of AD in the presence of apoE, et al. 2002). Entry of peripheral T cells was dependent on
and supporting the idea that activated microglia exacerbate TNFa produced by microglia (Liu et al. 2010). Limited
cerebral amyloidosis. expression of interferon (IFN)-c in the brain promoted
Activated microglia secrete multiple cytokines and T-cell infiltration into the cerebral parenchyma and
chemokines such as IL-1b, IL-6, TNF-a, IL-8, TGF-b, and microglia-mediated Ab clearance in the APP-transgenic
macrophage inflammatory protein-1a (MIP-1a), and the mouse (Monsonego et al. 2006). Further, the stimulation of
levels are altered in AD patients compared to control Ab-reactive T cells at sites of Ab plaques resulted in IFN-
(Sastre et al. 2006). Animal models of AD, including c-induced chemotaxis of leukocytes and therapeutic
Tg2567 mice, show elevated levels of TNF-a, IL-1b, IL- clearance of Ab (Fisher et al. 2010). In contrast, recent data
1a, chemoattractant protein-1, COX-2, and complement indicate that IFN-c produced by T-cells infiltrating the
component 1q (Benzing et al. 1999; Matsuoka et al. 2001). brain in AD tend to elevate microglial activation, Ab
In addition, polymorphisms of proinflammatory cytokines deposition and impaired cognitive functions, and these
are associated with an increased risk of AD (Ribizzi et al. effects were ameliorated by systemic administration of
2010; Solito & Sastre 2012). anti-IFN-c antibody in mice with AD (Browne et al. 2013).
Several studies showed that some molecules can mod- The role of T regulatory cells (Tregs) in AD brains has been
ulate microglial activation and Ab phagocytosis by studied. The adoptive transference of Tregs significantly
microglia. CD40 ligand increased TNF-a production in the ameliorates impaired cognition and reduces the Ab-deposits
Ab-stimulated microglia, promoting injury of primary and microglial activation in a mouse model of AD [25].
cortical neurons (Townsend et al. 2005). In addition, a Moreover, higher Tregs function has been associated with
study showed that deletion of CD40 ligand gene in APP/ lower cognitive manifestation in AD patients [68]. Contrary to
PS1 mice attenuated amyloid deposition and microgliosis Tregs, the current evidence indicates that inflammatory Th1
(Laporte et al. 2006). Overexpression of the proinflam- and Th17 CD4-positive T-cells play an important role in the
matory cytokine-like molecule human S100B in the physiopathology of AD, favoring the disease progression
Tg2576 mouse exacerbated cerebral b-amyloid accumula- (Browne et al. 2013; Goldeck et al. 2013).
tion and promoted Ab-induced microglial activation (Mori Taken together, the current evidence indicates that
et al. 2010). TREM2 is expressed in microglia and is a inflammatory Th1 and Th17 CD4-positive T-cells play an
genetic risk factor for AD (Guerreiro et al. 2013). Studies important role in the physiopathology of AD, favoring the
suggest that TREM2 risk variants have impaired function disease progression. Nevertheless, other important
of TREM2 and reduced phagocytic clearance of amyloid encephalitogenic T-cell subsets that play important
proteins or cellular microglia in the central nervous system inflammatory or anti-inflammatory roles in MS have not
(Jonsson et al. 2013). Deficiency of CCR2 in Tg2576 mice yet been studied in AD. Furthermore, studies implementing

123
Involvement of inflammation in Alzheimer’s disease pathogenesis and therapeutic potential…

the genetic deletion of specific subsets of inflammatory or delays dementia onset, slows down disease progression,
anti-inflammatory T-cells would be very helpful in order to and mitigates severity of cognitive dysfunction (Andersen
clarify the specific roles and relevance of these different et al. 1995; Breitner et al. 1994; Gasparini et al. 2004;
T-cell subsets in the development and progression of AD. Stewart et al. 1997). NSAIDs significantly reduced beta-
Therefore, similar to the case for PD, future efforts are amyloid (Ab) secretion from cultured cells and amyloid
required to evaluate the participation of important T-cell peptide accumulation in the AD animal models (Gasparini
subsets (Gonzalez & Pacheco 2014). et al. 2004; Lim et al. 2001; Yan et al. 2003). Among
NSAIDs, ibuprofen and flurbiprofen potently ameliorated
MicroRNAs and AD Ab burden via modulation of c-secretase activity and/or
anti-inflammatory effect (Carreras et al. 2013). Ibuprofen
MicroRNA (miRNA) species can contribute to homeostatic attenuated the level of both Ab accumulation and tau
brain gene expression by regulating translation of specific phosphorylation, and improved cognitive function in the
mRNAs. Among miRNAs, miRNA-146a is the most studied triple transgenic mouse model of AD that develops Ab and
small non-coding RNAs in the field of human neurobiology, tau pathology (McKee et al. 2008). Flurbiprofen reduced
and this miRNA modulates the innate response, inflammation, cognitive impairment and Ab deposition in AD animal
and the microglial activation state (Alexandrov et al. 2014). models (Kukar et al. 2007).
This miRNA is selectively increased in affected brain regions Beneficial effects of NSAIDs on AD experimental
by AD (Lukiw 2007; Sethi & Lukiw 2009) and can be induced models might involve inhibition of cyclooxygenase (COX),
by inflammatory cytokines and Ab, or combinations of these which is the key regulatory enzyme of the eicosanoid
factors (Lukiw et al. 2008). A significant increase of the levels biosynthetic pathway producing a variety of pro-inflam-
of miRNA-9, miRNA-125b, miRNA-146a, and miRNA-155 matory mediators (Breitner et al. 2011). COX-2 is con-
was found in the cerebrospinal fluid of AD patients compared sidered the most appropriate target for anti-inflammatory
with age-matched controls (Alexandrov et al. 2012). These drugs due to its induction by inflammatory stimuli, but
miRNAs are NF-jB-sensitive proinflammatory miRNAs, recent data indicate that COX-2 can mediate neuroprotec-
upregulated in AD brains and associated with progressive tion and that COX-1 is a major player in the neuroin-
propagation of neuroinflammation (Guedes et al. 2013). In flammatory process (Choi et al. 2009). COX-1 is
addition, a recent study showed that a specific genotype of predominantly localized in microglia, and the enzyme
miRNA-146a is associated with an increased risk of AD as might be the major player in neuroinflammation. There-
well as cognitive decline (Cui et al. 2014). Aberrant expres- fore, COX-1 suppression by NSAIDs could protect neurons
sion of miRNA-146a was also found in an animal model of from immune-derived challenge in the early stages of AD
AD (Li et al. 2011). MiRNA-146a increased the inflammatory (Rogers et al. 1993; Sung et al. 2004). In contrast, brain
responses in the brains by down-regulating complement factor COX-2 is abundant in neuronal dendrites, where it is
H and interleukin-1 receptor-associated kinase-1, which are essential for transduction of post-synaptic signals from
critical suppressors of innate immunity acting on the cerebral NMDA-type glutamate receptors (Kaufmann et al. 1997).
inflammation response (Olivieri et al. 2013). COX-2 inhibition by conventional COX-1/COX-2 dual-
Apart from miRNA-146a, miRNA-34a might be related inhibitors or by COX-2 selective inhibitors could thus be
to AD development by upregulating neuroinflammation in deleterious to patients with these conditions (Breitner et al.
the brain (Zhao et al. 2013). MiRNA-34a is a NF-jB- 2011). Furthermore, a recent study demonstrated that
sensitive miRNA and reduces TREM2 expression, result- administration of a COX-1 selective inhibitor, SC-560, in
ing in increased immune response and decreased phago- triple transgenic mice reduced neuroinflammation and
cytic responses (Bhattacharjee et al. 2014). Although the neuropathology and improved cognitive performance (Choi
analysis of miRNAs in AD is a relatively new research et al. 2013). Besides inhibiting cyclooxygenase, NSAIDs
area, the data reported so far strongly indicate that such as indomethacin (Netland et al. 1998) and flurbiprofen
inflammatory microRNAs could play a key role in AD. (Prosperi et al. 2004) reduced microglial activation in AD
brains. The benefits of NSAIDs may result from a variety
of mechanisms independent of COX inhibition. For
Anti-inflammatory agents and their therapeutic example, researchers have shown that action mechanisms
potentials for AD of NSAIDs are related to modulation of c-secretase activity
(Weggen et al. 2003), inhibition of RhoA (Fu et al. 2007),
Nonsteroidal anti-inflammatory drugs (NSAIDs) and activation of peroxisome proliferator-activated recep-
tor (PPAR)-c (Kummer & Heneka 2008).
Multiple epidemiological studies suggest that long-term Several randomized clinical trials have been performed
usage of NSAIDs decreases risk of AD development, to assess the possible therapeutic efficacy of NSAIDs (Lee

