You are on page 1of 11

Neuropharmacology xxx (2017) 1e11

Contents lists available at ScienceDirect

Neuropharmacology
journal homepage: www.elsevier.com/locate/neuropharm

Invited review

Crosstalk between endoplasmic reticulum stress and brain


inflammation in Alzheimer's disease
Luis E. Santos a, Sergio T. Ferreira a, b, *
a
Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
b
Institute of Biophysics Carlos Chagas Filho, 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: While most often noted for its cognitive symptoms, Alzheimer's disease (AD) is, at its core, a disease of
Received 29 September 2017 protein misfolding/aggregation, with an intriguing inflammatory component. Defective clearance and/or
Accepted 8 November 2017 abnormal production of the amyloid-b peptide (Ab), and its ensuing accumulation and aggregation,
Available online xxx
underlie two hallmark features of AD: brain accumulation of insoluble protein deposits known as am-
yloid or senile plaques, and buildup of soluble Ab oligomers (AbOs), diffusible toxins linked to synapse
Keywords:
dysfunction and memory impairment. In neurons, as in typical eukaryotic cells, the endoplasmic retic-
Alzheimer's disease
ulum (ER) serves as a main compartment for the folding, maturation, trafficking and quality control of
Endoplasmic reticulum stress
Unfolded protein response
newly synthesized proteins. The ER lumen, a calcium-rich, oxidizing environment, provides favorable
Inflammation conditions for these physiological functions to occur. These conditions, however, also favor protein ag-
gregation. Several stressors, including metabolic/nutrient stress and certain pathologies, may upset the
ER homeostasis, e.g., by affecting calcium levels or by causing the accumulation of unfolded or misfolded
proteins. Whatever the underlying cause, the result is what is commonly known as “ER stress”. This, in
turn, triggers a conserved cellular response mechanism known as the “unfolded protein response” (UPR).
The UPR comprises three pathways involving transcriptional or translational regulators aimed at
normalizing ER function, and each of them results in pro-inflammatory signaling. A positive feedback
loop exists between ER stress and inflammation, with clear implications for neurodegeneration and AD.
Here, we explore recent findings on the role of ER stress and the UPR in inflammatory processes leading
to synapse failure and memory impairment in AD.
© 2017 Elsevier Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2. The UPR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3. The UPR in AD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4. The inflammatory component of AD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5. ER stress intersects with neuroinflammation and AD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6. Nitric oxide regulation of the UPR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
7. Potential therapeutic approaches targeting ER stress/inflammation in AD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
8. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

1. Introduction
* Corresponding author. Institute of Biophysics Carlos Chagas Filho, Federal
University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil. Protein synthesis, folding, maturation and trafficking are highly
E-mail address: ferreira@bioqmed.ufrj.br (S.T. Ferreira).

https://doi.org/10.1016/j.neuropharm.2017.11.016
0028-3908/© 2017 Elsevier Ltd. All rights reserved.

Please cite this article in press as: Santos, L.E., Ferreira, S.T., Crosstalk between endoplasmic reticulum stress and brain inflammation in
Alzheimer's disease, Neuropharmacology (2017), https://doi.org/10.1016/j.neuropharm.2017.11.016
2 L.E. Santos, S.T. Ferreira / Neuropharmacology xxx (2017) 1e11

regulated processes, which readily respond to metabolic state, exact process by which BiP removal leads to such activation is not
physiological signals and pathological stressors. In eukaryotic cells, fully understood but, at least for PERK and IRE-1, appears to rely on
the endoplasmic reticulum (ER) is responsible for a significant their oligomerization and cross-phosphorylation (Bertolotti et al.,
portion of these functions, processing all membrane-bound and 2000; Janssens et al., 2014). Notably, in addition to this canonical
secretory proteins following their synthesis at adjacent ribosomes. mechanism based entirely on BiP, more recent reports have begun
Any perturbation of ER homeostasis may result in the accumulation to describe novel sources of UPR sensor activation. These include
of unfolded or misfolded proteins in its lumen - a condition known the direct binding of unfolded proteins (Gardner and Walter, 2011)
as ER stress (Hetz and Glimcher, 2009; Hotamisligil, 2010; and lipids (Volmer et al., 2013) to the UPR sensors, and the
Chaudhari et al., 2014; Hetz and Mollereau, 2014). participation of an additional ER protein, CNPY2, which may be
The ever-growing list of pathologies linked to ER stress includes required for full PERK activation after BiP release (Hong et al., 2017).
several amyloidoses, a family of diseases defined wholly or in part IRE-1 signaling is mediated by its endoribonuclease activity.
by protein misfolding and aggregation, which includes type 2 dia- Phosphorylated IRE-1 catalyzes splicing of the mRNA encoding
betes (T2D), prion diseases, Parkinson's disease, and Alzheimer's XBP1 (X box-binding protein 1). The alternatively spliced version of
disease (Lindholm et al., 2006; Mukherjee et al., 2015; Torres et al., this transcription factor, often termed XBP1s, finds its way into the
2015; Chiti and Dobson, 2017; Dogan, 2017). Interestingly, however, nucleus to upregulate the expression of several ER chaperones and
ER stress has also been implicated in the development of many enzymes involved in ERAD, contributing to restore normal ER
conditions with weaker or as yet unknown ties to protein mis- function (Lee et al., 2003).
folding and deposition. A notable example is major depressive Once active, the second UPR sensor, PERK, inhibits overall pro-
disorder (Bown et al., 2000; Nevell et al., 2014; Timberlake and tein synthesis by phosphorylating the alpha subunit of eukaryotic
Dwivedi, 2016), a condition associated with brain inflammation. initiation factor 2 (eIF2a), which delays formation of the ternary
Indeed, it is becoming increasingly clear that the connection be- complex comprising eIF2/tRNAMet/GTP and the assembly of func-
tween ER stress and related pathologies may go well beyond pro- tional ribosomes (Jackson et al., 2010). Attenuated translation is
teostasis, resting to some extent on its overlap with inflammation essential to lower the unfolded protein burden in the ER during
(Gold et al., 2013). stress conditions, and promotes cell survival, at least in the short-
Although a complex inflammatory component in Alzheimer's term (Ron and Walter, 2007). PERK is one of the four known
disease (AD) has been recognized for decades, whether it is a eIF2a kinases, and the only one resident in the ER. The other three,
consequence of neurodegeneration, a contributing factor, or part of HRI (heme-regulated inhibitor), GCN2 (general control non-
the underlying mechanism of pathogenesis remains to be deter- derepressible 2) and PKR (double-stranded RNA-dependent pro-
mined (Griffin et al., 1989, 1995; Holmes et al., 2009; Glass et al., tein kinase), have links to ER stress but are also subject to inde-
2010; De Felice and Ferreira, 2014; Ferreira et al., 2014). Mounting pendent activation pathways (Donnelly et al., 2013). For this reason,
evidence also points to a role for ER stress in AD, which, as argued they are considered part of what is known as the integrated stress
in the following sections, may be non-dissociable from the in- response (ISR; Harding et al., 2003; Pakos-Zebrucka et al., 2016).
flammatory response in causing neuronal/synapse dysfunction and The last branch of the UPR to be identified operates via trans-
memory failure in AD. location of ATF6 to the Golgi under ER stress. Releasing BiP unmasks
Golgi localization signals present in the ATF6 protein, which guide
2. The UPR its trafficking to the Golgi. Once in the Golgi apparatus, ATF6 is
processed by two local proteases, site-1 (S1P) and site-2 (S2P),
The unfolded protein response (UPR) is a conserved adaptive releasing its active form, known as ATF6 fragment (ATF6f) or 50kD-
mechanism triggered by ER stress and aimed at restoring proper ER ATF6, in reference to its cleaved, shorter size (Shen et al., 2002). Like
function. Through the activation of one or more of its three XBP1s, ATF6f is an active transcription factor that directs the
branches (see below), the early UPR attenuates protein synthesis, expression of genes involved in protein folding and ERAD (Janssens
while facilitating protein folding, degradation and autophagy. et al., 2014).
However, upon sustained activation by prolonged stress, the UPR
becomes maladaptive and may promote apoptosis (Hetz, 2012). 3. The UPR in AD
Several foldases and chaperones populate the ER (Jansen et al.,
2012), driving the protein maturation steps that take place in its AD, the most common form of old-age dementia, a group of
lumen: folding, quality control, ER-associated protein degradation conditions affecting nearly 47 million people worldwide (Prince
(ERAD) and export. The most abundant ER chaperone is BiP et al., 2015), is a prototypical amyloidosis: misfolded and aggre-
(immunoglobulin heavy chain binding protein), sometimes gated proteins compose its defining neuropathological lesions,
referred to as GRP78 (78-kDa glucose-regulated protein), one the namely amyloid-b (Ab) plaques (Glenner and Wong, 1984) and
first proteins identified as ER stress responsive (Kozutsumi et al., neurofibrillary tangles made up of hyperphosphorylated tau
1988; Lenny and Green, 1991; Li et al., 1994). BiP binds exposed (Grundke-Iqbal et al., 1986a, 1986b; Kosik et al., 1986). Clinically, AD
hydrophobic domains in partially folded proteins, preventing their presents with gradual cognitive decline, accompanied by debili-
misfolding and aggregation. An ATPase domain in the N-terminal of tating behavioral symptoms, including apathy, depression and de-
BiP regulates its affinity for protein targets, allowing proteins to be lusions, among others (Lyketsos et al., 2011; Santos et al., 2016). At
bound and released in a cyclic manner as they fold (Naidoo, 2009). the neuronal level, it involves excitotoxicity, neuronal oxidative
In animals, three transmembrane ER protein sensors serve as stress, deregulation of intracellular signaling pathways, and syn-
the main regulators of the UPR: IRE-1 (inositol requiring protein 1; apse damage/loss (Haass and Selkoe, 2007; Ferreira and Klein,
Cox et al., 1993; Morl et al., 1993), PERK (PKR-like ER kinase; 2011; Ferreira et al., 2015; Selkoe and Hardy, 2016).
Harding et al., 1999), and ATF6 (activating transcription factor 6; In 1991, before any of the UPR sensors had been formally
Haze et al., 1999). Under normal conditions, all three of these described, Hamos and co-workers reported the first measurements
sensors are bound to BiP in their luminal domains. This prevents of BiP expression in post-mortem AD brains (Hamos et al., 1991).
competing interactions, and keeps them inactive. However, upon They found BiP was upregulated in AD brains, especially in the
ER stress, BiP sequestration by accumulating misfolded/partially hippocampus and entorhinal cortex, but only in neurons retaining
folded proteins releases the sensors to trigger UPR signaling. The normal morphology. Another UPR-related chaperone, HSP72,

Please cite this article in press as: Santos, L.E., Ferreira, S.T., Crosstalk between endoplasmic reticulum stress and brain inflammation in
Alzheimer's disease, Neuropharmacology (2017), https://doi.org/10.1016/j.neuropharm.2017.11.016
L.E. Santos, S.T. Ferreira / Neuropharmacology xxx (2017) 1e11 3

