Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Brain Research 1821 (2023) 148589

Contents lists available at ScienceDirect

Brain Research
journal homepage: www.elsevier.com/locate/brainres

Alzheimer’s disease: The role of T lymphocytes in neuroinflammation


and neurodegeneration
Moses O. Asamu a, c, 1, Oladapo O. Oladipo b, c, *, 1, Oluseun A. Abayomi c, d, Afeez A. Adebayo b, c
a
Department of Anatomy, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
b
Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
c
College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
d
Olabisi Onabanjo University Teaching Hospital (OOUTH), Sagamu, Ogun State, Nigeria

A R T I C L E I N F O A B S T R A C T

Keywords: Alzheimer’s disease, the leading cause of progressive cognitive decline globally, has been reported to be
Alzheimer’s disease enhanced by neuroinflammation. Brain-resident innate immune cells and adaptive immune cells work together
Microglia to produce neuroinflammation. Studies over the past decade have established the neuroimmune axis present in
T lymphocyte
Alzheimer’s disease; the crosstalk between adaptive and innate immune cells within and outside the brain is
Neuroinflammation
Neurodegeneration
crucial to the onset and progression of Alzheimer’s disease. Although the role of the adaptive immune system in
Alzheimer’s disease is not fully understood, it has been hypothesized that the brain’s immune homeostasis is
significantly disrupted, which greatly contributes to neuroinflammation. Brain-infiltrating T cells possess
proinflammatory phenotypes and activities that directly contribute to neuroinflammation. The pro-inflammatory
activities of the adaptive immune system in Alzheimer’s disease are characterized by the upregulation of effector
T cell activities and the downregulation of regulatory T cell activities in the brain, blood, and cerebrospinal fluid.
In this review, we discuss the major impact of T lymphocytes on the pathogenesis and progression of Alzheimer’s
disease. Understanding the role and mechanism of action of T cells in Alzheimer’s disease would significantly
contribute to the identification of novel biomarkers for diagnosing and monitoring the progression of the disease.
This knowledge could also be crucial to the development of immunotherapies for Alzheimer’s disease.

1. Introduction 131 million in the incidence of AD across the globe (Albrecht et al.,
2021). Despite several scientific studies on AD’s pathogenesis, etiology,
Alzheimer’s disease (AD) is a multifactorial, neurodegenerative progression, and features, there is no established cure. Thus, AD is a
condition characterized by progressive memory loss and impairment in global concern that significantly threatens human life expectancy.
cognition (Reddy et al., 2018). Reports of the incidence and mortality of AD results in brain alterations that cumulatively manifest in its
AD in the USA presented it as the seventh leading cause of death, with an development and clinical presentation (Alonso Adel et al., 2004). Ag­
incidence of about 6.5 million and a projected 13.8 million by 2060 gregation and buildup of intracellular neurofibrillary tangles (NFT, also
([xxxx, 0000]). Likewise, the 2016 World Alzheimer’s Report projected tau tangles) and abnormal extracellular clumps (amyloid plaques) have
that from 2016 to 2050, there would be an increase from 46.8 million to been reported to be the main defining structural changes occurring in

Abbreviations: AD, Alzheimer’s disease; NFT, neurofibrillary tangles; Aβ, beta-amyloid; PS-1 and PS-2, presenilins; APP, amyloid precursor protein; BBB, blood-
brain barrier; ApoE, apolipoprotein E; BACE1, beta-site AβPP-cleaving enzyme; CSF, cerebrospinal fluid; ZO1, zonaoccludens protein 1; MCI, mild cognitive decline;
CAA, Cerebral amyloid angiopathy; CNS, central nervous system; Treg, regulatory T cell; Teff, effector T cells; Th-1, helper T cell-1; Th-17, helper T cell-17; RORγt,
retinoic acid receptor-related orphan nuclear receptor γt; MHC, major histocompatibility complex; EAE, experimental autoimmune encephalomyelitis; CTLA-4,
cytotoxic T-lymphocyte-associated protein-4; PD-1, programmed cell death-1; CCR2, C-C motif chemokine receptor 2; CCR5, C-C chemokine receptor type 5; CXCR2,
C-X-C chemokine receptor 2; ICAM-1, intercellular adhesion molecule 1; VCAM-1, Vascular cell adhesion molecule 1; M1, pro-inflammatory microglia; M2, anti-
inflammatory microglia; MIP-1 α, macrophage inflammatory proteins-1α; CX3CR1, CX3C motif chemokine receptor 1; RAGE, receptor for advanced glycation end
products.
* Corresponding author.
E-mail address: oladipooladapoo@gmail.com (O.O. Oladipo).
1
Moses O. Asamu and Oladapo O. Oladipo made equal contribution to the manuscript.

https://doi.org/10.1016/j.brainres.2023.148589
Received 8 June 2023; Received in revised form 3 September 2023; Accepted 18 September 2023
Available online 20 September 2023
0006-8993/© 2023 Elsevier B.V. All rights reserved.
M.O. Asamu et al. Brain Research 1821 (2023) 148589

AD (Alonso et al., 2001; Amor and Woodroofe, 2014; Anderson et al., accounted for by known mutations (Bondi et al., 2017).
2014). Aggregation of both proteins, beta-amyloid (Aβ) and tau, syn­ The etiology of AD has yet to be clearly understood, although risk
ergistically contributes to the loss of neuronal connections at synapses factors like aging, vascular diseases, head injuries, genetic factors, in­
and damage to the neurons in the brain tissue. These, in turn, induce and fections and environmental factors have been identified (Breijyeh and
promote cognitive decline in people with AD (Agnes et al., 2013). In AD Karaman, 2020). Brain aging, a complex and irreversible process char­
degeneration, microtubule motor proteins, phosphatase and kinase acterized by atrophy, synaptic loss, enlargement of the ventricles,
mechanisms are dysregulated (Apostolova, 2016; Ashutosh et al., 2011). deposition of NFTs, and cognitive decline, among other characteristics,
Other dysregulated proteins or genes in AD include presenilins (PS-1 and is a significant risk factor (Breijyeh and Karaman, 2020; Browne et al.,
PS-2), amyloid precursor protein (APP), and secretases (Ávila-Villa­ 2013). Genetic factors also play significant roles in AD etiology. Ac­
nueva et al., 2022). Each of these factors either upregulates tau hyper­ cording to twin and family studies, up to 80% of all AD cases had
phosphorylation into NFTs or increases Aβ production, which results in phenotypic variance that was correlated to genetic factors (Cao and
the neurodegenerative changes observed in AD (Apostolova, 2016; Zheng, 2018). The late-onset AD is assumed to be caused by both
Ashutosh et al., 2011; Ávila-Villanueva et al., 2022). environmental (about 30%) and genetic factors (about 70%) (Carrano
The role of T lymphocytes – the adaptive immune cells – in the et al., 2012). High levels of NFTs and Aβ plaques have also been iden­
pathogenesis of AD has not been extensively explored and reported. tified as the pathological hallmarks of AD, and several therapeutic
Unlike the innate immune cells, astrocytes and microglia, which are studies on AD have been based on these hypotheses. However, there is
activated by the protein buildup in AD to produce inflammatory and still no accepted theory for the pathogenesis of AD.
neurotoxic factors that, in turn, increase abnormal amyloid precursor The pre-clinical stage of AD, which is also known as the pre-
protein (APP) cleavage, production of Aβ and the phosphorylation of tau symptomatic stage, does not present any symptom of AD nor impair­
according to mice study (Baek et al., 2016). An alteration in the function ment in the performance of day-to-day activities but is marked by mild
of lymphocytes in AD may contribute to disease progression (Baik et al., memory loss and the onset of pathological impairment in the cortex and
2014). A study reported the presence of T-cells in the brain during hippocampus (Carrano et al., 2011; Cavanagh and Wong, 2018). This
postmortem examination of AD patients (Bartholomäus et al., 2009). stage is followed by a mild or early stage AD, which presents symptoms
Following an increase in the blood–brain barrier’s (BBB) permeability, like a mild decline in concentration and memory, mood swings and
Dai and Shen (2021) stated that there is an increased T-cell influx into depression, and disorientation of place and time (Cebrián et al., 2014).
the brain’s parenchyma, promoted by the increased expression of che­ These symptoms lead to difficulties in carrying out daily activities and
mokine receptors by peripheral T-cells (Baruch et al., 2016). These re­ are followed by the spread of the disease to areas of the cerebral cortex
ceptors include C-C motif chemokine receptor type 2 and 5 (CCR2 and that result in increased impairment in memory, speech, writing, reading,
CCR5) and C-X-C motif chemokine receptor 2 (CXCR2). Aβ also pro­ and loss of impulse control (Breijyeh and Karaman, 2020). This stage is
motes the infiltration of T-cells through BBB and dysfunction in T-cell known as moderate AD and progresses into severe AD or late stage. The
activation and presentation of antigens (Baruch et al., 2015), further late-stage AD is characterized by the spread of the disease to all cortical
contributing to the inability to clear Aβ by the immune cells (Baik et al., areas, severe accumulation of NFTs and senile plaques that result in
2014). progressive cognitive and functional decline and, eventually, death
The effect of chronic T-cells activation on neuroinflammation has (Carrano et al., 2011; Chabran et al., 2020).
been well studied [12, see below]. T-cells dysfunction in AD contributes
to neuroinflammation by secreting pro-inflammatory mediators, which 3. Aβ, Tau-Protein buildup and neuroinflammation
have been reported to be an essential modulator in the pathogenesis of
this neurodegenerative disorder (Baruch et al., 2016). Increased CD4 + Aβ and tau-protein are the pathological hallmarks of AD. This is
T cell count has also been reported to upregulate the risk of developing portrayed by the accumulation of amyloid plaques and NFT deposits in
AD (Bateman et al., 2012). In this review, we discussed NFT and Aβ the cerebral cortex and specific subcortical regions, which result in
plaques as the defining agents of AD, and we also discussed the struc­ parieto-temporal degeneration that extends to part of the cingulate
tural and functional changes in the BBB associated with AD. We gyrus and frontal cortex (Cheng et al., 2014). Masters and Selkoe (2012)
reviewed T lymphocytes’ role in AD pathogenesis, including the role of reiterated that the cerebral deposit of Aβ in the areas of the brain
CD4 + and CD8 + T lymphocytes. Understanding these concepts is responsible for cognition and memory is a defining characteristic of AD
germane in managing AD and its therapeutic research. (Chui and Dorovini-Zis, 2010). Also, several studies have suggested that
AD pathological diagnosis is defined by the deposit and accumulation of
2. Onset, Etiology, and progression of AD extracellular Aβ plaques (with Aβ42 amyloid protein being the main
constituent) and intracellular NFT of hyperphosphorylated tau-protein,
AD incidence and risk are directly associated with aging, increasing which binds to microtubules (Fig. 1) (Ciccocioppo et al., 2019; Cipollini
exponentially after age 65 (Bettcher et al., 2021). The onset of AD is et al., 2019; Cohen and Torres, 2019). Predominant evidence-based
often classified into two. Defined as the onset of AD before age 65, early- hypotheses on the aetiology of AD have been based on amyloid
onset AD comprises about 5% to 6% of AD cases (Blair et al., 2015), cascade and tau-protein hyperphosphorylation, while others include
while late-onset AD, i.e. occurring above age 65, makes up the majority Apolipoprotein E and neuroinflammation hypotheses (Baruch et al.,
of AD cases. The majority of the early-onset cases of AD are familial; the 2016). In healthy humans, APP cleaved by enzymes like beta-site AβPP-
incidence of AD in two or more members of a generation. The early- cleaving enzyme (BACE1) and gamma-secretase is involved in signal
onset AD has been identified to pose an increased incidence of trau­ transduction, migration of cells, and elongation of axons. At the same
matic brain injury, impaired mental functions, reduced impairment in time, its C-terminus is critical to gene expression and survival of the
memory, and a more aggressive course than late-onset AD (Blair et al., neuronal cell (Ávila-Villanueva et al., 2022). However, APP mutations
2015). Other distinctions between early-onset AD and late-onset AD or mutations in either of two APP cleavage enzymes, presenilins 1 (PS1)
include the increased incidence of tau burden, alterations in the or presenilin 2 (PS2), are associated with familial early-onset AD (Cic­
connection of the frontoparietal network, and atrophy in the posterior cocioppo et al., 2019). By identifying this mutation, there was a hy­
neocortex. This classification, which is still used to date in both research pothesis that Aβ causes AD, forming the basis of the amyloid cascade
and clinical trials, may not mirror the underlying etiology of AD. Ac­ experiment (Crews et al., 2010). This discovery drove research attention
cording to the review by Reitz et al. (2020), this gap in correlation is to Aβ. The amyloid cascade hypothesis includes a sequence of events, of
evidenced by the incidence of late-onset cases in multiplex families with which abnormal APP cleavage is key (Croese et al., 2021). The abnormal
early-onset AD and only a small percentage of early-onset AD being cleavage of this amyloidogenic transmembrane protein results in the

2
M.O. Asamu et al. Brain Research 1821 (2023) 148589

Fig. 1. Tau-protein aids microtubule stabilization by promoting tubulin packaging into the microtubule in the healthy neuron (Dai and Shen, 2021). However, with
increase in the aggregation of Aβ plaques in Alzheimer’s disease, tau-protein becomes hyperphosphorylated, dissociate from microtubule, and aggregates into
neurofibrillary tangle (Dionisio-Santos et al., 2019). Due to these alterations, synaptic damage and neuronal death is induced. Also, there is atrophy in the cerebral
cortex and hippocampus (Dong and Benveniste, 2001), as well as a resultant enlargement of the ventricles in Alzheimer’s disease.

production of excessive Aβ and the formation of oligomers and subse­ patients with low Aβ and high tau-protein levels (Dionisio-Santos et al.,
quently result in the buildup and aggregation of Aβ in the brain tissue 2019; Dong and Benveniste, 2001). This hypothesis made hyper­
(and CNS). Aβ oligomerization induces a sequence of events that causes phosphorylated tau and associated NFT budding targets in AD thera­
loss of synaptic communication, dysfunction of cholinergic neurons, and peutic research in addition to the initial research focus on Aβ (Dionisio-
synaptic degeneration. These further cause neuroinflammation, oxida­ Santos et al., 2019).
tive stress, activation of tauopathy, death of neurons, and eventually, Aβ and NFT are associated with activated microglia cells, markers of
impairment in cognition (Dá Mesquita et al., 2016). neuroinflammation (Dong et al., 2019; Dorszewska et al., 2016).
In the review by Medeiros et al. (2010), tau-protein was described as Elevated inflammatory cytokines like interleukin 6, 8 and tumor ne­
a protein associated with microtubules, found majorly in the axons crosis factor-α are released in response to glial activation as observed in
interacting with and aiding the assemblage of tubulin into the micro­ postmortem studies (Dubois et al., 2016; Duffy et al., 2018; Dutra et al.,
tubule for stabilization (Fig. 1) (Dai and Shen, 2021). Also reiterated was 2013). It has been suggested that these pro-inflammatory cytokine levels
that the phosphorylation of the protein prevents acute neuronal can be elevated by Aβ and tau-protein, according to in vitro studies
apoptosis by stabilizing β-catenin. Tauopathies in neurodegeneration (Edwards, 2019; El Khoury et al., 2007).
are a result of the alteration in the ratio of 3R and 4R tau (existing as 1:1 A study aiming to assess the early relationship between brain Aβ, tau-
in the normal human brain), leading to impairment in the ability of tau protein levels and neuroinflammation in cognitively normal older adults
to interact with microtubules, and there is abnormal phosphorylation of using a comparative analysis of cerebrospinal fluid inflammatory
tau residues (Dai and Shen, 2021). With the dysregulation of microtu­ markers and PET Aβ and tau-protein levels suggested, in agreement with
bule motor proteins, phosphatase, and kinase mechanisms, the hyper­ several lines of previous research, that AD hallmark proteins and neu­
phosphorylation of tau-protein, which upregulates its resistance to (and roinflammation share a dynamic relationship (Engelhardt et al., 2017).
degradation by ubiquitin-proteosome pathway) neutral proteases acti­ Several studies have reported that postmortem and cerebrospinal fluid
vated by calcium, results in fibrillization and accumulation into NFTs (CSF) markers of activated microglia are increased in patients with AD
(Apostolova, 2016; Ashutosh et al., 2011; Dani et al., 2018; Dansokho (Engelhardt et al., 2017; Erickson and Banks, 2013; Erickson et al.,
et al., 2016; Deczkowska and Schwartz, 2018; DeMaio et al., 2022). 2014; Fan et al., 2017).It has been suggested that CSF levels of both
There is evidence that tau-hyperphosphorylation is induced by Aβ, activated microglia and phosphorylated tau are associated (Fani et al.,
glucose impairment in the brain, and inflammation (Dai and Shen, 2021). At the same time, tau-protein and Aβ are usually colocalized with
2021). In Aβ-induced dysfunction in synapses, including dendrite dys­ these activated glial cells, evidenced in postmortem tissue, AD models,
regulation, abnormal tau-protein phosphorylation is an important and in vivo positron emission tomography studies (Engelhardt et al.,
mediator (De-Paula et al., 2012; DeTure and Dickson, 2019). With the 2017; Ferretti et al., 2016; Fiala et al., 2005; Fisher et al., 2014;
increase in Aβ, a sequence of events, which forms the basis of the tau Frackowiak et al., 1992; Fransen et al., 2020; Garbuz et al., 2021). This
hypothesis, is initiated. Hyperphosphorylation of tau-protein is induced, inference correlates with a positron emission tomography study of 43
and the hyperphosphorylated tau dissociates from microtubules, accu­ mild cognitive impaired (MCI) subjects over 2 years, which revealed that
mulates into NFTs and induces neuronal dysfunction and, ultimately, low baseline MCI cases with increasing Aβ load had correlated microglia
death of the neurons (Dionisio-Santos et al., 2019). The buildup of activation levels that subsequently decline as Aβ reaches AD levels
hyperphosphorylated tau correlates better with impairment in cognition (Dong et al., 2019). However, it was also reported that with the for­
compared to Aβ buildup, inferred from a study that showed a higher- mation of tau tangles in Aβ positive MCI subjects, there was a corre­
paced decline in cognition and dysfunctional synaptic plasticity in AD sponding increase in observed neuroinflammation (Dong et al., 2019).

