You are on page 1of 8

Molecular Neurobiology

https://doi.org/10.1007/s12035-018-0983-2

Neuroinflammation, Gut Microbiome, and Alzheimer’s Disease


Li Lin 1 & Li Juan Zheng 1 & Long Jiang Zhang 1

Received: 28 July 2017 / Accepted: 28 February 2018


# Springer Science+Business Media, LLC, part of Springer Nature 2018

Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease that develops insidiously and causes dementia finally.
There are also clinical complications in advanced dementia, such as eating problems, infections, which will lead to the decline of
patients’ life quality, and the rising cost of care for AD to our society. AD will be important public health challenge. Early
detection of AD may be a key issue to prevent, delay, and stop the disease. Gut microbiome and neuroinflammation are closely
related with nervous system diseases, although the specific mechanism is not clear. This review introduces the relationship
between neuroinflammation, gut microbiome, and AD.

Keywords Alzheimer’s disease . Gut microbiome . Neuroinflammation

Introduction resulting from AD will reach 700,000 in 2016, and many of


them die from the complications caused by AD [4].
Alzheimer’s disease (AD) is a neurodegenerative disease. It is Thus, early detection of AD may be a key issue to prevent,
the most common cause of dementia and develops insidiously. delay, and stop the disease. However, the pathogenesis of AD
Extracellular plaques containing amyloid beta (Aβ) and intra- is still unclear, and it is still difficult to diagnose AD before the
cellular neurofibrillary tangles containing hyperphosphorylated symptoms appear. There are great challenges in finding mea-
tau protein, along with synaptic and neuronal losses are the sures to prevent AD. Recent studies indicated the close corre-
main features of AD [1]. Patients with AD often suffer from a lation between neuroinflammation, gut microbiome, and AD,
decline in memory, aphasia, performance disorders, and per- which shed the light on the better understanding the pathogen-
sonality and behavior changes. There are also clinical compli- esis of AD. In this review, we will discuss the pathogenesis of
cations in advanced dementia, such as eating problems and AD and the possibility of early diagnosis, effective preven-
infections [2]. These clinical complications will lead to the tion, and treatment based on existing knowledge.
decline of patients’ life quality and the rising cost of care for
AD, which will be important public health challenges.
The surveys showed that there are about 35 million people Neuroinflammation and AD
suffering from dementia, and the number is increasing in the
elderly people in the world [3]. Recent report showed that Mechanism of Neuroinflammation
from 2000 to 2013, deaths from stroke reduced 23%, heart
disease declined 14%, and prostate cancer dropped 11%; how- So far, the exact mechanism of AD remains uncovered. The
ever, deaths caused by AD increased 71% [3, 4]. In 2013, current mainstream doctrine is the amyloid cascade hypothe-
official report showed that 84,767 persons died from AD, sis that Aβ deposition is the main reason of neurofibrillary
and it becomes the sixth main reason of death and the fifth tangles (NFTs), synaptic dysfunction, and dementia [5].
main reason of death in individuals age ≥ 65 years in the USA Honig et al. found that Solanezumab (designed to increase
[3, 4]. It is speculated that deaths of Americans age ≥ 65 years the clearance from the brain of soluble Aβ) at a dose of
400 mg administered every 4 weeks in patients with mild
Alzheimer’s disease did not significantly affect cognitive de-
* Long Jiang Zhang
kevinzhlj@163.com
cline, which challenges the theory of amyloid protein in
Alzheimer’s disease; however, the results may be subject to
1
Department of Medical Imaging, Jinling Hospital, Medical School of some factors, such as the small bioavailability caused by the
Nanjing University, Nanjing, Jiangsu 210002, China blood-brain barrier (BBB) [1]. Recently, an increasing number
Mol Neurobiol