123
S. Shadfar et al.

et al. 2010). However, the outcomes from those trials were suppression of the production of pro-inflammatory media-
disappointing (Table 1). Clinical trials with NSAIDs in tors (Heneka et al. 2001; Landreth & Heneka 2001). In
patients with established dementia did not significantly addition, PPARc activation is reported to improve mito-
attenuate progression of dementia (Soininen et al. 2007; chondrial metabolism and biogenesis (Alaynick 2008).
Thal et al. 2005). In a study with rofecoxib and naproxen, Ameliorative effects of PPARc agonists on Ab-induced
the drugs were not able to halt or slow the progression of neuroinflammation and neurotoxicity were first reported in
AD (Aisen et al. 2003). Another study also showed that 2000 (Combs et al. 2000), followed by a number of studies
naproxen and celecoxib were not able to prevent AD on actions of PPARc agonists in AD animal models
development (Group et al. 2007). A randomized controlled (Mandrekar-Colucci and Landreth 2011) and several clin-
study showed that ibuprofen treatment had no effect on ical trials with PPARc agonists (Nicolakakis &
worsening of cognition in AD patients (Pasqualetti et al. Hamel 2010; Tobinick & Gross 2008).
2009). One study suggested that NSAIDs are even detri- The actions of PPARc and its agonists in in vitro and
mental in later stages of AD pathogenesis (Breitner et al. animal models of AD have been extensively reviewed
2011). A few studies demonstrated that NSAIDs are (Mandrekar-Colucci & Landreth 2011; Mandrekar-Colucci
effective only in the ApoE 34 carrier subpopulation et al. 2013; Sodhi et al. 2011). Administration of PPARc
(Hayden et al. 2007; Szekely et al. 2008). It is well known agonists improved cognitive function, suppressed inflam-
that the ApoE 34 allele confers an inflammatory state in the mation, and reduced amyloid levels in many mouse models
brain compared to ApoE 33 (Vitek et al. 2009). of AD (Camacho et al. 2004; Combs et al. 2000; Sastre
The failure of NSAIDs in clinical studies with AD et al. 2003; Searcy et al. 2012). In a rat AD model, injec-
patients may be related to the inhibitory effects on tion of both ciglitazone and ibuprofen or oral pioglitazone
microglia. One of the physiologic functions of microglial administration potently suppressed Ab-caused microglial
cells is to clear Ab plaques by phagocytosis as well as inflammatory reactions (Heneka et al. 2003). The effects of
secretion of various proteases (Lee et al. 2010). Evidence pioglitazone and ibuprofen have been investigated in
for the role of microglia and their inflammatory mediators 12-month-old Tg2576 mice and Ab42 levels were only
in modulating neurogenesis is also compelling (Ekdahl significantly lowered in ibuprofen-treated animals, but a
et al. 2009). The clearance function declines with age, so trend was observed for pioglitazone (Yan et al. 2003). Oral
accumulation of plaques continues in spite of increasing treatment with ibuprofen or pioglitazone significantly
microglial numbers (Hickman et al. 2008). An alternative reduced microglial activation and neuroinflammatory
explanation for the unfortunate results is that current ani- markers in 10-month-old APPV717I mice (Heneka et al.
mal models of AD reflect early latent phases of disease 2005b). Interestingly, intraventricular injection of an
rather than progressed disease states (Zahs & Ashe 2010). antagonist of PPARc, GW9662, increased cerebellar dys-
If true, NSAID regimens should be initiated during latent function and Ab accumulation in APP/PS1 transgenic mice
disease phase in predisposed individuals (Cudaback et al. (Du et al. 2009).
2014). It remains unclear how PPARc agonists ameliorate
cognitive impairment in AD models (Table 2). One pro-
Proliferator-activated receptor gamma (PPARc) posed mechanism is that the robust anti-inflammatory
agonists effects of PPARc suppress the levels of proinflammatory
cytokines that have been linked to cognitive impairment
PPARc is expressed in cells of macrophagic lineage and its (Mandrekar-Colucci & Landreth 2011). Apart from the
activation has anti-inflammatory effects, resulting from anti-inflammatory mechanism, other properties of PPARc

Table 1 Effects of NSAIDs in AD patients


Drug Duration of treatment Dose (mg/day) Main effects on cognitive function Reference

Rofecoxib 1 year 25 Not significant Aisen et al. 2003


Naproxen 1 year 440 Not significant Aisen et al. 2003
Rofecoxib 4 years 25 Not significant Thal et al. 2005
Celecoxib 1 year 400 Not significant Soininen et al. 2007
Ibuprofen 1 year 400 Not significant Pasqualetti et al. 2009
NSAIDs N/A N/A Worsen in later stage Breitner et al. 2011
N/A not applicable

123
Involvement of inflammation in Alzheimer’s disease pathogenesis and therapeutic potential…

agonists might be responsible for reversal of the cognitive neurons and glial cells and regulate learning and memory,
deficits. For instance, PPARc agonists are reported to emotion, motor control, feeding and nociception (Howlett
stimulate Wnt signaling, resulting in neuroprotection in AD 2005). CB1 receptors are mostly localized in neuron ter-
models (Inestrosa & Toledo 2008). Recent works demon- minals where they modulate excitatory and inhibitory
strated that rosiglitazone-induced activation of ERK path- neurotransmission (Aso & Ferrer 2014). CB2 receptors are
way is related to cognitive enhancement of Tg2576 mice found in the immune system where they are involved in
(Denner et al. 2012; Jahrling et al. 2014). In addition, regulating immune cell migration and cytokine release. In
PPARc agonists improve peripheral insulin sensitivity, particular, CB2 receptors are mainly detected in microglial
which may enhance cognitive function in AD mice (Craft cells of the nervous system. They inhibit microglia-medi-
et al. 2013). ated neurotoxicity, and the main interest in the role of
Cognitive enhancement by PPARc agonists was cannabinoids as anti-inflammatory agents in several dis-
demonstrated in several clinical studies. Daily treatment eases involving inflammation has focused on compounds
with 15-30 mg pioglitazone improved cognition and cere- acting on CB2 receptors (Cabral & Griffin-Thomas 2009).
bral blood flow in mild AD (Sato et al. 2011). In contrast, Abundant evidence indicates that expression of CB2
no significant therapeutic effects were observed in a ran- receptors in microglia surrounding senile plaques is ele-
domized pilot clinical trial of the safety of pioglitazone vated (Solas et al. 2013). Intriguingly, the expression level
(Geldmacher et al. 2011). Preliminary data suggested that correlates with Ab42 levels and plaque deposition, but not
rosiglitazone preserved cognition in patients with early AD with severity of cognitive impairment, suggesting that
and amnestic mild cognitive impairment during treatment alterations in Ab metabolism induce expression of CB2
(Watson et al. 2005). Administration of rosiglitazone receptors.
(8 mg/day) improved cognitive function only in ApoE Agonists of CB2 receptors reduced the neuroinflamma-
epsilon4 non-carriers, while ApoE epsilon4 allele carriers tory response to Ab insults in different models of AD (Aso
showed no improvement or some decline (Risner et al. & Ferrer 2014). Activation of CB2 receptor by the receptor
2006). However, rosiglitazone failed to show efficacy for agonists significantly decreased neuroinflammatory
cognition enhancement in AD patients in the largest ran- responses in the brains that received the injection of Ab
domized, double-blind, placebo-controlled trials conducted (Fakhfouri et al. 2012; Martin-Moreno et al. 2011). Fur-
to date (Gold et al. 2010). In this study, no beneficial effect thermore, similar anti-neuroinflammatory effects by
of the drug was detected even in ApoE epsilon4 non-car- cannabinoids were observed in APP transgenic and tau-
riers. Edema seemed to be a common adverse effect of both pathy mice (Aso et al. 2013; Casarejos et al. 2013; Martin-
pioglitazone and rosiglitazone in the studies, despite their Moreno et al. 2012). In addition to CB2 receptors, CB1
generally high tolerability. Although overall results from receptors appeared to downregulate neuroinflammation, as
clinical trials assessing pioglitazone or rosiglitazone seem Aso et al. showed that chronic treatment with the selective
positive for treatment of AD at this moment, pharma- CB1 receptor agonist, arachidonyl-2-chloroethylamide,
coepidemiologic studies are warranted before their use can reduced the astrocytic expression of the pro-inflammatory
be recommended for AD treatment (Miller et al. 2011). cytokine interferon-c in APP/PS1 transgenic mice (Aso
et al. 2012). Recently, Stumm et al. demonstrated that
Cannabinoids deficiency in CB1 receptors exacerbated cognitive impair-
ment in spite of reduced amyloid deposition (Stumm et al.
Cannabinoids exert their physiological effect through two 2013). Cannabidiol, a dual agonist for CB1 and CB2
subtypes of cannabinoid G-protein-coupled receptors (CB1 receptors, also exhibited anti-inflammatory properties in
and CB2). CB1 receptors are abundantly expressed in both AD models (Martin-Moreno et al. 2011). These data

Table 2 Effects of PPARc agonists in AD patients


Drug Duration of Dose (mg/day) Main effects on Reference
treatment (year) cognitive function

Pioglitazone 0.5 15–30 Enhancive Sato et al. 2011


Pioglitazone 1.6 45 Not significant Geldmacher et al. 2011
Rosiglitazone 0.5 4 Preserved cognition Watson et al. 2005
Rosiglitazone 2 2, 4 or 8 Enhancive in subgroup Risner et al. 2006
Rosiglitazone 2 2 or 8 Not significant Gold et al. 2010
N/A not applicable