which is expressed in the cytoplasm rather than in the ER (Gupta IRE1 was the latest ER sensor to be linked to AD. In a case control
et al., 2010), was also notably increased in AD, and was often study involving 276 AD subjects (diagnosed using NINCDS-ADRDA
associated with Ab plaques and tau tangles (Hamos et al., 1991). criteria) and 254 healthy age-matched controls, an association was
That same year, an independent group reported increased levels of found between an XBP1 polymorphism and AD risk (Liu et al.,
HSP72 and HSP73 (another cytoplasmic chaperone) in the temporal 2013). Establishing the clinical relevance of this early report, an
cortex of AD patients (Perez et al., 1991). important role of XBP1 in memory formation was recently
Those early results were recently revisited and expanded by demonstrated (reviewed by Cisse ! et al. (2016)). Martinez et al.
other authors. The increase in BiP expression was confirmed, and (2016) used conditional knockout mice to show that lack of XBP1
phosphorylated forms of PERK, IRE1a and eIF2a were found to be in the central nervous system (CNS) impairs long-term potentiation
markedly increased in AD brains compared to age-matched con- (LTP) and memory, while overexpression of XBP1s is beneficial, and
trols (Hoozemans et al., 2005, 2009; Yoon et al., 2012; Stutzbach that these effects were connected to BDNF expression. Shortly after,
et al., 2013; Duran-Aniotz et al., 2017). Interestingly, Stutzbach Cisse! et al. (2016a) showed that directed expression of XBP1s in the
et al. (2013) further reported that UPR activation correlates with hippocampus restores cognitive deficits in a triple transgenic
age, even in the absence of pathology. Consistent with the initial mouse model of AD. This effect depended on activation of the
report by Hamos et al. (1991), increased levels of UPR markers have Kalirin-7 pathway, which is downstream to NMDA receptor acti-
been found mostly in morphologically intact neurons (Hoozemans vation, linked to synaptic plasticity, and known to be impaired in
et al., 2009, 2005). The UPR thus appears to be an early event AD patients and animal models.
preceding overt protein deposition in AD and may be neuro- Interestingly, in a recent development, the consequences of
protective, at least in the short-term (Hoozemans et al., 2012). direct IRE-1 deficiency on the AD-like phenotype of 5xFAD mice
On the other hand, pro-apoptotic components of the UPR have were assessed. While results similar to those seen with conditional
also been found to be elevated in AD brains, possibly as a conse- XBP1 knockouts might have been expected, AD mice harboring an
quence of prolonged ER stress. A transcription factor known as IRE-1 variant that lacked its ribonuclease domain were surprisingly
CHOP/GADD153 (CCAAT-enhancer-binding protein homologous found to have lower amyloid burden, improved cognition and to
protein/Growth arrest and DNA damage-inducible gene 153), form and sustain LTP at wild-type levels (Duran-Aniotz et al., 2017).
which is possibly the main link between ER stress and apoptosis, is This result underscores the complex role of the UPR sensors in AD,
increased in the brains of AD patients (Yoon et al., 2012), along with and suggests that XBP1 may have unknown functions that are in-
downstream effectors such as caspase-12 (Lee et al., 2010) and dependent of IRE-1. Indeed, the authors of the latter study noted
GADD34 (Honjo et al., 2015). CHOP/GADD153 is induced to some that, unlike XBP1 knockouts, mice deficient in IRE-1 had no alter-
extent by all three UPR branches, but is mostly associated with ations in hippocampal BDNF levels (Duran-Aniotz et al., 2017).
PERK. While eIF2a phosphorylation by activated PERK or other ki-
nases impairs general translation, it promotes synthesis of specific 4. The inflammatory component of AD
proteins, including ATF4, a transcription factor that drives CHOP
expression (Chaudhari et al., 2014). Stress-induced apoptosis may The notion that neuroinflammation is a component of AD pa-
have physiological roles, such as eliminating cells that become thology is arguably as old as the disease's first description. In Alz-
unresponsive to the UPR, but in AD it is a likely cause of neuro- heimer's seminal report, in 1907 (translated and commented in
degeneration (Baleriola et al., 2014). Stelzmann et al., 1995), he mentions glial alterations in the
In mouse models of AD, where specific aspects of the disease can affected brain, including “numerous fibers”, which most likely re-
be studied in isolation and correlations put to test, robust data flected reactive gliosis - activated astrocytes and microglia. The
support similar conclusions. AD-like pathology induced by precise role of neuroinflammation in AD, however, is yet to be
administration of the Ab peptide or its genetic overproduction was completely understood. A range of hypotheses currently suggest
shown to increase levels of BiP (Barbero-Camps et al., 2014), inflammation may be a beneficial response, a driver of symptoms,
phosphorylated eIF2a and CHOP (Baleriola et al., 2014; Yoon et al., or even a primary causal factor of the disease (Heneka et al., 2015;
2012), GADD34 and cleaved caspase-12 (Barbero-Camps et al., Santos et al., 2016).
2014; Xu et al., 2015). Notably, neurodegeneration associated The latter possibility, sometimes referred to as the ‘inflamma-
with intrahippocampal injections of Ab was shown to require tory hypothesis of AD’, has gained significant traction in later years.
activation of the ATF4-CHOP pathway (Baleriola et al., 2014). Evidence in its support includes notable correlations between
Three eIF2a kinases have been implicated in synapse loss and increased incidence of AD and conditions involving local or
cognitive deficits in AD mouse models. Ma et al. (2013) crossed PERK generalized inflammation (De Felice, 2013). These include major
and GCN2 knockouts with APPswePS1DE9 transgenic mice, a ge- depression (Green et al., 2003; Lyketsos and Olin, 2002), traumatic
netic model of the disease, in order to test whether those kinases brain injury (Breunig et al., 2013), periodontitis (Ide et al., 2016),
could be potential targets for therapy in AD. Interestingly, they found infections (Honjo et al., 2009), and metabolic disorders such as type
that eliminating either enzyme reduced brain levels of Ab and pre- 2 diabetes and obesity (De Felice and Ferreira, 2014; Ferreira et al.,
vented memory deficits associated with the APPswePS1DE9 2014). As the list of these AD-related conditions becomes more
phenotype (Ma et al., 2013). Around the same time, work by our diverse, neuroinflammation and the release of inflammatory me-
group showed that PKR-mediated phosphorylation of eIF2a un- diators - either systemically or in the brain - increasingly seems to
derlies synapse damage and memory impairment in the same be their common denominator.
APPswePS1DE9 transgenic mice as well as in mice that received an Even during low-grade insults, prostaglandins or cytokines such
intracerebroventricular infusion of Ab oligomers (Lourenco et al., as TNF-a and IL-1b may cross the blood-brain-barrier and prime
2013). brain cells to respond to inflammatory stimuli in a dysfunctional
These results are consistent with the established role of eIF2a in manner (Shie et al., 2005; Teeling et al., 2007; Maes et al., 2012;
the physiology of memory, and the potential of ER stress and the UPR Norden et al., 2015). While such primed cells do not actively
to interfere with it. Indeed, protein synthesis is required for late-LTP release cytokines, they do, however, upregulate genes related to
and long term memory, and a disruption in translation due to eIF2a antigen recognition, cytokine production and phagocytosis, which
phosphorylation is expected to impair synaptic plasticity and overall exaggerate their responses when a secondary pro-inflammatory
memory performance (Costa-Mattioli et al., 2009, 2007, 2005). stimulus arrives, often resulting in cytotoxicity (Perry and

Please cite this article in press as: Santos, L.E., Ferreira, S.T., Crosstalk between endoplasmic reticulum stress and brain inflammation in
Alzheimer's disease, Neuropharmacology (2017), https://doi.org/10.1016/j.neuropharm.2017.11.016
4 L.E. Santos, S.T. Ferreira / Neuropharmacology xxx (2017) 1e11

Holmes, 2014). Microglia, the resident macrophages of the brain, restricted to the cytoplasm. Phosphorylated IkB is prone to ubiq-
are typically described as the only CNS cell population susceptible uitination and degradation, leaving NF-kB free to translocate to the
to priming, perhaps, at least in part, because this concept was first nucleus and drive gene expression (Hinz and Scheidereit, 2014; Hu
noted in peripheral macrophages (Cherry et al., 2014). However, et al., 2006; Kaneko et al., 2003; Leonardi, 2002). The TRAF2-IRE-1
recent findings suggest a similar phenomenon may also occur in complex may also bind ASK-1 (apoptosis signal-regulating kinase
astrocytes (Hennessy et al., 2015). 1) to activate the JNK (c-Jun N-terminal kinase) pathway, promot-
Abnormally elevated responses to inflammatory stimuli have ing inflammatory signaling through the transcription factor known
been described in a transgenic mouse model of AD (Sly et al., 2001), as AP-1 (activator protein-1; Nishitoh et al., 2002) (Fig. 1). Addi-
as well as in microglia isolated from AD patients (Lue et al., 2001). tionally, in the recent report that examined deletion of the IRE-1
Additionally, microglial priming was shown to be, to some extent, ribonuclease domain in 5xFAD mice, the authors found that AD
an aspect of normal aging (Luo et al., 2010), suggesting that mice harboring the IRE-1 mutation had significantly reduced
increased susceptibility to neuroinflammation may explain, at least astrogliosis in the hippocampus (Duran-Aniotz et al., 2017).
in part, why age is the main risk factor for AD. Although this result was not explored in depth, it suggested that
There is also evidence for a causal connection between micro- XBP1s may also have a direct or indirect role in driving inflam-
glial activation and both cognitive and non-cognitive behavioral mation in AD.
symptoms (e.g., mood alterations) in AD (Henry et al., 2008; Maes As mentioned above, PERK (and, subsequently, eIF2a) phos-
et al., 2012; Sarlus and Heneka, 2017). While mood alterations are phorylation leads to overall inhibition of protein synthesis, with
not unique to Alzheimer's, the link to activated microglia is well- certain mRNAs being preferentially translated (Baleriola et al.,
established both in and out of the AD context, having been 2014; Hetz, 2012). IkB is amongst the proteins with inhibited
demonstrated in models of inflammation-induced ‘sickness translation, and its reduced expression under ER stress underlies an
behavior’ (Henry et al., 2008; Maes et al., 2012), mouse models of additional mechanism for NF-kB activation (Deng et al., 2004). In
AD (Hong et al., 2016; Ledo et al., 2016, 2013; Spangenberg et al., the mouse brain, activated PERK in astrocytes was also shown to
2016), and in dementia patients (Femminella et al., 2016). produce inflammation by increasing IL-6 expression through the
By genetically labeling and long-term tracking of a subset of JAK1-STAT3 pathway (Meares et al., 2014). Further, PERK may
cortical microglia, a recent report elegantly showed that APPswe/ directly phosphorylate GSK-3b (Glycogen Synthase Kinase 3b;
PS1L166P mice undergo a threefold expansion of the microglial Baltzis et al., 2007; Resende et al., 2008), an enzyme not only well-
population throughout the course of amyloid pathology (Füger described as a regulator of inflammatory pathways, including NF-
et al., 2017). This observation confirmed earlier results reached kB (Maixner and Weng, 2013), but also thoroughly linked to AD
with more limited techniques (Radde et al., 2006). Further, under pathology, where it influences Ab synthesis and functions as one of
physiological conditions, half of the microglia in the mouse brain the major tau kinases (Llorens-Martín et al., 2014).
likely survive for the entire lifespan of the animal (Füger et al., Finally, the ATF6 branch of the UPR was also shown to promote
2017). Such a long turnover is compatible with the long-lasting inflammation in certain contexts, through the CREBH transcription
priming effects suggested to link inflammatory conditions to late- factor (Zhang et al., 2006) or protein kinase B(Akt)/NF-kB activation
onset AD (Green et al., 2003; Santos et al., 2016). (Yamazaki et al., 2009). Direct evidence for a proinflammatory ac-
As a corollary to the inflammatory hypothesis, therapies tar- tion of ATF6 in brain pathology, however, is still lacking.
geting inflammation hold potential in AD prevention or treatment. In parallel to the three UPR branches, recent work has revealed
For example, observational studies have suggested beneficial ef- another potential feed-forward mechanism linking inflammation
fects of non-steroidal anti-inflammatory drugs (NSAIDs) on AD and stress signaling through eIF2a phosphorylation in AD. In cell
incidence, although randomized controlled trials have not culture and in AD mouse models, we demonstrated that microglia-
confirmed this effect (Wang et al., 2015). This apparent discrepancy derived TNF-a drives cognitive and non-cognitive aspects of AD
may be due to the fact that NSAIDs, as most current approaches, (Ledo et al., 2016, 2013). Moreover, we showed that, downstream of
lack the specificity to preserve any desirable type of inflammatory TNFR1 signaling, activation of PKR and eIF2a phosphorylation in
response. Some extent of microglial activation appears, in fact, to be neurons is required for AD-related memory deficits (Lourenco et al.,
beneficial and required for encapsulating amyloid plaques 2013). Given that eIF2a phosphorylation may induce NF-kB acti-
(Condello et al., 2015) and for proper amyloid clearance (Minter vation (Deng et al., 2004), leading to further TNF-a expression
et al., 2016), rendering treatment duration critical, and therapeu- (Chung et al., 2009), chronically activated UPR or ISR in AD could
tic windows narrow. Nonetheless, based on currently available form a self-activating loop. Of relevance to this proposed mecha-
data, additional studies are warranted to determine whether the nism, it was recently shown that systemic inflammation induces
inflammatory component of AD is a valuable therapeutic target. TNF-a expression in a PKR-dependent manner in the mouse brain
(Carret-Rebillat et al., 2015).
5. ER stress intersects with neuroinflammation and AD A second pathway that may reciprocally link ER stress and
inflammation in AD is oxidative signaling. The ER lumen is a strong
When active, each of the UPR branches has been shown to in- oxidative environment, where the ratio of GSH to GSSH is one to
crease production and release of pro-inflammatory mediators. The two orders of magnitude lower than in the rest of the cell
three pathways converge on activating nuclear factor-kB (NF-kB; (Chakravarthi et al., 2006; Hwang et al., 1992). This environment is
Salminen et al., 2009; Zhang and Kaufman, 2008), a transcription required for proper oxidative folding of proteins, and reductive
factor that drives expression of cytokines such as TNFa, IL-1, IL-6 signals have long been known to induce ER stress by impairing
and IL-8 (Bo€ hrer et al., 1997; Brand et al., 1996). These, in turn, may protein folding (DuRose et al., 2006). More recently, however,
further activate the NF-kB pathway, in a chronic feed-forward loop oxidative insults, which may result from prolonged UPR activation,
that has been implicated in AD (Chung et al., 2009). have also been shown to elicit ER stress (Eletto et al., 2014). A key
At the cytoplasmic side, activated IRE-1 was shown to bind the player in this oxidative regulatory mechanism is nitric oxide (NO), a
transducer protein TRAF-2 (TNF receptor-associated factor 2; Urano proinflammatory mediator deeply involved in AD pathogenesis
et al., 2000) and recruit the IKK (IkB kinase) complex. This signaling (Uehara et al., 2006), as discussed below.
hub integrates several pathways and activates NF-kB by phos-
phorylating IkB, which normally keeps NF-kB inactive and

Please cite this article in press as: Santos, L.E., Ferreira, S.T., Crosstalk between endoplasmic reticulum stress and brain inflammation in
Alzheimer's disease, Neuropharmacology (2017), https://doi.org/10.1016/j.neuropharm.2017.11.016
L.E. Santos, S.T. Ferreira / Neuropharmacology xxx (2017) 1e11 5

Fig. 1. e Common pathways link ER stress and the UPR to neuroinflammation in Alzheimer's disease and other disorders. UPR pathways converge on increased production of
inflammatory cytokines through the NF-kB pathway. These mechanisms have been observed in AD and in several conditions, including metabolic disorders (type 2 diabetes,
obesity), depression, traumatic brain injury (TBI), infections and, notably, aging. These conditions are considered risk factors for AD and share ER stress and neuroinflammation as
drivers of symptoms. Inflammatory mediators such as TNF-a may result from prolonged UPR activation, and further activate UPR pathways and the cellular integrated stress
response, e.g., via activation of PKR. Additionally, S-nitrosylation, a protein modification prominently found in AD and metabolic disorders, modulates ER stress sensors such as PERK
and IRE-1, leading to a dysfunctional UPR. These mechanisms amount to an ER stress-inflammation loop, that may underlie the connection between AD and its comorbidities.