of evidences show that neuroinflammation plays an important are linked to changes in astrocyte function [21]. Altered astro-
role in the pathogenesis of AD. Experimental evidence dem- cyte morphology in aging and diseased brain is complex and
onstrates that neuroinflammation is an active player in the region-specific [22]; however, the accumulation of activated
development and progression of AD [6, 7]. In the inflamma- astrocytes, often in clusters around amyloid plaques [23], cor-
tory response, microglia and astroglia are the essential relates strongly with Braak staging in AD [24]. Importantly,
members. astrocyte activation, together with the presence of phosphory-
Microglia originate from myeloid and account for about lated tau in synapses, is one of the best biological correlates of
15% of the nonneuronal cells in the brain [8] and are important cognitive decline in AD [25]. In many transgenic mouse
players in the development and progression of neuroinflam- models of AD, before the appearance of plaque and/or tangle
mation. Researchers categorized activated microglia as a clas- pathology, researchers found activated astrocytes accumulated
sic, proinflammatory (M1) phenotype and others as a nonin- in affected brain regions as well [26], suggesting that astrocyte
flammatory, alternative activation (M2) phenotype [9]. activation is involved in disease pathogenesis. One research
Increased concentrations of proinflammatory cytokines are suggested that astrocytes played a role in disease progression
the features of classic M1 activation, which include tumor in AD, probably due to the interaction of Aβ with astrocytes
necrosis factor-α (TNF-α), interleukin 1, interleukin 6, inter- inducing a pro-inflammatory profile [27].
leukin 12, and interleukin 18, with impaired phagocytic ca- Astrocytes, which far more than microglia in the brain, is
pacity accompanied [10]. Secretion of the anti-inflammatory considered to play a more significant and continuous role than
cytokines is the character of the M2 state, including interleu- microglia in the enduring neuroinflammatory [28, 29], and
kin 4, interleukin 10, interleukin 13, and transforming growth compared to microglia, astrocytes can eliminate and degrade
factor-β (TGF-β), with phagocytic capacity increased, where- Aβ without mediators or stimuli [30]. Astrocytes induce cen-
as toxic nitric oxide produced not [11, 12]. tral nervous system (CNS) inflammation of AD via acting as
In AD, by the way of cell-surface receptors, microglia bind immune cells, releasing cytokines and chemokines to affect
to soluble Aβ oligomers and Aβ fibrils [13, 14]. Through effector cells, regulating the blood-brain barrier and shaping
sequential cleavages by two membrane-bound proteases, amy- glial scars [31]. Astrocytes play a role in the occurrence and
loid precursor protein (APP) produces the Aβ peptide. development of AD mainly through elevating the expression
Microglia begin to clean up Aβ fibrils via phagocytosis after of proinflammatory cytokines and chemokines and modulat-
the receptor. As a consequence, these fibrils enter the ing the generation, internalization, and degradation of Aβ
endolysosomal pathway [14]. Compared to fibrillar Aβ, solu- [30]. Besides, Aβ could upregulate the expression of cyto-
ble Aβ can be degraded by various proteases outside the cell, kines and chemokines in astrocytes, as a result that astrocytes
which is mostly resistant to enzymatic degradation [14, 15]. reactivated [30]. Furthermore, astrocytes are closely related to
However, in AD, several mechanisms compromise cessation the oxidative stress response in AD, and it has been confirmed
of inflammation, such as progressing formation of Aβ and that activation of astrocytes are associated with intracellular
positive feedback loops between inflammation and APP pro- neurofibrillary tangles [32, 33].
cessing. On the contrary, further accumulation of Aβ, neuronal
debris, and, most probably, further activating factors establish Neuroinflammatory Imaging
chronic, nonresolving inflammation [14]. Chronic neuroin-
flammation can damage the BBB, causing activated microglia Recently, the developed positron emission tomography (PET)
to release proinflammatory cytokines [16]. The classical pro- imaging agents targeting neuroinflammatory processes offer
inflammatory cytokine TNF-α produced by microglia, which the possibility to in vivo track diverse brain inflammatory
are early cellular markers of AD pathology, was shown to in- events. Specifically, neuroinflammatory events such as
duce neuronal cell cycle events [17]. In addition, in sporadic microglial activation, reactive astrocytosis, and increased
cases of AD, inefficient clearance of Aβ has been confirmed as phospholipase activity can be quantified by using PET radio-
a major pathogenic pathway [14, 18]. By the way of downreg- tracers [34–36].
ulating the expression of Aβ phagocytosis receptors to increase Currently, molecular agents targeting the 18 kDa
cytokine concentrations, which is considered to be the reason translocator protein (TSPO), the formerly name of the periph-
of insufficient microglial phagocytic capacity [14, 19] (Fig. 1). eral benzodiazepine receptor (PBR), can be used for PET im-
Except for microglia, astrocytes are major participants in aging of microglial activation [37]. A lot of studies showed
neuroinflammation, and which is closely related to AD. It is that TSPO is a sensitive marker of reactive microglia and
estimated that astrocytes represent between 30 and 50% of inflammation secondary to neurodegeneration, including of
human neural cells, and they have long been acknowledged the AD type [38, 39].
to provide crucial trophic and metabolic support for neurons [11C] PK11195 and [18F] DPA-714 are currently used as
[20]. In AD brain, there are alterations in astrocyte morphol- radiotracers for PET, both based on TSPO as ligand [39].
ogy, gene expression, protein composition, and activity that However, [11C]PK11195 imaging agent itself has some
Mol Neurobiol

Fig. 1 Pathological mechanism


of microglia activation. Microglia
has the function of surveillance
and synaptic remodeling. In the
acute inflammatory response,
microglia bind TLR to help clear
and restore tissue homoeostasis.
However, microglia activated by
amyloid beta deposits and
persistently exposed to
proinflammatory cytokines,
which result in chronic
neuroinflammation. As a result,
synaptic remodeling was reduced
and caused neuronal death. TLR
Toll-like receptor. With
permission, from [14]

drawbacks, such as low permeability in the brain, nonspecific very high doses directly bind to Aβ, but Szekely et al. showed
binding and nuclide [11C] shorter half-life (20 min), being they were no more effective than other NSAIDs in reducing
difficult to prepare, etc., affecting its wide clinical application. the risk of AD [44, 47]. A systematic review pointed that it
Wiley et al. [40] found that [11C]PK11195 imaging agent on requires NSAID use lasting over 2 years to reduce risk esti-
the detection of mild to moderate AD patients with microglia mates [48], while the obvious protective effects of NSAIDs
activation level is not sensitive. Compared to the first genera- are reduced among elderly people, even disappeared [45, 49].
tion of ligand [11C]PK11195 imaging agent, [18F] DPA-714 Whether NSAIDs are effective and which is the best for AD is
imaging agent as a new generation of TSPO specific ligand still unclear. However, a recent study found that IL-33 ame-
showed a great potential [14]. Recent studies have found that liorated AD-like pathology and cognitive decline [50]. By the
[18F] DPA-714 PET can accurately assess neuritis in early and way of promoting the recruitment and phagocytic activity of
preclinical AD (Fig. 2) [41]. In general, PET has been used to microglia, IL-33 decreased soluble Aβ and amyloid plaque
assess neuroinflammation of early and preclinical AD, al- load. In addition, IL-33 injection regulated the immune re-
though there are still some shortcomings. sponse of amyloid precursor protein/presenilin 1 (APP/PS1)
mice and polarized microglia/macrophages toward an anti-
inflammatory phenotype. Collectively, these results suggest
Neuroinflammation as a Therapeutic Target in AD that IL-33 may be a potential treatment of AD.