123
S. Shadfar et al.

suggest that both CB1 and CB2 receptors could attenuate In particular, 4-O-methylhonokiol ameliorated memory
neuroinflammatory responses in AD models. impairment in several different animal models including a
The precise mechanisms by which cannabinoids exert systemic LPS-induced dementia model (Lee et al. 2012a),
anti-neuroinflammatory and neuroprotective effects remain presenilin 2 mutant transgenic mice (Lee et al. 2011a), and
to be elucidated. One study indicated that PPARc activa- Tg2576 mice (Lee et al. 2012b). Obovatol blocked Ab
tion by cannabidiol is associated with its ameliorative aggregation and enhanced learning and memory in Tg2576
effects on microglial activation (Esposito et al. 2011). A mice by blocking neuroinflammatory responses and Ab
cannabinoid derivative attenuated LPS-mediated micro- production (Choi et al. 2012a, b).
glial activation by inhibiting NF-jB signaling pathway Compounds isolated from the Magnolia family seem to
(More et al. 2013). Besides their anti-neuroinflammatory have multiple action mechanisms. Previous studies
mechanisms, cannabinoids possess neuroprotective prop- revealed that 4-O-methylhonokiol potently inhibited neu-
erties via diminishing excitotoxicity in postsynaptic neu- roinflammatory responses and was associated with cogni-
rons (Marsicano et al. 2003), enhancing vasodilation tive enhancement in AD animal models. The anti-
through CB1 in vascular smooth muscle, and the inhibition inflammatory activity was mediated by diminishing NF-jB
of endothelin-1 (Ronco et al. 2007). signaling pathway (Lee et al. 2012a). Interestingly,
A few clinical trials with cannabinoids including dron- Schuehly et al. demonstrated that 4-O-methylhonokiol acts
abinol and nabilone showed their effects in AD patients. like a CB2 receptor ligand (Schuehly et al. 2011). They
Dronabinol treatment decreased severity of altered behav- suggest that the effects of the compound on CB2 receptors
ior and increased body weight in AD patients who were are associated with anti-neuroinflammatory effects, since
previously refusing food (Volicer et al. 1997). Two studies the receptors are primarily associated with a broad range of
described that dronabinol reduced night-time agitation and inflammatory processes (Gertsch & Anavi-Goffer 2012).
behavioral disturbances without adverse effects during the Similar to 4-O-methylhonokiol, Magnolia polyphenols or
trial period (Walther et al. 2006; Walther et al. 2011). obovatol blunted microglial activation in vitro (Chuang
Similarly, nabilone reduced the severe agitation and et al. 2013; Ock et al. 2010) and in vivo (Choi et al. 2012b).
aggressiveness exhibited by an advanced AD patient who No clinical trials with magnolia extract or pure com-
was refractory to anti-psychotic and anxiolytic medications pounds from Magnolia family to assess their therapeutic
(Passmore 2008). However, they did not evaluate cognitive effects in AD have been reported to date. Although their
or neurodegenerative markers in those studies. Importantly, efficacy remains to be tested in AD patients, various studies
the revision in 2009 of the Cochrane Dementia and Cog- demonstrated therapeutic potential as determined by
nitive Improvement Group Specialized Register found no reduction in pathology and improvement in cognitive
evidence of cannabinoid effectiveness in the improvement function. Compounds from the Magnolia family such as
of behavior and other parameters of dementia as depicted 4-O-methylhonokiol and obovatol may be strong candi-
in Table 3 (Krishnan et al. 2009). Therefore, these clinical dates for intervention in AD.
results suggest that more controlled trials are needed to
assess the effectiveness of cannabinoids in the treatment of Agents that inhibit TNFa activity
dementia.
Increasing evidence indicates that TNFa may play a central
Magnolia extract, 4-O-methylhonokiol and obovatol role in the pathogenesis of AD (Trepanier & Milgram
2010). Inhibition of TNFa activity ameliorated AD-like
Magnolia extract contains at least 255 different ingredients pathology and cognitive impairment in AD animal models.
such as 4-O-methylhonokiol and obovatol. Compounds Thalidomide, a TNFa inhibitor, significantly attenuated
isolated from Magnolia family have been shown to possess neuroinflammation, Ab deposition, and tau phosphoryla-
multiple neuropharmacologic activities (Lee et al. 2011b). tion (He et al. 2013) and improved memory function

Table 3 Effects of cannabinoid and TNFa antagonist in AD patients


Drug Duration of Dose (mg/day) Main effects on Reference
treatment cognitive function

Cannabinoids N/A N/A Not significant Krishnan et al. 2009


Etanercept 0.5 year 25–30 Enhancive Tobinick et al. 2006
Etanercept Single dose 25 Enhancive Tobinick and Gross 2008
N/A not applicable

123
Involvement of inflammation in Alzheimer’s disease pathogenesis and therapeutic potential…

(Gabbita et al. 2012). Infliximab, the antibody against References


TNFa, reduced the levels of TNFa, amyloid plaques, and
tau phosphorylation in transgenic mice (Shi et al. 2011a). Aisen, P.S., K.A. Schafer, M. Grundman, E. Pfeiffer, M. Sano, K.L.
Davis, M.R. Farlow, S. Jin, R.G. Thomas, and L.J. Thal. 2003.
Interestingly, the same group reported that intrathecal
Effects of rofecoxib or naproxen vs placebo on Alzheimer
injection of infliximab enhanced cognitive function in disease progression: A randomized controlled trial. Journal of
women with AD, while the antibody increased the con- the American Medical Association 289: 2819–2826.
centration of Ab1-42 and phospho-tau but not Ab1–40 in the Alaynick, W.A. 2008. Nuclear receptors, mitochondria and lipid
metabolism. Mitochondrion 8: 329–337.
cerebrospinal fluid and blood 7 days after treatment (Shi
Alexandrov, P.N., P. Dua, J.M. Hill, S. Bhattacharjee, Y. Zhao, and
et al. 2011b). Another potent inhibitor of TNFa is etaner- W.J. Lukiw. 2012. microRNA (miRNA) speciation in Alzhei-
cept which is currently approved for rheumatoid arthritis. mer’s disease (AD) cerebrospinal fluid (CSF) and extracellular
In a pilot study, perispinal administration of etanercept fluid (ECF). International Journal of Biochemistry and Molec-
ular Biology 3: 365–373.
(25–50 mg, once weekly) for 6 months significantly
Alexandrov, P.N., P. Dua, and W.J. Lukiw. 2014. Up-Regulation of
improved all primary efficacy variables relative to baseline miRNA-146a in progressive, age-related inflammatory neurode-
(Tobinick et al. 2006). In addition, the same group reported generative disorders of the human CNS. Frontiers in Neurology
significant effects of etanercept on most neuropsychologi- 5: 181.
Andersen, K., L.J. Launer, A. Ott, A.W. Hoes, M.M. Breteler, and A.
cal measures and notable improvements in verbal learning,
Hofman. 1995. Do nonsteroidal anti-inflammatory drugs
memory and fluency of 12 patients with mild-to-severe AD decrease the risk for Alzheimer’s disease? The Rotterdam study.
as described in Table 3 (Tobinick & Gross 2008). Neurology 45: 1441–1445.
Although these small pilot studies show promise for TNF-a Aso, E., and I. Ferrer. 2014. Cannabinoids for treatment of
Alzheimer’s disease: Moving toward the clinic. Frontiers in
inhibition as a potential therapeutic for AD, these studies
Pharmacology 5: 37.
were limited by small sample sizes, open-label design, and Aso, E., S. Juves, R. Maldonado, and I. Ferrer. 2013. CB2
lack of placebo groups. Therefore, a well-designed study cannabinoid receptor agonist ameliorates Alzheimer-like pheno-
with large scale and placebo group are warranted for type in AbetaPP/PS1 mice. Journal of Alzheimers Disease 35:
847–858.
conclusive results of etanercept in AD treatment.
Aso, E., E. Palomer, S. Juves, R. Maldonado, F.J. Munoz, and I.
Ferrer. 2012. CB1 agonist ACEA protects neurons and reduces
the cognitive impairment of AbetaPP/PS1 mice. Journal of
Conclusion Alzheimer’s Disease 30: 439–459.
Benzing, W.C., J.R. Wujek, E.K. Ward, D. Shaffer, K.H. Ashe, S.G.
Younkin, and K.R. Brunden. 1999. Evidence for glial-mediated
Based on accumulating epidemiological and preclinical data, inflammation in aged APP(SW) transgenic mice. Neurobiology
the hypothesis has been drawn that inflammatory responses are of Aging 20: 581–589.
highly related to early stage AD. But most clinical trials with Bhattacharjee, S., Y. Zhao, and W.J. Lukiw. 2014. Deficits in the
miRNA-34a-regulated endogenous TREM2 phagocytosis sen-
anti-inflammatory agents have not been successful to date and
sor-receptor in Alzheimer’s disease (AD); an update. Frontiers
do not fully support the hypothesis. A number of competing in Aging Neuroscience 6: 116.
hypotheses can explain the failure of preclinical studies to Breitner, J.C., L.D. Baker, T.J. Montine, C.L. Meinert, C.G. Lyketsos,
translate into the clinical setting and we cannot presently K.H. Ashe, J. Brandt, S. Craft, D.E. Evans, R.C. Green, M.S.
Ismail, B.K. Martin, M.J. Mullan, M. Sabbagh, P.N. Tariot, and
confirm which of these explanations is correct. More preclin-
A.R. Group. 2011. Extended results of the Alzheimer’s disease
ical studies of the effects of anti-inflammatories on cognition anti-inflammatory prevention trial. Alzheimer’s & Dementia 7:
and the use of more predictive animal models should facilitate 402–411.
the outcome of true positives detected clinically. The identi- Breitner, J.C., B.A. Gau, K.A. Welsh, B.L. Plassman, W.M.
McDonald, M.J. Helms, and J.C. Anthony. 1994. Inverse
fication of genetic markers or other biomarkers for the disease
association of anti-inflammatory treatments and Alzheimer’s
will enable an earlier point of intervention, providing a more disease: Initial results of a co-twin control study. Neurology 44:
effective means of preventing the inflammatory response. The 227–232.
development of more selective inhibitors that target specific Browne, T.C., K. McQuillan, R.M. McManus, J.A. O’Reilly, K.H.
Mills, and M.A. Lynch. 2013. IFN-gamma Production by
mediators in the inflammatory cascade, such as etanercept for
amyloid beta-specific Th1 cells promotes microglial activation
TNF-a, may also offer new therapeutic avenues over nonse- and increases plaque burden in a mouse model of Alzheimer’s
lective anti-inflammatories. disease. The Journal of Immunology 190: 2241–2251.
Cabral, G.A., and L. Griffin-Thomas. 2009. Emerging role of the
Acknowledgments This work was fully supported by 2015 cannabinoid receptor CB2 in immune regulation: Therapeutic
Yeungnam University Research Grant. prospects for neuroinflammation. Expert Reviews in Molecular
Medicine 11: e3.
Compliance with ethical standards Camacho, I.E., L. Serneels, K. Spittaels, P. Merchiers, D. Dominguez,
and B. De Strooper. 2004. Peroxisome-proliferator-activated
Conflict of interest The authors of the present article declare that receptor gamma induces a clearance mechanism for the amyloid-
there is no conflict of interest. beta peptide. Journal of Neuroscience 24: 10908–10917.

