Вы находитесь на странице: 1из 6

Journal of Clinical Neuroscience 59 (2019) 6–11

Contents lists available at ScienceDirect

Journal of Clinical Neuroscience


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

Review article

Influence of microglia and astrocyte activation in the neuroinflammatory


pathogenesis of Alzheimer’s disease: Rational insights for the
therapeutic approaches
Mir Hilal Ahmad, Mahino Fatima, Amal Chandra Mondal ⇑
Cellular and Molecular Neurobiology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India

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

Article history: Alzheimer’s disease (AD), the most common progressive neurodegenerative disorder is characterized by
Received 23 May 2018 the formation of extracellular amyloid plaques and intracellular neurofibrillary tangles (NFTs). Increasing
Accepted 5 October 2018 evidences suggest a link between neuroinflammation and neuronal dysfunction in AD, orchestrated by
the progressive activation of microglial cells and astrocytes with the consequent overproduction of proin-
flammatory molecules. The concomitant release of anti-inflammatory mediators antagonizes the inflam-
Keywords: matory processes and leading to the severity of the AD pathology. The simultaneous detection of these
Alzheimer’s disease
inflammatory molecules in the pre-symptomatic stage may help in the early diagnosis of the AD. We have
Neuroinflammation
Cytokines
discussed the impact of microglia and astrocytic cells, the principal agents in the neuroinflammation pro-
Microglia cess, in relation to the progression of the AD. Modulation of the risk factors and targeting of these
Astrocytes immune mechanisms could lead to better therapeutic or preventive strategies for the AD. Further studies
need to determine, how the inhibition of inflammatory factors could be used for the AD alternative
therapies.
Ó 2018 Elsevier Ltd. All rights reserved.

1. Introduction Neuroinflammation in the AD is considered as a double-edged


sword where the immune cells of the brain have beneficial effects
AD, the most common chronic neurodegenerative dementia by degrading Ab accumulation as well as the adverse effects by
[1–3] is characterized by the presence of intra and extracellular producing cytotoxic substances that exacerbate Ab deposition
protein aggregates [2]. Extracellular protein aggregates correspond resulting in the neuronal dysfunctions [6,11]. High expression of
to the amyloid plaques, mainly composed of a 39 to 42 amino acids inflammatory mediators has been reported around the Ab plaques
peptide called b-amyloid (Ab), generated by abnormal proteolytic and NFTs exemplifying the relationship between neuroinflamma-
cleavage of the amyloid precursor protein (APP) by beta and tion and AD pathology [2]. In the early stages of the AD, neuropro-
gamma secretases [4]. These amyloid plaques accumulate around tective pathways, such as amyloid plaque clearance and
neurons which cause abnormalities in synaptic functioning, dis- antioxidant mechanisms against reactive oxygen species (ROS)
rupt the mitochondrial functioning and decreased levels one of generation, are very effective [12]. But with the progression of
the key neurotransmitters such as acetylcholine [5]. Intracellular AD, the elevated degree of oxidative stress up-regulates cellular
protein aggregates correspond to the NFTs produced by the accu- mediators of the immune system, resulting in the overproduction
mulation of paired helical filaments (PHF), generated by the aber- of pro-inflammatory molecules, and inflammation in the brain
rantly phosphorylated tau proteins [6–8]. Deposition of NFTs leads [13]. Cells in the CNS such as microglia, astrocytes and neurons
to the neuronal cell death, destabilization of microtubules and constitute the basis for neuroinflammation [14]. Microglial activa-
membrane degeneration [7–9]. Alongside the amyloid plaques, tion is one of the early events in the AD pathology which results in
NFTs and atrophy of the brain, the neuroinflammation triggered the increased pro-inflammatory effects and leading cause of neuro-
by the central nervous system (CNS) as innate the immune toxicity [15–17].
response, plays a significant role in the pathogenesis of AD [2,4]. Activated microglia can results in the AD progression through a
The activity in the inflammatory cascades can be a response to sequence of overactivation and up-regulation of proinflammatory
AD neuropathology [10]. factors, oxidative stress and ultimately neuroinflammation
[18–20]. The induction of microglia-induced inflammatory
⇑ Corresponding author. response results in the release of neurotoxic cytokines [21]. The
E-mail address: acmondal@mail.jnu.ac.in (A.C. Mondal). persistent microglia activation and the subsequent release of

https://doi.org/10.1016/j.jocn.2018.10.034
0967-5868/Ó 2018 Elsevier Ltd. All rights reserved.
M.H. Ahmad et al. / Journal of Clinical Neuroscience 59 (2019) 6–11 7