Lots of epidemiological studies have pointed that the inci-


dence of AD reduced after patients use nonsteroidal anti-
inflammatory drugs (NSAIDs) [42]. Four large epidemiolog- Gut Microbiome and Central Nervous System
ical studies have investigated how NSAID affects individuals
with AD. The Baltimore longitudinal study [43, 44] showed The microbiome of the human gastrointestinal tract is the larg-
that people who use NSAID greater than 2-year duration est reservoir of microbes in humans, containing approximately
spared approximately 60% [44, 45]; the Rotterdam study 1014 microorganisms from at least 1000 distinct microbial
showed a sparing of 80% in AD patients using NSAID was species that vary in diversity and stability between individuals
confirmed via prescription records [44]; and the MIRAGE and outnumber human host cells by about 100 to 1. The bac-
study showed a sparing of 36% [44, 46]. Some NSAIDs at terial density of human gut microbiota is up to 1012/mL,
Mol Neurobiol

Fig. 2 Image of 18F-DPA-714 PET (SUVr) in AD. a Patients with representations and (ii and iv) sagittal, coronal, and axial views (x = 65,
prodromal Alzheimer’s disease (p-AD) and controls. b Alzheimer’s y = 44, and z = 37) with corresponding z-values. With permission, from [41]
disease dementia (d-AD) and controls. (i and iii) BGlass brain^

which is the highest density in any known microbial ecosys- absence of the microbiota, mice harbor microglia with signif-
tem [51, 52]. The latest researches showed that changes in icantly altered developmental states. These microglia display
gut microbiota could affect the brain’s physiological, be- morphological characteristics and a gene expression profile
havioral, and cognitive function [53–57]. Pinto-Sanchez that indicate an arrest in their developmental maturation and
et al. found that the probiotic BL (Bifidobacterium subsequently are maintained in an immature status [63, 64].
longum NCC3001) reduces depression in patients with Notably, microglia derived from GF mice display limited re-
irritable bowel syndrome, which were associated with sponses toward viral infection and microbially associated mo-
changes in brain activation patterns that indicate that this lecular patterns. Such defective responses can be rescued by
probiotic reduces limbic reactivity [58]. Another func- administration of short-chain fatty acids [63, 64].
tional MR imaging study found less hippocampal activity In addition, the importance of gut microbiome and mi-
viewing negative valences images in participants with crobial metabolites in the formation of the BBB has been
high Prevotella and greater prominence for gray matter exemplified in GF mice [60, 65]. In the absence of gut
(cerebellum, frontal regions, and the hippocampus) in microorganisms, the BBB is more permeable to macro-
participants with high Bacteroides [59]. molecules compared to conventionally raised animals,
mediated by decreased expression of key tight-junction
proteins in the brain endothelium. Furthermore, perme-
Gut Microbiome Affects Neurodevelopment
ability decreased upon colonization of GF animals or, al-
ternatively, upon administration of the short-chain fatty
Neurogenesis is influenced by the presence of microorgan-
acid butyrate that is produced as a result of bacterial fer-
isms. Specifically, neurogenesis in the dorsal hippocampus
mentation in the gut [60, 65]. Correspondingly, the BBB
of adult germ-free (GF) mice is increased compared to con-
in the sterile fetus is permeable, compared to the adult
ventional mice [60, 61]. Interestingly, colonization of GF mice
BBB [60].
at weaning could not reverse this phenotype, indicating that
microbial signals very early in life reduce rates of
neurogenesis in the hippocampus. Moreover, adult GF mice Gut Microbiome and Mental Illness
exhibited increased volume of the amygdala and hippocampus
(specifically CA2/3) and differed in dendrite morphology, Microbes which resident in our gastrointestinal tract play an
while no differences in total brain volume were recorded be- important role in nervous system, and the disruption of the
tween GF and specific pathogen-free (SPF) animals [60, 62]. microbes may cause the development or exacerbation of men-
By tapping into pathways that govern neuronal differentiation tal disorders. For example, major depressive disorder is typi-
and survival, via neurotrophins and their receptors, gut mi- cally influenced by the microbiome [66]. In schizophrenia,
crobes can influence the fate of neurons in various regions dysbiosis and psychopathogenesis are similar [67, 68]. More
of the brain and subsequently neurodevelopment and health. than half of all patients with irritable bowel syndrome are
A central role for the microbiota in the development and mat- affected by mood disorders [69], and antidepressants are often
uration of the microglia has recently emerged [63, 64]. In the used to treat irritable bowel syndrome [69, 70]. As we know,
Mol Neurobiol

