Abstract
Aging is an inevitable process that all individuals experience, of which the extent differs among individuals. It has been recognized as the risk factor of neurodegenerative diseases by affecting gut microbiota compositions, microglia, and cognition abilities. Aging‐induced changes in gut microbiota compositions have a critical role in orchestrating the morphology and functions of microglia through the gut‐brain axis. Gut microbiota communicates with microglia by its secreted metabolites and neurotransmitters. This is highly associated with age‐related cognitive declines. Here, we review the main composition of microbiota in the aged individuals, outline the changes of the brain in age‐related cognitive decline from a neuroinflammation perspective, especially the changes of morphology and functions of microglia, discuss the crosstalk between microbiota and microglia in the aged brain and further highlight the role of microbiota‐microglia connections in neurodegenerative diseases (Alzheimer's disease and Parkinson's disease).
Keywords: cognitive aging, gut microbiota, microglia, neuroinflammation
Microbiota‐microglia connection has been recognized in recent years. Aging‐induced changes of gut microbiota composition have a critical role in mediating the morphology and functions of microglia through gut‐brain axis, which is tightly related to age‐related cognitive decline. Here, we review the age‐related changes of gut microbiota and brain, and highlight the interaction between microbiota and microglia in the aged brain.
Abbreviations
- Ach
acetylcholine
- AD
Alzheimer’s disease
- Aβ
amyloid‐beta
- BBB
blood–brain barrier
- BMDMs
bone marrow‐derived macrophages
- CCL3
C‐C motif ligand 3
- CNS
central nervous system
- CSF1R
colony stimulating factor 1 receptor
- DAMPs
danger‐associated molecular patterns
- ERK
extracellular signal‐regulated kinase
- FFAR2
SCFA receptor‐free fatty acid receptor 2
- FTL
ferritin light chain
- GABA
gamma‐aminobutyric acid
- GI
gastrointestinal
- HFD
high‐fat diet
- HiPR‐FISH
high‐phylogenetic‐resolution microbiota mapping by fluorescence in situ hybridization
- IL‐17A
interleukin‐17A
- IL‐1R1
interleukin‐1 receptor 1
- JNK
c‐Jun N‐terminal kinase
- LPS
lipopolysaccharide
- MAMPs
microbial‐associated molecular patterns
- MCTs
monocarboxylate transporters
- MDP
muramyl dipeptide
- MHC‐II
major histocompatibility complex class II
- NE
norepinephrine
- NF‐kB
nuclear factor‐kappa B
- NFTs
neurofibrillary tangles
- NLR3
NLR family pyrin domain containing 3
- PAMPs
pathogen‐associated molecular patterns
- PD
Parkinson’s disease
- PET
positron emission tomography
- PRPs
pattern recognition receptors
- ROS
reactive oxygen species
- SCFAs
short‐chain fatty acids
- Smad3
mothers against decapentaplegic homolog 3
- SMCT
sodium‐dependent monocarboxylate transporter
- ST‐seq
spatial transcriptomic sequencing
- TGF‐β1
transforming growth factor β
- TLRs
toll‐like receptors
- Tregs
regulatory T cells
1. INTRODUCTION
Aging is defined as time‐dependent decrease of functions, which all the mammalian on earth will go through. Since the establishment of modern society, human beings, either from western or eastern societies, have struggled to discover a way to achieve longer life. In the past decades, the lifespan of citizens has been largely extended. Following this, the growing speed of the aging population is much faster than before, and the proportion of aged people over 60 years will increase to 22% at the end of 2050 (Organization, 2015).
Due to progressive loss of physiological function during aging, the human body will undergo a variety of biological and psychological changes, such as genomic instability, mitochondrial dysfunction, and athletic ability decline (Green et al., 2011; Moskalev et al., 2013; Stones, 2019). Brain, as the main part of the central nervous system (CNS), also suffers structural and molecular changes with increasing age (Fjell & Walhovd, 2010). The hallmarks of brain aging include impaired neuronal network activity, dysregulated glial cell activation, stem cell exhaustion, and dysregulated metabolic activity (Mattson & Arumugam, 2018). These accelerated alterations induce cognitive decline in old people, which might lead to neurodegenerative diseases, such as Parkinson's disease (PD) and Alzheimer's disease (AD) (Baldwin & Greenwood, 2020; Goldberg et al., 2018).
Microglia are a group of neuroglia that account for 5–15% of total brain cells (Pelvig et al., 2008). As the resident‐macrophage cells, microglia function as the main immune defense in the CNS. To sustain brain homeostasis, microglia continually surveille the brain microenvironment through their connections with neighboring cells and factors (D. Li et al., 2018). During aging, microglia switch from resting state to activated state and contribute to the development of neurogenerative diseases (Johnson et al., 2020). Activated microglia perform in an ameboid morphology, produce pro‐inflammatory cytokines, and participate in regulating blood–brain barrier (BBB) integrity and synaptic plasticity in aged brain (Daria et al., 2017; Phi T Nguyen et al., 2020; Yousef et al., 2019).