pro-inflammatory cytokines induce a cascade of neurotoxic factor-a (TNF-a), interleukin (IL)-6, and nitric oxide (NO, which
changes leading to the AD onset/progression [22]. Astrocytes are confer the AD onset/progression [21].
also suggested to play a key role in the AD progression. Ab plaques In the past decades, many studies have focused on the under-
also activate astrocytes which results in the overexpression of standing of mechanisms of microglial activation in response to
cytokines (e.g., IL-1b or IL-6) and a higher degree of oxidative stress neuropathological conditions in vivo and in vitro. During AD pro-
[13]. The dysfunctions between the astrocytes and the surrounding gression, the microglia respond to various stimuli, including amy-
neurons disrupt the synaptic connections and initiate a cascade of loid beta peptides and NFTs [36]. The persistent classical form of
neuronal injury [23–24]. microglial activation occurs by many receptors, including scav-
Astrocytes engulf the oligomer forms of the Ab, however, they enger receptor class A type 1, MARCO, scavenger receptor class B
get accumulated resulting in the extracellular annular protofibrils member 1, CD36, and the receptor for advanced glycation end pro-
which cause elevated oxidative stress and loss of neurons duct [37–38]. G protein-coupled receptors formyl peptide receptor
[25–26]. Neurons are also suggested to be involved in neuroinflam- 2 and chemokine-like receptor 1, TLR2 [39–40] toll-like receptors
mation, by increasing the expression and production of the (TLRs) [41] TLR4, and the CD14 co-receptor, and a6b1 integrin [42].
inflammatory molecules [27–28]. Neurons produce an inflamma- The association between Ab deposits and these receptors results
tory response by exacerbating local inflammatory reactions in the production of inflammatory mediators such as cytokines
resulting in their own destruction [29]. In this review, we have (interleukin [IL]-1a, IL-1b, IL-6, IL-8, IL-12, IL-18, and IL-23, inter-
addressed on how neuroinflammatory processes are directly feron (IFN)-c, TNF-a, and granulocyte-macrophage colony-
related to AD progression. Moreover, we have discussed the stimulating-factor (GMCSF) [43–44], chemokines (monocyte
implications of the involvement of microglia and astrocytes in both chemotactic protein 1 (MCP1), MCP-113, fractalkine) [45–46],
the inflammatory and neuro-immunomodulatory processes during CD36 receptors [47], and NOD-like receptors (NLRs). The TNF-a
the AD progression level is found proximal to amyloid plaques in an elevated level in
the AD patients which reflecting of disease severity and contribut-
2. Microglial activation in the AD progression ing to the inflammatory milieu [48]. IL-1b levels elevated during
the progression of AD especially in response to the tau protein
Microglia are the brain’s resident immune cells which are found over-expression [49]. Exposure of cultured human brain endothe-
throughout the brain, spinal cord, retina and optic nerves but lial cells to Ab1–40 elicited expression of inflammatory molecules
mainly in the hippocampus and substantia nigra [2,15]. Functions IL-1b and IL-6 during AD progression [50]. The levels of IL-18
of the microglial cells in the CNS appear to be complex as they increased significantly in different regions of AD brains, where it
exhibit both neuroprotective and neurotoxic effects. It is precisely co-localized with Ab-plaques, IL-8 also increases expression of
this delicate balance between the neurotoxic and neuroprotective glycogen synthase kinase-3b (GSK-3b) and cyclin-dependent
and between proinflammatory and anti-inflammatory which kinase 5 (cdk5), involved in hyperphosphorylation of tau-protein
determines the role of microglia in AD onset/progression. In their [51]. Additionally, IL-12 and IL-23, which are known to be pro-
resting form, they perform neurogenesis, neuroprotection and duced by leucocytes, are produced by microglia in the mouse mod-
synaptic pruning, which is complement dependent [30–31], but els of AD [52] and inhibition of these cytokines reduces AD
their persistent over activation results in the ineffective Ab deposi- pathology [53]. This classical activation of microglia leads to the
tion clearance by microglia resulting in a proinflammatory state adaptive immune response by expressing major histocompatibility
[32–34]. Activated microglia produce and secrete several proin- class II (MHC-II) molecules and interaction with the T-cells [54].
flammatory mediators, [18–20,35] including tumor necrosis Besides the proinflammatory molecules, activated microglia also

Fig. 1. Possible role of microglia/astrocytes in the onset/progression of the AD pathology through cytokines mediated neuro-inflammation.
8 M.H. Ahmad et al. / Journal of Clinical Neuroscience 59 (2019) 6–11

Fig. 2. Possible influence of the pro-inflammatory cytokines released by activated microglia/astrocytes in the neuroinflammation, neuronal death and neurodegeneration
during the AD pathology.