gut–brain axis is the most popular hypothesis, which includes wild-type controls [83]. It is presumed that neuroinflammation
a bidirectional communication network that monitors and in- is one of the major reasons in the pathology of neurodegener-
tegrates gut functions and links them to cognitive and emo- ative diseases [84, 85]. Proinflammatory cytokines produced
tional centers of the brain. It consists of the central, autonomic both in the brain and periphery nervous system modulate neu-
and enteric nervous systems, the neuroendocrine, ronal function and can initiate pathologic cell death [86, 87].
enteroendocrine and neuroimmune systems [69, 71, 72]. Given the importance of microglia functions in both the pre-
Although the exact mechanism of gut-brain axis has not yet vention and promotion of neurodegenerative processes, it is
been fully clarified, the evidences from animals and human tempting to speculate that the gut microbiota may influence
studies have shown that gut microbiota can play an important these inflammatory diseases of the aging brain.
role in brain behavior and cognitive development by produc- It is possible that changes of the gut microbiota facilitate
ing hormones, immune factors, and metabolites, which also inflammatory processes that may contribute to the neuroin-
indicated that altering the gut microbiota may improve or even flammation in AD. Bacteria populating the gut microbiome
cure brain diseases [53]. can excrete large quantities of lipopolysaccharides (LPSs) and
Increasing evidences point toward a role for beneficial ef- amyloids. These LPSs and amyloids may lead to the patho-
fects of probiotics that extend beyond the modification of the genesis of AD during aging, when both the gastrointestinal
composition of the microbiota, including a beneficial impact tract epithelium and blood–brain barrier become more perme-
on behavior, mood, and cognition [73]. Gareau et al. found able. It has been proposed that LPS and amyloids may directly
that probiotics could not only dampen the hypothalamic- pass through a compromised gastrointestinal tract or blood–
pituitary-adrenal axis (HPA-axis) but they could also restore brain barrier and/or indirectly pass through these protective
neuronal activation indicated by nuclear c-Fos and brain- physiological barriers via LPS/amyloid triggered cytokines
derived neurotrophic factor (BDNF) expression in the CA1 or other small proinflammatory molecules that are normally
region of the hippocampus which were decreased following transited [88–90]. A recent study reported that Bacteroides
exposure to acute stress in infected mice [74]. This study sug- fragilis lipopolysaccharide (BF-LPS) exposure to human pri-
gested that probiotics could influence expression of key me- mary brain cells was extremely potent inducer of the pro-
diators of cognitive behavior within structures of the limbic inflammatory transcription factor NFκB (p50/p65) complex,
system. Follow-up studies revealed a role for the adaptive a known trigger involved in inflammatory neurodegeneration
immune system in modulating the microbiota–gut–brain axis. in the AD brain [91].
In healthy Balb/c mice, administration of Bifidobacterium In addition to LPS, a significant quantity of functional am-
(B. longum but not B. breve) could improve cognitive function yloid can be generated by many bacterial strains, including
as measured using the NOR task and the Barnes maze com- Escherichia coli, Bacillus subtilis, Salmonella typhimurium,
pared to vehicle, highlighting a strain-specific beneficial effect Salmonella enterica, Mycobacterium tuberculosis, and
[75]. Feeding mice a Western-style diet can also induce weight Staphylococcus aureus, through the accumulation of protein-
gain and cognitive deficits, which can be ameliorated by ad- aceous misfolded Aβ oligomers and fibrils, which may con-
ministration of L. helveticus R0052 [76]. The beneficial im- tribute to the pathology of AD [92, 93]. It has been hypothe-
pact of probiotic administration on cognitive function in sized that functional amyloids produced by gut bacteria may
humans is also being investigated. These studies, although be the source of misfolding of neuronal proteins such as alpha-
small in size, suggest that consumption of probiotic- synuclein, Aβ via cross-seeding, priming of the innate im-
containing products may have a beneficial impact on overall mune system, and activation of neuroinflammation [94, 95].
cognitive function in healthy adult populations [77, 78].

Future Directions
Gut Microbiome, Neuroinflammation,
and Alzheimer’s Disease Gut microbiome and neuroinflammation are closely related
with nervous system diseases, although the specific mecha-
For a long time, researchers have studied possible infectious nism remains unclear. Researches showed that the amyloid
reasons of neurodegenerative diseases. In AD, it has reported plaques, neurofibrillary tangles, gut microbiome, and neuro-
the connections to Herpes simplex virus 1, spirochetal, and inflammation play important roles in AD. Nowadays, imaging
Chlamydophila pneumoniae infections [79, 80]. Recently, one biomarkers of brain structure and function have been applied
research found fungal infection in the brains of AD and amyo- to diagnose AD. However, it is greatly significant to find
trophic lateral sclerosis (ALS) patients [81, 82]. Preliminary whether gut microbiome has influences on neuroinflammation
observations in the APP/PS1 mouse model of AD indicated in aging and AD. Thus, in future, we hypothesized whether
that these animals had decreased Allobaculum and we could diagnose AD early in combination with the imaging
Akkermansia, with an increase in Rikenellaceae compared to findings of neuroinflammation and the inflammatory markers
Mol Neurobiol