Recent studies suggested that the alterations of gut microbiota in the aged are associated with neurodegenerative diseases (Fujii et al., 2019). Gut‐brain axis indicates the complicated connections between gut and brain, which is crucial for microglial maturation and function (Erny et al., 2015; Fung et al., 2017). These findings pave a new way in attenuating and even reversing cognitive aging through microbiota‐microglia axis intervention. In this review, we will review the composition of gut microbiota in aged individuals, depict the changes of microglia associated with aging and discuss neuroinflammation in the aged brain. We then summarize the mechanism of microbiota in regulating microglial function in the aged brain and highlight the role of microbiota‐microglia connections in neurodegenerative diseases. This knowledge may enrich our understanding of the crosstalk between aging‐related cognitive decline and the microbiota‐microglia axis, facilitating the discovery of novel targets in restoring aging‐related cognitive decline.
2. THE COMPOSITION OF GUT MICROBIOTA IN THE AGED
2.1. Perturabation of Gut microbiota in the Adult
Currently, 2172 microorganism species, which are classified into 12 different phyla, have been identified in human beings (Hugon et al., 2015). Among them, 93.5% are occupied by Proteobacteria, Firmicutes, Actinobacteria, and Bacteroidetes (Thursby & Juge, 2017). In adults, gut microbiota communities remain relatively stable, while it could be disturbed by mental state, diet, pharmaceuticals, geography, and lifecycle stages.
The altered mental state is recognized as one of the main factors which are associated with gut dysbiosis. Recent evidence showed that anxiety is linked with unbalanced gut microorganisms and intestinal barrier disruption (Bruce R Stevens et al., 2018). Collected feces from human showed that Acidaminococcaceae, Enterobacteriaceae, Rikenellaceae, and Coriobacteriaceae families abundance are increased in patients with depression (Jiang et al., 2015; Stevens et al., 2020). Another systematic review showed that nine genera of bacteria were significantly abundant (Anaerostipes, Blautia, Clostridium, Klebsiella, Lachnospiraceae incertae sedis, Parabacteroides, Parasutterella, Phascolarctobacterium, and Streptococcus) in major depression disorders (Cheung et al., 2019). These species are mainly involved in butyrate and gamma‐aminobutyric acid (GABA) degradation pathways in depression (Valles‐Colomer et al., 2019). In the rodent model, chronic psychological stress induces translocation of E. Coli into enterocytes by more than 30 folds and enhanced permeability of HRP in enterocytes (Velin et al., 2004). Besides, psychological stress combined with filamentous bacteria inducing interleukin‐17A (IL‐17A) increases the gut permeability and promotes vaso‐occlusive episodes in patients with sickle cell disease (Xu et al., 2020). Antidepressant minocycline alleviates depressive behavior and increases abundance of Lachnospiraceae and Clostridiales Family XIII Families, which is known for butyrate production (Schmidtner et al., 2019). Moreover, gut microbiota dysbiosis‐induced damage of the intestinal barrier may make the gastrointestinal tract vulnerable to inflammation, which results in a feed‐forward cycle between gastrointestinal inflammation and barrier dysfunction and may induce the development of depression. A human study indicated that plasma level of lipopolysaccharide (LPS) endotoxin secreted by microbiota was elevated in patients with anxiety and depression and correlated with gastrointestinal barrier dysfunction, which might lead to anxiety and depression (Bruce R. Stevens et al., 2018).
The diet is another factor that alters the composition of gut microbiota. A high‐fat diet (HFD) is defined as a diet rich in fats, especially saturated fats, and is becoming a common lifestyle in human beings. Though numerous studies including rodent models and human have investigated the influence of HFD on microbiota communities, a meta‐analysis indicated that only the ratio of Firmicutes to Bacteroidetes was continuously increased in the 27 enrolled studies (Bisanz et al., 2019). Interestingly, a murine study showed that increased soluble fiber rather than fat in the diet significantly results in loss of the phylum Bacteroidetes and rise of Clostridia and Proteobacteria (Morrison et al., 2020).
2.2. Changed composition of gut microbiota in the elderly
The gastrointestinal tract in the elderly is more vulnerable to physiological changes and risk factors (Camilleri et al., 2000). Given the potential of gut microbiota to increase gastrointestinal permeability and influence gut inflammation through its secretion, considerable interest has appeared in the variation of gut microbiota composition at an advanced age. Generally, the diversity and stability of gut microbiota are declined during aging (Table 1), indicating gut dysbiosis in the elderly (Kong et al., 2019; Lynch & Pedersen, 2016). In the elderly, phylum Bacteroidetes, Faecalibacterium spp., and Clostridium IV are relatively abundant, while phylum Firmicutes, especially Clostridium XIVa, are more prevalent in the younger (Marcus J Claesson et al., 2011).