release cytotoxic factors such as superoxide radicals (O2 ), NO and astrocytes observed in the brains of patients with dementia were
ROS [55–56], therefore activated microglia play a crucial role in the initially analyzed by Dr. Alois Alzheimer, which found abundant
innate immunity and are the main source of proinflammatory glial cells in the neuritic plaques [2]. A more detailed analysis of
molecules leading to the neuroinflammation during the AD pathol- astrogliosis in the brains obtained from elderly patients (with
ogy. The possible mechanism of activated microglia in the neuroin- and without AD confirmed) has shown a correlation between the
flammation during the AD pathology is given in Figs. 1, 2, and degree of astrogliosis and cognitive impairment.
Table 1. Ab deposits can activate the astrocytes, leads to the over-
expression of cytokines (e.g., IL-1b or IL-6) resulting in oxidative
3. Astroglial activation in the AD onset/progression stress. Astrocytes damage and dysfunctions between astrocytes
and the surrounding neurons can destroy the synaptic homeostasis
Astrocytes located in the CNS support the endothelial cells of and initiate the cascade for neuronal injury [23–24]. These mech-
the blood-brain barrier (BBB), maintain ion balance, and provide anisms could result in enhanced ROS production and loss of astro-
nutrients to the neurons [2]. In AD brains, there are clear signs of cytes and eventually the death of neurons [59–60].
astrogliosis (increased number of astrocytes) around Ab deposits During AD pathology, astrocytes start to express BACE 1, thus
[57–58]. Like microglia, astrocytes release cytokines, ILs, NO, acquiring Ab producing ability [61]. Brains at the later stages of
and other potentially cytotoxic molecules after exposure to Ab, the AD, astrogliosis was observed in both the human tissues and
thereby exacerbating the neuroinflammatory response. Pathological in the brains isolated from AD animal models [57,62]. Large
M.H. Ahmad et al. / Journal of Clinical Neuroscience 59 (2019) 6–11 9

Table 1
The impact of microglia and astrocytes in the AD pathogenesis mediated by pro-inflammatory cytokines.

CNS origin Pro-inflammatory cytokines Effect on AD progression References


Microglia TNF-a "tau hyperphosphorylation [85–86]
Astrocyte " nitric oxide
"Ab synthesis; ;Ab clearance
Microglia IL-1b " tau phosphorylation [32,68,87–89]
Astrocytes ; tau pathology
" APP transcription and transduction in neurons
"Ab synthesis
;Ab related pathology
Microglia IL-6, IL-8 "tau hyperphosphorylation [90–92]
Astrocytes "production of APP
"Ab production
Microglia IL-18, IL-23 "tau hyperphosphorylation [51,93–94]
Astrocytes " GSK-3b expression
"Cdk5 expression
"production of APP
"Ab production