caused by intestinal microbes, and whether we could find new 9. Wang C, Wang Q, Lou Y, Xu J, Feng Z, Chen Y, Tang Q, Zheng G
et al (2018) Salidroside attenuates neuroinflammation and improves
methods of treating AD basing on AD-related neuroinflam-
functional recovery after spinal cord injury through microglia po-
mation and intestinal microbial changes, such as NSAIDs for larization regulation. J Cell Mol Med 22(2):1148–1166
the treatment of neuroinflammatories and probiotics that im- 10. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A (2002)
prove intestinal microbes. The recognition of the potential Macrophage polarization: tumor-associated macrophages as a par-
adigm for polarized M2 mononuclear phagocytes. Trends Immunol
contribution of microbiome-generated LPS and signaling
23(11):549–555
pathways implicated in neuroinflammation, brain Aβ deposi- 11. Koenigsknecht-Talboo J, Landreth GE (2005) Microglial phagocyto-
tion, and AD pathogenesis regulated by amyloids are relative- sis induced by fibrillar beta-amyloid and IgGs are differentially reg-
ly recent discoveries. As a result, scientists might find the ulated by proinflammatory cytokines. J Neurosci 25(36):8240–8249
biomarkers according to these advances, making the early 12. Zelcer N, Khanlou N, Clare R, Jiang Q, Reed-Geaghan EG,
Landreth GE, Vinters HV, Tontonoz P (2007) Attenuation of neu-
diagnosis of AD, novel therapeutics, and preventive strategies roinflammation and Alzheimer’s disease pathology by liver x re-
possible. ceptors. Proc Natl Acad Sci U S A 104(25):10601–10606
In general, gut microbiome may initiate AD with the com- 13. Minter MR, Taylor JM, Crack PJ (2016) The contribution of neuro-
bination of the amyloid plaques, neurofibrillary tangles, and inflammation to amyloid toxicity in Alzheimer’s disease. J
Neurochem 136(3):457–474
neuroinflammation. To study the relationship between gut
14. Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F,
microbiome, neuroinflammation, and AD, new methods need Feinstein DL, Jacobs AH, Wyss-Coray T et al (2015)
to be developed to assist in the clinical diagnosis of AD. For Neuroinflammation in Alzheimer’s disease. Lancet Neurol 14(4):
the effects of gut microbiome, it may be able to find a new 388–405
way to help clinicians make the early diagnosis, prevention 15. Lee CY, Landreth GE (2010) The role of microglia in amyloid
clearance from the AD brain. J Neural Transm (Vienna) 117(8):
and treatment of AD. However, these assumptions are also 949–960
lack of experimental verification; we expect that there will 16. Swanson A, Wolf T, Sitzmann A (2018) Neuroinflammation in
be more studies to verify this conjecture. Alzheimer’s disease: pleiotropic roles for cytokines and neuronal
pentraxins. Behav Brain Res S0166-4328(17):31862–31864
Acknowledgements Supported by grants from the National Natural 17. Bhaskar K, Maphis N, Xu G, Varvel NH, Kokiko-Cochran ON,
Science Foundation of China (grant nos. 81322020 and 81230032 to Weick JP, Staugaitis SM, Cardona A et al (2014) Microglial derived
L.J.Z.). tumor necrosis factor-alpha drives Alzheimer’s disease-related neu-
ronal cell cycle events. Neurobiol Dis 62:273–285
18. Mawuenyega KG, Sigurdson W, Ovod V, Munsell L, Kasten T,
Compliance with Ethical Standards Morris JC, Yarasheski KE, Bateman RJ (2010) Decreased clearance
of CNS beta-amyloid in Alzheimer’s disease. Science 330(6012):1774
Conflict of Interest The authors declare that they have no conflict of 19. Hickman SE, Allison EK, El Khoury J (2008) Microglial dysfunc-
interest. tion and defective beta-amyloid clearance pathways in aging
Alzheimer’s disease mice. J Neurosci 28(33):8354–8360
20. Clarke LE, Liddelow SA, Chakraborty C, Münch AE, Heiman M,
Barres BA (2018) Normal aging induces A1-like astrocyte reactiv-
References ity. Proc Natl Acad Sci U S A 115(8):E1896–E1905
21. Sofroniew MV, Vinters HV (2010) Astrocytes: biology and pathol-
1. Honig LS, Vellas B, Woodward M, Boada M, Bullock R, Borrie M, ogy. Acta Neuropathol 119(1):7–35
Hager K, Andreasen N et al (2018) Trial of solanezumab for mild 22. Rodriguez JJ, Yeh CY, Terzieva S, Olabarria M, Kulijewicz-Nawrot
dementia due to Alzheimer’s disease. N Engl J Med 378(4):321–330 M, Verkhratsky A (2014) Complex and region-specific changes in
2. Mitchell SL (2015) CLINICAL PRACTICE. Advanced dementia. astroglial markers in the aging brain. Neurobiol Aging 35(1):15–23
N Engl J Med 372(26):2533–2540 23. Walsh DM, Selkoe DJ (2004) Deciphering the molecular basis of
3. Association A (2015) 2015 Alzheimer’s disease facts and figures. memory failure in Alzheimer’s disease. Neuron 44(1):181–193
Alzheimers Dement 11(3):332–384 24. Simpson JE, Ince PG, Lace G, Forster G, Shaw PJ, Matthews F,
4. Association A (2016) 2016 Alzheimer’s disease facts and figures. Savva G, Brayne C et al (2010) Astrocyte phenotype in relation to
Alzheimers Dement 12(4):459–509 Alzheimer-type pathology in the ageing brain. Neurobiol Aging
5. Selkoe DJ, Hardy J (2016) The amyloid hypothesis of Alzheimer’s 31(4):578–590
disease at 25 years. EMBO Mol Med 8(6):595–608 25. Perez-Nievas BG, Stein TD, Tai HC, Dols-Icardo O, Scotton TC,
6. Zhang B, Gaiteri C, Bodea LG, Wang Z, McElwee J, Barroeta-Espar I, Fernandez-Carballo L, de Munain EL et al (2013)
Podtelezhnikov AA, Zhang C, Xie T et al (2013) Integrated sys- Dissecting phenotypic traits linked to human resilience to
tems approach identifies genetic nodes and networks in late-onset Alzheimer's pathology. Brain 136(8):2510–2526
Alzheimer’s disease. Cell 153(3):707–720 26. Schwab C, Klegeris A, McGeer PL (2010) Inflammation in trans-
7. Trovato Salinaro A, Pennisi M, Di Paola R, Scuto M, Crupi R, genic mouse models of neurodegenerative disorders. Biochim
Cambria MT, Ontario ML, Tomasello M et al (2018) Biophys Acta 1802(10):889–902
Neuroinflammation and neurohormesis in the pathogenesis of 27. Batarseh YS, Duong QV, Mousa YM, Al Rihani SB, Elfakhri K,
Alzheimer’s disease and Alzheimer-linked pathologies: modulation Kaddoumi A (2016) Amyloid-β and astrocytes interplay in
by nutritional mushrooms. Immun Ageing 15:8 amyloid-β related disorders. Int J Mol Sci 17(3):338
8. Varley J, Brooks DJ, Edison P (2015) Imaging neuroinflammation 28. Li C, Zhao R, Gao K, Wei Z, Yin MY, Lau LT, Chui D, Yu AC
in Alzheimer’s disease and other dementias: recent advances and (2011) Astrocytes: Implications for neuroinflammatory pathogene-
future directions. Alzheimers Dement 11(9):1110–1120 sis of Alzheimer’s disease. Curr Alzheimer Res 8(1):67–80
Mol Neurobiol