6. Nitric oxide regulation of the UPR Several pathways that result in iNOS expression involve NF-kB
activation, as it is one of the transcription factors driving the iNOS
NO is a highly diffusible signaling molecule, produced in vivo by gene in mice and humans (Aktan, 2004). Although it is reasonable
NO synthase (NOS), an enzyme found throughout the brain in both to assume that prolonged ER stress will at some point lead to iNOS
constitutively expressed and inducible isoforms. While neuronal expression through NF-kB and/or other pro-inflammatory path-
NOS (nNOS) is typically found in select neurons and has numerous ways, few reports have examined this directly, and the diversity of
physiological roles, inducible NOS (iNOS) is expressed by activated experimental models and approaches employed do not yet warrant
astrocytes and microglia, downstream of proinflammatory signals definite conclusions in this regard (Guo et al., 2010; Hosoi et al.,
(Pacher et al., 2007). 2013). On the other hand, NO-induced ER stress and activation of
Due to its chemical nature, NO defies typical ligand-receptor UPR pathways is well-established, having been described in
paradigms. While soluble guanylyl cyclase may be deserving of numerous cell types (Oyadomari et al., 2001; Gotoh et al., 2002;
the ‘NO-receptor’ moniker, many of the effects of NO, both physi- Nakato et al., 2015). Events downstream of activated IRE-1 (Kim
ological and pathological, are mediated by protein modification. In et al., 2010; Meares et al., 2011), ATF6 (Xu et al., 2004), PERK, and
pathology, reactions between NO or NO-derived molecules and PKR (Tong et al., 2011) have been observed in response to NO do-
biological targets are part of what is termed nitrative stress (Foster nors, while NOS inhibitors were proposed to reduce ischemic
et al., 2009; Jaffrey et al., 2001; Pacher et al., 2007). damage by mitigating the UPR-CHOP pathway (Iadecola et al., 1997;
In AD, as in virtually any CNS pathology, levels of NO-related Kim et al., 2008; Kohno et al., 1997). Nitration of tyrosines on sarco/
protein modifications are found to be increased, particularly at in- endoplasmic reticulum Caþþ-ATPase (SERCA), a calcium pump
flammatory loci. Post mortem assessment of AD brains showed that present in the ER membrane and involved in regulation of luminal
dysmorphic neurons, those found near amyloid deposits or con- calcium stores (Dickhout et al., 2005; Mekahli et al., 2011), is one of
taining Tau tangles, have increased immunoreactivity for nitrated the mechanisms mediating this link.
tyrosine (3-NT; Good et al., 1996; Butterfield et al., 2001). Being a Most notably, recent work showed that cysteine S-nitrosylation,
largely irreversible protein modification, 3-NT is useful as a marker another NO-related modification involved in AD pathology
of nitrative stress, and is a common consequence of iNOS expres- (Nakamura et al., 2013), is a powerful modulator of ER stress.
sion during inflammation. Glial iNOS was further shown to be Nakato et al. (2015) showed that the ribonuclease activity of IRE1 is
increased around neurofibrillary tangles and amyloid lesions in AD inhibited by S-nitrosylation, while PERK activity is increased by the
brains, as well as in Ab-stimulated glial cultures, in the latter case in same mechanism. Further, site-directed mutagenesis of a target
an NF-kB dependent manner (Akama and Van Eldik, 2000; De La cysteine residue on IRE1 rendered cells resistant to NO-induced
Monte et al., 2000; Pacher et al., 2007). Notably, 3-NT is also death (Nakato et al., 2015). This work built upon a seminal report
found increased in the CSF of AD patients, and its buildup is an early by the same group, which showed that protein disulphide isom-
event, detected in those diagnosed with mild cognitive impair- erase (PDI), a major ER chaperone, may also be functionally down-
ment, a preliminary condition that often progresses to AD regulated by S-nitrosylation, with negative implications to ER stress
(Butterfield et al., 2007; Pacher et al., 2007; Reed et al., 2009; and brain disease (Uehara et al., 2006).
Reynolds et al., 2006; Tohgi et al., 1999). Finally, although S-nitrosylation of UPR sensors has not yet been

Please cite this article in press as: Santos, L.E., Ferreira, S.T., Crosstalk between endoplasmic reticulum stress and brain inflammation in
Alzheimer's disease, Neuropharmacology (2017), https://doi.org/10.1016/j.neuropharm.2017.11.016
6 L.E. Santos, S.T. Ferreira / Neuropharmacology xxx (2017) 1e11

directly observed in AD patients or models, it was recently systemic inflammatory disease (Tobinick and Gross, 2008). Eta-
described in mouse models of obesity (Yang et al., 2015). That study nercept has further been tested in recent clinical trials in AD
reported that iNOS-mediated nitrosylation of IRE-1 was involved in (NCT01068353, NCT01716637, NCT00203359, NCT00203320;
several aspects of the obese phenotype, such as glucose intolerance, ClinicalTrials.gov). Although a recent phase 2 trial of Etanercept
and used a nitrosylation-resistant IRE-1 variant to provide evidence showed promising trends in terms of mood, cognition, and daily
for a causal link (Yang et al., 2015). These results underscore a functioning, no statistically significant results were found in that
possible role of ER stress, and particularly its regulation by NO, in study, possibly due to the small sample size (41 patients divided
the link between AD and inflammation-driven metabolic disorders, between treated and control groups) (Butchart et al., 2015). The
such as type 2 diabetes and obesity. drug, however, was confirmed to be well-tolerated, and larger trials
are warranted.
7. Potential therapeutic approaches targeting ER stress/ Another cytokine antibody currently in clinical use for periph-
inflammation in AD eral inflammatory diseases is Canakinumab, which targets IL-1b.
Given the evidence for IL-1b involvement in AD-related inflam-
Microglia are central to the inflammatory hypothesis of AD. As mation, including increased expression in patient brains, correla-
such, they have long been targets of potential therapeutic ap- tion to amyloid deposition and polymorphisms linked to higher
proaches, which sought to curb overall microglial responses, or incidence of AD, Canakinumab may be a promising approach for
shift them towards a more benign inflammatory profile (Choi et al., treating AD (Hallegua and Weisman, 2002). Not unlike TNF-a, IL-1b
2007; Fan et al., 2017; Kobayashi et al., 2013; Ledo et al., 2016; expression is regulated by NF-kB, and it may be induced by ER
Mandrekar and Landreth, 2010; Spangenberg et al., 2016). How- stress (Kim et al., 2014). Non-antibody alternatives, such as the drug
ever, as noted above, these approaches have had only limited suc- Anakinra, an IL-1 receptor antagonist, also exist for neutralizing IL-
cess, in part due to the complexity of inflammatory responses and b signaling. Albeit at relatively low concentrations, Anakinra has
the challenges in pharmacologically isolating their maladaptive been reported to reach the CNS following intravenous adminis-
aspects (Jiang et al., 2015; Wang et al., 2015). Additionally, it is tration (Galea et al., 2011; Goldbach-Mansky, 2011), while brain
increasingly clear that ER stress and inflammation coexist in the AD bioavailability remains a challenge for large, immunoglobulin-
brain, and are intertwined. The involvement of secreted cytokines based molecules such as Infliximab or Canakinumab (Hallegua
and, particularly, of NO, which readily crosses biological mem- and Weisman, 2002; Tweedie et al., 2007). For those larger mole-
branes, means that ER stress and inflammation may interact with cules, beneficial effects in brain disease following peripheral
each other even across individual cells and different cellular types. administration could stem from lower levels of circulating cyto-
ER stress and modulation of UPR sensors may also play a direct kines, leading to a “sink effect” that would reduce CNS concentra-
role in microglial priming. Although the concept of ‘ER stress tions (Dantzer and Kelley, 2007; Ferreira et al., 2014). Notably,
priming’ (i.e., lasting changes in ER stress proteins following an neither Anakira nor Canakinumab have yet been clinically tested in
insult) is itself virtually unexplored, Kim et al. (2014) showed that AD.
chemical induction of ER stress is sufficient to prime myeloid As noted above, a recent meta-analysis of the literature on over a
lineage cells to release IL-1b upon a subsequent stimulus. Other dozen different drugs showed that the use of NSAIDs in AD pre-
reports further suggest that ER stress may function as a key inter- vention is currently supported by observational studies, but this
mediate in toll-like receptor-mediated inflammatory priming support could not be confirmed by randomized trials (Wang et al.,
(Lebeaupin et al., 2015; Rao et al., 2014). Notably, the recent 2015). Such results highlight our incomplete understanding of the
observation that systemic LPS administration requires PKR mechanism of action of these drugs in AD. For instance, the effects
expression to increase hippocampal levels of TNF-a, IL-6 and Iba-1 of NSAIDs in ER stress are particularly complex. While several au-
(a microglial marker) in mice strongly supports a role for the in- thors have shown that NSAIDs induce apoptosis through UPR
tegrated stress response system in microglial priming (Carret- activation, others have described protective mechanisms, including
Rebillat et al., 2015). Given the increasingly recognized relevance CHOP inhibition, with outcomes appearing to depend greatly on
of microglial priming events in long-term epidemiological con- the drugs and model organism used (Hosoi et al., 2009; Kitamura,
nections between AD and other co-morbidities, including mood 2008; Tsutsumi et al., 2006, 2004; Yamazaki et al., 2006). Perhaps
alterations (Santos et al., 2016), involvement of ER stress in this a deeper understanding of the role of NSAIDs in ER stress will allow
process may represent new therapeutic targets. Thus, anti- for a better selection of candidate drugs. Not all NSAIDs affect ER
inflammatory approaches known to effectively modulate the UPR stress in the same manner (Kitamura, 2008), and this may deci-
may hold promise in the so far disappointing landscape of AD sively influence future choices of drugs and treatment regimens.
clinical trials. Minocycline, a tetracycline antibiotic with interesting anti-
Recent work from our group points to TNF-a as a prominent inflammatory properties, has been suggested as a promising
target candidate in this regard. The human monoclonal antibody approach to modulate microglia in AD. This is largely due to the
Infliximab, which neutralizes TNF-a and has been in use for auto- selectivity it shows towards neurotoxic inflammatory profiles of
immune and inflammatory diseases since the late 1990s, was suc- microglia, while preserving more benign, phagocytic phenotypes,
cessful in preventing eIF2a phosphorylation and memory loss in a which are essential for amyloid clearance (Kobayashi et al., 2013;
mouse model of AD (Lourenco et al., 2013). Further, Infliximab Santos et al., 2016). In mouse models of AD, minocycline was
prevented depressive-like behavior, a common non-cognitive shown to block AbO-mediated TNF-a release from microglia,
symptom of AD that is recapitulated in AbO-infused mice (Ledo positively impacting behavior (Ledo et al., 2016) and memory
et al., 2016), suggesting a broad range of effects. These findings (Biscaro et al., 2012; Garcez et al., 2017). Most notably, Choi et al.
are supported by pre-clinical studies showing that interference (2007) showed that, while preventing neuronal death and mem-
with TNF-a signaling using thalidomide attenuates cognitive defi- ory impairment in transgenic and AbO-infused mouse models of
cits in rodent models of AD (Alkam et al., 2008; Gabbita et al., 2012; AD, minocycline attenuated the upregulation of p-eIF2a observed
He et al., 2013; Tweedie et al., 2012). The concept of neutralizing in those mice. Whether direct or indirect, such an effect of mino-
TNF-a in AD patients is not new, and was pointed out in early case cycline on the UPR/ISR lends additional support to its use as a po-
reports employing Infliximab (Shi et al., 2011) and Etanercept, a tential anti-AD drug. The MADE (Minocycline in AD Efficacy,
decoy TNFR-Fc fusion protein also in clinical use for chronic ISRCTN16105064) trial is a phase 2, randomized, placebo controlled