123
S. Shadfar et al.

Carreras, I., A.C. McKee, J.K. Choi, N. Aytan, N.W. Kowall, B.G. transgenic mice. Biochemical and Biophysical Research Com-
Jenkins, and A. Dedeoglu. 2013. R-flurbiprofen improves tau, munications 384: 357–361.
but not Ass pathology in a triple transgenic model of Ekdahl, C.T., Z. Kokaia, and O. Lindvall. 2009. Brain inflammation
Alzheimer’s disease. Brain Research 1541: 115–127. and adult neurogenesis: the dual role of microglia. Neuroscience
Casarejos, M.J., J. Perucho, A. Gomez, M.P. Munoz, M. Fernandez- 158: 1021–1029.
Estevez, O. Sagredo, J. Fernandez Ruiz, M. Guzman, J.G. de El Khoury, J., M. Toft, S.E. Hickman, T.K. Means, K. Terada, C.
Yebenes, and M.A. Mena. 2013. Natural cannabinoids improve Geula, and A.D. Luster. 2007. Ccr2 deficiency impairs
dopamine neurotransmission and tau and amyloid pathology in a microglial accumulation and accelerates progression of Alzhei-
mouse model of tauopathy. Journal of Alzheimer’s Disease 35: mer-like disease. Nature Medicine 13: 432–438.
525–539. Esposito, G., C. Scuderi, M. Valenza, G.I. Togna, V. Latina, D. De
Choi, D.Y., J.W. Lee, G. Lin, Y.K. Lee, Y.H. Lee, I.S. Choi, S.B. Filippis, M. Cipriano, M.R. Carratu, T. Iuvone, and L. Steardo.
Han, J.K. Jung, Y.H. Kim, K.H. Kim, K.W. Oh, J.T. Hong, and 2011. Cannabidiol reduces Abeta-induced neuroinflammation
M.S. Lee. 2012a. Obovatol attenuates LPS-induced memory and promotes hippocampal neurogenesis through PPARgamma
impairments in mice via inhibition of NF-kappaB signaling involvement. PLoS ONE 6: e28668.
pathway. Neurochemistry International 60: 68–77. Etminan, M., S. Gill, and A. Samii. 2003. Effect of non-steroidal anti-
Choi, D.Y., J.W. Lee, J. Peng, Y.J. Lee, J.Y. Han, Y.H. Lee, I.S. Choi, inflammatory drugs on risk of Alzheimer’s disease: Systematic
S.B. Han, J.K. Jung, W.S. Lee, S.H. Lee, B.M. Kwon, K.W. Oh, review and meta-analysis of observational studies. British
and J.T. Hong. 2012b. Obovatol improves cognitive functions in Medical Journal 327: 128.
animal models for Alzheimer’s disease. Journal of Neurochem- Fakhfouri, G., A. Ahmadiani, R. Rahimian, A.A. Grolla, F. Moradi,
istry 120: 1048–1059. and A. Haeri. 2012. WIN55212-2 attenuates amyloid-beta-
Choi, D.Y., Y.J. Lee, J.T. Hong, and H.J. Lee. 2012c. Antioxidant induced neuroinflammation in rats through activation of cannabi-
properties of natural polyphenols and their therapeutic potentials noid receptors and PPAR-gamma pathway. Neuropharmacology
for Alzheimer’s disease. Brain Research Bulletin 87: 144–153. 63: 653–666.
Choi, S.H., S. Aid, and F. Bosetti. 2009. The distinct roles of Fisher, Y., A. Nemirovsky, R. Baron, and A. Monsonego. 2010. T
cyclooxygenase-1 and -2 in neuroinflammation: Implications for cells specifically targeted to amyloid plaques enhance plaque
translational research. Trends in Pharmacological Sciences 30: clearance in a mouse model of Alzheimer’s disease. PLoS ONE
174–181. 5: e10830.
Choi, S.H., S. Aid, L. Caracciolo, S.S. Minami, T. Niikura, Y. Frautschy, S.A., and G.M. Cole. 2010. Why pleiotropic interventions
Matsuoka, R.S. Turner, M.P. Mattson, and F. Bosetti. 2013. are needed for Alzheimer’s disease. Molecular Neurobiology 41:
Cyclooxygenase-1 inhibition reduces amyloid pathology and 392–409.
improves memory deficits in a mouse model of Alzheimer’s Fu, Q., J. Hue, and S. Li. 2007. Nonsteroidal anti-inflammatory drugs
disease. Journal of Neurochemistry 124: 59–68. promote axon regeneration via RhoA inhibition. Journal of
Chuang, D.Y., M.H. Chan, Y. Zong, W. Sheng, Y. He, J.H. Jiang, A. Neuroscience 27: 4154–4164.
Simonyi, Z. Gu, K.L. Fritsche, J. Cui, J.C. Lee, W.R. Folk, D.B. Gabbita, S.P., M.K. Srivastava, P. Eslami, M.F. Johnson, N.K.
Lubahn, A.Y. Sun, and G.Y. Sun. 2013. Magnolia polyphenols Kobritz, D. Tweedie, N.H. Greig, F.P. Zemlan, S.P. Sharma, and
attenuate oxidative and inflammatory responses in neurons and M.E. Harris-White. 2012. Early intervention with a small
microglial cells. Journal of Neuroinflammation 10: 15. molecule inhibitor for tumor necrosis factor-alpha prevents
Combs, C.K., D.E. Johnson, J.C. Karlo, S.B. Cannady, and G.E. cognitive deficits in a triple transgenic mouse model of
Landreth. 2000. Inflammatory mechanisms in Alzheimer’s Alzheimer’s disease. Journal of Neuroinflammation 9: 99.
disease: Inhibition of beta-amyloid-stimulated proinflammatory Gasparini, L., E. Ongini, and G. Wenk. 2004. Non-steroidal anti-
responses and neurotoxicity by PPARgamma agonists. Journal inflammatory drugs (NSAIDs) in Alzheimer’s disease: Old and
of Neuroscience 20: 558–567. new mechanisms of action. Journal of Neurochemistry 91:
Craft, S., B. Cholerton, and L.D. Baker. 2013. Insulin and 521–536.
Alzheimer’s disease: Untangling the web. Journal of Alzheimer’s Geldmacher, D.S., T. Fritsch, M.J. McClendon, and G. Landreth.
Disease 33: S263–S275. 2011. A randomized pilot clinical trial of the safety of
Crespo-Biel, N., C. Theunis, and F. Van Leuven. 2012. Protein tau: Prime pioglitazone in treatment of patients with Alzheimer disease.
cause of synaptic and neuronal degeneration in Alzheimer’s disease. Archives of Neurology 68: 45–50.
International Journal of Alzheimer’s Disease 2012: 251426. Gertsch, J., and S. Anavi-Goffer. 2012. Methylhonokiol attenuates
Cudaback, E., N.L. Jorstad, Y. Yang, T.J. Montine, and C.D. Keene. neuroinflammation: A role for cannabinoid receptors? Journal of
2014. Therapeutic implications of the prostaglandin pathway in Neuroinflammation 9: 135.
Alzheimer’s disease. Biochemical Pharmacology 88: 565–572. Giannakopoulos, P., F.R. Herrmann, T. Bussiere, C. Bouras, E.
Cui, L., Y. Li, G. Ma, Y. Wang, Y. Cai, S. Liu, Y. Chen, J. Li, Y. Xie, Kovari, D.P. Perl, J.H. Morrison, G. Gold, and P.R. Hof. 2003.
G. Liu, B. Zhao, and K. Li. 2014. A functional polymorphism in Tangle and neuron numbers, but not amyloid load, predict
the promoter region of microRNA-146a is associated with the cognitive status in Alzheimer’s disease. Neurology 60:
risk of Alzheimer disease and the rate of cognitive decline in 1495–1500.
patients. PLoS ONE 9: e89019. Gold, M., C. Alderton, M. Zvartau-Hind, S. Egginton, A.M. Saunders,
Denner, L.A., J. Rodriguez-Rivera, S.J. Haidacher, J.B. Jahrling, J.R. M. Irizarry, S. Craft, G. Landreth, U. Linnamagi, and S.
Carmical, C.M. Hernandez, Y. Zhao, R.G. Sadygov, J.M. Sawchak. 2010. Rosiglitazone monotherapy in mild-to-moderate
Starkey, H. Spratt, B.A. Luxon, T.G. Wood, and K.T. Dineley. Alzheimer’s disease: Results from a randomized, double-blind,
2012. Cognitive enhancement with rosiglitazone links the placebo-controlled phase III study. Dementia and Geriatric
hippocampal PPARgamma and ERK MAPK signaling pathways. Cognitive Disorders 30: 131–146.
Journal of Neuroscience 32: 16725–16735. Goldeck, D., A. Larbi, M. Pellicano, I. Alam, I. Zerr, C. Schmidt, T.
Du, J., B. Sun, K. Chen, L. Fan, and Z. Wang. 2009. Antagonist of Fulop, and G. Pawelec. 2013. Enhanced chemokine receptor
peroxisome proliferator-activated receptor gamma induces cere- expression on leukocytes of patients with Alzheimer’s disease.
bellar amyloid-beta levels and motor dysfunction in APP/PS1 PLoS ONE 8: e66664.