29. Savchenko VL, McKanna JA, Nikonenko IR, Skibo GG (2000) 47. Szekely CA, Green RC, Breitner JC, Østbye T, Beiser AS, Corrada
Microglia and astrocytes in the adult rat brain: comparative immu- MM, Dodge HH, Ganguli M et al (2008) No advantage of A beta
nocytochemical analysis demonstrates the efficacy of lipocortin 1 42-lowering NSAIDs for prevention of Alzheimer dementia in six
immunoreactivity. Neuroscience 96(1):195–203 pooled cohort studies. Neurology 70(24):2291–2298
30. Zou C, Shi Y, Ohli J, Schuller U, Dorostkar MM, Herms J (2016) 48. Szekely CA, Thorne JE, Zandi PP, Ek M, Messias E, Breitner JC,
Neuroinflammation impairs adaptive structural plasticity of dendrit- Goodman SN (2004) Nonsteroidal anti-inflammatory drugs for the
ic spines in a preclinical model of Alzheimer’s disease. Acta prevention of Alzheimer's disease: a systematic review.
Neuropathol 131(2):235–246 Neuroepidemiology 23(4):159–169
31. González-Reyes RE, Nava-Mesa MO, Vargas-Sánchez K, Ariza- 49. Szekely CA, Breitner JC, Fitzpatrick AL, Rea TD, Psaty BM,
Salamanca D, Mora-Muñoz L (2017) Involvement of astrocytes Kuller LH, Zandi PP (2008) NSAID use and dementia risk in the
in Alzheimer’s disease from a neuroinflammatory and oxidative Cardiovascular Health Study: role of APOE and NSAID type.
stress perspective. Front Mol Neurosci 10:427 Neurology 70(1):17–24
32. Kitazawa M, Oddo S, Yamasaki TR, Green KN, LaFerla FM (2005) 50. Fu AK, Hung KW, Yuen MY, Zhou X, Mak DS, Chan IC, Cheung
Lipopolysaccharide-induced inflammation exacerbates tau pathol- TH, Zhang B et al (2016) IL-33 ameliorates Alzheimer’s disease-
ogy by a cyclin-dependent kinase 5-mediated pathway in a trans- like pathology and cognitive decline. Proc Natl Acad Sci U S A
genic model of Alzheimer’s disease. J Neurosci 25(39):8843–8853 113(19):E2705–E2713
33. Saha RN, Pahan K (2006) Signals for the induction of nitric oxide 51. Hill JM, Bhattacharjee S, Pogue AI, Lukiw WJ (2014) The gastro-
synthase in astrocytes. Neurochem Int 49(2):154–163 intestinal tract microbiome and potential link to Alzheimer’s dis-
34. Edison P, Archer HA, Gerhard A, Hinz R, Pavese N, Turkheimer ease. Front Neurol 5:43
FE, Hammers A, Tai YF et al (2008) Microglia, amyloid, and cog- 52. Kim BS, Jeon YS, Chun J (2013) Current status and future promise
nition in Alzheimer’s disease: an [11C](R)PK11195-PET and of the human microbiome. Pediatr Gastroenterol Hepatol Nutr
[11C]PIB-PET study. Neurobiol Dis 32(3):412–419 16(2):71–79
35. Carter SF, Schöll M, Almkvist O, Wall A, Engler H, Långström B, 53. Wang HX, Wang YP (2016) Gut microbiota-brain axis. Chin Med J
Nordberg A (2012) Evidence for astrocytosis in prodromal 129(19):2373–2380
Alzheimer disease provided by 11C-deuterium-L-deprenyl: a 54. Jenkins TA, Nguyen JC, Polglaze KE, Bertrand PP (2016)
multitracer PET paradigm combining 11C-Pittsburgh compound Influence of tryptophan and serotonin on mood and cognition with
B and 18F-FDG. J Nucl Med 53(1):37–46 a possible role of the gut-brain Axis. Nutrients 8(1)
36. Esposito G, Giovacchini G, Liow JS, Bhattacharjee AK, Greenstein 55. Schmidt C (2015) Mental health: thinking from the gut. Nature
D, Schapiro M, Hallett M, Herscovitch P et al (2008) Imaging 518(7540):S12–S15
neuroinflammation in Alzheimer’s disease with radiolabeled ara- 56. Smith PA (2015) The tantalizing links between gut microbes and
chidonic acid and PET. J Nucl Med 49(9):1414–1421 the brain. Nature 526(75573):312–314
37. Minett T, Classey J, Matthews FE, Fahrenhold M, Taga M, 57. Mayer EA, Knight R, Mazmanian SK, Cryan JF (2014) Gut mi-