Please cite this article in press as: Santos, L.E., Ferreira, S.T., Crosstalk between endoplasmic reticulum stress and brain inflammation in
Alzheimer's disease, Neuropharmacology (2017), https://doi.org/10.1016/j.neuropharm.2017.11.016
L.E. Santos, S.T. Ferreira / Neuropharmacology xxx (2017) 1e11 7

trial, currently ongoing with around 500 recruited patients, which https://doi.org/10.1074/jbc.M704491200.
Barbero-Camps, E., Ferna !ndez, A., Baulies, A., Martinez, L., Fern! andez-Checa, J.C.,
may soon shed light on the potential of minocycline in AD.
Colell, A., 2014. Endoplasmic reticulum stress mediates amyloid b neurotoxicity
Interestingly, treatment with serotonin (5-HT) reduced TNF-a via mitochondrial cholesterol trafficking. Am. J. Pathol. 184, 2066e2081. https://
release in cultured primary microglia exposed to AbOs and in the doi.org/10.1016/j.ajpath.2014.03.014.
brains of AbO-infused mice, and prevented behavioral deficits Bertolotti, A., Zhang, Y., Hendershot, L.M., Harding, H.P., Ron, D., 2000. Dynamic
interaction of BiP and ER stress transducers in the unfolded-protein response.
associated with AD in mice (Ledo et al., 2016). Indeed, 5-HT is Nat. Cell Biol. 2, 326e332. https://doi.org/10.1038/35014014.
known to bind microglial receptors and to have anti-inflammatory Biscaro, B., Lindvall, O., Tesco, G., Ekdahl, C.T., Nitsch, R.M., 2012. Inhibition of
properties, which have been proposed as part of the mechanism of microglial activation protects hippocampal neurogenesis and improves cogni-
tive deficits in a transgenic mouse model for Alzheimer's disease. Neuro-
action of serotonergic antidepressants (Walker, 2013). Among these degener. Dis 9, 187e198. https://doi.org/10.1159/000330363.
drugs are the widely prescribed selective 5-HT reuptake inhibitors Bo
€hrer, H., Qiu, F., Zimmermann, T., Zhang, Y., Jllmer, T., Ma €nnel, D., Bo€ttiger, B.W.,
(SSRIs), which, in the past decade, have been proposed as modu- Stern, D.M., Waldherr, R., Saeger, H.D., Ziegler, R., Bierhaus, a, Martin, E.,
Nawroth, P.P., 1997. Role of NFkappaB in the mortality of sepsis. J. Clin. Invest.
lators of a number of AD-related mechanisms, from amyloid pro- 100, 972e985. https://doi.org/10.1172/JCI119648.
cessing to its inflammatory response (e.g., Cirrito et al., 2011; Ledo Bown, C., Wang, J.F., MacQueen, G., Young, L.T., 2000. Increased temporal cortex ER
et al., 2013). Intriguingly, SSRIs were suggested to be promoters of stress proteins in depressed subjects who died by suicide. Neuro-
psychopharmacology 22, 327e332. https://doi.org/10.1016/S0893-133X(99)
CHOP-mediated apoptosis (Glo € ckel et al., 2008); however, as shown
00091-3.
by Zschocke et al. (2011) and, more recently, by Omi et al. (2014), Brand, K., Page, S., Rogler, G., Bartsch, A., Brandl, R., Knuechel, R., Page, M.,
these results may differ amongst neural cell populations, or may Kaltschmidt, C., Baeuerle, P.A., Neumeier, D., 1996. Activated transcription factor
nuclear factor-kappa B is present in the atherosclerotic lesion. J. Clin. Invest. 97,
not apply to all SSRIs. Similarly to NSAIDs, pre-selecting SSRIs on
1715e1722. https://doi.org/10.1172/JCI118598.
the basis of their modulation of the stress response may help Breunig, J.J., Guillot-Sestier, M.V., Town, T., 2013. Brain injury, neuroinflammation
improve their outcomes as prospective AD therapies. and Alzheimer's disease. Front. Aging Neurosci. https://doi.org/10.3389/
fnagi.2013.00026.
Butchart, J., Brook, L., Hopkins, V., Teeling, J., Püntener, U., Culliford, D., Sharples, R.,
8. Conclusion Sharif, S., McFarlane, B., Raybould, R., Thomas, R., Passmore, P., Perry, V.H.,
Holmes, C., 2015. Etanercept in Alzheimer disease: a randomized, placebo-
Although the UPR is a vital response mechanism that evolved to controlled, double-blind, phase 2 trial. Neurology 84, 2161e2168. https://
doi.org/10.1212/WNL.0000000000001617.
protect cells from stressors, its prolonged activation is known to Butterfield, D.A., Reed, T.T., Perluigi, M., De Marco, C., Coccia, R., Keller, J.N.,
underlie pathology. Since the first description of an ER stress sensor Markesbery, W.R., Sultana, R., 2007. Elevated levels of 3-nitrotyrosine in brain
25 years ago (Cox et al., 1993; Morl et al., 1993), several lines of from subjects with amnestic mild cognitive impairment: implications for the
role of nitration in the progression of Alzheimer's disease. Brain Res. 1148,
evidence have linked aberrant UPR to neurodegeneration and, in 243e248. https://doi.org/10.1016/j.brainres.2007.02.084.
particular, to AD (e.g., Ma et al., 2013; Lourenco et al., 2013; Duran- Butterfield, D. a, Drake, J., Pocernich, C., Castegna, a, 2001. Evidence of oxidative
Aniotz et al., 2017; Hetz and Mollereau, 2014; Hoozemans et al., damage in Alzheimer's disease brain: central role for amyloid beta-peptide.
Trends Mol. Med. 7, 548e554. https://doi.org/10.1016/s1471-4914(01)02173-6.
2005). Given the extensive links between inflammation and ER Carret-Rebillat, A.-S., Pace, C., Gourmaud, S., Ravasi, L., Montagne-Stora, S.,
stress described above, it may no longer be possible to consider any Longueville, S., Tible, M., Sudol, E., Chang, R.C.-C., Paquet, C., Mouton-Liger, F.,
inflammatory hypothesis of disease without taking ER stress into Hugon, J., 2015. Neuroinflammation and Ab accumulation linked to systemic
inflammation are decreased by genetic PKR down-regulation. Sci. Rep. 5, 8489.
account, and vice-versa. From early microglial priming to late
https://doi.org/10.1038/srep08489.
neuronal death, UPR responses are part of the mechanisms by Chakravarthi, S., Jessop, C.E., Bulleid, N.J., 2006. The role of glutathione in disulphide
which inflammation influences AD incidence and progression. bond formation and endoplasmic-reticulum-generated oxidative stress. EMBO
Rep. 7, 271e275. https://doi.org/10.1038/sj.embor.7400645.
Novel pharmacological approaches targeting the inflammatory
Chaudhari, N., Talwar, P., Parimisetty, A., Lefebvre d'Hellencourt, C., Ravanan, P.,
component of AD may be more likely to succeed if they can prove 2014. A molecular web: endoplasmic reticulum stress, inflammation, and
effective in controlling a dysfunctional UPR, thus breaking the ER oxidative stress. Front. Cell. Neurosci. 8, 213. https://doi.org/10.3389/
stress-inflammation loop. fncel.2014.00213.
Cherry, J.D., Olschowka, J.A., O'Banion, M.K., 2014. Neuroinflammation and M2
microglia: the good, the bad, and the inflamed. J. Neuroinflammation 11, 98.
Acknowledgements https://doi.org/10.1186/1742-2094-11-98.
Chiti, F., Dobson, C.M., 2017. Protein misfolding, amyloid formation, and human
disease: a summary of progress over the last decade. Annu. Rev. Biochem. 86,
Work in the authors' laboratory is supported by grants from the 27e68. https://doi.org/10.1146/annurev-biochem-061516-045115.
National Institute for Translational Neuroscience/Brazil, Coor- Choi, Y., Kim, H.-S., Shin, K.Y., Kim, E.-M., Kim, M., Kim, H.-S., Park, C.H., Jeong, Y.H.,
denaça~o de Aperfeiçoamento de Pessoal de Nível Superior, Con- Yoo, J., Lee, J.-P., Chang, K.-A., Kim, S., Suh, Y.-H., 2007. Minocycline attenuates
neuronal cell death and improves cognitive impairment in Alzheimer's disease
selho Nacional de Desenvolvimento Científico e Tecnolo ! gico and
models. Neuropsychopharmacology 32, 2393e2404. https://doi.org/10.1038/
Fundaça~o Carlos Chagas Filho de Amparo a # Pesquisa do Estado do sj.npp.1301377.
Rio de Janeiro. L.E.S. is a postdoctoral fellow supported by CAPES. Chung, H.Y., Cesari, M., Anton, S., Marzetti, E., Giovannini, S., Seo, A.Y., Carter, C.,
Yu, B.P., Leeuwenburgh, C., 2009. Molecular inflammation: underpinnings of
aging and age-related diseases. Ageing Res. Rev. 8, 18e30. https://doi.org/
References 10.1016/j.arr.2008.07.002.
Cirrito, J.R., Disabato, B.M., Restivo, J.L., Verges, D.K., Goebel, W.D., Sathyan, A.,
Akama, K.T., Van Eldik, L.J., 2000. Beta-amyloid stimulation of inducible nitric-oxide Hayreh, D., D'Angelo, G., Benzinger, T., Yoon, H., Kim, J., Morris, J.C.,
synthase in astrocytes is interleukin-1beta- and tumor necrosis factor-alpha Mintun, M.A., Sheline, Y.I., 2011. Serotonin signaling is associated with lower
(TNFalpha)-dependent, and involves a TNFalpha receptor-associated factor- amyloid-{beta} levels and plaques in transgenic mice and humans. Proc. Natl.
and NFkappaB-inducing kinase-dependent signaling mech. J. Biol. Chem. 275, Acad. Sci. U. S. A. 108, 14968e14973. https://doi.org/10.1073/pnas.1107411108.
7918e7924. Cisse
!, M., Duplan, E., Checler, F., 2016. The Transcription Factor XBP1 in Memory and
Aktan, F., 2004. iNOS-mediated nitric oxide production and its regulation. Life Sci. Cognition: Implications in Alzheimer’s Disease. Mol. Med 22, 1. https://doi.org/
https://doi.org/10.1016/j.lfs.2003.10.042. 10.2119/molmed.2016.00229.
Alkam, T., Nitta, A., Mizoguchi, H., Saito, K., Seshima, M., Itoh, A., Yamada, K., Cisse
!, M., Duplan, E., Lorivel, T., Dunys, J., Bauer, C., Meckler, X., Gerakis, Y.,
Nabeshima, T., 2008. Restraining tumor necrosis factor-alpha by thalidomide Lauritzen, I., Checler, F., 2016a. The transcription factor XBP1s restores hippo-
prevents the Amyloid beta-induced impairment of recognition memory in campal synaptic plasticity and memory by control of the Kalirin-7 pathway in
mice. Behav. Brain Res. 189, 100e106. https://doi.org/10.1016/j.bbr.2007.12.014. Alzheimer model. Mol. Psychiatry 1e14. https://doi.org/10.1038/mp.2016.152.
Baleriola, J., Walker, C.A., Jean, Y.Y., Crary, J.F., Troy, C.M., Nagy, P.L., Hengst, U., 2014. Condello, C., Yuan, P., Schain, A., Grutzendler, J., 2015. Microglia constitute a barrier
Axonally synthesized ATF4 transmits a neurodegenerative signal across brain that prevents neurotoxic protofibrillar Ab42 hotspots around plaques. Nat.
regions. Cell 158, 1159e1172. https://doi.org/10.1016/j.cell.2014.07.001. Commun. 6, 6176. https://doi.org/10.1038/ncomms7176.
Baltzis, D., Pluquet, O., Papadakis, A.I., Kazemi, S., Qu, L.-K., Koromilas, A.E., 2007. Costa-Mattioli, M., Gobert, D., Harding, H., Herdy, B., Azzi, M., Bruno, M.,
The eIF2a kinases PERK and PKR activate glycogen synthase kinase 3 to pro- Bidinosti, M., Ben Mamou, C., Marcinkiewicz, E., Yoshida, M., Imataka, H.,
mote the proteasomal degradation of p53. J. Biol. Chem. 282, 31675e31687. Cuello, A.C., Seidah, N., Sossin, W., Lacaille, J.-C., Ron, D., Nader, K.,

Please cite this article in press as: Santos, L.E., Ferreira, S.T., Crosstalk between endoplasmic reticulum stress and brain inflammation in
Alzheimer's disease, Neuropharmacology (2017), https://doi.org/10.1016/j.neuropharm.2017.11.016
8 L.E. Santos, S.T. Ferreira / Neuropharmacology xxx (2017) 1e11