numbers of astrocytes associated with Ab deposits in AD brain have shown that pharmacological inhibition of (cyclooxigenase-2)
regions suggesting that these lesions can generate chemotactic COX-2 and inducible NO synthase have shown promising results
molecules that induce recruitment of astrocytes. It has been found [78–82]. Ibuprofen reduced amyloid beta deposition and tau hyper
that astrocytes throughout the entorhinal cortex of AD patients phosphorylation in the hippocampus in transgenic (Tg) mouse
gradually accumulate Ab1–42 positive material and the amount of models [82]. Likewise, a clinical trial with naproxen reduced the
this material is positively correlated with the extent of local AD tau phosphorylation and Ab levels in cerebrospinal fluid and
pathology. Ab1–42 within these astrocytes appears to be of neuronal plasma [83]. NSAIDs may decrease the AD progression reducing
origin, possibly accumulated by phagocytosis of locally degener- the inflammatory processes in the brain, by inhibiting the inflam-
ated dendrites and synapses, especially in the cortical molecular matory response of microglial and/or astrocytes, reducing cell
layer [63]. In support of this finding, recent evidences suggest that death due to excitotoxicity mediated by glutamate [2]. Lastly,
astroglial cells are able to phagocytize Ab peptides, a process which modulating the GSK-3b activity can be potential alternative thera-
may depend on their ApoE status, suggesting that ApoE polymor- peutic target to consider [84].
phisms may influence the risk to develop AD by affecting astroglial
Ab phagocytosis [64]. With the established fact that Ab deposition
5. Conclusions and future research directions
is a potent glial activator, therefore astrocytes and microglial acti-
vation could be an early event in the disease process, occurring
The production of neuro-inflammation which appears to be an
even in the absence of focal Ab deposition [65]. AD is associated
early biomarker significantly contributes to the progression of
with specific damage to astroglia, which may occur at the early
the AD. The concomitant release of anti-inflammatory mediators
stages of the disease and contribute to cognitive abnormalities
sometimes antagonizes the inflammatory processes and leading
[57]. Additionally, synchronous hyperactivity and intercellular
to the severity of the disease. Strategies, such as the interruption
Ca2+ waves have been observed in astrocytes in response to AD,
of defective inflammatory pathways or microglial stimulation,
both in vivo and in vitro [66–67]. A possible mechanism of astro-
which may increase the Ab clearance, could be successful. Several
cytes in inflammation in AD pathogenesis is given in Figs. 1, 2,
cytokines and other inflammatory molecules have been described
and Table 1.
as possible target candidates for AD therapies, but additional stud-
ies are needed to clarify, how they could contribute to alternative
4. Therapeutic strategies for targeting neuroinflammation in AD therapies of different neurodegenerative diseases. Blocking gene
pathology expression of targeted cytokines, their receptors, or better regulat-
ing the functioning of cells implicated in the neuroinflammation
The Ab accumulation and tau kinase activity can be decreased are strategies still in exploration. Some therapeutic strategies or
by using IL-1b or TNF-a/TNF-a, receptor peroxisome proliferator- dietary recommendations can be implemented as a preventive
activated receptor gamma (PPARc) agonists, cannabinoids, measure which successfully targets the ongoing inflammation.
minocycline or nicergoline and tyrosine kinase inhibitors. Another crucial factor is the comprehension of the role of each cel-
[4,43,68–71]. For example, galantamine, an acetylcholinesterase lular component in the inflammatory process, for example, the
inhibitor engages the nicotinic acetylcholine receptor a7 and can identification of cell-specific biomarkers. Indeed, specifically clari-
thereby increase the microglial uptake of Ab [72]. Likewise, PPARc fying changes in both immune system and inflammatory machin-
represents another factor that can be targeted by existing drugs, ery would make available different pathways for pharmacological
like thiazolidinedione class of antidiabetics [73]. The treatment of manipulations aimed at delaying the onset and/or the progression
APP/PS1 mice with PPAR-c agonists rapidly removes Ab by micro- of the AD. Finally, it is important to recognize each phase to AD
glia and, presumably astrocytes [74]. The possible mechanisms for progression and to clarify which processes are protective and
this may be the direct up-regulation of microglial CD36 expression which ones are detrimental. This will help to achieve more focused
[75] and lipidation of apolipoprotein E dependent on transcription and functional therapeutic strategies against AD progression.
factors of the LXR family [74]. Similarly, minocycline has been
demonstrated to prevent the buildup of Ab deposition, reducing Acknowledgements
caspase-3 activation and BACE1 levels in mice model of AD [76].
Various epidemiological studies demonstrated that the use of ACM gratefully acknowledges the financial support from JNU
non-steroidal anti-inflammatory drugs (NSAIDs) showed the pro- UPE-II (Project Id-247), DST-PURSE-II, DBT (BT/PR16164/
tective effect in AD patients [7,77]. Both in vitro and in vivo studies NER/95/88/2015), Ministry of Science and Technology (Govt. of
10 M.H. Ahmad et al. / Journal of Clinical Neuroscience 59 (2019) 6–11