Brayne C, Ince PG, Nicoll JA et al (2016) Microglial crobes and the brain: paradigm shift in neuroscience. J Neurosci
immunophenotype in dementia with Alzheimer’s pathology. J 34(46):15490–15496
Neuroinflammation 13(1):1–10 58. Pinto-Sanchez MI, Hall GB, Ghajar K, Nardelli A, Bolino C, Lau
38. Ching AS, Kuhnast B, Damont A, Roeda D, Tavitian B, Dolle F JT, Martin FP, Cominetti O et al (2017) Probiotic Bifidobacterium
(2012) Current paradigm of the 18-kDa translocator protein (TSPO) longum NCC3001 reduces depression scores and alters brain activ-
as a molecular target for PET imaging in neuroinflammation and ity: A pilot study in patients with irritable bowel syndrome.
neurodegenerative diseases. Insights Imaging 3(1):111–119 Gastroenterology 153(2):448–459
39. Jacobs AH, Tavitian B (2012) Noninvasive molecular imaging of 59. Tillisch K, Mayer E, Gupta A, Gill Z, Brazeilles R, Le Nevé B, van
neuroinflammation. J Cereb Blood Flow Metab 32(7):1393–1415 Hylckama Vlieg JET, Guyonnet D et al (2017) Brain structure and
40. Wiley CA, Lopresti BJ, Venneti S, Price J, Klunk WE, DeKosky response to emotional stimuli as related to gut microbial profiles in
ST, Mathis CA (2009) Carbon 11-labeled Pittsburgh compound B healthy women. Psychosom Med 79(8):905–913
and carbon 11-labeled (R)-PK11195 positron emission tomograph- 60. Sharon G, Sampson TR, Geschwind DH, Mazmanian SK (2016)
ic imaging in Alzheimer disease. Arch Neurol 66(1):60–67 The central nervous system and the gut microbiome. Cell 167(4):
41. Hamelin L, Lagarde J, Dorothée G, Leroy C, Labit M, Comley RA, 915–932
de Souza LC, Corne H et al (2016) Early and protective microglial 61. Ogbonnaya ES, Clarke G, Shanahan F, Dinan TG, Cryan JF,
activation in Alzheimer’s disease: a prospective study using 18F- O'Leary OF (2015) Adult hippocampal neurogenesis is regulated
DPA-714 PET imaging. Brain 139(Pt 4):1252–1264 by the microbiome. Biol Psychiatry 78(4):e7–e9
42. Imbimbo BP, Solfrizzi V, Panza F (2010) Are NSAIDs useful to 62. Luczynski P, Whelan SO, O'Sullivan C, Clarke G, Shanahan F,
treat Alzheimer’s disease or mild cognitive impairment? Front Dinan TG, Cryan JF (2016) Adult microbiota-deficient mice have
Aging Neurosci 2:19 distinct dendritic morphological changes: differential effects in the
43. Stewart WF, Kawas C, Corrada M, Metter EJ (1997) Risk of amygdala and hippocampus. Eur J Neurosci 44(9):2654–2666
Alzheimer’s disease and duration of NSAID use. Neurology 63. Erny D, Hrabě de Angelis AL, Jaitin D, Wieghofer P, Staszewski O,
48(3):626–632 David E, Keren-Shaul H, Mahlakoiv T et al (2015) Host microbiota
44. McGeer PL, Rogers J, McGeer EG (2016) Inflammation, constantly control maturation and function of microglia in the CNS.
antiinflammatory agents, and Alzheimer’s disease: the last 22 Nat Neurosci 18(7):965–977
years. J Alzheimers Dis 54(3):853–857 64. Matcovitch-Natan O, Winter DR, Giladi A, Vargas Aguilar S,
45. Zandi PP, Anthony JC, Hayden KM, Mehta K, Mayer L, Spinrad A, Sarrazin S, Ben-Yehuda H, David E et al (2016)
Breitner JC (2002) Reduced incidence of AD with NSAID but Microglia development follows a stepwise program to regulate
not H2 receptor antagonists: the Cache County study. brain homeostasis. Science 353(6301):aad8670
Neurology 59(6):880–886 65. Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Tóth
46. Yip AG, Green RC, Huyck M, Cupples LA, Farrer LA (2005) M, Korecka A, Bakocevic N et al (2014) The gut microbiota influ-
Nonsteroidal anti-inflammatory drug use and Alzheimer’s disease ences blood-brain barrier permeability in mice. Sci Transl Med
risk: the MIRAGE study. BMC Geriatr 5:2 6(263):263ra158
Mol Neurobiol