Sonenberg, N., 2005. Translational control of hippocampal synaptic plasticity the central nervous system within a therapeutic time window: results of a
and memory by the eIF2alpha kinase GCN2. Nature 436, 1166e1173. https:// dose-ranging study. J. Cereb. Blood Flow. Metab. 31, 439e447. https://doi.org/
doi.org/10.1038/nature03897. 10.1038/jcbfm.2010.103.
Costa-Mattioli, M., Gobert, D., Stern, E., Gamache, K., Colina, R., Cuello, C., Sossin, W., Garcez, M.L., Mina, F., Bellettini-Santos, T., Carneiro, F.G., Luz, A.P., Schiavo, G.L.,
Kaufman, R., Pelletier, J., Rosenblum, K., Krnjevic, K., Lacaille, J.C., Nader, K., Andrighetti, M.S., Scheid, M.G., Bolfe, R.P., Budni, J., 2017. Minocycline reduces
Sonenberg, N., 2007. eIF2 alpha phosphorylation bidirectionally regulates the inflammatory parameters in the brain structures and serum and reverses
switch from short- to long-term synaptic plasticity and memory. Cell 129, memory impairment caused by the administration of amyloid b (1-42) in mice.
195e206. Prog. Neuro-Psychopharmacology Biol. Psychiatry 77, 23e31. https://doi.org/
Costa-Mattioli, M., Sossin, W., Klann, E., Sonenberg, N., 2009. Translational control 10.1016/j.pnpbp.2017.03.010.
of long-lasting synaptic plasticity and memory. Neuron 61, 10e26. https:// Gardner, B.M., Walter, P., 2011. Unfolded proteins are ire1-activating ligands that
doi.org/10.1016/j.neuron.2008.10.055. directly induce the unfolded protein response. Science (80-. ) 333, 1891e1894.
Cox, J.S., Shamu, C.E., Walter, P., 1993. Transcriptional induction of genes encoding https://doi.org/10.1126/science.1209126.
endoplasmic reticulum resident proteins requires a transmembrane protein Glass, C.K., Saijo, K., Winner, B., Marchetto, M.C., Gage, F.H., 2010. Mechanisms
kinase. Cell 73, 1197e1206. https://doi.org/10.1016/0092-8674(93)90648-A. underlying inflammation in neurodegeneration. Cell 140, 918e934. https://
Dantzer, R., Kelley, K.W., 2007. Twenty years of research on cytokine-induced doi.org/10.1016/j.cell.2010.02.016.
sickness behavior. Brain. Behav. Immun. 21, 153e160. https://doi.org/10.1016/ Glenner, G.G., Wong, C.W., 1984. Alzheimer's disease: initial report of the purifi-
j.bbi.2006.09.006. cation and characterization of a novel cerebrovascular amyloid protein. Bio-
De Felice, F.G., 2013. Alzheimer's disease and insulin resistance: translating basic chem. Biophys. Res. Commun. 120, 885e890. https://doi.org/10.1016/S0006-
science into clinical applications. J. Clin. Invest. 123, 531e539. https://doi.org/ 291X(84)80190-4.
10.1172/JCI64595. Glo
€ckel, C., Thurner, P., Freissmuth, M., 2008. Selective serotonin reuptake inhibitors
De Felice, F.G., Ferreira, S.T., 2014. Inflammation, defective insulin signaling, and induce cell death via the unfolded protein response. BMC Pharmacol. 8, A45.
mitochondrial dysfunction as common molecular denominators connecting https://doi.org/10.1186/1471-2210-8-S1-A45.
type 2 diabetes to alzheimer disease. Diabetes 63, 2262e2272. https://doi.org/ Gold, P.W., Licinio, J., Pavlatou, M.G., 2013. Pathological parainflammation and
10.2337/db13-1954. endoplasmic reticulum stress in depression: potential translational targets
De La Monte, S.M., Lu, B.X., Sohn, Y.K., Etienne, D., Kraft, J., Ganju, N., Wands, J.R., through the CNS insulin, klotho and PPAR-g systems. Mol. Psychiatry 18,
2000. Aberrant expression of nitric oxide synthase III in Alzheimer's disease: 154e165. https://doi.org/10.1038/mp.2012.167.
relevance to cerebral vasculopathy and neurodegeneration. Neurobiol. Aging Goldbach-Mansky, R., 2011. Current status of understanding the pathogenesis and
309e319. https://doi.org/10.1016/S0197-4580(99)00108-6. management of patients with NOMID/CINCA. Curr. Rheumatol. Rep. 13,
Deng, J., Lu, P.D., Zhang, Y., Scheuner, D., Kaufman, R.J., Sonenberg, N., Harding, H.P., 123e131. https://doi.org/10.1007/s11926-011-0165-y.
Ron, D., 2004. Translational repression mediates activation of nuclear factor Good, P.F., Werner, P., Hsu, A., Olanow, C.W., Perl, D.P., 1996. Evidence of neuronal
kappa B by phosphorylated translation initiation factor 2. Mol. Cell. Biol. 24, oxidative damage in Alzheimer's disease. Am. J. Pathol. 149, 21e28.
10161e10168. https://doi.org/10.1128/MCB.24.23.10161-10168.2004. Gotoh, T., Oyadomari, S., Mori, K., Mori, M., 2002. Nitric oxide-induced apoptosis in
Dickhout, J.G., Hossain, G.S., Pozza, L.M., Zhou, J., Lhot! $ Austin, R.C., 2005.
ak, S., RAW 264.7 Macrophages is mediated by endoplasmic reticulum stress pathway
Peroxynitrite causes endoplasmic reticulum stress and apoptosis in human involving ATF6 and CHOP. J. Biol. Chem. 277, 12343e12350. https://doi.org/
vascular endothelium: implications in atherogenesis. Arterioscler. Thromb. 10.1074/jbc.M107988200.
Vasc. Biol. 25, 2623e2629. https://doi.org/10.1161/ Green, R.C., Cupples, L.A., Kurz, A., Auerbach, S., Go, R., Sadovnick, D., Duara, R.,
01.ATV.0000189159.96900.d9. Kukull, W.A., Chui, H., Edeki, T., Griffith, P.A., Friedland, R.P., Bachman, D.,
Dogan, A., 2017. Amyloidosis: insights from proteomics. Annu. Rev. Pathol. Mech. Farrer, L., 2003. Depression as a risk factor for Alzheimer disease: the MIRAGE
Dis. 12, 277e304. https://doi.org/10.1146/annurev-pathol-052016-100200. Study. Arch. Neurol. 60, 753e759. https://doi.org/10.1001/archneur.60.5.753.
Donnelly, N., Gorman, A.M., Gupta, S., Samali, A., 2013. The eIF2a kinases: their Griffin, W.S., Sheng, J.G., Roberts, G.W., Mrak, R.E., 1995. Interleukin-1 expression in
structures and functions. Cell. Mol. Life Sci. 70, 3493e3511. https://doi.org/ different plaque types in Alzheimer's disease. J. Neuropathol. Exp. Neurol. 54,
10.1007/s00018-012-1252-6. 276e281. https://doi.org/10.1097/00005072-199503000-00014.
Duran-Aniotz, C., Cornejo, V.H., Espinoza, S., Ardiles, Alvaro O., Medinas, D.B., Griffin, W.S., Stanley, L.C., Ling, C., White, L., MacLeod, V., Perrot, L.J., White, C.L.,
Salazar, C., Foley, A., Gajardo, I., Thielen, P., Iwawaki, T., Scheper, W., Soto, C., Araoz, C., 1989. Brain interleukin 1 and S-100 immunoreactivity are elevated in
Palacios, A.G., Hoozemans, J.J.M., Hetz, C., 2017. IRE1 signaling exacerbates Down syndrome and Alzheimer disease. Proc. Natl. Acad. Sci. U. S. A. 86,
Alzheimer's disease pathogenesis. Acta Neuropathol. 1e18. https://doi.org/ 7611e7615. https://doi.org/10.1073/pnas.86.19.7611.
10.1007/s00401-017-1694-x. Grundke-Iqbal, I., Iqbal, K., Quinlan, M., Tung, Y.C., Zaidi, M.S., Wisniewski, H.M.,
DuRose, J.B., Tam, A.B., Niwa, M., 2006. Intrinsic capacities of molecular sensors of 1986a. Microtubule-associated protein tau. A component of Alzheimer paired
the unfolded protein response to sense alternate forms of endoplasmic retic- helical filaments. J. Biol. 261, 6084e6089. https://doi.org/10.1074/
ulum stress. Mol. Biol. Cell 17, 3095e3107. https://doi.org/10.1091/mbc.E06-01- jbc.M111.271320.
0055. Grundke-Iqbal, I., Iqbal, K., Tung, Y.C., Quinlan, M., Wisniewski, H.M., Binder, L.I.,
Eletto, D., Chevet, E., Argon, Y., Appenzeller-Herzog, C., 2014. Redox controls UPR to 1986b. Abnormal phosphorylation of the microtubule-associated protein tau
control redox. J. Cell Sci. 127, 3649e3658. https://doi.org/10.1242/jcs.153643. (tau) in Alzheimer cytoskeletal pathology. Proc. Natl. Acad. Sci. U. S. A. 83,
Fan, Z., Brooks, D.J., Okello, A., Edison, P., 2017. An early and late peak in microglial 4913e4917. https://doi.org/10.1097/00002093-198701030-00020.
activation in Alzheimer's disease trajectory. Brain. https://doi.org/10.1093/ Guo, F.J., Lin, E.A., Liu, P., Lin, J., Liu, C., 2010. XBP1U inhibits the XBP1S-mediated
brain/aww349 aww349. upregulation of the iNOS gene expression in mammalian ER stress response.
Femminella, G.D., Ninan, S., Atkinson, R., Fan, Z., Brooks, D.J., Edison, P., 2016. Does Cell. Signal 22, 1818e1828. https://doi.org/10.1016/j.cellsig.2010.07.006.
microglial activation influence hippocampal volume and neuronal function in Gupta, S., Deepti, A., Deegan, S., Lisbona, F., Hetz, C., Samali, A., 2010. HSP72 protects
Alzheimer's disease and Parkinson's disease Dementia? J. Alzheimer’s Dis. 51, cells from ER stress-induced apoptosis via enhancement of IRE1a-XBP1
1275e1289. https://doi.org/10.3233/JAD-150827. signaling through a physical interaction. PLoS Biol. 8, e1000410 https://doi.org/
Ferreira, S.T., Clarke, J.R., Bomfim, T.R., De Felice, F.G., 2014. Inflammation, defective 10.1371/journal.pbio.1000410.
insulin signaling, and neuronal dysfunction in Alzheimer's disease. Alzheimer’s Haass, C., Selkoe, D.J., 2007. Soluble protein oligomers in neurodegeneration: les-
Dement. 10, S76eS83. https://doi.org/10.1016/j.jalz.2013.12.010. sons from the Alzheimer's amyloid beta-peptide. Nat. Rev. Mol. Cell Biol. 8,
Ferreira, S.T., Klein, W.L., 2011. The Ab oligomer hypothesis for synapse failure and 101e112. https://doi.org/10.1038/nrm2101.
memory loss in Alzheimer's disease. Neurobiol. Learn. Mem. 96, 529e543. Hallegua, D.S., Weisman, M.H., 2002. Potential therapeutic uses of interleukin 1
https://doi.org/10.1016/j.nlm.2011.08.003. receptor antagonists in human diseases. Ann. Rheum. Dis. 61, 960e967. https://
Ferreira, S.T., Lourenco, M.V., Oliveira, M.M., De Felice, F.G., 2015. Soluble amyloid-b doi.org/10.1136/ard.61.11.960.
oligomers as synaptotoxins leading to cognitive impairment in Alzheimer's Hamos, J.E., Oblas, B., Pulaski-Salo, D., Welch, W.J., Bole, D.G., Drachman, D.A., 1991.
disease. Front. Cell. Neurosci. 9, 1e17. https://doi.org/10.3389/fncel.2015.00191. Expression of heat shock proteins in Alzheimer's disease. Neurology 41,
Foster, M.W., Hess, D.T., Stamler, J.S., 2009. Protein S-nitrosylation in health and 345e350. https://doi.org/10.1212/WNL.41.3.345.
disease: a current perspective. Trends Mol. Med. 15, 391e404. https://doi.org/ Harding, H.P., Zhang, Y., Ron, D., 1999. Protein translation and folding are coupled by
10.1016/j.molmed.2009.06.007. an endoplasmic-reticulum-resident kinase. Nature 397, 271e274. https://
Füger, P., Hefendehl, J.K., Veeraraghavalu, K., Wendeln, A.-C., Schlosser, C., doi.org/10.1038/16729.
Obermüller, U., Wegenast-Braun, B.M., Neher, J.J., Martus, P., Kohsaka, S., Harding, H.P., Zhang, Y., Zeng, H., Novoa, I., Lu, P.D., Calfon, M., Sadri, N., Yun, C.,
Thunemann, M., Feil, R., Sisodia, S.S., Skodras, A., Jucker, M., 2017. Microglia Popko, B., Paules, R., Stojdl, D.F., Bell, J.C., Hettmann, T., Leiden, J.M., Ron, D.,
turnover with aging and in an Alzheimer's model via long-term in vivo single- 2003. An integrated stress response regulates amino acid metabolism and
cell imaging. Nat. Neurosci. https://doi.org/10.1038/nn.4631. resistance to oxidative stress. Mol. Cell 11, 619e633. https://doi.org/10.1016/
Gabbita, S.P., Srivastava, M.K., Eslami, P., Johnson, M.F., Kobritz, N.K., Tweedie, D., S1097-2765(03)00105-9.
Greig, N.H., Zemlan, F.P., Sharma, S.P., Harris-White, M.E., 2012. Early inter- Haze, K., Yoshida, H., Yanagi, H., Yura, T., Mori, K., 1999. Mammalian transcription
vention with a small molecule inhibitor for tumor nefosis factor-a prevents factor ATF6 is synthesized as a transmembrane protein and activated by pro-
cognitive deficits in a triple transgenic mouse model of Alzheimer's disease. teolysis in response to endoplasmic reticulum stress. Mol. Biol. Cell 10,
J. Neuroinflammation 9, 578. https://doi.org/10.1186/1742-2094-9-99. 3787e3799. https://doi.org/10.1091/mbc.10.11.3787.
Galea, J., Ogungbenro, K., Hulme, S., Greenhalgh, A., Aarons, L., Scarth, S., He, P., Cheng, X., Staufenbiel, M., Li, R., Shen, Y., 2013. Long-term treatment of
Hutchinson, P., Grainger, S., King, A., Hopkins, S.J., Rothwell, N., Tyrrell, P., 2011. thalidomide ameliorates amyloid-like pathology through inhibition of b-sec-
Intravenous Anakinra can achieve experimentally effective concentrations in retase in a mouse model of Alzheimer's disease. PLoS One 8, e55091. https://

Please cite this article in press as: Santos, L.E., Ferreira, S.T., Crosstalk between endoplasmic reticulum stress and brain inflammation in
Alzheimer's disease, Neuropharmacology (2017), https://doi.org/10.1016/j.neuropharm.2017.11.016
L.E. Santos, S.T. Ferreira / Neuropharmacology xxx (2017) 1e11 9

doi.org/10.1371/journal.pone.0055091. 710e723. https://doi.org/10.1074/mcp.M111.016550.