3
M.O. Asamu et al. Brain Research 1821 (2023) 148589

Hence, with diverse research identifying neuroinflammation as a key worsens with disease progression (Guerreiro and Bras, 2015).
factor in the pathogenesis of AD, it gives new direction to explore in
therapeutic AD research (Gate et al., 2020). 5. Role of brain resident and peripheral innate immune cells in
neuroinflammation and AD
4. Vascular dysfunction and BBB disruption in AD
Microglia is the most abundant immune cell in the brain and con­
In the brain of AD patients, there are several reported structural and stitutes more than 80% of brain-resident immune cells. Microglia are
functional changes. The hippocampus and other brain areas involved in indispensable to cerebral circuit development and synaptic hemody­
spatial orientation and cognition have received significant research namics in the normal brain (Ittner and Götz, 2011; Ittner et al., 2010).
attention in the study of AD (Dionisio-Santos et al., 2019). Neocortical These cells play essential roles in presynaptic microenvironment im­
atrophy (which commences at the hippocampus and entorhinal cortex), mune surveillance and modulate phagocytic and proteolytic processes,
with increased synaptic and neuronal loss, is a marked pathological which control axonal and dendritic terminal pruning (Iturria-Medina
change observed in AD (Gatz et al., 2006). It spreads into the association et al., 2016). Through their numerous receptors, microglial cells sense
cortices in the temporal, parietal and frontal lobes. Atrophy of the brain, exogenous and endogenous insults to the CNS and respond by initiating
focusing on the medial temporal lobe and local changes in brain me­ immune processes. They also maintain brain homeostasis and the plas­
tabolites, are structural changes in observed AD (Gendelman and Mos­ ticity of neural pathways by remodeling synapses (Jahn, 2013). Micro­
ley, 2015; Goverman, 2009). glia carry out these processes by releasing trophic factors such as brain-
In addition to tau and Aβ aggregation, vascular dysfunctions derived neurotrophic factors (Janelidze et al., 2018). Microglia become
contribute to neuronal loss and decline in cognition apparent in AD activated, migrate to the lesion site and initiate an immune response
(Guerreiro and Bras, 2015). There are reports that vascular dysfunction when exposed to pathological triggers (protein aggregates or neuronal
promotes AD onset and progression (Hamelin et al., 2016). Vascular death) (Ji et al., 2013). Activated microglia takes one of two phenotypes:
biomarkers observed before marked cognitive decline in AD are a the pro-inflammatory (M1) phenotype that secretes pro-inflammatory
decrease in Aβ42 and an increase in tau levels in the CSF (Harry, 2013). cytokines such as interleukin-12, interleukin-1β, and tumor necrosis
The neurovascular unit comprises the blood–brain barrier (BBB), factor-α, or the anti-inflammatory (M2) phenotype that secretes anti-
smooth muscle cells in the walls of the blood vessels, pericytes, neurons, inflammatory cytokines such as interleukin-10, interleukin-13 and
and neuroglia (Hamelin et al., 2016). Decrease in the expression of interleukin-14 (Kawakami et al., 2004).
zonaoccludens protein 1 (ZO1) and occludin, a sealing protein molecule Microglia express immune-related receptors such as chemokine,
of tight junction that anchors it with the adjacent plasma membrane, in pattern recognition, and cytokine receptors, which interact with pro-
vascular endothelial cells progressively increases BBB permeability in inflammatory signals and activate microglia (Kebir et al., 2007).
AD (Heneka et al., 2015; Heneka et al., 2010; Herrera CA, Prince M, Several studies have linked microglia activation with the onset and
Knapp M, Karagiannidou M, and Guerchet M.World Alzheimer Report, progression of neurodegenerative disorders (Janelidze et al., 2018;
2016). BBB disruption at the hippocampus was observed in an MRI Kawakami et al., 2004; Kelly et al., 2013). Studies in experimental AD
image of a patient with mild cognitive decline (MCI) in a blood–brain- models have revealed microglia’s protective tendency in neuro­
CSF directional pathway of the contrast agent (Hickman et al., 2018). degeneration. These studies reported the ability of microglia to surround
In comparison, BBB disruption was observed before the atrophy of amyloid plaque through chemotaxis and clear the plaque by suppressing
the hippocampus (Höftberger et al., 2004). This observation suggests its growth and accumulation in extracellular spaces (Kessler et al., 1998;
that BBB disruption occurs before the observed neurodegenerative Kierdorf and Prinz, 2013). However, this effect is transient as the con­
decline in AD. Cerebral amyloid angiopathy (CAA), a vascular stant alteration in brain homeostasis characteristic of AD leads to the
dysfunction marked by the deposit of Aβ in cerebrovasculature, and accumulation of Aβ. Accumulated Aβ can gain resistance to microglia-
neuroinflammationare characteristics of AD pathology (Hamelin et al., induced elimination and degradation (Kivisäkk et al., 2006). The accu­
2016). Neuroinflammation is stimulated by microglial and macrophages mulated Aβ primes and activate microglia; activated microglia take an
of the innate immune cells, with contributions from the circulating inflammatory phenotype and amplify the ongoing neuroinflammation
immune cells (Hamelin et al., 2016). CAA is an important cause of BBB (Knezevic et al., 2018; Koch et al., 2016). The constant exposure to in­
disruption. Factors contributing to BBB disruption, like degeneration of flammatory factors and Aβ promotes the functional impairment of
endothelial cells, smooth muscle and pericytes, have been linked to CAA microglia in AD (Kessler et al., 1998). Accumulated Aβ initiates neuro­
(Hopperton et al., 2018). Aβ binds to receptors for advanced glycation inflammatory and neurodegeneration by priming microglia and sup­
end product in a process that produces reactive oxygen species and pressing the release of anti-inflammatory cytokines (Koch et al., 2016;
causes disruption in the tight junction between endothelial cells to Konjevic Sabolek et al., 2019).
disrupt the integrity of the BBB (Iqbal et al., 2009). Thus, it makes Aβ Microglia activation is a typical and key hallmark of the patho­
toxic to the endothelial cells of the brain, in vitro. Blair et al. (2015) physiology of AD (Koch et al., 2016; Konjevic Sabolek et al., 2019). In
posited that tau-protein dysfunction also contributes to BBB disruption. CK-p25 mice, the activation of microglia has been shown to correspond
The report further explains that the infiltration of perivascular tau- with neurodegeneration through single-cell level RNA sequencing
protein to the region of hippocampal blood vessels correlates with the (Korin et al., 2017). Through the upregulation of the expression of pro-
disruption of BBB (Ismail et al., 2020). Multiple neurodegenerative inflammatory factors such as macrophage migration inhibitory factor
pathways can be activated by disruption in the continuous pericyte and and tumor-necrosis factor-α, microglia enhanced inflammation as early
astrocyte sheathed mono-layered endothelium of the BBB, which will as Aβ production. This shows the potential of microglia activation dur­
permit the influx of neurotoxic cells and other molecules, including ing neurodegeneration to initiate and propagate neuronal loss and
microbes, debris, and others, involved in neuroinflammatory and im­ cognitive dysfunction (Korin et al., 2017). Chronic microglia activation
mune responses into the brain (Guerreiro and Bras, 2015). can also cause the accumulation of p-Tau within nerves (Kovac et al.,
Generally, in AD pathology, the disruption of BBB modulates the 2011). This accumulation of p-Tau initiates a form of positive feedback
clearance of Aβ; causes impairment of tight junction, pericyte, and in which the accumulation of p-Tau enhances the activation and pro-
endothelial cells; activates neuroglia cells; and permits the influx of inflammatory phenotype of microglia (Krishnan et al., 2019; Kumar
leukocytes, including cytotoxic T-cells, into the brain (Hamelin et al., et al., 2012). This process eventually activates a vicious cycle of neu­
2016). While this BBB breakdown ultimately results in inflammation, rodegeneration, neuroinflammation, pro-inflammatory activation of
damage to the neurons and synapses, and loss of neuronal connection. In microglia, and accumulation of p-Tau through the induction of neuronal
AD neuropathology, vascular dysfunction is a hallmark disease that p38-mediated mitogen-activated protein kinase signaling (Kuyumcu

4
M.O. Asamu et al. Brain Research 1821 (2023) 148589

et al., 2012). T cells can activate both microglia and astrocyte. T cell in astrocytes. J Immunol., 2010; Machado-Santos et al., 2018). The
participates in the activation of astrocyte and promotes neuro­ infiltration of monocytes into the brain is facilitated by C-C motif che­
inflammation. Interferon- γ secreted by T cells induces astrocyte pro­ mokine receptor 2 (CCR2). Blockade of the chemokine receptor has been
liferation and brain-reactive astrogliosis (Lassmann, 2018). Also, the shown to limit the protective effect of monocyte. In a transgenic mouse
interleukin-17 secreted by Th-17 cells activates astrocytes by regu­ model, the blockade of CCR2 increased Aβ accumulation and promoted
lating several signaling pathways in astrocytes. The cytokine controls memory loss (Machhi et al., 2021; Machhi et al., 2016). Monocytes are
the expression of macrophage inflammatory proteins-1α (MIP-1 α) crucial to the clearance of vascular Aβ in AD. Monocytes infiltrate the
through PI3K/Akt and NF-κB signaling pathways, stimulates the acti­ luminal walls of Aβ + veins, internalize Aβ, and return to circulation
vation of inducible nitric oxide synthase, and promotes the signaling of (Machhi et al., 2020). Evidence suggests a protective role of monocyte
interleukin-6 (Laurent et al., 2017; Laurent et al., 2018). and a therapeutic tendency in targeting monocyte. However, the pro-
Neutrophils which serve as the body’s first line of immune defense, inflammatory landscape of AD limits phagocytic capacity and directly
have been shown to infiltrate the brain of AD patients and animal impairs the monocyte’s ability to eliminate cerebral deposits of Aβ
models of AD (Leng et al., 2023; Li et al., 2009; Liu and Wang, 2009). (Machhi et al., 2017). This constraint has been observed in human
Neutrophil: lymphocyte ratio in peripheral blood has also been linked studies of AD where the Aβ uptake and phagocytosis capacity of pe­
with cognitive decline in AD (Lowther and Hafler, 2012; Lue et al., ripheral monocytes was impaired (Machhi et al., 2017).
2001). Neutrophils contribute to AD via several mechanisms. Neutro­
phils are involved in the BBB impairment, intravascular adhesion, and 6. Role of T lymphocytes in AD
brain infiltration characteristic of AD (Leng et al., 2023). Infiltrating
neutrophils produce interleukin-17, which promotes neurodegeneration The brain is immunologically active in the physiological state and
and BBB impairment (Lueg et al., 2015). Neutrophils also contribute to adequately protected by resident and infiltrating peripheral immune
neurotoxicity by forming neutrophils extracellular traps and producing cells (Ismail et al., 2020). Although present in low quantities at the
myeloperoxidase (Leng et al., 2023). The role of neutrophils in AD was physiological state, adaptive immune cells (T and B cells) can infiltrate
further buttressed when depletion of neutrophil infiltration was shown the meninges and reside in the dura (Fig. 2). These adaptive immune
to suppress microglial activation and amyloid plaque burden in mouse cells play critical roles in brain homeostasis and are involved in spatial
models of AD (5XFAD and 3xTg mice). This study also reported better learning and neuronal development (Malm et al., 2010). The cytokines
performance in the cognitive ability test – the Y-maze spontaneous (interleukin-4, interleukin-17, and interferon-γ) released by these cells
alternation task (Leng et al., 2023). play a crucial role in brain homeostasis (Malm et al., 2005). The adap­
Monocytes are another innate immune cell that has been implicated tive cells located in the choroid plexus, meninges, and other brain tissues
in AD pathogenesis. Monocytes have been shown to infiltrate the brain, serve a neuroprotective purpose or instigate neurodegeneration
suppress Aβ accumulation and improve cognitive function in transgenic depending on the local environment (Marco and Skaper, 2006; Masters
mouse models – APP/PS1 and 5xFAD (Ma X, Reynolds SL, Baker BJ, Li X, and Selkoe, 2012). Brain-resident immune cells, microglia, were initially
Benveniste EN, Qin H. IL-17 enhancement of the IL-6 signaling cascade thought to be solely responsible for the neuropathology of AD, but

Fig 2. The role of adaptive immune systems in brain homeostasis. There is homeostasis in healthy brain tissue. This homeostasis is characterized by a balance in the
activities of neurons, peripheral immune cells and brain resident immune cells (microglia and astrocytes). The brain resident immune cells are quiescent and are
called resting microglia and resting astrocytes. The peripheral immune cells such as antigen-presenting cells are also quiescent. Due to this balance, the brain is
devoid of inflammation and functions optimally (Ismail et al., 2020; Malm et al., 2010).