66. O'Brien SM, Scott LV, Dinan TG (2004) Cytokines: abnormalities 82. Alonso R, Pisa D, Marina AI, Morato E, Rábano A, Rodal I,
in major depression and implications for pharmacological treat- Carrasco L (2015) Evidence for fungal infection in cerebrospinal
ment. Hum Psychopharmacol 19(6):397–403 fluid and brain tissue from patients with amyotrophic lateral scle-
67. Dinan TG, Borre YE, Cryan JF (2014) Genomics of schizophre- rosis. Int J Biol Sci 11(5):546–558
nia: time to consider the gut microbiome? Mol Psychiatry 83. Harach T, Marungruang N, Duthilleul N, Cheatham V, Mc Coy KD,
19(12):1252–1257 Frisoni G, Neher JJ, Fåk F et al (2017) Reduction of Abeta amyloid
68. Nemani K, Hosseini Ghomi R, McCormick B, Fan X (2015) pathology in APPPS1 transgenic mice in the absence of gut micro-
Schizophrenia and the gut-brain axis. Prog Neuro- biota. Sci Rep 7:41802
Psychopharmacol Biol Psychiatry 56:155–160 84. Cappellano G, Carecchio M, Fleetwood T, Magistrelli L, Cantello
69. Rogers GB, Keating DJ, Young RL, Wong ML, Licinio J, R, Dianzani U, Comi C (2013) Immunity and inflammation in
Wesselingh S (2016) From gut dysbiosis to altered brain function neurodegenerative diseases. Am J Neurodegener Dis 2(2):89–107
and mental illness: mechanisms and pathways. Mol Psychiatry 85. Glass CK, Saijo K, Winner B, Marchetto MC, Gage FH (2010)
21(6):738–748 Mechanisms underlying inflammation in neurodegeneration. Cell
70. Neufeld KA, Foster JA (2009) Effects of gut microbiota on the 140(6):918–934
brain: implications for psychiatry. J Psychiatry Neurosci 34(3): 86. Koprich JB, Reske-Nielsen C, Mithal P, Isacson O (2008)
230–231 Neuroinflammation mediated by IL-1beta increases susceptibility
71. Grenham S, Clarke G, Cryan JF, Dinan TG (2011) Brain-gut- of dopamine neurons to degeneration in an animal model of
microbe communication in health and disease. Front Physiol 2:94 Parkinson's disease. J Neuroinflammation 5:8
72. Mayer EA, Tillisch K (2011) The brain-gut axis in abdominal pain
87. McCoy MK, Tansey MG (2008) TNF signaling inhibition in the
syndromes. Annu Rev Med 62(1):381–396
CNS: implications for normal brain function and neurodegenerative
73. Gareau MG (2016) Cognitive function and the microbiome. Int Rev
disease. J Neuroinflammation 5:45
Neurobiol 131:227–246
74. Gareau MG, Wine E, Rodrigues DM, Cho JH, Whary MT, Philpott 88. Zhao Y, Lukiw WJ (2015) Microbiome-generated amyloid and po-
DJ, Macqueen G, Sherman PM (2011) Bacterial infection causes tential impact on amyloidogenesis in Alzheimer’s disease (AD). J
stress-induced memory dysfunction in mice. Gut 60(3):307–317 Nat Sci 1(7)
75. Savignac HM, Kiely B, Dinan TG, Cryan JF (2014) Bifidobacteria 89. Zhao Y, Dua P, Lukiw WJ (2015) Microbial sources of amyloid and
exert strain-specific effects on stress-related behavior and physiol- relevance to amyloidogenesis and Alzheimer’s disease (AD). J
ogy in BALB/c mice. Neurogastroenterol Motil 26(11):1615–1627 Alzheimers Dis Parkinsonism 5(1):177
76. Ohland CL, Kish L, Bell H, Thiesen A, Hotte N, Pankiv E, Madsen 90. Köhler CA, Maes M, Slyepchenko A, Berk M, Solmi M, Lanctôt
KL (2013) Effects of Lactobacillus helveticus on murine behavior KL, Carvalho AF (2016) The gut-brain axis, including the
are dependent on diet and genotype and correlate with alterations in microbiome, leaky gut and bacterial translocation: mechanisms
the gut microbiome. Psychoneuroendocrinology 38(9):1738–1747 and pathophysiological role in Alzheimer’s disease. Curr Pharm
77. Tillisch K, Labus J, Kilpatrick L, Jiang Z, Stains J, Ebrat B, Des 22(40):6152–6166
Guyonnet D, Legrain-Raspaud S et al (2013) Consumption of 91. Lukiw WJ (2016) Bacteroides fragilis lipopolysaccharide and
fermented milk product with probiotic modulates brain activity. inflammatory signaling in Alzheimer’s disease. Front Microbiol
Gastroenterology 144:1394–1401 1401.e1–4 7:1544
78. Messaoudi M, Lalonde R, Violle N, Javelot H, Desor D, Nejdi A, 92. Schwartz K, Boles BR (2013) Microbial amyloids—functions and
Bisson JF, Rougeot C et al (2011) Assessment of psychotropic-like interactions within the host. Curr Opin Microbiol 16(1):93–99
properties of a probiotic formulation (Lactobacillus helveticus 93. Hufnagel DA, Tukel C, Chapman MR (2013) Disease to dirt: the
R0052 and Bifidobacterium longum R0175) in rats and human biology of microbial amyloids. PLoS Pathog 9(11):e1003740
subjects. Br J Nutr 105(5):755–764 94. Friedland RP (2015) Mechanisms of molecular mimicry involv-
79. Maheshwari P, Eslick GD (2015) Bacterial infection and Alzheimer’s ing the microbiota in neurodegeneration. J Alzheimers Dis
disease: a meta-analysis. J Alzheimers Dis 43(3):957–966 45(2):349–362
80. Agostini S, Clerici M, Mancuso R (2014) How plausible is a link 95. Chen SG, Stribinskis V, Rane MJ, Demuth DR, Gozal E,
between HSV-1 infection and Alzheimer’s disease? Expert Rev Roberts AM, Jagadapillai R, Liu R et al (2016) Exposure to
Anti-Infect Ther 12(3):275–278 the functional bacterial amyloid protein curli enhances alpha-
81. Alonso R, Pisa D, Marina AI, Morato E, Rabano A, Carrasco L synuclein aggregation in aged Fischer 344 rats and
(2014) Fungal infection in patients with Alzheimer’s disease. J Caenorhabditis elegans. Sci Rep 6:34477
Alzheimers Dis 41(1):301–311

You might also like