123
Involvement of inflammation in Alzheimer’s disease pathogenesis and therapeutic potential…

Gonzalez, H., and R. Pacheco. 2014. T-cell-mediated regulation of Jaworski, T., B. Lechat, D. Demedts, L. Gielis, H. Devijver, P.
neuroinflammation involved in neurodegenerative diseases. Borghgraef, H. Duimel, F. Verheyen, S. Kugler, and F. Van
Journal of Neuroinflammation 11: 201. Leuven. 2011. Dendritic degeneration, neurovascular defects,
Group, A.R., C.G. Lyketsos, J.C. Breitner, R.C. Green, B.K. Martin, and inflammation precede neuronal loss in a mouse model for
C. Meinert, S. Piantadosi, and M. Sabbagh. 2007. Naproxen and tau-mediated neurodegeneration. American Journal of Pathology
celecoxib do not prevent AD in early results from a randomized 179: 2001–2015.
controlled trial. Neurology 68: 1800–1808. Jonsson, T., H. Stefansson, S. Steinberg, I. Jonsdottir, P.V. Jonsson, J.
Guedes, J., A.L. Cardoso, and M.C. Pedroso de Lima. 2013. Snaedal, S. Bjornsson, J. Huttenlocher, A.I. Levey, J.J. Lah, D.
Involvement of microRNA in microglia-mediated immune Rujescu, H. Hampel, I. Giegling, O.A. Andreassen, K. Engedal,
response. Clinical and Developmental Immunology 2013: 186872. I. Ulstein, S. Djurovic, C. Ibrahim-Verbaas, A. Hofman, M.A.
Guerreiro, R., A. Wojtas, J. Bras, M. Carrasquillo, E. Rogaeva, E. Ikram, C.M. van Duijn, U. Thorsteinsdottir, A. Kong, and K.
Majounie, C. Cruchaga, C. Sassi, J.S. Kauwe, S. Younkin, L. Stefansson. 2013. Variant of TREM2 associated with the risk of
Hazrati, J. Collinge, J. Pocock, T. Lashley, J. Williams, J.C. Alzheimer’s disease. New England Journal of Medicine 368:
Lambert, P. Amouyel, A. Goate, R. Rademakers, K. Morgan, J. 107–116.
Powell, P. St George-Hyslop, A. Singleton, and J. Hardy. 2013. Kaufmann, W.E., K.I. Andreasson, P.C. Isakson, and P.F. Worley.
TREM2 variants in Alzheimer’s disease. New England Journal 1997. Cyclooxygenases and the central nervous system.
of Medicine 368: 117–127. Prostaglandins 54: 601–624.
Hartlage-Rubsamen, M., U. Zeitschel, J. Apelt, U. Gartner, H. Franke, Kettenmann, H., U.K. Hanisch, M. Noda, and A. Verkhratsky. 2011.
T. Stahl, A. Gunther, R. Schliebs, M. Penkowa, V. Bigl, and S. Physiology of microglia. Physiological Reviews 91: 461–553.
Rossner. 2003. Astrocytic expression of the Alzheimer’s disease Kim, W.G., R.P. Mohney, B. Wilson, G.H. Jeohn, B. Liu, and J.S.
beta-secretase (BACE1) is stimulus-dependent. Glia 41: Hong. 2000. Regional difference in susceptibility to lipopolysac-
169–179. charide-induced neurotoxicity in the rat brain: Role of microglia.
Hayden, K.M., P.P. Zandi, A.S. Khachaturian, C.A. Szekely, M. Journal of Neuroscience 20: 6309–6316.
Fotuhi, M.C. Norton, J.T. Tschanz, C.F. Pieper, C. Corcoran, Kitazawa, M., S. Oddo, T.R. Yamasaki, K.N. Green, and F.M.
C.G. Lyketsos, J.C. Breitner, K.A. Welsh-Bohmer, and I. Cache LaFerla. 2005. Lipopolysaccharide-induced inflammation exac-
County. 2007. Does NSAID use modify cognitive trajectories in erbates tau pathology by a cyclin-dependent kinase 5-mediated
the elderly? The Cache County study. Neurology 69: 275–282. pathway in a transgenic model of Alzheimer’s disease. Journal
He, P., X. Cheng, M. Staufenbiel, R. Li, and Y. Shen. 2013. Long- of Neuroscience 25: 8843–8853.
term treatment of thalidomide ameliorates amyloid-like pathol- Koenigsknecht-Talboo, J., and G.E. Landreth. 2005. Microglial
ogy through inhibition of beta-secretase in a mouse model of phagocytosis induced by fibrillar beta-amyloid and IgGs are
Alzheimer’s disease. PLoS ONE 8: e55091. differentially regulated by proinflammatory cytokines. Journal
Heneka, M.T., V. Gavrilyuk, G.E. Landreth, M.K. O’Banion, G. of Neuroscience 25: 8240–8249.
Weinberg, and D.L. Feinstein. 2003. Noradrenergic depletion Krishnan, S., R. Cairns, and R. Howard. 2009. Cannabinoids for the
increases inflammatory responses in brain: Effects on IkappaB treatment of dementia. Cochrane Database of Systematic
and HSP70 expression. Journal of Neurochemistry 85: 387–398. Reviews 15(2): CD007204.
Heneka, M.T., G.E. Landreth, and D.L. Feinstein. 2001. Role for Kukar, T., S. Prescott, J.L. Eriksen, V. Holloway, M.P. Murphy, E.H.
peroxisome proliferator-activated receptor-gamma in Alzhei- Koo, T.E. Golde, and M.M. Nicolle. 2007. Chronic administra-
mer’s disease. Annals of Neurology 49: 276. tion of R-flurbiprofen attenuates learning impairments in trans-
Heneka, M.T., E. Reyes-Irisarri, M. Hull, and M.P. Kummer. 2011. genic amyloid precursor protein mice. BMC Neuroscience 8: 54.
Impact and Therapeutic Potential of PPARs in Alzheimer’s Kummer, M.P., and M.T. Heneka. 2008. PPARs in Alzheimer’s
Disease. Current Neuropharmacology 9: 643–650. Disease. PPAR Research 2008: 403896.
Heneka, M.T., M. Sastre, L. Dumitrescu-Ozimek, I. Dewachter, J. Landreth, G.E., and M.T. Heneka. 2001. Anti-inflammatory actions of
Walter, T. Klockgether, and F. Van Leuven. 2005a. Focal glial peroxisome proliferator-activated receptor gamma agonists in
activation coincides with increased BACE1 activation and Alzheimer’s disease. Neurobiology of Aging 22: 937–944.
precedes amyloid plaque deposition in APP[V717I] transgenic Laporte, V., G. Ait-Ghezala, C.H. Volmar, and M. Mullan. 2006.
mice. Journal of Neuroinflammation 2: 22. CD40 deficiency mitigates Alzheimer’s disease pathology in
Heneka, M.T., M. Sastre, L. Dumitrescu-Ozimek, A. Hanke, I. transgenic mouse models. Journal of Neuroinflammation 3: 3.
Dewachter, C. Kuiperi, K. O’Banion, T. Klockgether, F. Van Lee, Y.J., D.Y. Choi, I.S. Choi, K.H. Kim, Y.H. Kim, H.M. Kim, K.
Leuven, and G.E. Landreth. 2005b. Acute treatment with the Lee, W.G. Cho, J.K. Jung, S.B. Han, J.Y. Han, S.Y. Nam, Y.W.
PPARgamma agonist pioglitazone and ibuprofen reduces glial Yun, J.H. Jeong, K.W. Oh, and J.T. Hong. 2012a. Inhibitory
inflammation and Abeta1-42 levels in APPV717I transgenic effect of 4-O-methylhonokiol on lipopolysaccharide-induced
mice. Brain 128: 1442–1453. neuroinflammation, amyloidogenesis and memory impairment
Hickman, S.E., E.K. Allison, and J. El Khoury. 2008. Microglial via inhibition of nuclear factor-kappaB in vitro and in vivo
dysfunction and defective beta-amyloid clearance pathways in models. Journal of Neuroinflammation 9: 35.
aging Alzheimer’s disease mice. Journal of Neuroscience 28: Lee, Y.J., D.Y. Choi, Y.K. Lee, Y.M. Lee, S.B. Han, Y.H. Kim, K.H.
8354–8360. Kim, S.Y. Nam, B.J. Lee, J.K. Kang, Y.W. Yun, K.W. Oh, and
Howlett, A.C. 2005. Cannabinoid receptor signaling. Handbook of J.T. Hong. 2012b. 4-O-methylhonokiol prevents memory impair-
Experimental Pharmacology 168: 53–79. ment in the Tg2576 transgenic mice model of Alzheimer’s
Inestrosa, N.C., and E.M. Toledo. 2008. The role of Wnt signaling in disease via regulation of beta-secretase activity. Journal of
neuronal dysfunction in Alzheimer’s Disease. Molecular Neu- Alzheimer’s Disease 29: 677–690.
rodegeneration 3: 9. Lee, Y.J., I.S. Choi, M.H. Park, Y.M. Lee, J.K. Song, Y.H. Kim, K.H.
Jahrling, J.B., C.M. Hernandez, L. Denner, and K.T. Dineley. 2014. Kim, D.Y. Hwang, J.H. Jeong, Y.P. Yun, K.W. Oh, J.K. Jung,
PPARgamma recruitment to active ERK during memory S.B. Han, and J.T. Hong. 2011a. 4-O-Methylhonokiol attenuates
consolidation is required for Alzheimer’s disease-related cogni- memory impairment in presenilin 2 mutant mice through
tive enhancement. Journal of Neuroscience 34: 4054–4063. reduction of oxidative damage and inactivation of astrocytes