5
M.O. Asamu et al. Brain Research 1821 (2023) 148589

recent research has shown crosstalk between microglia and peripheral subtypes of CD4 + T cells participate in neurohomeostasis and
immune cells in AD neuropathology (Mathys et al., 2017; Mattson, contribute significantly to its maintenance. However, the subtypes play
2002; McGeer and McGeer, 2013; Medana et al., 2000; Medeiros et al., diverse roles during neurodegeneration, which may either facilitate
2011; Melzer et al., 2009). The interaction between innate and adaptive neuroprotection or neurodegeneration (Monteiro et al., 2016).
immunity is critical in neuroprotection, neuroinflammation, and neu­
rodegeneration (Medana et al., 2000). Microglia interacts with both the 6.1. Role of CD4 + T lymphocytes in AD
pro-inflammatory and anti-inflammatory arms of the adaptive immune
system, and this crosstalk fine-tunes the activities of microglia in return Both astrocytes and microglia are involved in the activation and
(Medana et al., 2000; Croese et al., 2021). differentiation of T cells. They regulate the differentiation of CD4 + T
As the adaptive immune system can facilitate both neuroprotection cells by modulating cytokines and molecules involved in the process.
and neuropathology, there is a need to balance its pro-inflammatory and They modulate the expression of molecules such as interleukin-6,
anti-inflammatory signaling to ensure brain homeostasis and protection. interleukin-10, interleukin-1, tumor necrosis factor-α, and trans­
While in the brain, T lymphocytes (CD4 + and CD8 + T cells) have two forming growth factor-β responsible for the differentiation of regulatory
fates. If the T cells fail to recognize an antigen and are not activated, they T cell (Treg) and effector T cells (Teff) (Morales et al., 2013; Morris et al.,
migrate back to the periphery through the CNS lymphatic system. Due to 2019). While Teff secretes pro-inflammatory cytokines such as inter­
their failure to recognize the cognate antigen, the T cells do not cross the feron-γ, granzyme B, and interleukin-17, which promote pro-
BBB and eventually undergo apoptosis (Mendez, 2019; Merlini et al., inflammatory microglia phenotypes, Treg secretes anti-inflammatory
2018). However, if they are activated by the binding of a cognate anti­ cytokines and promotes neuroprotection (Fig. 3) (Lueg et al., 2015;
gen with their receptor, they initiate an effector immune reaction Mount et al., 2007). Treg is crucial to immune tolerance. This subtype of
(Meuth et al., 2009; Michaud et al., 2013). The effector immune reaction T cells prevents effector immune action against a wide array of antigens
develops into a neuroinflammatory condition that alters the secretory such as self-antigens, environmental antigens, and bacterial antigens
ability and permeability of the BBB (Mietelska-Porowska and Wojda, (Mundt et al., 2019; Murphy and LeVine, 2010). In addition to sup­
2017). The compromised BBB alters cytokine responses and facilitates pressing effector immune action, Treg regulates the proliferation of Teff
the massive infiltration of immune cells into the brain, amplifying the via the secretion of immunosuppressive cytokines that initiates
ongoing neuroinflammation (Masters and Selkoe, 2012; Mittal et al., apoptosis and cytotoxicity.
2019; Mokhtar et al., 2013; Mondragón-Rodríguez et al., 2020). T cells Several studies and cumulative evidence have suggested the role of T
control neurohomeostasis via a cascade of secretory molecules and im­ lymphocytes in neurodegeneration, such as AD (Bartholomäus et al.,
mune signals (Montagne et al., 2015). At the physiological level, all 2009; Naert and Rivest, 2011; Nazem and Mansoori, 2008). Post-

Fig 3. The role of CD4 + T cells in Alzheimer’s disease. Activated antigen presenting cells present misfolded self-antigens to CD4 + T cells through major histo­
compatibility complex. Upon the recognition cognate antigen, CD4 + T cells differentiate into effector or regulatory subtype. The effector subtype, such as Th-1, Th-2
and Th-17, produced proinflammatory and neurotoxic cytokines that promote the transition of resting microglia into activated state. This phenomenon sustains and
enhances neuroinflammation and progression of Alzheimer’s disease. On the contrary, the regulatory subtype, Treg, produces anti-inflammatory cytokine to inhibit
antigen presentation and neuroinflammation (Mundt et al., 2019; Parbo et al., 2017; Parkhurst et al., 2013; Prinz and Priller, 2017).

6
M.O. Asamu et al. Brain Research 1821 (2023) 148589

mortem analysis of the brain of AD patients revealed a high number of (M2) phenotype. This process effectively suppresses neuroinflammation,
CD4 + and CD8 + T cells compared with control patients (Bartholomäus which is highly upregulated in AD (Rossi et al., 2011). Studies in mouse
et al., 2009; Naert and Rivest, 2011; Nazem and Mansoori, 2008). The models of AD have also shown the critical role played by CD4 + CD25 +
infiltration of a high number of T cells into the brain has been reported in Treg in the progression of AD (Rossi et al., 2021; Salvador et al., 2021).
animal models of AD with mutations of APP and Aβ (Nilson et al., 2017; Treg coordinates immune tolerance, modulates immune responses,
Nordengen et al., 2019; Oberstein et al., 2018). There was also an suppresses AD neurodegeneration, and regulates the response of
elevation in the expression of endothelial adhesion molecules, ICAM-1, microglia to amyloid deposition (Rizzo et al., 2018). Studies in mouse
and vascular cell adhesion protein 1 (VCAM-1) (Baruch et al., 2015; models of AD have shown that the immunosuppressive role of Treg also
Nordengen et al., 2019; Oberstein et al., 2018). A study conducted in the affects the phagocytic action of innate immune cells in the CNS and the
3xTg mouse model reported high expression of α4-integrins on the clearance of amyloid plaques (Rossi et al., 2021; Salvador et al., 2021).
surface of peripherally circulating CD4 + T cells compared to the wild In APP/PS1 mouse model, the depletion of Treg impaired the recruit­
type (Parbo et al., 2017). The α4β1 + CD4 + T cells were also shown to ment of microglia for the clearance of Aβ plaque and promoted the
infiltrate close to the VCAM-1 + cerebral vessels (Park and Kupper, associated cognitive impairment. The subsequent treatment with low-
2015). This shows the significance of α4β1 integrin expression and the dose peripheral administration of interleukin-2 restored the recruit­
signaling of VCAM to the infiltration of CD4 + T cells in the neuropa­ ment of microglia and improved cognitive capacity (Salvador et al.,
thology of AD. The signaling blockade between the two was also shown 2021).
to impair leukocyte adhesion to blood vessels in the brain and improve According to the study conducted by Baruch et al. 2015 there seems
cognitive capacity (Park and Kupper, 2015). The alteration in BBB seen to be a dichotomy between the role of Treg in brain tissues and those
in AD occurs during the early stages and facilitates the infiltration of all circulating in peripheral blood in CNS disorders (Rossi et al., 2021). The
subtypes of CD4 + T cells. The Treg infiltrating the brain confers neu­ study reported a decrease in the circulating Treg while those in brain
roprotection during the early stages of AD but becomes less potent as the tissues increased. However, the anti-inflammatory function of Treg in
disease advances. During the advanced stages of AD, Teff, such as helper AD was conserved. Based on the report from the study, the group sug­
T cell-1 (Th-1) and helper T cell-17 (Th-17), secrete interferon-γ and gested transient depletion or pharmacological inhibition of peripheral
interleukin-17, respectively. These cytokines reduce Treg’s immuno­ Treg to suppress neuroinflammation, reduce plaque formation, enhance
suppressive capacity and cause immune tolerance breakdown (Mundt Aβ clearance, and improve cognitive capacity (Rossi et al., 2021).
et al., 2019; Parbo et al., 2017; Parkhurst et al., 2013). Another study reported that depletion of peripheral circulating Treg
T cells also influence neuroinflammation by regulating the secretion impaired the recruitment of activated microglia to amyloid deposits but
of pro-inflammatory factors by myeloid cells such as dendritic cells and did not affect the clearance of Aβ by microglia (Salvador et al., 2021). In
macrophages. T cells control the secretion of tumor necrosis factor- α, the 5XFAD mouse model, the depletion of Treg promoted Aβ plaque
interleukin-1β, and interleukin-6 by these cells (Nilson et al., 2017). In clearance and recruitment of immune cells via the choroid plexus (Rossi
addition, T cells can initiate neuroinflammation directly in an MHC-II- et al., 2021). These studies and reports suggest that a reduction in the
based manner. To achieve this, parenchymal T cells are activated and population of circulating Treg may serve a neuroprotective function and
infiltrate the brain after conventional dendritic cells present myelin help to suppress the neuropathology of AD. There is a need for further
antigens to them (Pellicanò et al., 2012). The population of retinoic acid studies to explain the exact roles of Treg at each stage of AD neuropa­
receptor-related orphan nuclear receptor γt+ (RORγt + ) Th-17 T cells thology, the mechanisms employed for these effects, and the potential
increase significantly in AD compared to mild cognitive impairment and Treg-based therapeutic modulations.
age-matched healthy subjects (Perry and Holmes, 2014; Pietronigro Brain immune homeostasis and neuropathology during neuro­
et al., 2019; Pirker-Kees et al., 2013; Prinz et al., 2011). RORγt + Th-17 degeneration are controlled by the crosstalk and interaction between
cells majorly secrete interleukin-17, which has been shown to weaken microglia and T cells (Sarlus and Heneka, 2017). The crosstalk between
the tight junctions that form the BBB (Fig. 3). The compromised BBB has infiltrating CD4 + T cells and microglia has been reported to serve a
altered permeability and allows for the mass migration of peripheral crucial role in the immunoregulatory mechanisms involved in the
immune cells into the brain tissues. Upon infiltrating the brain, the neuropathology of AD (Fig. 3) (Baek et al., 2016; Saunders et al., 2012).
immune cells produce inflammatory cytokines such as tumor necrosis Aβ-specific Th-1 cells induce major histocompatibility complex class II
factor-α, interleukin-1β, and interleukin-6 that constitute an inflamma­ (MHC II) + microglia, suppressing AD neuropathology in the 5XFAD
tory response in the brain (Prinz and Priller, 2017). The report of studies mouse model (Saunders et al., 2012). This effect was proposed as
in mouse models of AD has shown that Aβ promotes the infiltration of interferon- γ signaling dependent (Fig. 3) (Saunders et al., 2012).
Th-17 cells into the brain (Prinz and Priller, 2017; Rahman and Lendel, However, Aβ-specific Th-1 and Th-17 cells have a contrasting effect on
2021; Ransohoff, 2016). More Th-17 cells infiltrated the hippocampus, microglia in APP/PS1 mouse model. In this setting, Th-1 and Th-17 cells
and there was a corresponding elevation in the expression of interleukin- increased β–amyloid plaque burden, neuroinflammation, microgliosis,
17 and interleukin-22 in the hippocampus, CSF, and peripheral blood and cognitive impairment (Baek et al., 2016). The effect seen in APP/
when Aβ was injected into the hippocampus (Prinz and Priller, 2017; PS1 mouse model may be due to the secretion of interferon-γ by Th-1
Rahman and Lendel, 2021; Ransohoff, 2016). The infiltration of Th-17 cells (Saunders et al., 2012). The interferon-γ secreted by Th-1 cells
cells also caused neuronal apoptosis (Ransohoff, 2016). can activate microglia and astrocytes, offsetting the brain’s immune
Due to its regulatory capacity, Treg has been linked to several neu­ homeostasis, promoting Aβ deposition, and impairing cognitive func­
roinflammatory and neurodegenerative diseases, including AD. How­ tions [142; 163]. These reports suggest that the role of Th-1 cells in AD is
ever, the role of Treg in neuroprotection and neurodegeneration is based on the disease stage, and this could mean that manipulating Th-1
controversial. Treg population is eit6her reduced or normal in AD and cells at a specific disease stage could be a viable therapeutic approach
has a proven neuroprotective function (Pirker-Kees et al., 2013; Prinz for AD.
et al., 2011; Reddy et al., 2018). Treg’s presence suppresses AD’s pro­ The infiltration of T cells into the AD brain correlates with the
gression, and its absence leads to an accelerated decline in cognitive abundance of p-tau. This has been observed in the medial frontal gyrus,
capacity (Fig. 3) (Reitz et al., 2020). The abundance of Treg has been inferior parietal lobule, and middle temporal gyrus of AD patients
shown to regulate the population and activities of microglia (Rizzo et al., (Schlüter et al., 2001). Tau-dependent infiltration of T cells has also been
2018). In APP/PS1 mice, a high level of circulating Treg slowed down seen in the frontotemporal gyrus and hippocampus (Schwartz et al.,
the decline in cognitive capacity and increased the microglia population 2022). These observations confirm Tau’s contribution to AD patho­
in the plaques. Treg also induced and promoted the conversion of physiology and the infiltration of T cells into the brain. However, the
microglia from pro-inflammatory (M1) phenotype to anti-inflammatory molecular mechanisms responsible for this process have not been

7
M.O. Asamu et al. Brain Research 1821 (2023) 148589

identified. initial lesions (Stadelmann et al., 2008). The initial tissue damage in AD,
caused by inflammatory CD4 + T cells and neurotoxic Aβ or phos­
6.2. Role of Cd8 + T lymphocytes in AD phorylated tau protein, is detected by myeloid cells, majorly microglia
(Sengupta et al., 1998). The microglia, in turn, become activated and
Analysis of AD in patients and mouse models has implicated CD8 + T upregulated; and eventually produce pro-inflammatory cytokines and
cells in the neuropathology of AD (Fig. 4) (Baruch et al., 2015; Nazem chemokines that recruit CD8 + T cells (Sengupta et al., 1998; Starr et al.,
and Mansoori, 2008; Prinz et al., 2011; Sengupta et al., 1998). CD8 + T 2009; Steinbach et al., 2018). While in the brain tissues, CD8 + T cells
cells are the predominant T cell in brain tissues associated with cognitive can interact with several cell types with high expression of MHC-I in AD
functions. The CD8 + T cells are near neuronal processes and microglia (Stojić-Vukanić et al., 2020; Stojiljkovic et al., 2019). CD8 + T cells can
(Kumar et al., 2012; Machhi et al., 2021). A crucial link has also been act directly on neurons and impair neuronal integrity through the
established between the infiltration of CD8 + T cells and tau pathology perforin-granzyme-dependent or Fas ligand-Fas receptor mechanisms
in experimental animal models and patients (Nazem and Mansoori, (Streit et al., 2004). They can also destroy myelin sheath and oligo­
2008; Schwartz et al., 2022). Infiltration of CD8 + T cells into the AD- dendrocytes, which compromises the neurons. This phenomenon is
affected brain tissues has been reported in both human and animal termed “collateral injury” (Sweeney et al., 2018; Taipa et al., 2018). It is
model studies and has been linked with disease progression (Fig. 4). worth noting that the activities of CD8 + T cells are also capable of a
These reports suggest the involvement of CD8 + T cells in AD neuro­ “spillover” effect. A phenomenon in which “spillover” of cytotoxic
pathology (Bartholomäus et al., 2009; Sheng et al., 2001). Higher per­ molecules from immunological synapses occurs after CD8 + T cells have
centage of HLA-DR + CD8 + T cells actively secreting pro-inflammatory attacked and killed off several target cells. This phenomenon leads to the
cytokines have been revealed in the blood immune profiling of AD pa­ unintended death of neighboring cells (Tang and Le, 2016).
tients compared with healthy people (Prinz et al., 2011; Shipton et al., Another mechanism of action of CD8 + T cells is the silencing of
2011). This suggests the activation of circulating CD8 + T cells in the neural electrical signaling. CD8 + T cells can cause massive perforin
blood of AD patients. insertion, forming an ion channel (Sweeney et al., 2018; Tiemessen
The CD8 + T cells observed during the analysis of AD-affected et al., 2007). This leads to an influx of Ca2+ ions and an elevation of
hippocampi possess granules containing granzyme A (Prinz et al., intracellular Ca2+ ion levels. The high intracellular Ca2+ ion level may
2011). The peripheral blood of patients with AD or AD-like mild neu­ be further coupled with the release of glutamate by the activated CD8 +
rocognitive disorder has been shown to have a high number of CD8 + T-cells (Sweeney et al., 2018; Tiemessen et al., 2007). CD8 + T cells can
TEMRA cells that produce either interferon-γ or tumor necrosis factor-α also cause neuronal damage and cognitive impairment by secretion of
(Prinz et al., 2011). Subsets of CD8 + T cells that produce interleukin-17 pro-inflammatory cytokines such as interferon-γ, tumor necrosis factor-
have also been detected in the peripheral blood of AD patients. These α, and interleukin-17. These cytokines are well capable of compromising
findings illustrate the active role of CD8 + T cells in AD pathogenesis and the BBB by impairing its permeability. With increased BBB permeability,
cognitive impairment (Smolders et al., 2018). One of the significant many T cells infiltrate the CNS parenchyma (Togo et al., 2002; Town
contributions of CD8 + T cells to neuropathology is the propagation of et al., 2005). The cytokines also upregulate MHC-I and sensitize

Fig 4. The role of CD8 + T cells in Alzheimer’s disease. Upon presentation of cognate antigen to CD8 + T cells, they become activated, differentiate and expand.
Activated CD8 + T cells produce proinflammatory cytokines; interact and activate microglia; and promote neuroinflammation (Sarlus and Heneka, 2017; Weiss
et al., 2011).