TABLE 1.
Taxon (Phylum/Genus) | Trend | Country | Subjects amount | Composition of subjects | Method (Fecal Samples) | References | |
---|---|---|---|---|---|---|---|
Unhealthy Aging | |||||||
Actinobacteria | Bifidobacterium | ↓ | Spain | 153 subjects | 49 adults (ages <50 years), 58 elderly (ages 50–65 years), 46 old (ages 66–80 (n = 19), and >80 (n = 27) years) subjects | qPCR | Salazar et al. (2019) |
↓ | Thailand | 120 subjects | 73 adults (ages 30–40 years) and 47 elderly (ages ≥65 years) subjects | 16S rRNA sequencing | La‐Ongkham et al. (2020) | ||
Bacteroidetes | Alistipes | ↑ | Ireland | 170 subjects | 161 elderly (ages ≥65 years), 9 control (ages 28–46 years) subjects | 16S rRNA sequencing | M. J. Claesson et al. (2011) |
Bacteroides | ↑ | Finland | 32 subjects | 9 elderly non‐steroidal anti‐inflammatory drugs (NSAID) users (ages 77–85 years), 9 elderly non‐users (ages 70–83 years), and 14 young adults (ages 21–39 years) | %G + C profiling/16S rDNA sequencing | Mäkivuokko et al. (2010) | |
↑ | Ireland | 170 subjects | 161 elderly (ages ≥65 years), 9 control (ages 28–46 years) subjects | 16S rRNA sequencing | M. J. Claesson et al. (2011) | ||
↑ | Thailand | 120 subjects | 73 adults (ages 30–40 years) and 47 elderly (ages ≥65 years) subjects | 16S rRNA sequencing | La‐Ongkham et al. (2020) | ||
↓ | India | 54 subjects | 12 young (ages 3–15 years), 18 elderly (ages 25–40 years) and 24 old (ages >50 years) | 16S rRNA sequencing | Chaudhari et al. (2020) | ||
↓ | Spain | 76 subjects | 38 old subjects in retirement homes (ages 77–95 years), 38 elderly subjects in their own homes (ages 57–67 years) | qPCR | Salazar et al. (2013) | ||
↓ | Spain | 153 subjects | 49 adults (ages <50 years), 58 elderly (ages 50–65 years), 46 old (ages 66–80 (n = 19), and >80 (n = 27) years) subjects | qPCR | Salazar et al. (2019) | ||
Parabacteroides | ↑ | Thailand | 120 subjects | 73 adults (ages 30–40 years) and 47 elderly (ages ≥65 years) subjects | 16S rRNA sequencing | La‐Ongkham et al. (2020) | |
Firmicutes/Bacteroidetes | ↓ | Thailand | 120 subjects | 73 adults (ages 30–40 years) and 47 elderly (ages ≥65 years) subjects | 16S rRNA sequencing | La‐Ongkham et al. (2020) | |
Firmicutes | Clostridium | ↑ | Finland | 32 subjects | 9 elderly NSAID users (ages 77–85 years), 9 elderly non‐users (ages 70–83 years), and 14 young adults (ages 21–39 years) | %G + C profiling/16S rDNA sequencing | Mäkivuokko et al. (2010) |
↓ | Spain | 76 subjects | 38 old subjects in retirement homes (ages 77–95 years), 38 elderly subjects in their own homes (ages 57–67 years) | qPCR | Salazar et al. (2013) | ||
Coprobacillus | ↓ | Finland | 32 subjects | 9 elderly NSAID users (ages 77–85 years), 9 elderly non‐users (ages 70–83 years), and 14 young adults (ages 21–39 years) | %G + C profiling/16S rDNA sequencing | Mäkivuokko et al. (2010) | |
Dialister | ↓ | Finland | 32 subjects | 9 elderly NSAID users (ages 77–85 years), 9 elderly non‐users (ages 70–83 years), and 14 young adults (ages 21–39 years) | %G + C profiling/16S rDNA sequencing | Mäkivuokko et al. (2010) | |
Dorea | ↓ | Thailand | 120 subjects | 73 adults (ages 30–40 years) and 47 elderly (ages ≥65 years) subjects | 16S rRNA sequencing | La‐Ongkham et al. (2020) | |
Faecalibacterium | ↑ | Ireland | 170 subjects | 161 elderly (ages ≥65 years), 9 control (ages 28–46 years) subjects | 16S rRNA sequencing | M. J. Claesson et al. (2011) | |
↓ | Spain | 153 subjects | 49 adults (ages <50 years), 58 elderly (ages 50–65 years), 46 old (ages 66–80 (n = 19), and >80 (n = 27) years) subjects | qPCR | Salazar et al. (2019) | ||
↓ | Spain | 76 subjects | 38 old subjects in retirement homes (ages 77–95 years), 38 elderly subjects in their own homes (ages 57–67 years) | qPCR | Salazar et al. (2013) | ||
Lactobacillus | ↑ | Finland | 32 subjects | 9 elderly NSAID users (ages 77–85 years), 9 elderly non‐users (ages 70–83 years), and 14 young adults (ages 21–39 years) | %G + C profiling/16S rDNA sequencing | Mäkivuokko et al. (2010) | |