India). M. Fatima acknowledges the financial support from SERB N- [31] Vinet J, van Weering HR, Heinrich A, Kälin RE, Wegner A, Brouwer N, et al.
Neuroprotective function for ramified microglia in hippocampal excitotoxicity.
PDF Grant No. (PDF/2016/000670).
J Neuroinflammation 2012;9:27.
[32] Hickman SE, Allison EK, El Khoury J. Microglial dysfunction and defective b-
amyloid clearance pathways in aging Alzheimer’s disease mice. J Neurosci
Appendix A. Supplementary data
2008;28:8354–60.
[33] Johnston H, Boutin H, Allan SM. Assessing the contribution of inflammation in
Supplementary data to this article can be found online at models of Alzheimer’s disease. Biochem Soc Trans 2011;886–890.
[34] D’Andrea MR, Cole GM, Ard MD. The microglial phagocytic role with specific
https://doi.org/10.1016/j.jocn.2018.10.034.
plaque types in the Alzheimer disease brain. Neurobiol Aging 2004;25:675–83.
[35] Liu G, Zhang S, Cai Z, Ma G, Zhang L, Jiang Y, et al. PICALM gene rs3851179
References polymorphism contributes to Alzheimer’s disease in an Asian population.
Neuromolecular Med 2013;15:384–8.
[36] Czeh M, Gressens P, Kaindl AM. The yin and yang of microglia. Dev Neurosci
[1] Prince M, Bryce R, Albanese E, Wimo A, Ribeiro W, Ferri CP. The global
2011;33:199–209.
prevalence of dementia: a systematic review and metaanalysis. Alzheimers
[37] Yu Y, Richard DY. Microglial Ab receptors in Alzheimer’s disease. Cell Mol
Dement 2013;9(63–75):e2.
Neurobiol 2015;35:71–83.
[2] Morales I, Guzmán-Martínez L, Cerda-Troncoso C, Farías GA, Maccioni RB.
[38] Cai Z, Liu N, Wang C, Qin B, Zhou Y, Xiao M, et al. Role of RAGE in Alzheimer’s
Neuroinflammation in the pathogenesis of Alzheimer’s disease. A rational
disease. J Cell Mol Med 2016;36:483–95.
framework for the search of novel therapeutic approaches. Front Cell Neurosci
[39] Le Y, Gong W, Tiffany HL, Tumanov A, Nedospasov S, Shen W, et al. Amyloid
2014;8:112.
(beta)42 activates a G-protein-coupled chemoattractant receptor, FPR-like-1.
[3] Joshi Y, Pratico D. Neuroinflammation and Alzheimer’s disease: lessons learned
Journal Neurosci. 2001;21. RC123-RC.
from 5-lipoxygenase. Transl Neurosci 2014;5:197–202.
[40] Peng L, Yu Y, Liu J, Li S, He H, Cheng N, et al. The chemerin receptor CMKLR1 is a
[4] Walters A, Phillips E, Zheng R, Biju M, Kuruvilla T. Evidence for
functional receptor for amyloid-b peptide. J Alzheimers Dis 2015;43:227–42.
neuroinflammation in Alzheimer’s disease. Prog Neurol Psychiatry
[41] Carty M, Bowie AG. Evaluating the role of Toll-like receptors in diseases of the
2016;20:25–31.
central nervous system. Biochem Pharmacol 2011;81:825–37.
[5] Bronzuoli MR, Iacomino A, Steardo L, Scuderi C. Targeting neuroinflammation
[42] Stewart CR, Stuart LM, Wilkinson K, Van Gils JM, Deng J, Halle A, et al. CD36
in Alzheimer’s disease. J Inflamm Res 2016;9:199.
ligands promote sterile inflammation through assembly of a Toll-like receptor
[6] Bhaskar M. Neuroinflammation in Alzheimer’s disease-A review. J Pharm Sci
4 and 6 heterodimer. Nat Immunol 2010;11:155.
Technol 2016;2:1–6.
[43] Von Bernhardi R, Tichauer JE, Eugenín J. Aging-dependent changes of
[7] Calsolaro V, Edison P. Neuroinflammation in Alzheimer’s disease: Current
microglial cells and their relevance for neurodegenerative disorders. J
evidence and future directions. Alzheimers Dement 2016;12:719–32.
Neurochem 2010;112:1099–114.
[8] Whittington RA, Planel E, Terrando N. Impaired resolution of inflammation in
[44] Li K, Liu S, Yao S, Wang B, Dai D, Yao L. Interaction between interleukin-8 and