8
M.O. Asamu et al. Brain Research 1821 (2023) 148589

neuronal and non-neuronal cells to CD8 + T cells (Trajkovic et al., adhesion molecules (Baruch et al., 2015). During AD, T cells are one of
2001). They can also facilitate the direct killing of neuronal cells the predominant cells infiltrating the brain through pathologically
(Tröscher et al., 2019). In animal models of AD, these cytokines have altered BBB (Zhang et al., 2022). Several factors can promote the infil­
been shown to promote synaptic plasticity and cognitive impairment by tration of T cells into the brain during AD. One of the factors is dendritic
upregulating the release of glutamate and the expression or phosphor­ cells that promote brain infiltration of Aβ-specific T cells (Zhao et al.,
ylation of glutamate receptors (Unger et al., 2020; Unger et al., 2018; 2003). Aβ can also promote brain infiltration of T cells by initiating the
Vass and Lassmann, 1990). secretion of tumor necrosis factor-α by microglia and expression of
CD8 + T cells communicate directly with several subsets of CD11b + transforming growth factor-β1 by astrocytes (Zhao et al., 2018; Zotova
myeloid cells, including microglia (Villegas-Mendez et al., 2012). While et al., 2013). Excessive secretion of transforming growth factor-β1 by
the action of CD8 + T cells is mainly towards a target cell, CD11b + astrocytes can induce two contrasting effects. Transforming growth
myeloid cells secrete a wide array of pro-inflammatory molecules factor-β1 can suppress the accumulation of Aβ and plaque formation. On
(reactive oxygen and nitrogen, and cytokines inclusive), which leads to the contrary, transforming growth factor-β1 can promote brain
an attack on neighboring cells (which could include neurons) (Sarlus amyloidosis and enhance neuroinflammation (Zotova et al., 2013).
and Heneka, 2017; Steinbach et al., 2018; Villegas-Mendez et al., 2012;
Wagner et al., 2020). This interaction between CD8 + T cells and 7. Concluding remarks
microglia that leads to the latter secreting pro-inflammatory cytokines
has been established in chronic infection (Wattmo et al., 2016). Mouse There is ample proof that neuroinflammation plays a part in the
model of experimental autoimmune encephalomyelitis (EAE) has been development and progression of Alzheimer’s disease. Apart from the
used to show that CD8 + T cells employ Fas ligand-Fas receptor signaling roles of brain resident innate immune cells, microglia, in neuro­
to interact with monocytes/monocyte-derived cells infiltrating the brain inflammation, the adaptive immune system has also been implicated in
(Webers et al., 2020). This phenomenon could also occur between CD8 this process. Recent studies have shown the involvement of adaptive
+ T cells and microglia, leading to their activation and secretion of pro- immune cells, particularly T lymphocytes, in neuroinflammation and
inflammatory mediators (Weiss et al., 2011). The interaction between neurodegeneration. The presentation and binding of T lymphocytes with
CD8 + T cells and myeloid cells in the brain tissues could be said to be a cognate antigens lead to their activation and clonal expansion into
hotspot in brain immunopathologies. In AD, the presence of CD8 + T effector subsets of CD4 + and CD8 + T cells that secrete pro-
cells in the brain tissues could enhance the activation of microglia and, inflammatory cytokines. The secretion of these cytokines exacerbates
in turn, promote AD progression (Wood et al., 1993). neuroinflammation, which promotes several physiological and
The presence of tissue-resident memory cell-like CD8 + T cells in morphological changes in the brain. T cell-induced neuroinflammation
hippocampi and subcortical white matter of animal models of AD and alters the permeability of the BBB and promotes the buildup of NFT and
AD patients suggests another form of CD8 + T cells in AD (Baruch et al., amyloid plaques. Understanding the role of adaptive immune cells and
2015; Nazem and Mansoori, 2008; Wyatt-Johnson and Brutkiewicz, the interplay between adaptive immune cells and brain-resident innate
2020; Xie and Yang, 2015). Tissue-resident memory cells highly express immune cells in the pathogenesis and progression of AD will play a
inhibitory receptors, cytotoxic T-lymphocyte-associated protein-4 crucial role in developing immunotherapies for AD. Targeting immune-
(CTLA-4) and programmed cell death-1 (PD-1), and have a reduced suppressive and anti-inflammatory subsets, such as Treg, could improve
capacity to produce cytotoxic enzymes (Mount et al., 2007; Murphy and immunoregulation and neuroprotection while preventing neuro­
LeVine, 2010; Naert and Rivest, 2011; Nazem and Mansoori, 2008). inflammation and neurodegeneration. Also, the presence of inflamma­
However, these cells can produce pro-inflammatory cytokines and may tory mediators in CSF and blood; and changes in peripheral lymphocytes
induce the activation of myeloid cells when their inhibition is surpassed can be used as biomarkers to diagnose AD early and monitor its
by stimulatory signals (Wyatt-Johnson and Brutkiewicz, 2020; Yang progression.
et al., 2013; Yang et al., 2013; Yong et al., 1991). This means tissue- There is little information on each AD stage’s immune composition
resident memory cells can influence the activational and functional and signature. This lack of characterization of the immune signature of
status of microglia and contribute to AD progression. However, there is AD stages is mainly due to the inadequate recruitment and availability of
little evidence on this concept, and further research is needed to explain patients for such studies. To develop better biomarkers and immuno­
this phenomenon. therapy for AD, there is a need to understand the immune mediators
present at the early, moderate, and late AD stages. Understanding the
6.3. Role of T cell in BBB disruption activities of immune cells in AD has been challenging, and most of the
current information is obtained from mouse models. AD studies would
The infiltration of T cells through the BBB is controlled by cytokines, greatly benefit from developing model systems that recreate the activ­
chemokines, and their receptors. Endothelial cells control the movement ities of peripheral immune cells and vascular systems in human AD. Such
of cytokines and chemokines between the brain and blood (Zenaro et al., models could incorporate peripheral immune cells and vascular systems,
2015). In AD, the expression of high levels of chemokine receptors such single-cell imaging, patient-derived cells, and in vitro three-dimensional
as CXCR2, CCR5, and MIP-1 α leads to the movement of T cells through models with higher cellular complexity. Microfluidics, induced plurip­
the tight junctions of brain endothelium. This phenomenon was otent stem cell-derived microglia and multicellular human models could
confirmed in an animal model of AD where the accumulation of Aβ also be employed in dissecting immune signalling and cell interaction
upregulated the expression of RAGE and CCR5, which promoted T cell involved in neurodegeneration.
infiltration into the brain (Park and Kupper, 2015). At the initial stage of
brain infiltration, immune cells bind with selectins present on endo­ CRediT authorship contribution statement
thelial cells before crossing the endothelium and engaging chemokine
receptor CX3CR1 on endothelial cells. After engaging CX3CR1, immune Moses O. Asamu: Conceptualization, Writing – review & editing.
cells circulate against blood flow (Zenaro et al., 2017; Zhang et al., Oladapo O. Oladipo: Conceptualization, Writing – review & editing.
2013). The binding of immune cells with endothelial CXCR1, they Oluseun A. Abayomi: Writing and Proofreading. Afeez A. Adebayo:
adhere to the endothelium by activating integrins expressed on their Writing and Proofreading.
surface. After the adhesion, immune cells undergo transendothelial
migration or extravasation and move directly to the site of inflammation Declaration of Competing Interest
(Zenaro et al., 2017). This process is enhanced during AD as brain
amyloidosis activates and upregulates the expression of brain vessel The authors declare that they have no known competing financial

9
M.O. Asamu et al. Brain Research 1821 (2023) 148589

interests or personal relationships that could have appeared to influence pathology. Nature Communications 18 (6), 7967. https://doi.org/10.1038/
ncomms8967.
the work reported in this paper.
Baruch, K., Deczkowska, A., Rosenzweig, N., Tsitsou-Kampeli, A., Sharif, A.M.,
Matcovitch-Natan, O., Kertser, A., David, E., Amit, I., Schwartz, M., 2016. PD-1
Data availability immune checkpoint blockade reduces pathology and improves memory in mouse
models of alzheimer’s disease. Nature Medicine 22 (2), 135–137. https://doi.org/
10.1038/nm.4022.
No data was used for the research described in the article. Bateman, R.J., Xiong, C., Benzinger, T.L.S., Fagan, A.M., Goate, A., Fox, N.C., Marcus, D.
S., Cairns, N.J., Xie, X., Blazey, T.M., Holtzman, D.M., Santacruz, A., Buckles, V.,
Oliver, A., Moulder, K., Aisen, P.S., Ghetti, B., Klunk, W.E., McDade, E., Martins, R.
Acknowledgements
N., Masters, C.L., Mayeux, R., Ringman, J.M., Rossor, M.N., Schofield, P.R.,
Sperling, R.A., Salloway, S., Morris, J.C., 2012. dominantly inherited alzheimer
None. network. Clinical and biomarker changes in dominantly inherited alzheimer’s
disease. The New England Journal of Medicine 367 (9), 795–804.
Bettcher, B.M., Tansey, M.G., Dorothée, G., Heneka, M.T., 2021. Peripheral and central
Statements Declaration of interest immune system crosstalk in alzheimer disease - a research prospectus. Nature
Reviews. Neurology 17 (11), 689–701. https://doi.org/10.1038/s41582-021-00549-
x.
None. Blair, L.J., Frauen, H.D., Zhang, B., Nordhues, B.A., Bijan, S., Lin, Y.C., Zamudio, F.,
Hernandez, L.D., Sabbagh, J.J., Selenica, M.L., Dickey, C.A., 2015. Tau depletion
prevents progressive blood-brain barrier damage in a mouse model of tauopathy.
Funding Acta NeuropatholCommun. 31 (3), 8. https://doi.org/10.1186/s40478-015-0186-2.
Bondi, M.W., Edmonds, E.C., Salmon, D.P., 2017. Alzheimer’s disease: past, present, and
Not applicable. future. J IntNeuropsychol Soc. 23 (9–10), 818–831. https://doi.org/10.1017/
S135561771700100X.
Breijyeh, Z., Karaman, R., 2020. Comprehensive review on alzheimer’s disease: Causes
Ethical approval and consent to participate and treatment. Molecules 25 (24), 5789. https://doi.org/10.3390/
molecules25245789.
Browne TC, McQuillan K, McManus RM, O’Reilly JA, Mills KH, Lynch MA. IFN-γ
Not applicable. Production by amyloid β-specific Th1 cells promotes microglial activation and
increases plaque burden in a mouse model of Alzheimer’s disease. J Immunol. 2013
Mar 1;190(5):2241-51. doi: 10.4049/jimmunol.1200947.
Consent for publication Cao, W., Zheng, H., 2018. Peripheral immune system in aging and alzheimer’s disease.
Molecular Neurodegeneration 13 (1), 51. https://doi.org/10.1186/s13024-018-
0284-2.
All authors agree to the final manuscript. Carrano, A., Hoozemans, J.J., van der Vies, S.M., Rozemuller, A.J., van Horssen, J., de
Vries, H.E., 2011. amyloid beta induces oxidative stress-mediated blood-brain
References barrier changes in capillary amyloid angiopathy. Antioxidants & Redox Signaling 15
(5), 1167–1178. https://doi.org/10.1089/ars.2011.3895.
Carrano, A., Hoozemans, J.J., van der Vies, S.M., van Horssen, J., de Vries, H.E.,
Agnes, P., Christiane, S., Peter, D., 2013. T-Cells show increased production of cytokines
Rozemuller, A.J., 2012. Neuroinflammation and blood-brain barrier changes in
and activation markers in alzheimer’s disease. Brain. Disord. Ther. 3, 3–112.
capillary amyloid angiopathy. Neuro-Degenerative Diseases 10 (1–4), 329–331.
Albrecht, D.S., Sagare, A., Pachicano, M., Sweeney, M.D., Toga, A., Zlokovic, B., Chui, H.,
https://doi.org/10.1159/000334916.
Joe, E., Schneider, L., Morris, J.C., Benzinger, T., Pa, J., 2021. Early
Cavanagh, C., Wong, T.P., 2018. Preventing synaptic deficits in alzheimer’s disease by
neuroinflammation is associated with lower amyloid and tau levels in cognitively
inhibiting tumor necrosis factor alpha signaling. IBRO Reports 4, 18–21. https://doi.
normal older adults. Brain, Behavior, and Immunity 94, 299–307.
org/10.1016/j.ibror.2018.01.003.
Alonso Adel, C., Mederlyova, A., Novak, M., Grundke-Iqbal, I., Iqbal, K., 2004. Promotion
Cebrián, C., Loike, J.D., Sulzer, D., 2014. Neuronal MHC-I expression and its implications
of hyperphosphorylation by frontotemporal dementia tau mutations. The Journal of
in synaptic function, axonal regeneration and parkinson’s and other brain diseases.
Biological Chemistry 279 (33), 34873–34881. https://doi.org/10.1074/jbc.
Frontiers in Neuroanatomy 8, 114. https://doi.org/10.3389/fnana.2014.00114.
M405131200.
Chabran, E., Noblet, V., Loureiro de Sousa, P., Demuynck, C., Philippi, N., Mutter, C.,
Alonso, A., Zaidi, T., Novak, M., Grundke-Iqbal, I., Iqbal, K., 2001.
Anthony, P., Martin-Hunyadi, C., Cretin, B., Blanc, F., 2020. Changes in gray matter
Hyperphosphorylation induces self-assembly of tau into tangles of paired helical
volume and functional connectivity in dementia with lewy bodies compared to
filaments/straight filaments. ProcNatlAcadSci U S A. 98 (12), 6923–6928. https://
alzheimer’s disease and normal aging: implications for fluctuations. Alzheimer’s
doi.org/10.1073/pnas.121119298.
Research & Therapy 6;12(1):9. https://doi.org/10.1186/s13195-019-0575-z.
Amor, S., Woodroofe, M.N., 2014. innate and adaptive immune responses in
Cheng, X., He, P., Yao, H., Dong, Q., Li, R., Shen, Y., 2014. Occludin deficiency with
neurodegeneration and repair. Immunology 141 (3), 287–291. https://doi.org/
BACE1 elevation in cerebral amyloid angiopathy. Neurology 82 (19), 1707–1715.
10.1111/imm.12134.
https://doi.org/10.1212/WNL.0000000000000403.
Anderson, K.M., Olson, K.E., Estes, K.A., Flanagan, K., Gendelman, H.E., Mosley, R.L.,
Chui, R., Dorovini-Zis, K., 2010. Regulation of CCL2 and CCL3 expression in human brain
2014. Dual destructive and protective roles of adaptive immunity in
endothelial cells by cytokines and lipopolysaccharide. Journal of Neuroinflammation
neurodegenerative disorders. Transl Neurodegener. 3 (1), 25. https://doi.org/
7, 1. https://doi.org/10.1186/1742-2094-7-1.
10.1186/2047-9158-3-25.
Ciccocioppo, F., Lanuti, P., Pierdomenico, L., Simeone, P., Bologna, G., Ercolino, E.,
Apostolova, L.G., 2016. Alzheimer disease: CONTINUUM: Lifelong Learning in
Buttari, F., Fantozzi, R., Thomas, A., Onofrj, M., Centonze, D., Miscia, S.,
Neurology 22 (2, Dementia), 419–434.
Marchisio, M., 2019. The characterization of regulatory t-Cell profiles in alzheimer’s
Ashutosh, Kou, W., Cotter, R., Borgmann, K., Wu, L.i., Persidsky, R., Sakhuja, N.,
disease and multiple sclerosis. Scientific Reports 9 (1), 8788. https://doi.org/
Ghorpade, A., 2011. CXCL8 protects human neurons from amyloid-β-induced
10.1038/s41598-019-45433-3.
neurotoxicity: Relevance to alzheimer’s disease. Biochemical and Biophysical
Cipollini, V., Anrather, J., Orzi, F., Iadecola, C., 2019. Th17 and cognitive impairment:
Research Communications 412 (4), 565–571.
possible mechanisms of action. Frontiers in Immunology 13, 95. https://doi.org/
Ávila-Villanueva, M., Marcos Dolado, A., Gómez-Ramírez, J., Fernández-Blázquez, M.,
10.3389/fnana.2019.00095.
2022. Brain structural and functional changes in cognitive impairment due to
Cohen, J., Torres, C., 2019. Astrocyte senescence: Evidence and significance. Aging Cell
alzheimer’s disease. Frontiers in Psychology 21 (13), 886619. https://doi.org/
18 (3), e12937.
10.3389/fpsyg.2022.886619.
Crews, L., Rockenstein, E., Masliah, E., 2010. APP transgenic modeling of alzheimer’s
Baek, H., Ye, M., Kang, G.H., Lee, C., Lee, G., Choi, D.B., Jung, J., Kim, H., Lee, S., Kim, J.
disease: mechanisms of neurodegeneration and aberrant neurogenesis. Brain
S., Lee, H.J., Shim, I., Lee, J.H., Bae, H., 2016. Neuroprotective effects of CD4+CD25
StructFunct. 214 (2–3), 111–126. https://doi.org/10.1007/s00429-009-0232-6.
+Foxp3+ regulatory T cells in a 3xTg-AD alzheimer’s disease model. Oncotarget 7
Croese, T., Castellani, G., Schwartz, M., 2021. Immune cell compartmentalization for
(43), 69347–69357. https://doi.org/10.18632/oncotarget.12469.
brain surveillance and protection. Nature Immunology 22 (9), 1083–1092. https://
Baik, S.H., Cha, M.Y., Hyun, Y.M., Cho, H., Hamza, B., Kim, D.K., Han, S.H., Choi, H.,
doi.org/10.1038/s41590-021-00994-2.
Kim, K.H., Moon, M., Lee, J., Kim, M., Irimia, D., Mook-Jung, I., 2014. Migration of
Dá Mesquita, S., Ferreira, A.C., Sousa, J.C., Correia-Neves, M., Sousa, N., Marques, F.,
neutrophils targeting amyloid plaques in alzheimer’s disease mouse model.
2016. Insights on the pathophysiology of alzheimer’s disease: the crosstalk between
Neurobiology of Aging 35 (6), 1286–1292. https://doi.org/10.1016/j.
amyloid pathology, neuroinflammation and the peripheral immune system.
neurobiolaging.2014.01.003.
Neuroscience and Biobehavioral Reviews 68, 547–562. https://doi.org/10.1016/j.
Bartholomäus, I., Kawakami, N., Odoardi, F., Schläger, C., Miljkovic, D., Ellwart, J.W.,
neubiorev.2016.06.014.
Klinkert, W.E., Flügel-Koch, C., Issekutz, T.B., Wekerle, H., Flügel, A., 2009. Effector
Dai, L., Shen, Y., 2021. insights into t-cell dysfunction in alzheimer’s disease. Aging Cell
T cell interactions with meningeal vascular structures in nascent autoimmune CNS
20 (12), e13511.
lesions. Nature 462 (7269), 94–98. https://doi.org/10.1038/nature08478.
Dani, M., Wood, M., Mizoguchi, R., Fan, Z., Walker, Z., Morgan, R., Hinz, R., Biju, M.,
Baruch, K., Rosenzweig, N., Kertser, A., Deczkowska, A., Sharif, A.M., Spinrad, A.,
Kuruvilla, T., Brooks, D.J., Edison, P., 2018. Microglial activation correlates in vivo
Tsitsou-Kampeli, A., Sarel, A., Cahalon, L., Schwartz, M., 2015. Breaking immune
tolerance by targeting Foxp3(+) regulatory T cells mitigates alzheimer’s disease