Heneka, M.T., Golenbock, D.T., Latz, E., 2015. Innate immunity in Alzheimer's dis- Janssens, S., Pulendran, B., Lambrecht, B.N., 2014. Emerging functions of the
ease. Nat. Immunol. 16, 229e236. https://doi.org/10.1038/ni.3102. unfolded protein response in immunity. Nat. Immunol. 15, 910e919. https://
Hennessy, E., Griffin, E.W., Cunningham, C., 2015. Astrocytes are primed by chronic doi.org/10.1038/ni.2991.
neurodegeneration to produce exaggerated chemokine and cell infiltration Jiang, T., Tan, L., Zhu, X.-C., Zhou, J.-S., Cao, L., Tan, M.-S., Wang, H.-F., Chen, Q.,
responses to acute stimulation with the cytokines IL-1 and TNF-. J. Neurosci. 35, Zhang, Y.-D., Yu, J.-T., 2015. Silencing of TREM2 exacerbates tau pathology,
8411e8422. https://doi.org/10.1523/JNEUROSCI.2745-14.2015. neurodegenerative changes, and spatial learning deficits in P301S tau trans-
Henry, C.J., Huang, Y., Wynne, A., Hanke, M., Himler, J., Bailey, M.T., Sheridan, J.F., genic mice. Neurobiol. Aging 36, 3176e3186. https://doi.org/10.1016/
Godbout, J.P., 2008. Minocycline attenuates lipopolysaccharide (LPS)-induced j.neurobiolaging.2015.08.019.
neuroinflammation, sickness behavior, and anhedonia. J. Neuroinflammation 5, Kaneko, M., Niinuma, Y., Nomura, Y., 2003. Activation signal of nuclear factor-kB in
15. https://doi.org/10.1186/1742-2094-5-15. response to endoplasmic reticulum stress is transduced via IRE1 and tumor
Hetz, C., 2012. The unfolded protein response: controlling cell fate decisions under necrosis factor receptor-associated factor 2. Biol. Pharm. Bull. 26, 931e935.
ER stress and beyond. Nat. Rev. Mol. Cell Biol. 13, 89e102. https://doi.org/ https://doi.org/10.1248/bpb.26.931.
10.1038/nrm3270. Kim, I., Xu, W., Reed, J.C., 2008. Cell death and endoplasmic reticulum stress: disease
Hetz, C., Glimcher, L.H., 2009. Fine-tuning of the unfolded protein response: relevance and therapeutic opportunities. Nat. Rev. Drug Discov. 7, 1013e1030.
assembling the IRE1alpha interactome. Mol. Cell 35, 551e561. https://doi.org/ https://doi.org/10.1038/nrd2755.
10.1016/j.molcel.2009.08.021. Kim, S., Joe, Y., Jeong, S.O., Zheng, M., Back, S.H., Park, S.W., Ryter, S.W., Chung, H.T.,
Hetz, C., Mollereau, B., 2014. Disturbance of endoplasmic reticulum proteostasis in 2014. Endoplasmic reticulum stress is sufficient for the induction of IL-1b
neurodegenerative diseases. Nat. Rev. Neurosci. 15, 233e249. https://doi.org/ production via activation of the NF-kB and inflammasome pathways. Innate
10.1038/nrn3689. Immun. 20, 799e815. https://doi.org/10.1177/1753425913508593.
Hinz, M., Scheidereit, C., 2014. The IkB kinase complex in NF-kB regulation and Kim, Y.H., Joo, H.S., Kim, D.-S., 2010. Nitric oxide induction of IRE1-alpha-dependent
beyond. EMBO Rep. 15, 46e61. https://doi.org/10.1002/embr.201337983. CREB phosphorylation in human glioma cells. Nitric Oxide 23, 112e120. https://
Holmes, C., Cunningham, C., Zotova, E., Woolford, J., Dean, C., Kerr, S., Culliford, D., doi.org/10.1016/j.niox.2010.04.009.
Perry, V.H., 2009. Systemic inflammation and disease progression in Alzheimer Kitamura, M., 2008. Endoplasmic reticulum stress and unfolded protein response in
disease. Neurology 73, 768e774. https://doi.org/10.1212/ renal pathophysiology: janus faces. AJP Ren. Physiol. 295, F323eF334. https://
WNL.0b013e3181b6bb95. doi.org/10.1152/ajprenal.00050.2008.
Hong, F., Liu, B., Wu, B.X., Morreall, J., Roth, B., Davies, C., Sun, S., Diehl, J.A., Li, Z., Kobayashi, K., Imagama, S., Ohgomori, T., Hirano, K., Uchimura, K., Sakamoto, K.,
2017. CNPY2 is a key initiator of the PERKeCHOP pathway of the unfolded Hirakawa, a, Takeuchi, H., Suzumura, a, Ishiguro, N., Kadomatsu, K., 2013.
protein response. Nat. Struct. Mol. Biol. https://doi.org/10.1038/nsmb.3458. Minocycline selectively inhibits M1 polarization of microglia. Cell Death Dis. 4,
Hong, S., Beja-Glasser, V.F., Nfonoyim, B.M., Frouin, A., Li, S., Ramakrishnan, S., e525 https://doi.org/10.1038/cddis.2013.54.
Merry, K.M., Shi, Q., Rosenthal, A., Barres, B.A., Lemere, C.A., Selkoe, D.J., Kohno, K., Higuchi, T., Ohta, S., Kohno, K., Kumon, Y., Sakaki, S., 1997. Neuro-
Stevens, B., 2016. Complement and microglia mediate early synapse loss in protective nitric oxide synthase inhibitor reduces intracellular calcium accu-
Alzheimer mouse models. Science (80-. ) 352, 712e716. https://doi.org/10.1126/ mulation following transient global ischemia in the gerbil. Neurosci. Lett. 224,
science.aad8373. 17e20.
Honjo, K., van Reekum, R., Verhoeff, N.P.L.G., 2009. Alzheimer's disease and infec- Kosik, K.S., Joachim, C.L., Selkoe, D.J., 1986. Microtubule-associated protein tau (tau)
tion: do infectious agents contribute to progression of Alzheimer's disease? is a major antigenic component of paired helical filaments in Alzheimer disease.
Alzheimer’s Dement. 5, 348e360. https://doi.org/10.1016/j.jalz.2008.12.001. Proc. Natl. Acad. Sci. U. S. A. 83, 4044e4048. https://doi.org/10.1097/00002093-
Honjo, Y., Ayaki, T., Tomiyama, T., Horibe, T., Ito, H., Mori, H., Takahashi, R., 198701030-00022.
Kawakami, K., 2015. Increased GADD34 in oligodendrocytes in Alzheimer's Kozutsumi, Y., Segal, M., Normington, K., Gething, M.-J., Sambrook, J., 1988. The
disease. Neurosci. Lett. 602, 50e55. https://doi.org/10.1016/ presence of malfolded proteins in the endoplasmic reticulum signals the in-
j.neulet.2015.06.052. duction of glucose-regulated proteins. Nature 332, 462e464. https://doi.org/
Hoozemans, J.J.M., van Haastert, E.S., Nijholt, D.A.T., Rozemuller, A.J.M., 10.1038/332462a0.
Eikelenboom, P., Scheper, W., 2009. The unfolded protein response is activated Lebeaupin, C., Proics, E., de Bieville, C.H.D., Rousseau, D., Bonnafous, S., Patouraux, S.,
in pretangle neurons in Alzheimer's disease Hippocampus. Am. J. Pathol. 174, Adam, G., Lavallard, V.J., Rovere, C., Le Thuc, O., Saint-Paul, M.C., Anty, R.,
1241e1251. https://doi.org/10.2353/ajpath.2009.080814. Schneck, A.S., Iannelli, A., Gugenheim, J., Tran, A., Gual, P., Bailly-Maitre, B., 2015.
Hoozemans, J.J.M., van Haastert, E.S., Nijholt, D.A.T., Rozemuller, A.J.M., Scheper, W., ER stress induces NLRP3 inflammasome activation and hepatocyte death. Cell
2012. Activation of the unfolded protein response is an early event in Alz- Death Dis. 6, e1879 https://doi.org/10.1038/cddis.2015.248.
heimer's and Parkinson's disease. Neurodegener. Dis. 10, 212e215. https:// Ledo, J.H., Azevedo, E.P., Beckman, D., Ribeiro, F.C., Santos, L.E., Razolli, D.S.,
doi.org/10.1159/000334536. Kincheski, G.C., Melo, H.M., Bellio, M., Teixeira, A.L., Velloso, L.A., Foguel, D., De
Hoozemans, J.J.M., Veerhuis, R., Van Haastert, E.S., Rozemuller, J.M., Baas, F., Felice, F.G., Ferreira, S.T., 2016. Cross talk between brain innate immunity and
Eikelenboom, P., Scheper, W., 2005. The unfolded protein response is activated serotonin signaling underlies depressive-like behavior induced by Alzheimer's
in Alzheimer's disease. Acta Neuropathol. 110, 165e172. https://doi.org/10.1007/ amyloid- oligomers in mice. J. Neurosci. 36, 12106e12116. https://doi.org/
s00401-005-1038-0. 10.1523/JNEUROSCI.1269-16.2016.
Hosoi, T., Honda, M., Oba, T., Ozawa, K., 2013. ER stress upregulated PGE2/IFNg- Ledo, J.H., Azevedo, E.P., Clarke, J.R., Ribeiro, F.C., Figueiredo, C.P., Foguel, D., De
induced IL-6 expression and down-regulated iNOS expression in glial cells. Sci. Felice, F.G., Ferreira, S.T., 2013. Amyloid-b oligomers link depressive-like
Rep. 3, 3388. https://doi.org/10.1038/srep03388. behavior and cognitive deficits in mice. Mol. Psychiatry 18, 1053e1054.
Hosoi, T., Sasaki, M., Baba, S., Ozawa, K., 2009. Effect of pranoprofen on endoplasmic https://doi.org/10.1038/mp.2012.168.
reticulum stress in the primary cultured glial cells. Neurochem. Int. 54, 1e6. Lee, A.-H., Iwakoshi, N.N., Glimcher, L.H., 2003. XBP-1 regulates a subset of endo-
https://doi.org/10.1016/j.neuint.2008.09.017. plasmic reticulum resident chaperone genes in the unfolded protein response.
Hotamisligil, G.S., 2010. Endoplasmic reticulum stress and the inflammatory basis of Mol. Cell. Biol. 23, 7448e7459. https://doi.org/10.1128/MCB.23.21.7448-
metabolic disease. Cell 140, 900e917. https://doi.org/10.1016/j.cell.2010.02.034. 7459.2003.
Hu, P., Han, Z., Couvillon, A.D., Kaufman, R.J., Exton, J.H., 2006. Autocrine tumor Lee, J.H., Won, S.M., Suh, J., Son, S.J., Moon, G.J., Park, U.-J., Gwag, B.J., 2010. Induction
necrosis factor alpha links endoplasmic reticulum stress to the membrane of the unfolded protein response and cell death pathway in Alzheimer's disease,
death receptor pathway through IRE1 -mediated NF- B activation and down- but not in aged Tg2576 mice. Exp. Mol. Med. 42, 386. https://doi.org/10.3858/
regulation of TRAF2 expression. Mol. Cell. Biol. 26, 3071e3084. https:// emm.2010.42.5.040.
doi.org/10.1128/MCB.26.8.3071-3084.2006. Lenny, N., Green, M., 1991. Regulation of endoplasmic reticulum stress proteins in
Hwang, C., Sinskey, A., Lodish, H., 1992. Oxidized redox state of glutathione in the COS cells transfected with immunoglobulin m heavy chain cDNA. J. Biol. Chem.
endoplasmic reticulum. Science (80-. ) 257, 1496e1502. https://doi.org/10.1126/ 266, 20532e20537.
science.1523409. Leonardi, A., 2002. Endoplasmic reticulum stress causes thyroglobulin retention in
Iadecola, C., Zhang, F., Casey, R., Nagayama, M., Ross, M.E., 1997. Delayed reduction this organelle and triggers activation of nuclear factor- B via tumor necrosis
of ischemic brain injury and neurological deficits in mice lacking the inducible factor receptor-associated factor 2. Endocrinology 143, 2169e2177. https://
nitric oxide synthase gene. J. Neurosci. 17, 9157e9164. doi.org/10.1210/en.143.6.2169.
Ide, M., Harris, M., Stevens, A., Sussams, R., Hopkins, V., Culliford, D., Fuller, J., Li, W.W., Sistonen, L., Morimoto, R.I., Lee, A.S., 1994. Stress induction of the
Ibbett, P., Raybould, R., Thomas, R., Puenter, U., Teeling, J., Perry, V.H., Holmes, C., mammalian GRP78/BiP protein gene: in vivo genomic footprinting and iden-
2016. Periodontitis and cognitive decline in Alzheimer's disease. PLoS One 11, tification of p70CORE from human nuclear extract as a DNA-binding compo-
e0151081. https://doi.org/10.1371/journal.pone.0151081. nent specific to the stress regulatory element. Mol. Cell. Biol. 14, 5533e5546.
Jackson, R.J., Hellen, C.U.T., Pestova, T.V., 2010. The mechanism of eukaryotic Lindholm, D., Wootz, H., Korhonen, L., 2006. ER stress and neurodegenerative dis-
translation initiation and principles of its regulation. Nat. Rev. Mol. Cell Biol. 11, eases. Cell Death Differ. 13, 385e392. https://doi.org/10.1038/sj.cdd.4401778.
113e127. https://doi.org/10.1038/nrm2838. Liu, S.-Y., Wang, W., Cai, Z.-Y., Yao, L.-F., Chen, Z.-W., Wang, C.-Y., Zhao, B., Li, K.-S.,
Jaffrey, S.R., Erdjument-Bromage, H., Ferris, C.D., Tempst, P., Snyder, S.H., 2001. 2013. Polymorphism "116C/G of human X-box-binding protein 1 promoter is
Protein S-nitrosylation: a physiological signal for neuronal nitric oxide. Nat. Cell associated with risk of Alzheimer's disease. CNS Neurosci. Ther. 19, 229e234.
Biol. 3, 193e197. https://doi.org/10.1038/35055104. https://doi.org/10.1111/cns.12064.
Jansen, G., Maattanen, P., Denisov, A.Y., Scarffe, L., Schade, B., Balghi, H., Dejgaard, K., Llorens-Martín, M., Jurado, J., Hern! andez, F., Avila, J., 2014. GSK-3b, a pivotal kinase
Chen, L.Y., Muller, W.J., Gehring, K., Thomas, D.Y., 2012. An interaction map of in Alzheimer disease. Front. Mol. Neurosci. 7, 46. https://doi.org/10.3389/
endoplasmic reticulum chaperones and foldases. Mol. Cell. Proteomics 11, fnmol.2014.00046.