Alzheimer’s disease: a review. Front Immunol 2017;8:1464.
methylenetetrahydrofolate reductase genes modulates Alzheimer’s disease
[9] Karran E, Mercken M, De Strooper B. The amyloid cascade hypothesis for
risk. Dement Geriatr Cogn Disord 2009;27:286–91.
Alzheimer’s disease: an appraisal for the development of therapeutics. Nat Rev
[45] Westin K, Buchhave P, Nielsen H, Minthon L, Janciauskiene S, Hansson O. CCL2
Drug Discov 2011;10:698.
is associated with a faster rate of cognitive decline during early stages of
[10] Grammas P, Martinez J, Miller B. Cerebral microvascular endothelium and the
Alzheimer’s disease. PLoS ONE 2012;7:e30525.
pathogenesis of neurodegenerative diseases. Expert Rev Mol Med 2011;13.
[46] Wu J, Bie B, Yang H, Xu JJ, Brown DL, Naguib M. Suppression of central
[11] Ramberg V. Neuroinflammation in Alzheimer’s disease: focus on NF-jB and C/
chemokine fractalkine receptor signaling alleviates amyloid-induced memory
EBP transcription factors. Depart of Neurochem, Stockholm Univ; 2011.
deficiency. Neurobiol Aging 2013;34:2843–52.
[12] McNaull BB, Todd S, McGuinness B, Passmore AP. Inflammation and anti-
[47] Bamberger ME, Harris ME, McDonald DR, Husemann J, Landreth GE. A cell
inflammatory strategies for Alzheimer’s disease – a mini-review. Gerontology
surface receptor complex for fibrillar b-amyloid mediates microglial
2010:3–14.
activation. J Neurosci 2003;23:2665–74.
[13] Bagyinszky E, Van Giau V, Shim K, Suk K, An SSA, Kim S. Role of inflammatory
[48] Wang W-Y, Tan M-S, Yu J-T, Tan L. Role of pro-inflammatory cytokines
molecules in the Alzheimer’s disease progression and diagnosis. J Neurol Sci
released from microglia in Alzheimer’s disease. Ann Transl Med 2015;3.
2017;376:242–54.
[49] Bonda DJ, Wang X, Perry G, Nunomura A, Tabaton M, Zhu X, et al. Oxidative
[14] Shastri A, Bonifati DM, Kishore U. Innate immunity and neuroinflammation.
stress in Alzheimer disease: a possibility for prevention. Neuropharmacol
Mediators Inflamm 2013;2013.
2010;59:290–4.
[15] Venneti S, Wiley CA, Kofler J. Imaging microglial activation during
[50] Vukic V, Callaghan D, Walker D, Lue L-F, Liu QY, Couraud P-O, et al. Expression
neuroinflammation and Alzheimer’s disease. J Neuroimmune Pharmacol
of inflammatory genes induced by beta-amyloid peptides in human brain
2009;4:227–43.
endothelial cells and in Alzheimer’s brain is mediated by the JNK-AP1
[16] Hanisch U-K, Kettenmann H. Microglia: active sensor and versatile effector
signaling pathway. Neurobiol Dis 2009;34:95–106.
cells in the normal and pathologic brain. Nat Neurosci 2007;10:1387.
[51] Sutinen EM, Pirttila T, Anderson G, Salminen A, Ojala JO. Pro-inflammatory
[17] Heneka MT, Kummer MP, Latz E. Innate immune activation in
interleukin-18 increases Alzheimer’s disease-associated amyloid-b production
neurodegenerative disease. Nat Rev Immunol 2014;14:463–77.
in human neuron-like cells. J Neuroinflammation 2012;9:199.
[18] Cai Z, Hussain MD, Yan L-J. Microglia, neuroinflammation, and beta-amyloid
[52] Vom Berg J, Prokop S, Miller KR, Obst J, Kälin RE, Lopategui-Cabezas I, et al.
protein in Alzheimer’s disease. Int J Neurosci 2014;124:307–21.
Inhibition of IL-12/IL-23 signaling reduces Alzheimer’s disease–like pathology
[19] Thameem Dheen S, Kaur C, Ling E-A. Microglial activation and its implications
and cognitive decline. Nat Med 2012;18:1812.
in the brain diseases. Curr Med Chem 2007;14:1189–97.
[53] Tan M-S, Yu J-T, Jiang T, Zhu X-C, Guan H-S, Tan L. IL12/23 p40 inhibition