10
M.O. Asamu et al. Brain Research 1821 (2023) 148589

with both tau and amyloid in alzheimer’s disease. Brain 141 (9), 2740–2754. Smolders, J., Huitinga, I., 2020. Tissue-resident memory T cells invade the brain
https://doi.org/10.1093/brain/awy188. PMID: 30052812. parenchyma in multiple sclerosis white matter lesions. Brain 143 (6), 1714–1730.
Dansokho, C., Ait Ahmed, D., Aid, S., Toly-Ndour, C., Chaigneau, T., Calle, V., https://doi.org/10.1093/brain/awaa117.
Cagnard, N., Holzenberger, M., Piaggio, E., Aucouturier, P., Dorothée, G., 2016. Garbuz DG, Zatsepina OG, Evgen’ev MB. [Beta Amyloid, Tau Protein, and
Regulatory T cells delay disease progression in alzheimer-like pathology. Brain 139 Neuroinflammation: An Attempt to Integrate Different Hypotheses of Alzheimer’s
(Pt 4), 1237–1251. https://doi.org/10.1093/brain/awv408. Disease Pathogenesis]. Mol Biol (Mosk). 2021 Sep-Oct;55(5):734-747. Russian. doi:
Deczkowska A, Schwartz M. Targeting neuro-immune communication in 10.31857/S0026898421050049. PMID: 34671002.
neurodegeneration: Challenges and opportunities. J Exp Med. 2018 Nov 5;215(11): Gate, D., Saligrama, N., Leventhal, O., Yang, A.C., Unger, M.S., Middeldorp, J., Chen, K.,
2702-2704. doi: 10.1084/jem.20181737. Lehallier, B., Channappa, D., De Los Santos, M.B., McBride, A., Pluvinage, J.,
DeMaio, A., Mehrotra, S., Sambamurti, K., Husain, S., 2022. the role of the adaptive Elahi, F., Tam, G.K., Kim, Y., Greicius, M., Wagner, A.D., Aigner, L., Galasko, D.R.,
immune system and T cell dysfunction in neurodegenerative diseases. Journal of Davis, M.M., Wyss-Coray, T., 2020. Clonally expanded CD8 T cells patrol the
Neuroinflammation 19 (1), 251. https://doi.org/10.1186/s12974-022-02605-9. cerebrospinal fluid in alzheimer’s disease. Nature 577 (7790), 399–404. https://doi.
De-Paula, V.J., Radanovic, M., Diniz, B.S., Forlenza, O.V., 2012. Alzheimer’s disease. org/10.1038/s41586-019-1895-7.
SubcellBiochem. 65, 329–352. https://doi.org/10.1007/978-94-007-5416-4_14. Gatz, M., Reynolds, C.A., Fratiglioni, L., Johansson, B., Mortimer, J.A., Berg, S., Fiske, A.,
DeTure, M.A., Dickson, D.W., 2019. The neuropathological diagnosis of alzheimer’s Pedersen, N.L., 2006. Role of genes and environments for explaining alzheimer
disease. MolNeurodegener. 14 (1), 32. https://doi.org/10.1186/s13024-019-0333-5. disease. Archives of General Psychiatry 63 (2), 168–174. https://doi.org/10.1001/
Dionisio-Santos, D.A., Olschowka, J.A., O’Banion, M.K., 2019. Exploiting microglial and archpsyc.63.2.168. PMID: 16461860.
peripheral immune cell crosstalk to treat alzheimer’s disease. Journal of Gendelman, H.E., Mosley, R.L., 2015. A perspective on roles played by innate and
Neuroinflammation 16, 74. https://doi.org/10.1186/s12974-019-1453-0. adaptive immunity in the pathobiology of neurodegenerative disorders. Journal of
Dong, Y., Benveniste, E.N., 2001. Immune function of astrocytes. Glia 36 (2), 180–190. Neuroimmune Pharmacology 10 (4), 645–650. https://doi.org/10.1007/s11481-
https://doi.org/10.1002/glia.1107. 015-9639-4.
Dong, X., Nao, J., Shi, J., Zheng, D., 2019. Predictive value of routine peripheral blood Goverman, J., 2009. Autoimmune T cell responses in the central nervous system. Nature
biomarkers in alzheimer’s disease. Frontiers in Aging Neuroscience 5 (11), 332. Reviews. Immunology 9 (6), 393–407. https://doi.org/10.1038/nri2550.
https://doi.org/10.3389/fnagi.2019.00332. Guerreiro, R., Bras, J., 2015. The age factor in alzheimer’s disease. Genome Medicine 20
Dorszewska, J., Prendecki, M., Oczkowska, A., Dezor, M., Kozubski, W., 2016. Molecular (7), 106. https://doi.org/10.1186/s13073-015-0232-5.
basis of familial and sporadic alzheimer’s disease. Current Alzheimer Research 13 Hamelin, L., Lagarde, J., Dorothée, G., Leroy, C., Labit, M., Comley, R.A., de Souza, L.C.,
(9), 952–963. https://doi.org/10.2174/1567205013666160314150501. PMID: Corne, H., Dauphinot, L., Bertoux, M., Dubois, B., Gervais, P., Colliot, O., Potier, M.
26971934. C., Bottlaender, M., Sarazin, M., 2016. Early and protective microglial activation in
Dubois, B., Hampel, H., Feldman, H.H., Scheltens, P., Aisen, P., Andrieu, S., alzheimer’s disease: a prospective study using 18 F-DPA-714 PET imaging. Brain 139
Bakardjian, H., Benali, H., Bertram, L., Blennow, K., Broich, K., Cavedo, E., (4), 1252–1264.
Crutch, S., Dartigues, J.-F., Duyckaerts, C., Epelbaum, S., Frisoni, G.B., Gauthier, S., Harry, G.J., 2013. Microglia during development and aging. Pharmacology &
Genthon, R., Gouw, A.A., Habert, M.-O., Holtzman, D.M., Kivipelto, M., Lista, S., Therapeutics 139 (3), 313–326. https://doi.org/10.1016/j.
Molinuevo, J.-L., O’Bryant, S.E., Rabinovici, G.D., Rowe, C., Salloway, S., pharmthera.2013.04.013.
Schneider, L.S., Sperling, R., Teichmann, M., Carrillo, M.C., Cummings, J., Jack, C. Heneka, M.T., O’Banion, M.K., Terwel, D., Kummer, M.P., 2010. Neuroinflammatory
R., 2016. Preclinical alzheimer’s disease: Definition, natural history, and diagnostic processes in alzheimer’s disease. Journal of Neural Transmission (Vienna) 117 (8),
criteria. alzheimer’s & Dementia 12 (3), 292–323. 919–947. https://doi.org/10.1007/s00702-010-0438-z.
Duffy, S.S., Keating, B.A., Perera, C.J., Moalem-Taylor, G., 2018. The role of regulatory T Heneka, M.T., Carson, M.J., Khoury, J.E., Landreth, G.E., Brosseron, F., Feinstein, D.L.,
cells in nervous system pathologies. Journal of Neuroscience Research 96 (6), Jacobs, A.H., Wyss-Coray, T., Vitorica, J., Ransohoff, R.M., Herrup, K., Frautschy, S.
951–968. https://doi.org/10.1002/jnr.24073. A., Finsen, B., Brown, G.C., Verkhratsky, A., Yamanaka, K., Koistinaho, J., Latz, E.,
Dutra, R.C., Moreira, E.L.G., Alberti, T.B., Marcon, R., Prediger, R.D., Calixto, J.B., 2013. Halle, A., Petzold, G.C., Town, T., Morgan, D., Shinohara, M.L., Perry, V.H.,
Spatial reference memory deficits precede motor dysfunction in an experimental Holmes, C., Bazan, N.G., Brooks, D.J., Hunot, S., Joseph, B., Deigendesch, N.,
autoimmune encephalomyelitis model: the role of kallikrein-kinin system. Brain, Garaschuk, O., Boddeke, E., Dinarello, C.A., Breitner, J.C., Cole, G.M., Golenbock, D.
Behavior, and Immunity 33, 90–101. https://doi.org/10.1016/j.bbi.2013.06.002. T., Kummer, M.P., 2015. Neuroinflammation in alzheimer’s disease. Lancet
Edwards, F.A., 2019. A unifying hypothesis for alzheimer’s disease: from plaques to Neurology 14 (4), 388–405.
neurodegeneration. Trends in Neurosciences 42 (5), 310–322. https://doi.org/ Herrera CA, Prince M, Knapp M, Karagiannidou M, and Guerchet M.World Alzheimer
10.1016/j.tins.2019.03.003. Report 2016: Improving healthcare for people living with dementia. Coverage,
El Khoury, J., Toft, M., Hickman, S.E., Means, T.K., Terada, K., Geula, C., Luster, A.D., quality and costs now and in the future. 2016 Sep. doi:10.13140/
2007. Ccr2 deficiency impairs microglial accumulation and accelerates progression RG.2.2.22580.04483.
of alzheimer-like disease. Nature Medicine 13 (4), 432–438. https://doi.org/ Hickman, S., Izzy, S., Sen, P., Morsett, L., El Khoury, J., 2018. Microglia in
10.1038/nm1555. neurodegeneration. Nature Neuroscience 21 (10), 1359–1369. https://doi.org/
Engelhardt, B., Vajkoczy, P., Weller, R.O., 2017. the movers and shapers in immune 10.1038/s41593-018-0242-x.
privilege of the CNS. Nature Immunology 18 (2), 123–131. https://doi.org/10.1038/ Höftberger, R., Aboul-Enein, F., Brueck, W., Lucchinetti, C., Rodriguez, M.,
ni.3666. Schmidbauer, M., Jellinger, K., Lassmann, H., 2004. Expression of major
Erickson, M.A., Banks, W.A., 2013. Blood-brain barrier dysfunction as a cause and histocompatibility complex class I molecules on the different cell types in multiple
consequence of alzheimer’s disease. Journal of Cerebral Blood Flow and Metabolism sclerosis lesions. Brain Pathology 14 (1), 43–50. https://doi.org/10.1111/j.1750-
33 (10), 1500–1513. https://doi.org/10.1038/jcbfm.2013.135. 3639.2004.tb00496.x.
Erickson, M.A., Morofuji, Y., Owen, J.B., Banks, W.A., 2014. Rapid transport of CCL11 Hopperton, K.E., Mohammad, D., Trépanier, M.O., Giuliano, V., Bazinet, R.P., 2018.
across the blood-brain barrier: regional variation and importance of blood cells. The Markers of microglia in post-mortem brain samples from patients with alzheimer’s
Journal of Pharmacology and Experimental Therapeutics 349 (3), 497–507. https:// disease: a systematic review. Molecular Psychiatry 23 (2), 177–198.
doi.org/10.1124/jpet.114.213074. Iqbal, K., Liu, F., Gong, C.X., Alonso Adel, C., Grundke-Iqbal, I., 2009. Mechanisms of
Fan, Z., Brooks, D.J., Okello, A., Edison, P., 2017. An early and late peak in microglial tau-induced neurodegeneration. Acta Neuropathologica 118 (1), 53–69. https://doi.
activation in alzheimer’s disease trajectory. Brain 140 (3), 792–803. https://doi.org/ org/10.1007/s00401-009-0486-3.
10.1093/brain/aww349. PMID: 28122877; PMCID: PMC5837520. Ismail, R., Parbo, P., Madsen, L.S., Hansen, A.K., Hansen, K.V., Schaldemose, J.L.,
Fani, L., Georgakis, M.K., Ikram, M.A., Ikram, M.K., Malik, R., Dichgans, M., 2021. Kjeldsen, P.L., Stokholm, M.G., Gottrup, H., Eskildsen, S.F., Brooks, D.J., 2020. The
Circulating biomarkers of immunity and inflammation, risk of alzheimer’s disease, relationships between neuroinflammation, beta-amyloid and tau deposition in
and hippocampal volume: a mendelian randomization study. Translational alzheimer’s disease: a longitudinal PET study. Journal of Neuroinflammation 17 (1),
Psychiatry 11 (1), 291. https://doi.org/10.1038/s41398-021-01400-z. 151. https://doi.org/10.1186/s12974-020-01820-6. PMID: 32375809; PMCID:
Ferretti, M.T., Merlini, M., Späni, C., Gericke, C., Schweizer, N., Enzmann, G., PMC7203856.
Engelhardt, B., Kulic, L., Suter, T., Nitsch, R.M., 2016. T-cell brain infiltration and Ittner, L.M., Götz, J., 2011. Amyloid-β and tau–a toxic pas de deux in alzheimer’s disease.
immature antigen-presenting cells in transgenic models of alzheimer’s disease-like Nature Reviews. Neuroscience 12 (2), 65–72. https://doi.org/10.1038/nrn2967.
cerebral amyloidosis. Brain, Behavior, and Immunity 54, 211–225. https://doi.org/ Ittner, L.M., Ke, Y.D., Delerue, F., Bi, M., Gladbach, A., van Eersel, J., Wölfing, H.,
10.1016/j.bbi.2016.02.009. Chieng, B.C., Christie, M.J., Napier, I.A., Eckert, A., Staufenbiel, M., Hardeman, E.,
Fiala M, Lin J, Ringman J, Kermani-Arab V, Tsao G, Patel A, Lossinsky AS, Graves MC, Götz, J., 2010. Dendritic function of tau mediates amyloid-beta toxicity in
Gustavson A, Sayre J, Sofroni E, Suarez T, Chiappelli F, Bernard G. Ineffective alzheimer’s disease mouse models. Cell 142 (3), 387–397. https://doi.org/10.1016/
phagocytosis of amyloid-beta by macrophages of Alzheimer’s disease patients. J j.cell.2010.06.036.
Alzheimers Dis. 2005 Jun;7(3):221-32; discussion 255-62. doi: 10.3233/jad-2005- Iturria-Medina, Y., Sotero, R.C., Toussaint, P.J., Mateos-Pérez, J.M., Evans, A.C.,
7304. Weiner, M.W., Aisen, P., Petersen, R., Jack, C.R., Jagust, W., Trojanowki, J.Q.,
Fisher Y, Strominger I, Biton S, Nemirovsky A, Baron R, Monsonego A. Th1 polarization Toga, A.W., Beckett, L., Green, R.C., Saykin, A.J., Morris, J., Shaw, L.M.,
of T cells injected into the cerebrospinal fluid induces brain immunosurveillance. J Khachaturian, Z., Sorensen, G., Kuller, L., Raichle, M., Paul, S., Davies, P., Fillit, H.,
Immunol. 2014 Jan 1;192(1):92-102. doi: 10.4049/jimmunol.1301707. Hefti, F., Holtzman, D., Mesulam, M.M., Potter, W., Snyder, P., Schwartz, A.,
Frackowiak, J., Wisniewski, H.M., Wegiel, J., Merz, G.S., Iqbal, K., Wang, K.C., 1992. Montine, T., Thomas, R.G., Donohue, M., Walter, S., Gessert, D., Sather, T.,
Ultrastructure of the microglia that phagocytose amyloid and the microglia that Jiminez, G., Harvey, D., Bernstein, M., Fox, N., Thompson, P., Schuff, N.,
produce beta-amyloid fibrils. Acta Neuropathologica 84 (3), 225–233. https://doi. Borowski, B., Gunter, J., Senjem, M., Vemuri, P., Jones, D., Kantarci, K., Ward, C.,
org/10.1007/BF00227813. Koeppe, R.A., Foster, N., Reiman, E.M., Chen, K., Mathis, C., Landau, S., Cairns, N.J.,
Fransen, N.L., Hsiao, C.C., van der Poel, M., Engelenburg, H.J., Verdaasdonk, K., Householder, E., Taylor-Reinwald, L., Lee, V., Korecka, M., Figurski, M.,
Vincenten, M.C.J., Remmerswaal, E.B.M., Kuhlmann, T., Mason, M.R.J., Hamann, J., Crawford, K., Neu, S., Foroud, T.M., Potkin, S., Shen, L.i., Faber, K., Kim, S., Nho, K.,