Please cite this article in press as: Santos, L.E., Ferreira, S.T., Crosstalk between endoplasmic reticulum stress and brain inflammation in
Alzheimer's disease, Neuropharmacology (2017), https://doi.org/10.1016/j.neuropharm.2017.11.016
10 L.E. Santos, S.T. Ferreira / Neuropharmacology xxx (2017) 1e11

Lourenco, M.V., Clarke, J.R., Frozza, R.L., Bomfim, T.R., Forny-Germano, L., Oyadomari, S., Takeda, K., Takiguchi, M., Gotoh, T., Matsumoto, M., Wada, I., Akira, S.,
Batista, A.F., Sathler, L.B., Brito-Moreira, J., Amaral, O.B., Silva, C.A., Freitas- Araki, E., Mori, M., 2001. Nitric oxide-induced apoptosis in pancreatic cells is
Correa, L., Espírito-Santo, S., Campello-Costa, P., Houzel, J.C., Klein, W.L., mediated by the endoplasmic reticulum stress pathway. Proc. Natl. Acad. Sci.
Holscher, C., Carvalheira, J.B., Silva, A.M., Velloso, L.A., Munoz, D.P., Ferreira, S.T., 98, 10845e10850. https://doi.org/10.1073/pnas.191207498.
De Felice, F.G., 2013. TNF-a mediates PKR-dependent memory impairment and Pacher, P., Beckman, J.S., Liaudet, L., 2007. Nitric oxide and peroxynitrite in health
brain IRS-1 inhibition induced by Alzheimer's b-amyloid oligomers in mice and and disease. Physiol. Rev. 87, 315e424, 87/1/315 [pii]10.1152/
monkeys. Cell Metab. 18, 831e843. https://doi.org/10.1016/j.cmet.2013.11.002. physrev.00029.2006.
Lue, L.F., Rydel, R., Brigham, E.F., Yang, L.B., Hampel, H., Murphy, G.M., Brachova, L., Pakos-Zebrucka, K., Koryga, I., Mnich, K., Ljujic, M., Samali, A., Gorman, A.M., 2016.
Yan, S., Du, Walker, D.G., Shen, Y., Rogers, J., 2001. Inflammatory repertoire of The integrated stress response. EMBO Rep. 17, 1374e1395. https://doi.org/
Alzheimer's disease and nondemented elderly microglia in vitro. Glia 35, 10.15252/embr.201642195.
72e79. https://doi.org/10.1002/glia.1072. Perez, N., Sugar, J., Charya, S., Johnson, G., Merril, C., Bierer, L., Perl, D.,
Luo, X.-G., Ding, J.-Q., Chen, S.-D., 2010. Microglia in the aging brain: relevance to Haroutunian, V., Wallace, W., 1991. Increased synthesis and accumulation of
neurodegeneration. Mol. Neurodegener. 5, 12. https://doi.org/10.1186/1750- heat shock 70 proteins in Alzheimer's disease. Mol. Brain Res. 11, 249e254.
1326-5-12. https://doi.org/10.1016/0169-328X(91)90033-T.
Lyketsos, C.G., Carrillo, M.C., Ryan, J.M., Khachaturian, A.S., Trzepacz, P., Amatniek, J., Perry, V.H., Holmes, C., 2014. Microglial priming in neurodegenerative disease. Nat.
Cedarbaum, J., Brashear, R., Miller, D.S., 2011. Neuropsychiatric symptoms in Rev. Neurol. 10, 217e224. https://doi.org/10.1038/nrneurol.2014.38.
Alzheimer's disease. Alzheimer's Dement. https://doi.org/10.1016/ Prince, M., Wimo, A., Guerchet, M., Gemma-Claire, A., Wu, Y.-T., Prina, M., 2015.
j.jalz.2011.05.2410. World alzheimer report 2015: the global impact of dementia - an analysis of
Lyketsos, C.G., Olin, J., 2002. Depression in Alzheimer's disease: overview and prevalence, incidence, cost and trends. Alzheimer’s Dis. Int. 84 https://doi.org/
treatment. Biol. Psychiatry 52, 243e252. https://doi.org/10.1016/S0006- 10.1111/j.0963-7214.2004.00293.x.
3223(02)01348-3. Radde, R., Bolmont, T., Kaeser, S.A., Coomaraswamy, J., Lindau, D., Stoltze, L.,
Ma, T., Trinh, M. a, Wexler, A.J., Bourbon, C., Gatti, E., Pierre, P., Cavener, D.R., Calhoun, M.E., J€ aggi, F., Wolburg, H., Gengler, S., Haass, C., Ghetti, B., Czech, C.,
Klann, E., 2013. Suppression of eIF2a kinases alleviates Alzheimer's disease- Ho €lscher, C., Mathews, P.M., Jucker, M., 2006. Ab42-driven cerebral amyloidosis
related plasticity and memory deficits. Nat. Neurosci. 16, 1299e1305. https:// in transgenic mice reveals early and robust pathology. EMBO Rep. 7, 940e946.
doi.org/10.1038/nn.3486. https://doi.org/10.1038/sj.embor.7400784.
Maes, M., Berk, M., Goehler, L., Song, C., Anderson, G., Gałecki, P., Leonard, B., 2012. Rao, J., Yue, S., Fu, Y., Zhu, J., Wang, X., Busuttil, R.W., Kupiec-Weglinski, J.W., Lu, L.,
Depression and sickness behavior are Janus-faced responses to shared inflam- Zhai, Y., 2014. ATF6 mediates a pro-inflammatory synergy between ER stress
matory pathways. BMC Med. 10, 66. https://doi.org/10.1186/1741-7015-10-66. and TLR activation in the pathogenesis of liver ischemia-reperfusion injury. Am.
Maixner, D.W., Weng, H.-R., 2013. No title. J. Pharm. Pharmacol. 1. doi:10.13188/ J. Transpl. 14, 1552e1561. https://doi.org/10.1111/ajt.12711.
2327-204X.1000001. Reed, T.T., Pierce, W.M., Turner, D.M., Markesbery, W.R., Allan Butterfield, D., 2009.
Mandrekar, S., Landreth, G.E., 2010. Microglia and inflammation in Alzheimer's Proteomic identification of nitrated brain proteins in early Alzheimer's disease
disease. CNS Neurol. Disord. Drug Targets 9, 156e167. https://doi.org/10.2174/ inferior parietal lobule. J. Cell. Mol. Med. 13, 2019e2029. https://doi.org/10.1111/
187152710791012071. j.1582-4934.2008.00478.x.
Martínez, G., Vidal, R.L., Mardones, P., Serrano, F.G., Ardiles, A.O., Wirth, C., Valde!s, P., Resende, R., Ferreiro, E., Pereira, C., Oliveira, C.R., 2008. ER stress is involved in
Thielen, P., Schneider, B.L., Kerr, B., Valde !s, J.L., Palacios, A.G., Inestrosa, N.C., Abeta-induced GSK-3beta activation and tau phosphorylation. J. Neurosci. Res.
Glimcher, L.H., Hetz, C., 2016. Regulation of Memory Formation by the Tran- 86, 2091e2099. https://doi.org/10.1002/jnr.21648.
scription Factor XBP1. Cell Rep 14, 1382e1394. https://doi.org/10.1016/ Reynolds, M.R., Reyes, J.F., Fu, Y., Bigio, E.H., Guillozet-Bongaarts, A.L., Berry, R.W.,
j.celrep.2016.01.028. Binder, L.I., 2006. Tau nitration occurs at tyrosine 29 in the fibrillar lesions of
Meares, G.P., Hughes, K.J., Naatz, A., Papa, F.R., Urano, F., Hansen, P.A., Alzheimer's disease and other tauopathies. J. Neurosci. 26, 10636e10645.
Benveniste, E.N., Corbett, J.A., 2011. IRE1-Dependent activation of AMPK in https://doi.org/10.1523/JNEUROSCI.2143-06.2006.
response to nitric oxide. Mol. Cell. Biol. 31, 4286e4297. https://doi.org/10.1128/ Ron, D., Walter, P., 2007. Signal integration in the endoplasmic reticulum unfolded
MCB.05668-11. protein response. Nat. Rev. Mol. Cell Biol. 8, 519e529. https://doi.org/10.1038/
Meares, G.P., Liu, Y., Rajbhandari, R., Qin, H., Nozell, S.E., Mobley, J.A., Corbett, J.A., nrm2199.
Benveniste, E.N., 2014. PERK-dependent activation of JAK1 and STAT3 contrib- Salminen, A., Kauppinen, A., Suuronen, T., Kaarniranta, K., Ojala, J., 2009. ER stress in
utes to endoplasmic reticulum stress-induced inflammation. Mol. Cell. Biol. 34, Alzheimer's disease: a novel neuronal trigger for inflammation and Alzheimer's
3911e3925. https://doi.org/10.1128/MCB.00980-14. pathology. J. Neuroinflammation 6, 41. https://doi.org/10.1186/1742-2094-6-41.
Mekahli, D., Bultynck, G., Parys, J.B., de Smedt, H., Missiaen, L., 2011. Endoplasmic- Santos, L.E., Beckman, D., Ferreira, S.T., 2016. Microglial dysfunction connects
reticulum calcium depletion and disease. Cold Spring Harb. Perspect. Biol. depression and Alzheimer's disease. Brain. Behav. Immun. 55, 151e165. https://
https://doi.org/10.1101/cshperspect.a004317. doi.org/10.1016/j.bbi.2015.11.011.
Minter, M.R., Taylor, J.M., Crack, P.J., 2016. The contribution of neuroinflammation to Sarlus, H., Heneka, M.T., 2017. Microglia in Alzheimer's disease. J. Clin. Invest. 127,
amyloid toxicity in Alzheimer's disease. J. Neurochem. https://doi.org/10.1111/ 3240e3249. https://doi.org/10.1172/JCI90606.
jnc.13411. Selkoe, D.J., Hardy, J., 2016. The amyloid hypothesis of Alzheimer's disease at 25
Morl, K., Ma, W., Gething, M.-J., Sambrook, J., 1993. A transmembrane protein with a years. EMBO Mol. Med. https://doi.org/10.15252/emmm.201606210.
cdc2þCDC28-related kinase activity is required for signaling from the ER to the Shen, J., Chen, X., Hendershot, L., Prywes, R., 2002. ER stress regulation of ATF6
nucleus. Cell 74, 743e756. https://doi.org/10.1016/0092-8674(93)90521-Q. localization by dissociation of BiP/GRP78 binding and unmasking of Golgi
Mukherjee, A., Morales-Scheihing, D., Butler, P.C., Soto, C., 2015. Type 2 diabetes as a localization signals. Dev. Cell 3, 99e111. https://doi.org/10.1016/S1534-5807(02)
protein misfolding disease. Trends Mol. Med. https://doi.org/10.1016/ 00203-4.
j.molmed.2015.04.005. Shi, J.Q., Wang, B.R., Jiang, W.W., Chen, J., Zhu, Y.W., Zhong, L.L., Zhang, Y.D., Xu, J.,
Naidoo, N., 2009. ER and agingdprotein folding and the ER stress response. Ageing 2011. Cognitive improvement with intrathecal administration of infliximab in a
Res. Rev. 8, 150e159. https://doi.org/10.1016/j.arr.2009.03.001. woman with Alzheimer's disease. J. Am. Geriatr. Soc. 59, 1142e1144. https://
Nakamura, T., Tu, S., Akhtar, M., Sunico, C., Okamoto, S.I., Lipton, S., 2013. Aberrant doi.org/10.1111/j.1532-5415.2011.03445.x.
Protein S-nitrosylation in neurodegenerative diseases. Neuron 78, 596e614. Shie, F.-S., Montine, K.S., Breyer, R.M., Montine, T.J., 2005. Microglial EP2 is critical to
https://doi.org/10.1016/j.neuron.2013.05.005. neurotoxicity from activated cerebral innate immunity. Glia 52, 70e77. https://
Nakato, R., Ohkubo, Y., Konishi, A., Shibata, M., Kaneko, Y., Iwawaki, T., Nakamura, T., doi.org/10.1002/glia.20220.
Lipton, S.A., Uehara, T., 2015. Regulation of the unfolded protein response via S- Sly, L., Krzesicki, R., Brashler, J., Buhl, A., McKinley, D., Carter, D., Chin, J., 2001.
nitrosylation of sensors of endoplasmic reticulum stress. Sci. Rep. 5, 14812. Endogenous brain cytokine mRNA and inflammatory responses to lipopoly-
https://doi.org/10.1038/srep14812. saccharide are elevated in the Tg2576 transgenic mouse model of Alzheimer's
Nevell, L., Zhang, K., Aiello, A.E., Koenen, K., Galea, S., Soliven, R., Zhang, C., disease. Brain Res. Bull. 56, 581e588. https://doi.org/10.1016/S0361-9230(01)
Wildman, D.E., Uddin, M., 2014. Elevated systemic expression of ER stress 00730-4.
related genes is associated with stress-related mental disorders in the detroit Spangenberg, E.E., Lee, R.J., Najafi, A.R., Rice, R.A., Elmore, M.R.P., Blurton-Jones, M.,
neighborhood health study. Psychoneuroendocrinology 43, 62e70. https:// West, B.L., Green, K.N., 2016. Eliminating microglia in Alzheimer's mice pre-
doi.org/10.1016/j.psyneuen.2014.01.013. vents neuronal loss without modulating amyloid-b pathology. Brain 139,
Nishitoh, H., Matsuzawa, A., Tobiume, K., Saegusa, K., Takeda, K., Inoue, K., Hori, S., 1265e1281. https://doi.org/10.1093/brain/aww016.
Kakizuka, A., Ichijo, H., 2002. ASK1 is essential for endoplasmic reticulum Stelzmann, R.A., Norman Schnitzlein, H., Reed Murtagh, F., 1995. An english trans-
stress-induced neuronal cell death triggered by expanded polyglutamine re- lation of alzheimer's 1907 paper. Clin. Anat. 8, 429e431. https://doi.org/
peats. Genes Dev. 16, 1345e1355. https://doi.org/10.1101/gad.992302. 10.1002/ca.980080612.
Norden, D.M., Muccigrosso, M.M., Godbout, J.P., 2015. Microglial priming and Stutzbach, L.D., Xie, S.X., Naj, A.C., Albin, R., Gilman, S., Lee, V.M.Y., Trojanowski, J.Q.,
enhanced reactivity to secondary insult in aging, and traumatic CNS injury, and Devlin, B., Schellenberg, G.D., 2013. The unfolded protein response is activated
neurodegenerative disease. Neuropharmacology 96, 29e41. https://doi.org/ in disease-affected brain regions in progressive supranuclear palsy and Alz-
10.1016/j.neuropharm.2014.10.028. heimer's disease. Acta Neuropathol. Commun. 1, 31. https://doi.org/10.1186/
Omi, T., Tanimukai, H., Kanayama, D., Sakagami, Y., Tagami, S., Okochi, M., 2051-5960-1-31.
Morihara, T., Sato, M., Yanagida, K., Kitasyoji, A., Hara, H., Imaizumi, K., Teeling, J.L., Felton, L.M., Deacon, R.M.J., Cunningham, C., Rawlins, J.N.P., Perry, V.H.,
Maurice, T., Chevallier, N., Marchal, S., Takeda, M., Kudo, T., 2014. Fluvoxamine 2007. Sub-pyrogenic systemic inflammation impacts on brain and behavior,
alleviates ER stress via induction of Sigma-1 receptor. Cell Death Dis. 5, e1332 independent of cytokines. Brain. Behav. Immun. 21, 836e850. https://doi.org/
https://doi.org/10.1038/cddis.2014.301. 10.1016/j.bbi.2007.01.012.