[20] Mandrekar-Colucci S, Landreth GE. Microglia and inflammation in Alzheimer’s
ameliorates Alzheimer’s disease-associated neuropathology and spatial
disease. CNS Neurol Disord Drug Targets 2010;9:156–67.
memory in SAMP8 mice. J Alzheimers Dis 2014;38:633–46.
[21] Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, et al. Inflammation
[54] Olson JK, Miller SD. Microglia initiate central nervous system innate and
and Alzheimer’s disease. Neurobiol Aging 2000;21:383–421.
adaptive immune responses through multiple TLRs. J Immunol
[22] Sudduth TL, Schmitt FA, Nelson PT, Wilcock DM. Neuroinflammatory
2004;173:3916–24.
phenotype in early Alzheimer’s disease. Neurobiol Aging 2013;34:1051–9.
[55] Colton CA, Wilcock DM. Assessing activation states in microglia. CNS Neurol
[23] Agostinho P, Cunha RA, Oliveira C. Neuroinflammation, oxidative stress and
Disord Drug Targets 2010;9:174–91.
the pathogenesis of Alzheimer’s disease. Curr Pharm Des 2010;16:2766–78.
[56] Ha SK, Moon E, Lee P, Ryu JH, Oh MS, Kim SY. Acacetin attenuates
[24] Avila-Muñoz E, Arias C. When astrocytes become harmful: functional and
neuroinflammation via regulation the response to LPS stimuli in vitro and
inflammatory responses that contribute to Alzheimer’s disease. Ageing Res Rev
in vivo. Neurochem. Res 2012;37:1560–7.
2014;18:29–40.
[57] Verkhratsky A, Olabarria M, Noristani HN, Yeh C-Y, Rodriguez JJ. Astrocytes in
[25] Sollvander S, Nikitidou E, Brolin R, Söderberg L, Sehlin D, Lannfelt L, et al.
Alzheimer’s disease. Neurotherapeutics 2010;7:399–412.
Accumulation of amyloid-b by astrocytes result in enlarged endosomes and
[58] Okabe Y, Takahashi T, Mitsumasu C, Kosai K-I, Tanaka E, Matsuishi T.
microvesicle-induced apoptosis of neurons. Mol Neurodegener 2016;11:38.
Alterations of gene expression and glutamate clearance in astrocytes derived
[26] Lasagna-Reeves CA, Kayed R. Astrocytes contain amyloid-b annular protofibrils
from an MeCP2-null mouse model of Rett syndrome. PLoS One 2012;7.
in Alzheimer’s disease brains. FEBS Lett 2011;585:3052–7.
[59] Sollvander S, Nikitidou E, Brolin R, Soderberg L, Sehlin D, Lannfelt L, et al.
[27] Ramberg V, Tracy LM, Samuelsson M, Nilsson LNG, Iverfeldt K. The CCAAT/
Accumulation of amyloid-Ab by astrocytes result in enlarged endosomes and
enhancer binding protein (C/EBP) d is differently regulated by fibrillar and
microvesicle-induced apoptosis of neurons. Mol Neurodegener 2016;11:38.
oligomeric forms of the Alzheimer amyloid-b peptide. J Neuroinflammation
[60] Lasagna-Reeves CA, Castillo-Carranza DL, Sengupta U, Clos AL, Jackson GR,
2011;8:34.
Kayed R. Tau oligomers impair memory and induce synaptic and
[28] Pan M-H, Lai C-S, Ho C-T. Anti-inflammatory activity of natural dietary
mitochondrial dysfunction in wild-type mice. Mol Neurodegener 2011;6:39.
flavonoids. Food Funct 2010;1:15–31.
[61] Rossner S, Lange-Dohna C, Zeitschel U, Perez-Polo JR. Alzheimer’s disease b
[29] Heneka MT, O’Banion MK, Terwel D, Kummer MP. Neuroinflammatory
secretase BACE1 is not a neuronal-specific enzyme. J Neurochem
processes in Alzheimer’s disease. J Neural Transm 2010;117:919–47.
2005;92:226–34.
[30] Sierra A, Encinas JM, Deudero JJ, Chancey JH, Enikolopov G, Overstreet-
[62] Rodriguez JJ, Olabarria M, Chvatal A, Verkhratsky A. Astroglia in dementia and
Wadiche LS, et al. Microglia shape adult hippocampal neurogenesis through
Alzheimer’s disease. Cell Death Differ 2009;16:378–85.
apoptosis-coupled phagocytosis. Cell Stem Cell 2010;7:483–95.
M.H. Ahmad et al. / Journal of Clinical Neuroscience 59 (2019) 6–11 11