123
S. Shadfar et al.

and the ERK pathway. Free Radical Biology and Medicine 50: levels and improves cognitive performance in Tg APP 2576
66–77. mice. Journal of Neuroinflammation 9: 8.
Lee, Y.J., S.B. Han, S.Y. Nam, K.W. Oh, and J.T. Hong. 2010. Martin-Moreno, A.M., D. Reigada, B.G. Ramirez, R. Mechoulam, N.
Inflammation and Alzheimer’s disease. Archives of Pharmacal Innamorato, A. Cuadrado, and M.L. de Ceballos. 2011.
Research 33: 1539–1556. Cannabidiol and other cannabinoids reduce microglial activation
Lee, Y.J., Y.M. Lee, C.K. Lee, J.K. Jung, S.B. Han, and J.T. Hong. in vitro and in vivo: Relevance to Alzheimer’s disease.
2011b. Therapeutic applications of compounds in the Magnolia Molecular Pharmacology 79: 964–973.
family. Pharmacology & Therapeutics 130: 157–176. Matsuoka, Y., M. Picciano, B. Malester, J. LaFrancois, C. Zehr, J.M.
Leuner, K., S. Hauptmann, R. Abdel-Kader, I. Scherping, U. Keil, Daeschner, J.A. Olschowka, M.I. Fonseca, M.K. O’Banion, A.J.
J.B. Strosznajder, A. Eckert, and W.E. Muller. 2007. Mitochon- Tenner, C.A. Lemere, and K. Duff. 2001. Inflammatory
drial dysfunction: The first domino in brain aging and responses to amyloidosis in a transgenic mouse model of
Alzheimer’s disease? Antioxidants & Redox Signaling 9: Alzheimer’s disease. American Journal of Pathology 158:
1659–1675. 1345–1354.
Li, Y.Y., J.G. Cui, J.M. Hill, S. Bhattacharjee, Y. Zhao, and W.J. McGeer, P.L., and E.G. McGeer. 2013. The amyloid cascade-
Lukiw. 2011. Increased expression of miRNA-146a in Alzhei- inflammatory hypothesis of Alzheimer disease: Implications for
mer’s disease transgenic mouse models. Neuroscience Letters therapy. Acta Neuropathologica 126: 479–497.
487: 94–98. McKee, A.C., I. Carreras, L. Hossain, H. Ryu, W.L. Klein, S. Oddo,
Liang, X., Q. Wang, T. Hand, L. Wu, R.M. Breyer, T.J. Montine, and F.M. LaFerla, B.G. Jenkins, N.W. Kowall, and A. Dedeoglu.
K. Andreasson. 2005. Deletion of the prostaglandin E2 EP2 2008. Ibuprofen reduces Abeta, hyperphosphorylated tau and
receptor reduces oxidative damage and amyloid burden in a memory deficits in Alzheimer mice. Brain Research 1207:
model of Alzheimer’s disease. Journal of Neuroscience 25: 225–236.
10180–10187. Miller, B.W., K.C. Willett, and A.R. Desilets. 2011. Rosiglitazone
Lim, D., A. Iyer, V. Ronco, A.A. Grolla, P.L. Canonico, E. Aronica, and pioglitazone for the treatment of Alzheimer’s disease.
and A.A. Genazzani. 2013. Amyloid beta deregulates astroglial Annals of Pharmacotherapy 45: 1416–1424.
mGluR5-mediated calcium signaling via calcineurin and Nf-kB. Monsonego, A., J. Imitola, S. Petrovic, V. Zota, A. Nemirovsky, R.
Glia 61: 1134–1145. Baron, Y. Fisher, T. Owens, and H.L. Weiner. 2006. Abeta-
Lim, G.P., F. Yang, T. Chu, E. Gahtan, O. Ubeda, W. Beech, J.B. induced meningoencephalitis is IFN-gamma-dependent and is
Overmier, K. Hsiao-Ashec, S.A. Frautschy, and G.M. Cole. associated with T cell-dependent clearance of Abeta in a mouse
2001. Ibuprofen effects on Alzheimer pathology and open field model of Alzheimer’s disease. Proceedings of the National
activity in APPsw transgenic mice. Neurobiology of Aging 22: Academy of Sciences USA 103: 5048–5053.
983–991. Monsonego, A., V. Zota, A. Karni, J.I. Krieger, A. Bar-Or, G. Bitan,
Liu, Q., H.G. Lee, K. Honda, S.L. Siedlak, P.L. Harris, A.D. Cash, X. A.E. Budson, R. Sperling, D.J. Selkoe, and H.L. Weiner. 2003.
Zhu, J. Avila, A. Nunomura, A. Takeda, M.A. Smith, and G. Increased T cell reactivity to amyloid beta protein in older
Perry. 2005. Tau modifiers as therapeutic targets for Alzheimer’s humans and patients with Alzheimer disease. Journal of Clinical
disease. Biochimica et Biophysica Acta 1739: 211–215. Investigation 112: 415–422.
Liu, Y.J., D.W. Guo, L. Tian, D.S. Shang, W.D. Zhao, B. Li, W.G. Morales, I., L. Guzman-Martinez, C. Cerda-Troncoso, G.A. Farias,
Fang, L. Zhu, and Y.H. Chen. 2010. Peripheral T cells derived and R.B. Maccioni. 2014. Neuroinflammation in the pathogen-
from Alzheimer’s disease patients overexpress CXCR2 con- esis of Alzheimer’s disease. A rational framework for the search
tributing to its transendothelial migration, which is microglial of novel therapeutic approaches. Frontiers in Cellular Neuro-
TNF-alpha-dependent. Neurobiology of Aging 31: 175–188. science 8: 112.
Lukiw, W.J. 2007. Micro-RNA speciation in fetal, adult and More, S.V., J.Y. Park, B.W. Kim, H. Kumar, H.W. Lim, S.M. Kang,
Alzheimer’s disease hippocampus. NeuroReport 18: 297–300. S. Koppula, S.H. Yoon, and D.K. Choi. 2013. Anti-neuroin-
Lukiw, W.J., Y. Zhao, and J.G. Cui. 2008. An NF-kappaB-sensitive flammatory activity of a novel cannabinoid derivative by
micro RNA-146a-mediated inflammatory circuit in Alzheimer inhibiting the NF-kappaB signaling pathway in lipopolysaccha-
disease and in stressed human brain cells. Journal of Biological ride-induced BV-2 microglial cells. Journal of Pharmaceutical
Chemistry 283: 31315–31322. Sciences 121: 119–130.
Mandrekar-Colucci, S., and G.E. Landreth. 2010. Microglia and Mori, T., N. Koyama, G.W. Arendash, Y. Horikoshi-Sakuraba, J. Tan,
inflammation in Alzheimer’s disease. CNS & Neurological and T. Town. 2010. Overexpression of human S100B exacer-
Disorders: Drug Targets 9: 156–167. bates cerebral amyloidosis and gliosis in the Tg2576 mouse
Mandrekar-Colucci, S., and G.E. Landreth. 2011. Nuclear receptors model of Alzheimer’s disease. Glia 58: 300–314.
as therapeutic targets for Alzheimer’s disease. Expert Opinion on Mucke, L., G.Q. Yu, L. McConlogue, E.M. Rockenstein, C.R.
Therapeutic Targets 15: 1085–1097. Abraham, and E. Masliah. 2000. Astroglial expression of human
Mandrekar-Colucci, S., A. Sauerbeck, P.G. Popovich, and D.M. alpha(1)-antichymotrypsin enhances alzheimer-like pathology in
McTigue. 2013. PPAR agonists as therapeutics for CNS trauma amyloid protein precursor transgenic mice. American Journal of
and neurological diseases. American Society for Neurochemistry Pathology 157: 2003–2010.
5: e00129. Nagele, R.G., J. Wegiel, V. Venkataraman, H. Imaki, K.C. Wang, and
Marsicano, G., S. Goodenough, K. Monory, H. Hermann, M. Eder, A. J. Wegiel. 2004. Contribution of glial cells to the development of
Cannich, S.C. Azad, M.G. Cascio, S.O. Gutierrez, M. van der amyloid plaques in Alzheimer’s disease. Neurobiology of Aging
Stelt, M.L. Lopez-Rodriguez, E. Casanova, G. Schutz, W. 25: 663–674.
Zieglgansberger, V. Di Marzo, C. Behl, and B. Lutz. 2003. CB1 Netland, E.E., J.L. Newton, R.E. Majocha, and B.A. Tate. 1998.
cannabinoid receptors and on-demand defense against excito- Indomethacin reverses the microglial response to amyloid beta-
toxicity. Science 302: 84–88. protein. Neurobiology of Aging 19: 201–204.
Martin-Moreno, A.M., B. Brera, C. Spuch, E. Carro, L. Garcia- Nicolakakis, N., and E. Hamel. 2010. The nuclear receptor
Garcia, M. Delgado, M.A. Pozo, N.G. Innamorato, A. Cuadrado, PPARgamma as a therapeutic target for cerebrovascular and
and M.L. de Ceballos. 2012. Prolonged oral cannabinoid brain dysfunction in Alzheimer’s disease. Frontiers in Aging
administration prevents neuroinflammation, lowers beta-amyloid Neuroscience 2: 21.