11
M.O. Asamu et al. Brain Research 1821 (2023) 148589

Thal, L., Buckholtz, N., Albert, M., Frank, R., Hsiao, J., Kaye, J., Quinn, J., Lind, B., Korin, B., Ben-Shaanan, T.L., Schiller, M., Dubovik, T., Azulay-Debby, H., Boshnak, N.T.,
Carter, R., Dolen, S., Schneider, L.S., Pawluczyk, S., Beccera, M., Teodoro, L., Koren, T., Rolls, A., 2017. High-dimensional, single-cell characterization of the
Spann, B.M., Brewer, J., Vanderswag, H., Fleisher, A., Heidebrink, J.L., Lord, J.L., brain’s immune compartment. Nature Neuroscience 20 (9), 1300–1309. https://doi.
Mason, S.S., Albers, C.S., Knopman, D., Johnson, K., Doody, R.S., Villanueva- org/10.1038/nn.4610.
Meyer, J., Chowdhury, M., Rountree, S., Dang, M., Stern, Y., Honig, L.S., Bell, K.L., Kovac A, Zilka N, Kazmerova Z, Cente M, Zilkova M, Novak M. Misfolded truncated
Ances, B., Carroll, M., Leon, S., Mintun, M.A., Schneider, S., Oliver, A., Marson, D., protein τ induces innate immune response via MAPK pathway. J Immunol. 2011 Sep
Griffith, R., Clark, D., Geldmacher, D., Brockington, J., Roberson, E., Grossman, H., 1;187(5):2732-9. doi: 10.4049/jimmunol.1100216. Epub 2011 Aug 3. PMID:
Mitsis, E., de Toledo-Morrell, L., Shah, R.C., Duara, R., Varon, D., Greig, M.T., 21813771.
Roberts, P., Albert, M., Onyike, C., D’Agostino, D., Kielb, S., Galvin, J.E., Krishnan, A., Kocab, A.J., Zacks, D.N., Marshak-Rothstein, A., GregoryKsander, M., 2019.
Cerbone, B., Michel, C.A., Rusinek, H., de Leon, M.J., Glodzik, L., De Santi, S., A small peptide antagonist of the fas receptor inhibits neuroinflammation and
Doraiswamy, P.M., Petrella, J.R., Wong, T.Z., Arnold, S.E., Karlawish, J.H., Wolk, D., prevents axon degeneration and retinal ganglion cell death in an inducible mouse
Smith, C.D., Jicha, G., Hardy, P., Sinha, P., Oates, E., Conrad, G., Lopez, O.L., model of glaucoma. Journal of Neuroinflammation 16, 184. https://doi.org/
Oakley, MaryAnn, Simpson, D.M., Porsteinsson, A.P., Goldstein, B.S., Martin, K., 10.1186/s12974-019-1576-3.
Makino, K.M., Ismail, M.S., Brand, C., Mulnard, R.A., Thai, G., Mc-Adams-Ortiz, C., Kumar, P., Pradhan, K., Karunya, R., Ambasta, R.K., Querfurth, H.W., 2012. Cross-
Womack, K., Mathews, D., Quiceno, M., Diaz-Arrastia, R., King, R., Weiner, M., functional E3 ligases parkin and c-terminus Hsp70-interacting protein in
Martin-Cook, K., DeVous, M., Levey, A.I., Lah, J.J., Cellar, J.S., Burns, J.M., neurodegenerative disorders. Journal of Neurochemistry 120 (3), 350–370. https://
Anderson, H.S., Swerdlow, R.H., Apostolova, L., Tingus, K., Woo, E., Silverman, D.H. doi.org/10.1111/j.1471-4159.2011.07588.x.
S., Lu, P.H., Bartzokis, G., Graff-Radford, N.R., Parfitt, F., Kendall, T., Johnson, H., Kuyumcu, M.E., Yesil, Y., Oztürk, Z.A., Kizilarslanoğlu, C., Etgül, S., Halil, M., Ulger, Z.,
Farlow, M.R., Hake, AnnMarie, Matthews, B.R., Herring, S., Hunt, C., van Dyck, C.H., Cankurtaran, M., Arıoğul, S., 2012. The evaluation of neutrophil-lymphocyte ratio in
Carson, R.E., MacAvoy, M.G., Chertkow, H., Bergman, H., Hosein, C., Black, S., alzheimer’s disease. Dementia and Geriatric Cognitive Disorders 34 (2), 69–74.
Stefanovic, B., Caldwell, C., Hsiung, G.-Y., Feldman, H., Mudge, B., Assaly, M., https://doi.org/10.1159/000341583.
Kertesz, A., Rogers, J., Bernick, C., Munic, D., Kerwin, D., Mesulam, M.-M., Lassmann, H., 2018. Multiple sclerosis pathology. Cold Spring Harbor Perspectives in
Lipowski, K., Wu, C.-K., Johnson, N., Sadowsky, C., Martinez, W., Villena, T., Medicine 8 (3), a028936.
Turner, R.S., Johnson, K., Reynolds, B., Sperling, R.A., Johnson, K.A., Marshall, G., Laurent, C., Dorothée, G., Hunot, S., Martin, E., Monnet, Y., Duchamp, M., Dong, Y.,
Frey, M., Lane, B., Rosen, A., Tinklenberg, J., Sabbagh, M.N., Belden, C.M., Légeron, F.P., Leboucher, A., Burnouf, S., Faivre, E., Carvalho, K., Caillierez, R.,
Jacobson, S.A., Sirrel, S.A., Kowall, N., Killiany, R., Budson, A.E., Norbash, A., Zommer, N., Demeyer, D., Jouy, N., Sazdovitch, V., Schraen-Maschke, S.,
Johnson, P.L., Allard, J., Lerner, A., Ogrocki, P., Hudson, L., Fletcher, E., Delarasse, C., Buée, L., Blum, D., 2017. Hippocampal T cell infiltration promotes
Carmichael, O., Olichney, J., DeCarli, C., Kittur, S., Borrie, M., Lee, T.-Y., Bartha, R., neuroinflammation and cognitive decline in a mouse model of tauopathy. Brain 140
Johnson, S., Asthana, S., Carlsson, C.M., Potkin, S.G., Preda, A., Nguyen, D., (1), 184–200. https://doi.org/10.1093/brain/aww270.
Tariot, P., Reeder, S., Bates, V., Capote, H., Rainka, M., Scharre, D.W., Kataki, M., Laurent, C., Buée, L., Blum, D., 2018. Tau and neuroinflammation: What impact for
Adeli, A., Zimmerman, E.A., Celmins, D., Brown, A.D., Pearlson, G.D., Blank, K., alzheimer’s disease and tauopathies? Biomed J. 41 (1), 21–33. https://doi.org/
Anderson, K., Santulli, R.B., Kitzmiller, T.J., Schwartz, E.S., Sink, K.M., 10.1016/j.bj.2018.01.003.
Williamson, J.D., Garg, P., Watkins, F., Ott, B.R., Querfurth, H., Tremont, G., Leng, F., Hinz, R., Gentleman, S., Hampshire, A., Dani, M., Brooks, D.J., Edison, P., 2023.
Salloway, S., Malloy, P., Correia, S., Rosen, H.J., Miller, B.L., Mintzer, J., Spicer, K., Neuroinflammation is independently associated with brain network dysfunction in
Bachman, D., Finger, E., Pasternak, S., Rachinsky, I., Drost, D., Pomara, N., alzheimer’s disease. Molecular Psychiatry 28 (3), 1303–1311.
Hernando, R., Sarrael, A., Schultz, S.K., Ponto, L.L.B., Shim, H., Smith, K.E., Li, M., Shang, D.S., Zhao, W.D., Tian, L., Li, B., Fang, W.G., Zhu, L., Man, S.M., Chen, Y.
Relkin, N., Chaing, G., Raudin, L., Smith, A., Fargher, K., Raj, B.A., Neylan, T., H., 2009. Amyloid beta interaction with receptor for advanced glycation end
Grafman, J., Davis, M., Morrison, R., Hayes, J., Finley, S., Friedl, K., Fleischman, D., products up-regulates brain endothelial CCR5 expression and promotes T cells
Arfanakis, K., James, O., Massoglia, D., Fruehling, J.J., Harding, S., Peskind, E.R., crossing the blood-brain barrier. Journal of Immunology 182 (9), 5778–5788.
Petrie, E.C., Li, G., Yesavage, J.A., Taylor, J.L., Furst, A.J., 2016. Alzheimer’s disease https://doi.org/10.4049/jimmunol.0803013.
neuroimaging initiative. Early role of vascular dysregulation on late-onset Liu, R., Wang, J.Z., 2009. Protein phosphatase 2A in alzheimer’s disease.
alzheimer’s disease based on multifactorial data-driven analysis. Nature Pathophysiology 16 (4), 273–277. https://doi.org/10.1016/j.
Communications 7 (1). https://doi.org/10.1038/ncomms11934. pathophys.2009.02.008.
Jahn, H., 2013. Memory loss in alzheimer’s disease. Dialogues ClinNeurosci. 15 (4), Lowther, D.E., Hafler, D.A., 2012. Regulatory T cells in the central nervous system.
445–454. https://doi.org/10.31887/DCNS.2013.15.4/hjahn. Immunology Reviews 248 (1), 156–169. https://doi.org/10.1111/j.1600-
Janelidze, S., Mattsson, N., Stomrud, E., Lindberg, O., Palmqvist, S., Zetterberg, H., 065X.2012.01130.x.
Blennow, K., Hansson, O., 2018. CSF biomarkers of neuroinflammation and Lue, L.F., Rydel, R., Brigham, E.F., Yang, L.B., Hampel, H., Murphy Jr, G.M.,
cerebrovascular dysfunction in early alzheimer disease. Neurology 91 (9), Brachova, L., Yan, S.D., Walker, D.G., Shen, Y., Rogers, J., 2001. inflammatory
e867–e877. repertoire of alzheimer’s disease and nondemented elderly microglia in vitro. Glia 35
Ji, K., Akgul, G., Wollmuth, L.P., Tsirka, S.E., Dunaevsky, A., 2013. Microglia actively (1), 72–79. https://doi.org/10.1002/glia.1072. PMID: 11424194.
regulate the number of functional synapses. PLoS One1 8 (2), e56293. Lueg, G., Gross, C.C., Lohmann, H., Johnen, A., Kemmling, A., Deppe, M., Groger, J.,
Kawakami, N.; Lassmann, S.; Li, Z.; Odoardi, F.; Ritter, T.; Ziemssen, T.; Klinkert, W.E.F.; Minnerup, J., Wiendl, H., Meuth, S.G., Duning, T., 2015. Clinical relevance of
Ellwart, J.W.; Bradl, M.; Krivacic, K.; et al. The Activation Status of Neuroantigen- specific t-cell activation in the blood and cerebrospinal fluid of patients with mild
Specific T Cells in the Target Organ Determines the Clinical Outcome of alzheimer’s disease. Neurobiology of Aging 36 (1), 81–89. https://doi.org/10.1016/
Autoimmune Encephalomyelitis. J. Exp. Med. 2004, 199, 185–197. doi: https://doi. j.neurobiolaging.2014.08.008.
org/10.1084/jem.20031064. Ma X, Reynolds SL, Baker BJ, Li X, Benveniste EN, Qin H. IL-17 enhancement of the IL-6
Kebir, H., Kreymborg, K., Ifergan, I., Dodelet-Devillers, A., Cayrol, R., Bernard, M., signaling cascade in astrocytes. J Immunol. 2010 May 1;184(9):4898-906. doi:
Giuliani, F., Arbour, N., Becher, B., Prat, A., 2007. Human TH17 lymphocytes 10.4049/jimmunol.1000142.
promote blood-brain barrier disruption and central nervous system inflammation. Machado-Santos, J., Saji, E., Tröscher, A.R., Paunovic, M., Liblau, R., Gabriely, G.,
Nature Medicine 13 (10), 1173–1175. https://doi.org/10.1038/nm1651. Bien, C.G., Bauer, J., Lassmann, H., 2018. the compartmentalized inflammatory
Kelly, R.J., Minogue, A.M., Lyons, A., Jones, R.S., Browne, T.C., Costello, D.A., response in the multiple sclerosis brain is composed of tissue-resident CD8+ T
Denieffe, S., O’Sullivan, C., Connor, T.J., Lynch, M.A., 2013. Glial activation in lymphocytes and B cells. Brain 141 (7), 2066–2082. https://doi.org/10.1093/brain/
AβPP/PS1 mice is associated with infiltration of IFNγ-Producing cells. Journal of awy151.
Alzheimer’s Disease 37 (1), 63–75. https://doi.org/10.3233/JAD-130539. Machhi, J., Sinha, A., Patel, P., Kanhed, A.M., Upadhyay, P., Tripathi, A., Parikh, Z.S.,
Kessler B, Hudrisier D, Schroeter M, Tschopp J, Cerottini JC, Luescher IF. Peptide Chruvattil, R., Pillai, P.P., Gupta, S., Patel, K., Giridhar, R., Yadav, M.R., 2016.
modification or blocking of CD8, resulting in weak TCR signaling, can activate CTL Neuroprotective potential of novel Multi-Targeted isoalloxazine derivatives in
for Fas- but not perforin-dependent cytotoxicity or cytokine production. J Immunol. rodent models of alzheimer’s disease through activation of canonical wnt/β-Catenin
(1998) 161:6939–46. signalling pathway. Neurotoxicity Research 29 (4), 495–513. https://doi.org/
Kierdorf, K., Prinz, M., 2013. Factors regulating microglia activation. Frontiers in 10.1007/s12640-016-9598-4. Epub 2016 Jan 21.
Cellular Neuroscience 23 (7), 44. https://doi.org/10.3389/fncel.2013.00044. Machhi, J., Prajapati, N., Tripathi, A., Parikh, Z.S., Kanhed, A.M., Patel, K., Pillai, P.P.,
Kivisäkk, P., Tucky, B., Wei, T., Campbell, J.J., Ransohoff, R.M., 2006. Human Giridhar, R., Yadav, M.R., 2017. Synthesis and biological evaluation of novel Multi-
cerebrospinal fluid contains CD4+ memory T cells expressing gut- or skin-specific target-Directed benzazepines against excitotoxicity. MolNeurobiol. 54 (9),
trafficking determinants: relevance for immunotherapy. BMC Immunology 7 (7), 14. 6697–6722. https://doi.org/10.1007/s12035-016-0184-9.
https://doi.org/10.1186/1471-2172-7-14. Machhi, J., Kevadiya, B.D., Muhammad, I.K., Herskovitz, J., Olson, K.E., Mosley, R.L.,
Knezevic, D., Verhoeff, N.P.LG., Hafizi, S., Strafella, A.P., Graff-Guerrero, A., Rajji, T., Gendelman, H.E., 2020. Harnessing regulatory T cell neuroprotective activities for
Pollock, B.G., Houle, S., Rusjan, P.M., Mizrahi, R., 2018. Imaging microglial treatment of neurodegenerative disorders. Molecular Neurodegeneration 15 (1), 32.
activation and amyloid burden in amnestic mild cognitive impairment. Journal of https://doi.org/10.1186/s13024-020-00375-7.
Cerebral Blood Flow and Metabolism 38 (11), 1885–1895. Machhi, J., Yeapuri, P., Lu, Y., Foster, E., Chikhale, R., Herskovitz, J., Namminga, K.L.,
Koch, G., Di Lorenzo, F., Del Olmo, M.F., Bonní, S., Ponzo, V., Caltagirone, C., Olson, K.E., Abdelmoaty, M.M., Gao, J., Quadros, R.M., Kiyota, T., Jingjing, L.,
Bozzali, M., Martorana, A., 2016. Reversal of LTP-Like cortical plasticity in Kevadiya, B.D., Wang, X., Liu, Y., Poluektova, L.Y., Gurumurthy, C.B., Mosley, R.L.,
alzheimer’s disease patients with Tau-Related faster clinical progression. Journal of Gendelman, H.E., 2021. CD4+ effector T cells accelerate alzheimer’s disease in mice.
Alzheimer’s Disease 50 (2), 605–616. https://doi.org/10.3233/JAD-150813. Journal of Neuroinflammation 18 (1), 272. https://doi.org/10.1186/s12974-021-
Konjevic Sabolek, M., Held, K., Beltrán, E., Niedl, A.G., Meinl, E., Hohlfeld, R., 02308-7.
Lassmann, H., Dornmair, K., 2019. Communication of CD8+ T cells with Malm, T.M., Koistinaho, M., Pärepalo, M., Vatanen, T., Ooka, A., Karlsson, S.,
mononuclear phagocytes in multiple sclerosis. Annals of Clinical Translational Koistinaho, J., 2005. Bone-marrow-derived cells contribute to the recruitment of
Neurology 6 (7), 1151–1164. https://doi.org/10.1002/acn3.783. microglial cells in response to beta-amyloid deposition in APP/PS1 double