[63] Nagele RG, D’Andrea MR, Lee H, Venkataraman V, Wang HY. Astrocytes [78] Scuderi C, Stecca C, Bronzuoli MR, Rotili D, Valente S, Mai A, et al. Sirtuin
accumulate Ab42 and give rise to astrocytic amyloid plaques in Alzheimer modulators control reactive gliosis in an in vitro model of Alzheimer’s disease.
disease brains. Brain Res 2003;971:197–209. Front Pharmacol 2014;5:89.
[64] Niino M, Iwabuchi K, Kikuchi S, Ato M, Morohashi T, Ogata A, et al. [79] Bicca M, Costa R, Loch-Neckel G, Figueiredo C, Medeiros R, Calixto J. B2
Amelioration of experimental autoimmune encephalomyelitis in C57BL/6 receptor blockage prevents Ab-induced cognitive impairment by
mice by an agonist of peroxisome proliferator-activated receptor-gamma. J neuroinflammation inhibition. Behav Brain Res 2015;278:482–91.
Neuroimmunol 2001;116:40–8. [80] Gan P, Zhang L, Chen Y, Zhang Y, Zhang F, Zhou X, et al. Anti-inflammatory
[65] Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj EK, et al. Oxidative effects of glaucocalyxin B in microglia cells. J Pharmacol Sci 2015;128:35–46.
damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol [81] Scuderi C, Steardo L. Neuroglial roots of neurodegenerative diseases:
2001;60:759–67. therapeutic potential of palmitoylethanolamide in models of Alzheimer’s
[66] Kuchibhotla KV, Lattarulo CR, Hyman BT, Bacskai BJ. Synchronous disease. CNS Neurol Disord Drug Targets 2013;12:62–9.
hyperactivity and intercellular calcium waves in astrocytes in Alzheimer [82] Choi SH, Aid S, Caracciolo L, Sakura Minami S, Niikura T, Matsuoka Y, et al.
mice. Science 2009;323:1211–5. Cyclooxygenase-1 inhibition reduces amyloid pathology and improves
[67] Abramov AY, Canevari L, Duchen MR. Changes in intracellular calcium and memory deficits in a mouse model of Alzheimer’s disease. J Neurochem
glutathione in astrocytes as the primary mechanism of amyloid neurotoxicity. 2013;124:59–68.
J Neurosci 2003;23:5088–95. [83] Breitner JC, Baker LD, Montine TJ, Meinert CL, Lyketsos CG, Ashe KH, et al.
[68] Kitazawa M, Cheng D, Tsukamoto MR, Koike MA, Wes PD, Vasilevko V, et al. Extended results of the Alzheimer’s disease anti-inflammatory prevention
Blocking IL-1 signaling rescues cognition, attenuates tau pathology, and trial. Alzheimer’s Dement 2011;7:402–11.
restores neuronal b-catenin pathway function in an Alzheimer’s disease [84] Green HF, Nolan YM. GSK-3 mediates the release of IL-1b, TNF-a and IL-10
model. J Immunol 2011;187:6539–49. from cortical glia. Neurochem Int 2012;61:666–71.
[69] Shi J-Q, Shen W, Chen J, Wang B-R, Zhong L-L, Zhu Y-W, et al. Anti-TNF-a [85] Park KM, Bowers WJ. Tumor necrosis factor-alpha mediated signaling in
reduces amyloid plaques and tau phosphorylation and induces CD11c-positive neuronal homeostasis and dysfunction. Cell Signal 2010;22:977–83.
dendritic-like cell in the APP/PS1 transgenic mouse brains. Brain Res [86] Zhao J, O’Connor T, Vassar R. The contribution of activated astrocytes to Ab-
2011;1368:239–47. production: implications for Alzheimer’s disease pathogenesis. J
[70] He P, Cheng X, Staufenbiel M, Li R, Shen Y. Long-term treatment of thalidomide Neuroinflammation 2011;8:150.
ameliorates amyloid-like pathology through inhibition of b-secretase in a [87] Kitazawa M, Cheng D, Tsukamoto MR, Koike MA, Wes PD, Vasilevko V, et al.
mouse model of Alzheimer’s disease. PLoS ONE 2013;8:e55091. Blocking IL-1 signaling rescues cognition, attenuates tau pathology, and
[71] Tweedie D, Ferguson RA, Fishman K, Frankola KA, Van Praag H, Holloway HW, restores neuronal b-catenin pathway function in an Alzheimer’s disease
et al. Tumor necrosis factor-a synthesis inhibitor 3,60 -dithiothalidomide model. J Immunol 2011;187:6539–49.
attenuates markers of inflammation, Alzheimer pathology and behavioral [88] Tachida Y, Nakagawa K, Saito T, Saido TC, Honda T, Saito Y, et al. Interleukin-1b
deficits in animal models of neuroinflammation and Alzheimer’s disease. J up-regulates TACE to enhance a-cleavage of APP in neurons: resulting
Neuroinflammation 2012;9:106. decrease in Ab production. J Neurochem 2008;104:1387–93.
[72] Takata K, Kitamura Y, Saeki M, Terada M, Kagitani S, Kitamura R, et al. [89] Griffin WST, Liu L, Li Y, Mrak RE, Barger SW. Interleukin-1 mediates Alzheimer
Galantamine-induced amyloid-b clearance mediated via stimulation of and Lewy body pathologies. J Neuroinflammation 2006;3:5.
microglial nicotinic acetylcholine receptors. J Biol Chem 2010;285:40180–91. [90] Spooren A, Kolmus K, Laureys G, Clinckers R, De Keyser J, Haegeman G, et al.
[73] Gorlovoy P, Larionov S, Pham TTH, Neumann H. Accumulation of tau induced Interleukin-6, a mental cytokine. Brain Res Rev 2011;67:157–83.
in neurites by microglial proinflammatory mediators. FASEB J [91] Dugan LL, Ali SS, Shekhtman G, Roberts AJ, Lucero J, Quick KL, et al. IL-6
2009;23:2502–13. mediated degeneration of forebrain GABAergic interneurons and cognitive
[74] Mandrekar-Colucci S, Karlo JC, Landreth GE. Mechanisms underlying the rapid impairment in aged mice through activation of neuronal NADPH oxidase. PLoS
peroxisome proliferator-activated receptor-c-mediated amyloid clearance and ONE 2009;4:e5518.
reversal of cognitive deficits in a murine model of Alzheimer’s disease. J [92] Chakrabarty P, Jansen-West K, Beccard A, Ceballos-Diaz C, Levites Y, Verbeeck
Neurosci 2012;32:10117–28. C, et al. Massive gliosis induced by interleukin-6 suppresses Ab deposition
[75] Yamanaka M, Ishikawa T, Griep A, Axt D, Kummer MP, Heneka MT. PPARc/ in vivo: evidence against inflammation as a driving force for amyloid
RXRa-induced and CD36-mediated microglial amyloid-b phagocytosis results deposition. FASEB J 2010;24:548–59.
in cognitive improvement in amyloid precursor protein/presenilin 1 mice. J [93] Curran B, O’Connor JJ. The pro-inflammatory cytokine interleukin-18 impairs
Neurosci 2012;32:17321–31. long-term potentiation and NMDA receptor-mediated transmission in the rat
[76] Birch AM, Katsouri L, Sastre M. Modulation of inflammation in transgenic hippocampus in vitro. Neuroscience 2001;108:83–90.
models of Alzheimer’s disease. J Neuroinflammation 2014;11:25. [94] Ojala JO, Sutinen EM, Salminen A, Pirttila T. Interleukin-18 increases
[77] Shadfar S, Hwang CJ, Lim M-S, Choi D-Y, Hong JT. Involvement of inflammation expression of kinases involved in tau phosphorylation in SH-SY5Y
in Alzheimer’s disease pathogenesis and therapeutic potential of anti- neuroblastoma cells. J Neuroimmunol 2008;205:86–93.
inflammatory agents. Arch Pharm Res 2015;38:2106–19.

Вам также может понравиться