Please cite this article in press as: Santos, L.E., Ferreira, S.T., Crosstalk between endoplasmic reticulum stress and brain inflammation in
Alzheimer's disease, Neuropharmacology (2017), https://doi.org/10.1016/j.neuropharm.2017.11.016
L.E. Santos, S.T. Ferreira / Neuropharmacology xxx (2017) 1e11 11

Timberlake, M.A., Dwivedi, Y., 2016. Altered expression of endoplasmic reticulum endoplasmic reticulum unfolded protein response transducers through their
stress associated genes in Hippocampus of learned helpless rats: relevance to transmembrane domains. Proc. Natl. Acad. Sci. U. S. A. 110, 4628e4633. https://
depression pathophysiology. Front. Pharmacol. 6 https://doi.org/10.3389/ doi.org/10.1073/pnas.1217611110.
fphar.2015.00319. Walker, F.R., 2013. A critical review of the mechanism of action for the selective
Tobinick, E.L., Gross, H., 2008. Rapid cognitive improvement in Alzheimer's disease serotonin reuptake inhibitors: do these drugs possess anti-inflammatory
following perispinal etanercept administration. J. Neuroinflammation 5, 2. properties and how relevant is this in the treatment of depression? Neuro-
https://doi.org/10.1186/1742-2094-5-2. pharmacology 67, 304e317. https://doi.org/10.1016/j.neuropharm.2012.10.002.
Tohgi, H., Abe, T., Yamazaki, K., Murata, T., Ishizaki, E., Isobe, C., 1999. Alterations of Wang, J., Tan, L., Wang, H.F., Tan, C.C., Meng, X.F., Wang, C., Tang, S.W., Yu, J.T., 2015.
3-nitrotyrosine concentration in the cerebrospinal fluid during aging and in Anti-inflammatory drugs and risk of Alzheimer's disease: an updated system-
patients with Alzheimer's disease. Neurosci. Lett. 269, 52e54. https://doi.org/ atic review and meta-analysis. J. Alzheimers Dis. 44, 385e396. https://doi.org/
10.1016/S0304-3940(99)00406-1. 10.3233/JAD-141506.
Tong, L., Heim, R.A., Wu, S., 2011. Nitric oxide: a regulator of eukaryotic initiation Xu, N., Xiao, Z., Zou, T., Huang, Z., 2015. Induction of GADD34 regulates the
factor 2 kinases. Free Radic. Biol. Med. 50, 1717e1725. https://doi.org/10.1016/ neurotoxicity of amyloid. Am. J. Alzheimers. Dis. Other Demen 30, 313e319.
j.freeradbiomed.2011.03.032. https://doi.org/10.1177/1533317514545616.
Torres, M., Matamala, J.M., Duran-Aniotz, C., Cornejo, V.H., Foley, A., Hetz, C., 2015. Xu, W., Liu, L., Charles, I.G., Moncada, S., 2004. Nitric oxide induces coupling of
ER stress signaling and neurodegeneration: at the intersection between Alz- mitochondrial signalling with the endoplasmic reticulum stress response. Nat.
heimer's disease and Prion-related disorders. Virus Res. 207, 69e75. https:// Cell Biol. 6, 1129e1134. https://doi.org/10.1038/ncb1188.
doi.org/10.1016/j.virusres.2014.12.018. Yamazaki, H., Hiramatsu, N., Hayakawa, K., Tagawa, Y., Okamura, M., Ogata, R.,
Tsutsumi, S., Gotoh, T., Tomisato, W., Mima, S., Hoshino, T., Hwang, H.-J., Huang, T., Nakajima, S., Yao, J., Paton, A.W., Paton, J.C., Kitamura, M., 2009.
Takenaka, H., Tsuchiya, T., Mori, M., Mizushima, T., 2004. Endoplasmic reticulum Activation of the Akt-NF-kappaB pathway by subtilase cytotoxin through the
stress response is involved in nonsteroidal anti-inflammatory drug-induced ATF6 branch of the unfolded protein response. J. Immunol. 183, 1480e1487.
apoptosis. Cell Death Differ. 11, 1009e1016. https://doi.org/10.1038/ https://doi.org/10.4049/jimmunol.0900017.
sj.cdd.4401436. Yamazaki, T., Muramoto, M., Oe, T., Morikawa, N., Okitsu, O., Nagashima, T.,
Tsutsumi, S., Namba, T., Tanaka, K.-I., Arai, Y., Ishihara, T., Aburaya, M., Mima, S., Nishimura, S., Katayama, Y., Kita, Y., 2006. Diclofenac, a non-steroidal anti-in-
Hoshino, T., Mizushima, T., 2006. Celecoxib upregulates endoplasmic reticulum flammatory drug, suppresses apoptosis induced by endoplasmic reticulum
chaperones that inhibit celecoxib-induced apoptosis in human gastric cells. stresses by inhibiting caspase signaling. Neuropharmacology 50, 558e567.
Oncogene 25, 1018e1029. https://doi.org/10.1038/sj.onc.1209139. https://doi.org/10.1016/j.neuropharm.2005.10.016.
Tweedie, D., Ferguson, R. a, Fishman, K., Frankola, K. a, Van Praag, H., Yang, L., Calay, E.S., Fan, J., Arduini, A., Kunz, R.C., Gygi, S.P., Yalcin, A., Fu, S.,
Holloway, H.W., Luo, W., Li, Y., Caracciolo, L., Russo, I., Barlati, S., Ray, B., Hotamisligil, G.S., 2015. S-Nitrosylation links obesity-associated inflammation
Lahiri, D.K., Bosetti, F., Greig, N.H., Rosi, S., 2012. Tumor necrosis factor-a syn- to endoplasmic reticulum dysfunction. Science (80-. ) 349, 500e506. https://
thesis inhibitor 3,60 -dithiothalidomide attenuates markers of inflammation, doi.org/10.1126/science.aaa0079.
Alzheimer pathology and behavioral deficits in animal models of neuro- Yoon, S.O., Park, D.J., Ryu, J.C., Ozer, H.G., Tep, C., Shin, Y.J., Lim, T.H., Pastorino, L.,
inflammation and Alzheimer's disease. J. Neuroinflammation 9, 106. https:// Kunwar, A.J., Walton, J.C., Nagahara, A.H., Lu, K.P., Nelson, R.J., Tuszynski, M.H.,
doi.org/10.1186/1742-2094-9-106. Huang, K., 2012. JNK3 perpetuates metabolic stress induced by Abeta peptides.
Tweedie, D., Sambamurti, K., Greig, N.H., 2007. TNF-alpha inhibition as a treatment Neuron 75, 824e837. https://doi.org/10.1016/j.neuron.2012.06.024.
strategy for neurodegenerative disorders: new drug candidates and targets. Zhang, K., Kaufman, R.J., 2008. From endoplasmic-reticulum stress to the inflam-
Curr. Alzheimer Res. 4, 378e385. matory response. Nature 454, 455e462. https://doi.org/10.1038/nature07203.
Uehara, T., Nakamura, T., Yao, D., Shi, Z.-Q., Gu, Z., Ma, Y., Masliah, E., Nomura, Y., Zhang, K., Shen, X., Wu, J., Sakaki, K., Saunders, T., Rutkowski, D.T., Back, S.H.,
Lipton, S.A., 2006. S-Nitrosylated protein-disulphide isomerase links protein Kaufman, R.J., 2006. Endoplasmic reticulum stress activates cleavage of CREBH
misfolding to neurodegeneration. Nature 441, 513e517. https://doi.org/10.1038/ to induce a systemic inflammatory response. Cell 124, 587e599. https://doi.org/
nature04782. 10.1016/j.cell.2005.11.040.
Urano, F., Wang, X., Bertolotti, A., Zhang, Y., Chung, P., Harding, H.P., Ron, D., 2000. Zschocke, J., Zimmermann, N., Berning, B., Ganal, V., Holsboer, F., Rein, T., 2011.
Coupling of stress in the ER to activation of JNK protein kinases by trans- Antidepressant drugs diversely affect autophagy pathways in astrocytes and
membrane protein kinase IRE1. Science (80-. ) 287, 664e666. https://doi.org/ neuronsddissociation from cholesterol homeostasis. Neuro-
10.1126/science.287.5453.664. psychopharmacology 36, 1754e1768. https://doi.org/10.1038/npp.2011.57.
Volmer, R., van der Ploeg, K., Ron, D., 2013. Membrane lipid saturation activates

Please cite this article in press as: Santos, L.E., Ferreira, S.T., Crosstalk between endoplasmic reticulum stress and brain inflammation in
Alzheimer's disease, Neuropharmacology (2017), https://doi.org/10.1016/j.neuropharm.2017.11.016

You might also like