123
Involvement of inflammation in Alzheimer’s disease pathogenesis and therapeutic potential…

Nilsson, L.N., K.R. Bales, G. DiCarlo, M.N. Gordon, D. Morgan, Sastre, M., I. Dewachter, G.E. Landreth, T.M. Willson, T. Klock-
S.M. Paul, and H. Potter. 2001. Alpha-1-antichymotrypsin gether, F. van Leuven, and M.T. Heneka. 2003. Nonsteroidal
promotes beta-sheet amyloid plaque deposition in a transgenic anti-inflammatory drugs and peroxisome proliferator-activated
mouse model of Alzheimer’s disease. Journal of Neuroscience receptor-gamma agonists modulate immunostimulated process-
21: 1444–1451. ing of amyloid precursor protein through regulation of beta-
Ock, J., H.S. Han, S.H. Hong, S.Y. Lee, Y.M. Han, B.M. Kwon, and secretase. Journal of Neuroscience 23: 9796–9804.
K. Suk. 2010. Obovatol attenuates microglia-mediated neuroin- Sastre, M., T. Klockgether, and M.T. Heneka. 2006. Contribution of
flammation by modulating redox regulation. British Journal of inflammatory processes to Alzheimer’s disease: Molecular
Pharmacology 159: 1646–1662. mechanisms. International Journal of Developmental Neuro-
Olivieri, F., M.R. Rippo, A.D. Procopio, and F. Fazioli. 2013. science 24: 167–176.
Circulating inflamma-miRs in aging and age-related diseases. Sato, T., H. Hanyu, K. Hirao, H. Kanetaka, H. Sakurai, and T.
Frontiers in Genetics 4: 121. Iwamoto. 2011. Efficacy of PPAR-gamma agonist pioglitazone
Ousman, S.S., and P. Kubes. 2012. Immune surveillance in the central in mild Alzheimer disease. Neurobiology of Aging 32:
nervous system. Nature Neuroscience 15: 1096–1101. 1626–1633.
Padmanabhan, J., M. Levy, D.W. Dickson, and H. Potter. 2006. Schuehly, W., J.M. Paredes, J. Kleyer, A. Huefner, S. Anavi-Goffer,
Alpha1-antichymotrypsin, an inflammatory protein overex- S. Raduner, K.H. Altmann, and J. Gertsch. 2011. Mechanisms of
pressed in Alzheimer’s disease brain, induces tau phosphoryla- osteoclastogenesis inhibition by a novel class of biphenyl-type
tion in neurons. Brain 129: 3020–3034. cannabinoid CB(2) receptor inverse agonists. Chemistry &
Panza, F., V. Frisardi, V. Solfrizzi, B.P. Imbimbo, G. Logroscino, A. Biology 18: 1053–1064.
Santamato, A. Greco, D. Seripa, and A. Pilotto. 2012. Searcy, J.L., J.T. Phelps, T. Pancani, I. Kadish, J. Popovic, K.L.
Immunotherapy for Alzheimer’s disease: From anti-beta-amyloid Anderson, T.L. Beckett, M.P. Murphy, K.C. Chen, E.M. Blalock,
to tau-based immunization strategies. Immunotherapy 4: 213–238. P.W. Landfield, N.M. Porter, and O. Thibault. 2012. Long-term
Pasqualetti, P., C. Bonomini, G. Dal Forno, L. Paulon, E. Sinforiani, pioglitazone treatment improves learning and attenuates patho-
C. Marra, O. Zanetti, and P.M. Rossini. 2009. A randomized logical markers in a mouse model of Alzheimer’s disease.
controlled study on effects of ibuprofen on cognitive progression Journal of Alzheimer’s Disease 30: 943–961.
of Alzheimer’s disease. Aging Clinical and Experimental Sethi, P., and W.J. Lukiw. 2009. Micro-RNA abundance and stability
Research 21: 102–110. in human brain: Specific alterations in Alzheimer’s disease
Passmore, M.J. 2008. The cannabinoid receptor agonist nabilone for temporal lobe neocortex. Neuroscience Letters 459: 100–104.
the treatment of dementia-related agitation. International Jour- Shi, J.Q., W. Shen, J. Chen, B.R. Wang, L.L. Zhong, Y.W. Zhu, H.Q.
nal of Geriatric Psychiatry 23: 116–117. Zhu, Q.Q. Zhang, Y.D. Zhang, and J. Xu. 2011a. Anti-TNF-
Prosperi, C., C. Scali, M. Barba, A. Bellucci, M.G. Giovannini, G. alpha reduces amyloid plaques and tau phosphorylation and
Pepeu, and F. Casamenti. 2004. Comparison between flurbipro- induces CD11c-positive dendritic-like cell in the APP/PS1
fen and its nitric oxide-releasing derivatives HCT-1026 and transgenic mouse brains. Brain Research 1368: 239–247.
NCX-2216 on Abeta(1-42)-induced brain inflammation and Shi, J.Q., B.R. Wang, W.W. Jiang, J. Chen, Y.W. Zhu, L.L. Zhong,
neuronal damage in the rat. International Journal of Y.D. Zhang, and J. Xu. 2011b. Cognitive improvement with
Immunopathology and Pharmacology 17: 317–330. intrathecal administration of infliximab in a woman with
Qiao, X., D.J. Cummins, and S.M. Paul. 2001. Neuroinflammation- Alzheimer’s disease. Journal of the American Geriatrics Society
induced acceleration of amyloid deposition in the APPV717F 59: 1142–1144.
transgenic mouse. European Journal of Neuroscience 14: 474–482. Simpson, J.E., P.G. Ince, G. Lace, G. Forster, P.J. Shaw, F. Matthews,
Ransohoff, R.M., and B. Engelhardt. 2012. The anatomical and G. Savva, C. Brayne, and S.B. Wharton. 2010. Astrocyte
cellular basis of immune surveillance in the central nervous phenotype in relation to Alzheimer-type pathology in the ageing
system. Nature Reviews Immunology 12: 623–635. brain. Neurobiology of Aging 31: 578–590.
Ribizzi, G., S. Fiordoro, S. Barocci, E. Ferrari, and M. Megna. 2010. Sodhi, R.K., N. Singh, and A.S. Jaggi. 2011. Neuroprotective
Cytokine polymorphisms and Alzheimer disease: Possible asso- mechanisms of peroxisome proliferator-activated receptor ago-
ciations. Neurological Sciences 31: 321–325. nists in Alzheimer’s disease. Naunyn-Schmiedeberg’s Archives
Risner, M.E., A.M. Saunders, J.F. Altman, G.C. Ormandy, S. Craft, of Pharmacology 384: 115–124.
I.M. Foley, M.E. Zvartau-Hind, D.A. Hosford, and A.D. Roses. Soininen, H., C. West, J. Robbins, and L. Niculescu. 2007. Long-term
2006. Efficacy of rosiglitazone in a genetically defined popula- efficacy and safety of celecoxib in Alzheimer’s disease.
tion with mild-to-moderate Alzheimer’s disease. Pharmacoge- Dementia and Geriatric Cognitive Disorders 23: 8–21.
nomics Journal 6: 246–254. Solas, M., P.T. Francis, R. Franco, and M.J. Ramirez. 2013. CB2
Rogers, J., L.C. Kirby, S.R. Hempelman, D.L. Berry, P.L. McGeer, receptor and amyloid pathology in frontal cortex of Alzheimer’s
A.W. Kaszniak, J. Zalinski, M. Cofield, L. Mansukhani, P. disease patients. Neurobiology of Aging 34: 805–808.
Willson, et al. 1993. Clinical trial of indomethacin in Solito, E., and M. Sastre. 2012. Microglia function in Alzheimer’s
Alzheimer’s disease. Neurology 43: 1609–1611. disease. Frontiers in Pharmacology 3: 14.
Ronco, A.M., M. Llanos, D. Tamayo, and S. Hirsch. 2007. Steele, M.L., and S.R. Robinson. 2012. Reactive astrocytes give
Anandamide inhibits endothelin-1 production by human cultured neurons less support: Implications for Alzheimer’s disease.
endothelial cells: A new vascular action of this endocannabinoid. Neurobiology of Aging 33: e1–e13.
Pharmacology 79: 12–16. Stewart, W.F., C. Kawas, M. Corrada, and E.J. Metter. 1997. Risk of
Rossner, S., J. Apelt, R. Schliebs, J.R. Perez-Polo, and V. Bigl. 2001. Alzheimer’s disease and duration of NSAID use. Neurology 48:
Neuronal and glial beta-secretase (BACE) protein expression in 626–632.
transgenic Tg2576 mice with amyloid plaque pathology. Journal Stumm, C., C. Hiebel, R. Hanstein, M. Purrio, H. Nagel, A. Conrad,
of Neuroscience Research 64: 437–446. B. Lutz, C. Behl, and A.B. Clement. 2013. Cannabinoid receptor
Rossner, S., C. Lange-Dohna, U. Zeitschel, and J.R. Perez-Polo. 2005. 1 deficiency in a mouse model of Alzheimer’s disease leads to
Alzheimer’s disease beta-secretase BACE1 is not a neuron- enhanced cognitive impairment despite of a reduction in amyloid
specific enzyme. Journal of Neurochemistry 92: 226–234. deposition. Neurobiology of Aging 34: 2574–2584.