12
M.O. Asamu et al. Brain Research 1821 (2023) 148589

transgenic alzheimer mice. Neurobiology of Disease 18 (1), 134–142. https://doi. Murphy, M.P., LeVine 3rd., H., 2010. Alzheimer’s disease and the amyloid-beta peptide.
org/10.1016/j.nbd.2004.09.009. Journal of Alzheimer’s Disease 19 (1), 311–323. https://doi.org/10.3233/JAD-
Malm, T., Koistinaho, M., Muona, A., Magga, J., Koistinaho, J., 2010. The role and 2010-1221.
therapeutic potential of monocytic cells in alzheimer’s disease. Glia 58 (8), 889–900. Naert, G., Rivest, S., 2011. CC chemokine receptor 2 deficiency aggravates cognitive
https://doi.org/10.1002/glia.20973. impairments and amyloid pathology in a transgenic mouse model of alzheimer’s
Marco, S., Skaper, S.D., 2006. Amyloid beta-peptide1-42 alters tight junction protein disease. The Journal of Neuroscience 31 (16), 6208–6220. https://doi.org/10.1523/
distribution and expression in brain microvessel endothelial cells. NeurosciLett. 401 JNEUROSCI.0299-11.2011.
(3), 219–224. https://doi.org/10.1016/j.neulet.2006.03.047. Nazem, A., Mansoori, G.A., 2008. Nanotechnology solutions for alzheimer’s disease:
Masters, C.L., Selkoe, D.J., 2012. Biochemistry of amyloid β-protein and amyloid deposits advances in research tools, diagnostic methods and therapeutic agents. Journal of
in alzheimer disease. Cold Spring HarbPerspect Med. 2 (6), a006262 https://doi.org/ Alzheimer’s Disease 13 (2), 199–223. https://doi.org/10.3233/jad-2008-13210.
10.1101/cshperspect.a006262. Nilson, A.N., English, K.C., Gerson, J.E., Barton Whittle, T., Nicolas Crain, C., Xue, J.,
Mathys, H., Adaikkan, C., Gao, F., Young, J.Z., Manet, E., Hemberg, M., De Jager, P.L., Sengupta, U., Castillo-Carranza, D.L., Zhang, W., Gupta, P., Kayed, R., 2017. Tau
Ransohoff, R.M., Regev, A., Tsai, L.H., 2017. Temporal tracking of microglia oligomers associate with inflammation in the brain and retina of tauopathy mice and
activation in neurodegeneration at Single-Cell resolution. Cell Reports 21 (2), in neurodegenerative diseases. Journal of Alzheimer’s Disease 55 (3), 1083–1099.
366–380. https://doi.org/10.1016/j.celrep.2017.09.039. https://doi.org/10.3233/JAD-160912. PMID: 27716675; PMCID: PMC5147514.
Mattson, M.P., 2002. Oxidative stress, perturbed calcium homeostasis, and immune Nordengen, K., Kirsebom, B.E., Henjum, K., Selnes, P., Gísladóttir, B., Wettergreen, M.,
dysfunction in alzheimer’s disease. Journal of Neurovirology 8 (6), 539–550. Torsetnes, S.B., Grøntvedt, G.R., Waterloo, K.K., Aarsland, D., Nilsson, L.N.G.,
https://doi.org/10.1080/13550280290100978. Fladby, T., 2019. Glial activation and inflammation along the alzheimer’s disease
McGeer, P.L., McGeer, E.G., 2013. The amyloid cascade-inflammatory hypothesis of continuum. Journal of Neuroinflammation 16 (1), 46. https://doi.org/10.1186/
alzheimer disease: implications for therapy. Acta Neuropathologica 126 (4), s12974-019-1399-2. PMID: 30791945; PMCID: PMC6383268.
479–497. https://doi.org/10.1007/s00401-013-1177-7. Oberstein, T.J., Taha, L., Spitzer, P., Hellstern, J., Herrmann, M., Kornhuber, J., Maler, J.
Medana, I.M., Gallimore, A., Oxenius, A., Martinic, M.M., Wekerle, H., Neumann, H., M., 2018. Imbalance of circulating Th17 and regulatory T cells in alzheimer’s
2000. MHC class i-restricted killing of neurons by virusspecific CD8+ T lymphocytes disease: A case control study. Frontiers in Immunology 4 (9), 1213. https://doi.org/
is effected through the Fas/FasL, but not the perforin pathway. European Journal of 10.3389/fimmu.2018.01213.
Immunology 30, 3623–3633. https://doi.org/10.1002/1521-4141(200012)30: Parbo, P., Ismail, R., Hansen, K.V., Amidi, A., Mårup, F.H., Gottrup, H., Brændgaard, H.,
12<3623::AID-IMMU3623>3.0.CO;2-F. Eriksson, B.O., Eskildsen, S.F., Lund, T.E., Tietze, A., Edison, P., Pavese, N.,
Medeiros, R., Baglietto-Vargas, D., LaFerla, F.M., 2011. The role of tau in alzheimer’s Stokholm, M.G., Borghammer, P., Hinz, R., Aanerud, J., Brooks, D.J., 2017. Brain
disease and related disorders. CNS NeurosciTher. 17 (5), 514–524. https://doi.org/ inflammation accompanies amyloid in the majority of mild cognitive impairment
10.1111/j.1755-5949.2010.00177.x. cases due to alzheimer’s disease. Brain 140 (7), 2002–2011. https://doi.org/
Melzer, N., Meuth, S.G., Wiendl, H., 2009. CD8+ T cells and neuronal damage: direct and 10.1093/brain/awx120. PMID: 28575151.
collateral mechanisms of cytotoxicity and impaired electrical excitability. The Park, C., Kupper, T., 2015. The emerging role of resident memory T cells in protective
FASEB Journal 23, 3659–3673. https://doi.org/10.1096/fj.09-136200. immunity and inflammatory disease. Nature Medicine 21, 688–697. https://doi.org/
Mendez, M.F., 2019. Early-onset alzheimer disease and its variants. Continuum 10.1038/nm.3883.
(MinneapMinn). 25 (1), 34–51. https://doi.org/10.1212/CON.0000000000000687. Parkhurst, C.N., Yang, G., Ninan, I., Savas, J.N., Yates 3rd, J.R., Lafaille, J.J.,
Merlini, M., Kirabali, T., Kulic, L., Nitsch, R.M., Ferretti, M.T., 2018. Extravascular CD3+ Hempstead, B.L., Littman, D.R., Gan, W.B., 2013. Microglia promote learning-
T cells in brains of alzheimer disease patients correlate with tau but not with amyloid dependent synapse formation through brain-derived neurotrophic factor. Cell 155
pathology: an immunohistochemical study. Neuro-Degenerative Diseases 18 (1), (7), 1596–1609. https://doi.org/10.1016/j.cell.2013.11.030.
49–56. https://doi.org/10.1159/000486200. Pellicanò, M., Larbi, A., Goldeck, D., Colonna-Romano, G., Buffa, S., Bulati, M.,
Meuth, S.G., Herrmann, A.M., Simon, O.J., Siffrin, V., Melzer, N., Bittner, S., Meuth, P., Rubino, G., Iemolo, F., Candore, G., Caruso, C., Derhovanessian, E., Pawelec, G.,
Langer, H.F., Hallermann, S., Boldakowa, N., Herz, J., Munsch, T., Landgraf, P., 2012. Immune profiling of alzheimer patients. Journal of Neuroimmunology 242
Aktas, O., Heckmann, M., Lessmann, V., Budde, T., Kieseier, B.C., Zipp, F., (1–2), 52–59. https://doi.org/10.1016/j.jneuroim.2011.11.005.
Wiendl, H., 2009. Cytotoxic CD8+ T cell-neuron interactions: perforin-dependent Perry, V.H., Holmes, C., 2014. Microglial priming in neurodegenerative disease. Nature
electrical silencing precedes but is not causally linked to neuronal cell death. The Reviews. Neurology 10 (4), 217–224. https://doi.org/10.1038/nrneurol.2014.38.
Journal of Neuroscience 29 (49), 15397–15409. https://doi.org/10.1523/ Pietronigro, E., Zenaro, E., Bianca, V.D., Dusi, S., Terrabuio, E., Iannoto, G., Slanzi, A.,
JNEUROSCI.4339-09.2009. Ghasemi, S., Nagarajan, R., Piacentino, G., Tosadori, G., Rossi, B., Constantin, G.,
Michaud, J.P., Bellavance, M.A., Préfontaine, P., Rivest, S., 2013. Real-time in vivo 2019. Blockade of α4 integrins reduces leukocyte-endothelial interactions in cerebral
imaging reveals the ability of monocytes to clear vascular amyloid beta. Cell Reports vessels and improves memory in a mouse model of alzheimer’s disease. Scientific
5 (3), 646–653. https://doi.org/10.1016/j.celrep.2013.10.010. Reports 9 (1), 12055. https://doi.org/10.1038/s41598-019-48538-x.
Mietelska-Porowska, A., Wojda, U., 2017. T lymphocytes and inflammatory mediators in Pirker-Kees, A., Schmied, C., Dal-Bianco, P., 2013. T-cells show increased production of
the interplay between brain and blood in alzheimer’s disease: Potential pools of new cytokines and activation markers in alzheimer’s disease. Brain Disord Ther. 3, 1.
biomarkers. Journal of Immunology Research 2017, 4626540. https://doi.org/ https://doi.org/10.4172/2168-975X.1000112.
10.1155/2017/4626540. Prinz, M., Priller, J., Sisodia, S.S., Ransohoff, R.M., 2011. Heterogeneity of CNS myeloid
Mittal, K., Eremenko, E., Berner, O., Elyahu, Y., Strominger, I., Apelblat, D., cells and their roles in neurodegeneration. Nature Neuroscience 14 (10), 1227–1235.
Nemirovsky, A., Spiegel, I., Monsonego, A., 2019. CD4 T Cells Induce A Subset of https://doi.org/10.1038/nn.2923.
MHCII-Expressing Microglia that Attenuates Alzheimer Pathology. iScience. 28 (16), Prinz, M., Priller, J., 2017. the role of peripheral immune cells in the CNS in steady state
298–311. https://doi.org/10.1016/j.isci.2019.05.039. and disease. Nature Neuroscience 20 (2), 136–144. https://doi.org/10.1038/
Mokhtar SH, Bakhuraysah MM, Cram DS, Petratos S. The Beta-amyloid protein of nn.4475.
Alzheimer’s disease: communication breakdown by modifying the neuronal Rahman, M.M., Lendel, C., 2021. Extracellular protein components of amyloid plaques
cytoskeleton. Int J Alzheimers Dis. 2013;2013:910502. doi: 10.1155/2013/910502. and their roles in alzheimer’s disease pathology. MolNeurodegener. 16 (1), 59.
Mondragón-Rodríguez, S., Salgado-Burgos, H., Peña-Ortega, F., 2020. Circuitry and https://doi.org/10.1186/s13024-021-00465-0.
synaptic dysfunction in alzheimer’s disease: A new tau hypothesis. Neural Plasticity Ransohoff, R.M., 2016. A polarizing question: do M1 and M2 microglia exist? Nature
1 (2020), 2960343. https://doi.org/10.1155/2020/2960343. Neuroscience 19 (8), 987–991. https://doi.org/10.1038/nn.4338.
Montagne, A., Barnes, S.R., Sweeney, M.D., Halliday, M.R., Sagare, A.P., Zhao, Z., Reddy, P.H., Yin, X., Manczak, M., Kumar, S., Pradeepkiran, J.A., Vijayan, M., Reddy, A.
Toga, A.W., Jacobs, R.E., Liu, C.Y., Amezcua, L., Harrington, M.G., Chui, H.C., P., 2018. Mutant APP and amyloid beta-induced defective autophagy, mitophagy,
Law, M., Zlokovic, B.V., 2015. Blood-brain barrier breakdown in the aging human mitochondrial structural and functional changes and synaptic damage in
hippocampus. Neuron 85 (2), 296–302. https://doi.org/10.1016/j. hippocampal neurons from alzheimer’s disease. Human Molecular Genetics 27 (14),
neuron.2014.12.032. 2502–2516. https://doi.org/10.1093/hmg/ddy154.
Monteiro, S., Ferreira, F.M., Pinto, V., Roque, S., Morais, M., de Sá-Calçada, D., Mota, C., Reitz, C., Rogaeva, E., Beecham, G.W., 2020. Late-onset vsnonmendelian early-onset
Correia-Neves, M., Cerqueira, J.J., 2016. Absence of IFNγ promotes hippocampal alzheimer disease: A distinction without a difference? Neurol Genet. 6 (5), e512.
plasticity and enhances cognitive performance. Translational Psychiatry 6 (1), e707. Rizzo, F.R., Musella, A., De Vito, F., Fresegna, D., Bullitta, S., Vanni, V., Guadalupi, L.,
Morales, I., Jiménez, J.M., Mancilla, M., Maccioni, R.B., 2013. Tau oligomers and fibrils Stampanoni Bassi, M., Buttari, F., Mandolesi, G., Centonze, D., Gentile, A., 2018.
induce activation of microglial cells. Journal of Alzheimer’s Disease 37 (4), 849–856. Tumor necrosis factor and Interleukin-1β modulate synaptic plasticity during
https://doi.org/10.3233/JAD-131843. neuroinflammation. Neural Plasticity 14 (2018), 8430123. https://doi.org/10.1155/
Morris, G., Berk, M., Maes, M., Puri, B.K., 2019. Could alzheimer’s disease originate in 2018/8430123.
the periphery and if so how so? Molecular Neurobiology 56 (1), 406–434. https:// Rossi B, Angiari S, Zenaro E, Budui SL, Constantin G. Vascular inflammation in central
doi.org/10.1007/s12035-018-1092-y. nervous system diseases: adhesion receptors controlling leukocyte-endothelial
Mount, M.P., Lira, A., Grimes, D., Smith, P.D., Faucher, S., Slack, R., Anisman, H., interactions. J Leukoc Biol. 2011 Apr;89(4):539-56. doi: 10.1189/jlb.0710432.
Hayley, S., Park, D.S., 2007. involvement of interferon-gamma in microglial- Rossi, B., Santos-Lima, B., Terrabuio, E., Zenaro, E., Constantin, G., 2021. Common
mediated loss of dopaminergic neurons. The Journal of Neuroscience 27 (12), peripheral immunity mechanisms in multiple sclerosis and alzheimer’s disease.
3328–3337. https://doi.org/10.1523/JNEUROSCI.5321-06.2007. Frontiers in Immunology 19 (12), 639369. https://doi.org/10.3389/
Mundt, S., Mrdjen, D., Utz, S.G., Greter, M., Schreiner, B., Becher, B., 2019. Conventional fimmu.2021.639369.
DCs sample and present myelin antigens in the healthy CNS and allow parenchymal Salvador, A.F., de Lima, K.A., Kipnis, J., 2021. Neuromodulation by the immune system:
T cell entry to initiate neuroinflammation. Science Immunology. https://doi.org/ a focus on cytokines. Nature Reviews. Immunology 21 (8), 526–541. https://doi.
10.1126/sciimmunol.aau8380. Jan 25;4(31):eaau8380. org/10.1038/s41577-021-00508-z.
Sarlus, H., Heneka, M.T., 2017. Microglia in alzheimer’s disease. The Journal of Clinical
Investigation 127 (9), 3240–3249.

13
M.O. Asamu et al. Brain Research 1821 (2023) 148589

Saunders, J.A., Estes, K.A., Kosloski, L.M., Allen, H.E., Dempsey, K.M., Torres- at sites of amyloid-β plaques: A potential role in shaping plaque pathology?
Russotto, D.R., Meza, J.L., Santamaria, P.M., Bertoni, J.M., Murman, D.L., Ali, H.H., Alzheimer’s & Dementia 14 (8), 1022–1037. https://doi.org/10.1016/j.
Standaert, D.G., Mosley, R.L., Gendelman, H.E., 2012. CD4+ regulatory and jalz.2018.02.017.
effector/memory T cell subsets profile motor dysfunction in parkinson’s disease. Unger, M.S., Li, E., Scharnagl, L., Poupardin, R., Altendorfer, B., Mrowetz, H., Hutter-
Journal of Neuroimmune Pharmacology 7 (4), 927–938. https://doi.org/10.1007/ Paier, B., Weiger, T.M., Heneka, M.T., Attems, J., Aigner, L., 2020. CD8+ t-cells
s11481-012-9402-z. infiltrate alzheimer’s disease brains and regulate neuronal- and synapse-related gene
Schlüter, D., Meyer, T., Strack, A., Reiter, S., Kretschmar, M., Wiestler, O.D., Hof, H., expression in APP-PS1 transgenic mice. Brain, Behavior, and Immunity 89, 67–86.
Deckert, M., 2001. Regulation of microglia by CD4+ and CD8+ T cells: selective https://doi.org/10.1016/j.bbi.2020.05.070.
analysis in CD45-congenic normal and toxoplasma gondii-infected bone marrow Vass, K., Lassmann, H., 1990. Intrathecal application of interferon gamma. Progressive
chimeras. Brain Pathology 11 (1), 44–55. https://doi.org/10.1111/j.1750- appearance of MHC antigens within the rat nervous system. AmJ Pathol. 137,
3639.2001.tb00380. 789–800.
Schwartz, M., Abellanas, M.A., Tsitsou-Kampeli, A., Suzzi, S., 2022. The brain-immune Villegas-Mendez A, Greig R, Shaw TN, de Souza JB, Gwyer Findlay E, Stumhofer JS,
ecosystem: Implications for immunotherapy in defeating neurodegenerative Hafalla JC, Blount DG, Hunter CA, Riley EM, Couper KN. IFN-γ-producing CD4+ T
diseases. Neuron 110 (21), 3421–3424. https://doi.org/10.1016/j. cells promote experimental cerebral malaria by modulating CD8+ T cell
neuron.2022.09.007. accumulation within the brain. J Immunol. 2012 Jul 15;189(2):968-79. doi:
Sengupta, A., Kabat, J., Novak, M., Wu, Q., Grundke-Iqbal, I., Iqbal, K., 1998. 10.4049/jimmunol.1200688.
Phosphorylation of tau at both thr 231 and ser 262 is required for maximal inhibition Wagner, C.A., Roqué, P.J., Mileur, T.R., Liggitt, D., Goverman, J.M., 2020. Myelin-
of its binding to microtubules. Arch BiochemBiophys. 357 (2), 299–309. https://doi. specific CD8+ T cells exacerbate brain inflammation in CNS autoimmunity. The
org/10.1006/abbi.1998.0813. Journal of Clinical Investigation 130, 203–213. https://doi.org/10.1172/
Sheng, J.G., Jones, R.A., Zhou, X.Q., McGinness, J.M., Van Eldik, L.J., Mrak, R.E., JCI132531.
Griffin, W.S.T., 2001. interleukin-1 promotion of MAPK-p38 overexpression in Wattmo, C., Minthon, L., Wallin, Å.K., 2016. Mild versus moderate stages of alzheimer’s
experimental animals and in alzheimer’s disease: potential significance for tau disease: three-year outcomes in a routine clinical setting of cholinesterase inhibitor
protein phosphorylation. Neurochemistry International 39 (5-6), 341–348. therapy. Alzheimer’s Research & Therapy 17 (8), 7. https://doi.org/10.1186/
Shipton, O.A., Leitz, J.R., Dworzak, J., Acton, C.E., Tunbridge, E.M., Denk, F., s13195-016-0174-1.
Dawson, H.N., Vitek, M.P., Wade-Martins, R., Paulsen, O., Vargas-Caballero, M., Webers, A., Heneka, M.T., Gleeson, P.A., 2020. The role of innate immune responses and
2011. Tau protein is required for amyloid {beta}-induced impairment of neuroinflammation in amyloid accumulation and progression of alzheimer’s disease.
hippocampal long-term potentiation. The Journal of Neuroscience 31 (5), Immunology and Cell Biology 98 (1), 28–41. https://doi.org/10.1111/imcb.12301.
1688–1692. https://doi.org/10.1523/JNEUROSCI.2610-10.2011. Weiss, R., Lifshitz, V., Frenkel, D., 2011. TGF-β1 affects endothelial cell interaction with
Smolders, J., Heutinck, K.M., Fransen, N.L., Remmerswaal, E.B.M., Hombrink, P., Ten macrophages and T cells leading to the development of cerebrovascular amyloidosis.
Berge, I.J.M., van Lier, R.A.W., Huitinga, I., Hamann, J., 2018. Tissue-resident Brain, Behavior, and Immunity 25 (5), 1017–1024. https://doi.org/10.1016/j.
memory T cells populate the human brain. Nature Communications 9 (1), 4593. bbi.2010.11.012.
https://doi.org/10.1038/s41467-018-07053-9. Wood, J.A., Wood, P.L., Ryan, R., Graff-Radford, N.R., Pilapil, C., Robitaille, Y.,
Stadelmann, C., Albert, M., Wegner, C., Bruck, W., 2008. Cortical pathology in multiple Quirion, R., 1993. Cytokine indices in alzheimer’s temporal cortex: no changes in
sclerosis. Current Opinion in Neurology 21, 229–234. https://doi.org/10.1097/01. mature IL-1 beta or IL-1RA but increases in the associated acute phase proteins IL-6,
wco.0000318863.65635.9a. alpha 2-macroglobulin and c-reactive protein. Brain Research 629 (2), 245–252.
Starr, J.M., Farrall, A.J., Armitage, P., McGurn, B., Wardlaw, J., 2009. Blood-brain https://doi.org/10.1016/0006-8993(93)91327-o. PMID: 7509248.
barrier permeability in alzheimer’s disease: a case-control MRI study. Psychiatry Wyatt-Johnson, S.K., Brutkiewicz, R.R., 2020. The complexity of microglial interactions
Research 171 (3), 232–241. https://doi.org/10.1016/j.pscychresns.2008.04.003. with innate and adaptive immune cells in alzheimer’s disease. Frontiers in Aging
Steinbach, K., Vincenti, I., Merkler, D., 2018. Tissue-restricted immune residentmemory Neuroscience 19 (12), 592359. https://doi.org/10.3389/fnagi.2020.592359.
T cells in responses: for better or worse? Frontiers in Immunology 9, 2827. https:// Xie, L., Yang, S.H., 2015. interaction of astrocytes and T cells in physiological and
doi.org/10.3389/fimmu.2018.02827. pathological conditions. Brain Research 14 (1623), 63–73. https://doi.org/10.1016/
Stojić-Vukanić, Z., Hadžibegović, S., Nicole, O., Nacka-Aleksić, M., Leštarević, S., j.brainres.2015.03.026.
Leposavić, G., 2020. CD8+ T Cell-Mediated mechanisms contribute to the 2022 Alzheimer’s disease facts and figures. Alzheimers Dement. 2022 Apr;18(4):700-
progression of neurocognitive impairment in both multiple sclerosis and alzheimer’s 789. doi: 10.1002/alz.12638. Epub 2022 Mar 14.
disease? Frontiers in Immunology 19 (11), 566225. https://doi.org/10.3389/ Yang, Y.M., Shang, D.S., Zhao, W.D., Fang, W.G., Chen, Y.H., 2013. Microglial TNF-
fimmu.2020.566225. α-dependent elevation of MHC class I expression on brain endothelium induced by
Stojiljkovic, M.R., Ain, Q., Bondeva, T., Heller, R., Schmeer, C., Witte, O.W., 2019. amyloid-beta promotes T cell transendothelial migration. Neurochemical Research
Phenotypic and functional differences between senescent and aged murine 38 (11), 2295–2304. https://doi.org/10.1007/s11064-013-1138-5.
microglia. Neurobiology of Aging 74, 56–69. https://doi.org/10.1016/j. Yang, H., Yang, H., Xie, Z., Wei, L., Bi, J., Fiorina, P., 2013. Systemic transplantation of
neurobiolaging.2018.10.007. human umbilical cord derived mesenchymal stem cells-educated T regulatory cells
Streit, W.J., Sammons, N.W., Kuhns, A.J., Sparks, D.L., 2004. Dystrophic microglia in the improved the impaired cognition in AβPPswe/PS1dE9 transgenic mice. PLoS One1 8
aging humn brain. Glia 45 (2), 208–212. https://doi.org/10.1002/glia.10319. (7), e69129.
Sweeney, M.D., Sagare, A.P., Zlokovic, B.V., 2018. Blood-brain barrier breakdown in Yong, V.W., Moumdjian, R., Yong, F.P., Ruijs, T.C., Freedman, M.S., Cashman, N.,
alzheimer disease and other neurodegenerative disorders. Nature Reviews. Antel, J.P., 1991. Gamma-interferon promotes proliferation of adult human
Neurology 14 (3), 133–150. https://doi.org/10.1038/nrneurol.2017.188. astrocytes in vitro and reactive gliosis in the adult mouse brain in vivo. Proceedings
Taipa, R., Ferreira, V., Brochado, P., Robinson, A., Reis, I., Marques, F., Mann, D.M., of the National Academy of Sciences of the United States of America 88 (16),
Melo-Pires, M., Sousa, N., 2018. inflammatory pathology markers (activated 7016–7020. https://doi.org/10.1073/pnas.88.16.7016.
microglia and reactive astrocytes) in early and late onset alzheimer disease: a post Zenaro, E., Pietronigro, E., Della Bianca, V., Piacentino, G., Marongiu, L., Budui, S.,
mortem study. Neuropathology and Applied Neurobiology 44 (3), 298–313. https:// Turano, E., Rossi, B., Angiari, S., Dusi, S., Montresor, A., Carlucci, T., Nanì, S.,
doi.org/10.1111/nan.12445. Epub 2017 Nov 27 PMID: 29044639. Tosadori, G., Calciano, L., Catalucci, D., Berton, G., Bonetti, B., Constantin, G., 2015.
Tang, Y., Le, W., 2016. Differential roles of M1 and M2 microglia in neurodegenerative Neutrophils promote alzheimer’s disease-like pathology and cognitive decline via
diseases. Molecular Neurobiology 53 (2), 1181–1194. https://doi.org/10.1007/ LFA-1 integrin. Nature Medicine 21 (8), 880–886. https://doi.org/10.1038/
s12035-014-9070-5. nm.3913.
Tiemessen, M.M., Jagger, A.L., Evans, H.G., van Herwijnen, M.J., John, S., Taams, L.S., Zenaro, E., Piacentino, G., Constantin, G., 2017. The blood-brain barrier in alzheimer’s
2007. CD4+CD25+Foxp3+ regulatory T cells induce alternative activation of disease. Neurobiology of Disease 107, 41–56. https://doi.org/10.1016/j.
human monocytes/macrophages. Proceedings of the National Academy of Sciences nbd.2016.07.007.
of the United States of America 104 (49), 19446–19451. https://doi.org/10.1073/ Zhang, J., Ke, K.-F., Liu, Z., Qiu, Y.-H., Peng, Y.-P., Vitorica, J., 2013. Th17 cell-mediated
pnas.0706832104. neuroinflammation is involved in neurodegeneration of aβ1-42-induced alzheimer’s
Togo, T., Akiyama, H., Iseki, E., Kondo, H., Ikeda, K., Kato, M., Oda, T., Tsuchiya, K., disease model rats. PLoS One1 8 (10), e75786.
Kosaka, K., 2002. Occurrence of T cells in the brain of alzheimer’s disease and other Zhang, Y., Wang, J., Zhang, Y.i., Wang, L., Wei, J., Chumnanvej, S., 2022. Characteristics
neurological diseases. Journal of Neuroimmunology 124 (1–2), 83–92. https://doi. scanning of brain structure and function changes in patients with different degrees of
org/10.1016/s0165-5728(01)00496-9. alzheimer’s disease. Contrast Media & Molecular Imaging 2022, 1–8.
Town, T., Tan, J., Flavell, R.A., Mullan, M., 2005. T-cells in alzheimer’s disease. Zhao, M., Cribbs, D.H., Anderson, A.J., Cummings, B.J., Su, J.H., Wasserman, A.J.,
Neuromolecular Medicine 7 (3), 255–264. https://doi.org/10.1385/NMM:7:3:255. Cotman, C.W., 2003. the induction of the TNFalpha death domain signaling pathway
Trajkovic, V., Stosic-Grujicic, S., Samardzic, T., Markovic, M., Miljkovic, D., Ramic, Z., in alzheimer’s disease brain. Neurochemical Research 28 (2), 307–318. https://doi.
Mostarica, S.M., 2001. interleukin-17 stimulates inducible nitric oxide synthase org/10.1023/a:1022337519035. PMID: 12608703.
activation in rodent astrocytes. Journal of Neuroimmunology 119 (2), 183–191. Zhao, D., Feng, F., Zhao, C., Wu, F., Ma, C., Bai, Y., Guo, J., Li, H., 2018. Role of perforin
https://doi.org/10.1016/s0165-5728(01)00391-5. secretion from CD8+ t-cells in neuronal cytotoxicity in multiple sclerosis.
Tröscher, A.R., Wimmer, I., Quemada-Garrido, L., Köck, U., Gessl, D., Verberk, S.G.S., Neurological Research 40 (1), 62–67. https://doi.org/10.1080/
Martin, B., Lassmann, H., Bien, C.G., Bauer, J., 2019. Microglial nodules provide the 01616412.2017.1398371.
environment for pathogenic T cells in human encephalitis. Acta Neuropathologica Zotova, E., Bharambe, V., Cheaveau, M., Morgan, W., Holmes, C., Harris, S., Neal, J.W.,
137 (4), 619–635. https://doi.org/10.1007/s00401-019-01958-5. Love, S., Nicoll, J.A., Boche, D., 2013. inflammatory components in human
Unger, M.S., Marschallinger, J., Kaindl, J., Klein, B., Johnson, M., Khundakar, A.A., alzheimer’s disease and after active amyloid-β42 immunization. Brain 136 (Pt 9),
Roßner, S., Heneka, M.T., Couillard-Despres, S., Rockenstein, E., Masliah, E., 2677–2696. https://doi.org/10.1093/brain/awt210.
Attems, J., Aigner, L., 2018. Doublecortin expression in CD8+ t-cells and microglia

14

You might also like