123
S. Shadfar et al.

Sung, S., H. Yang, K. Uryu, E.B. Lee, L. Zhao, D. Shineman, J.Q. Volicer, L., M. Stelly, J. Morris, J. McLaughlin, and B.J. Volicer.
Trojanowski, V.M. Lee, and D. Pratico. 2004. Modulation of 1997. Effects of dronabinol on anorexia and disturbed behavior
nuclear factor-kappa B activity by indomethacin influences A in patients with Alzheimer’s disease. International Journal of
beta levels but not A beta precursor protein metabolism in a Geriatric Psychiatry 12: 913–919.
model of Alzheimer’s disease. American Journal of Pathology Walther, S., R. Mahlberg, U. Eichmann, and D. Kunz. 2006. Delta-9-
165: 2197–2206. tetrahydrocannabinol for nighttime agitation in severe dementia.
Sy, M., M. Kitazawa, R. Medeiros, L. Whitman, D. Cheng, T.E. Lane, Psychopharmacology (Berl) 185: 524–528.
and F.M. Laferla. 2011. Inflammation induced by infection Walther, S., B. Schupbach, E. Seifritz, P. Homan, and W. Strik. 2011.
potentiates tau pathological features in transgenic mice. Amer- Randomized, controlled crossover trial of dronabinol, 2.5 mg,
ican Journal of Pathology 178: 2811–2822. for agitation in 2 patients with dementia. Journal of Clinical
Szekely, C.A., J.C. Breitner, A.L. Fitzpatrick, T.D. Rea, B.M. Psaty, Psychopharmacology 31: 256–258.
Z.H. Kuller, and P.P. Zandi. 2008. NSAID use and dementia risk Watson, G.S., B.A. Cholerton, M.A. Reger, L.D. Baker, S.R. Plymate,
in the Cardiovascular Health Study: Role of APOE and NSAID S. Asthana, M.A. Fishel, J.J. Kulstad, P.S. Green, D.G. Cook,
type. Neurology 70: 17–24. S.E. Kahn, M.L. Keeling, and S. Craft. 2005. Preserved
Thal, L.J., S.H. Ferris, L. Kirby, G.A. Block, C.R. Lines, E. Yuen, C. cognition in patients with early Alzheimer disease and amnestic
Assaid, M.L. Nessly, B.A. Norman, C.C. Baranak, and S.A. mild cognitive impairment during treatment with rosiglitazone:
Reines. 2005. A randomized, double-blind, study of rofecoxib in A preliminary study. The American Journal of Geriatric
patients with mild cognitive impairment. Neuropsychopharma- Psychiatry 13: 950–958.
cology 30: 1204–1215. Weggen, S., J.L. Eriksen, S.A. Sagi, C.U. Pietrzik, V. Ozols, A. Fauq,
Thounaojam, M.C., D.K. Kaushik, and A. Basu. 2013. MicroRNAs in T.E. Golde, and E.H. Koo. 2003. Evidence that nonsteroidal anti-
the brain: It’s regulatory role in neuroinflammation. Molecular inflammatory drugs decrease amyloid beta 42 production by
Neurobiology 47: 1034–1044. direct modulation of gamma-secretase activity. Journal of
Tobinick, E., H. Gross, A. Weinberger, and H. Cohen. 2006. TNF- Biological Chemistry 278: 31831–31837.
alpha modulation for treatment of Alzheimer’s disease: A Wenk, G.L. 2003. Neuropathologic changes in Alzheimer’s disease.
6-month pilot study. Medscape General Medicine 8: 25. Journal of Clinical Psychiatry 64: 7–10.
Tobinick, E.L., and H. Gross. 2008. Rapid cognitive improvement in Yan, Q., J. Zhang, H. Liu, S. Babu-Khan, R. Vassar, A.L. Biere, M.
Alzheimer’s disease following perispinal etanercept administra- Citron, and G. Landreth. 2003. Anti-inflammatory drug therapy
tion. Journal of Neuroinflammation 5: 2. alters beta-amyloid processing and deposition in an animal
Togo, T., H. Akiyama, E. Iseki, H. Kondo, K. Ikeda, M. Kato, T. Oda, model of Alzheimer’s disease. Journal of Neuroscience 23:
K. Tsuchiya, and K. Kosaka. 2002. Occurrence of T cells in the 7504–7509.
brain of Alzheimer’s disease and other neurological diseases. Yoshiyama, Y., M. Higuchi, B. Zhang, S.M. Huang, N. Iwata, T.C.
Journal of Neuroimmunology 124: 83–92. Saido, J. Maeda, T. Suhara, J.Q. Trojanowski, and V.M. Lee.
Townsend, K.P., T. Town, T. Mori, L.F. Lue, D. Shytle, P.R. Sanberg, 2007. Synapse loss and microglial activation precede tangles in a
D. Morgan, F. Fernandez, R.A. Flavell, and J. Tan. 2005. CD40 P301S tauopathy mouse model. Neuron 53: 337–351.
signaling regulates innate and adaptive activation of microglia in Zahs, K.R., and K.H. Ashe. 2010. ‘Too much good news’—Are
response to amyloid beta-peptide. European Journal of Alzheimer mouse models trying to tell us how to prevent, not
Immunology 35: 901–910. cure, Alzheimer’s disease? Trends in Neurosciences 33:
Trepanier, C.H., and N.W. Milgram. 2010. Neuroinflammation in 381–389.
Alzheimer’s disease: Are NSAIDs and selective COX-2 Zhao, Y., S. Bhattacharjee, B.M. Jones, P. Dua, P.N. Alexandrov,
inhibitors the next line of therapy? Journal of Alzheimer’s J.M. Hill, and W.J. Lukiw. 2013. Regulation of TREM2
Disease 21: 1089–1099. expression by an NF-small ka, CyrillicB-sensitive miRNA-34a.
Verkhratsky, A., M. Olabarria, H.N. Noristani, C.Y. Yeh, and J.J. NeuroReport 24: 318–323.
Rodriguez. 2010. Astrocytes in Alzheimer’s disease. Neurother-
apeutics 7: 399–412.
Vitek, M.P., C.M. Brown, and C.A. Colton. 2009. APOE genotype-
specific differences in the innate immune response. Neurobiol-
ogy of Aging 30: 1350–1360.

123

Você também pode gostar