↑ | Spain | 76 subjects | 38 old subjects in retirement homes (ages 77–95 years), 38 elderly subjects in their own homes (ages 57–67 years) | qPCR | Salazar et al. (2013) | ||
Lachnospiraceae | ↓ | Ireland | 170 subjects | 161 elderly (ages ≥65 years), 9 control (ages 28–46 years) subjects | 16S rRNA sequencing | M. J. Claesson et al. (2011) | |
Oscillospira | ↑ | Finland | 32 subjects | 9 elderly NSAID users (ages 77–85 years), 9 elderly non‐users (ages 70–83 years), and 14 young adults (ages 21–39 years) | %G + C profiling/16S rDNA sequencing | Mäkivuokko et al. (2010) | |
Roseburia | ↓ | Finland | 32 subjects | 9 elderly NSAID users (ages 77–85 years), 9 elderly non‐users (ages 70–83 years), and 14 young adults (ages 21–39 years) | %G + C profiling/16S rDNA sequencing | Mäkivuokko et al. (2010) | |
Ruminococcus | ↓ | Finland | 32 subjects | 9 elderly NSAID users (ages 77–85 years), 9 elderly non‐users (ages 70–83 years), and 14 young adults (ages 21–39 years) | %G + C profiling/16S rDNA sequencing | Mäkivuokko et al. (2010) | |
Sporobacter | ↑ | Finland | 32 subjects | 9 elderly NSAID users (ages 77–85 years), 9 elderly non‐users (ages 70–83 years), and 14 young adults (ages 21–39 years) | %G + C profiling/16S rDNA sequencing | Mäkivuokko et al. (2010) | |
Streptococcus | ↑ | Finland | 32 subjects | 9 elderly NSAID users (ages 77–85 years), 9 elderly non‐users (ages 70–83 years), and 14 young adults (ages 21–39 years) | %G + C profiling/16S rDNA sequencing | Mäkivuokko et al. (2010) | |
Verrucomicrobia | Akkermansia | ↑ | Spain | 76 subjects | 38 old subjects in retirement homes (ages 77–95 years), 38 elderly subjects in their own homes (ages 57–67 years) | qPCR | Salazar et al. (2013) |
The composition of gut microbiota in the elderly is disturbed by several factors, such as residence situation, exercise, nutrition, and age‐associated alterations. The residence location of the elderly may affect the composition of gut microbiota. The elderly who received long‐term residential care showed an increased proportion of phylum Bacteroidetes, genera Parabacteroides, Eubacterium, Anaerotruncus, Lactonifactor, and Coprobacillus compared with the community‐dwelling elderly (Claesson et al., 2012). Meanwhile, the movement was associated with the changed composition of the elderly with a reduced proportion of Ruminococcus and Prevotella and increased Oscillibacter (Claesson et al., 2012). Age‐induced deterioration in anatomical barriers and physiological functions also involves in the composition adjustment of gut microbiota in the elderly. In older individuals, elevated pro‐inflammatory factors, such as IL‐6, have a direct impact on intestinal permeability accompanied with decreased anti‐inflammatory factors (Nicoletti, 2015). The impaired intestinal barrier leads to an increased proportion of LPS‐secreting bacteria, such as Bacteroides and Proteobacteria (Kumar et al., 2016; Schiffrin et al., 2010). Apart from anatomical changes in the gastrointestinal (GI) tract during aging, deterioration in digestive function, such as occlusal force, muscle thickness, swallowing function, and secretive function, could lead to less fiber‐containing foods, which is a risk of malnutrition (Amarya et al., 2015). Malnutrition is reported to be related to an abundant subpopulation of Clostridiales (Jeffery et al., 2016). Immunity, as an important force in defending pathogens, has been shown a role in regulating gut microbiota (Vijay‐Kumar et al., 2010). Zhang et al. revealed that lack of adaptive immune system in mice resulted in elevated diversity and evenness of gut microbiota with an increase in age, which performed as an increased abundance of A. muciniphila colonization (H. Zhang et al., 2015). More evidence is required to reveal the role of the immune system in regulating gut microbiota during aging.
Centenarians are a group of people who have an extended lifespan than the normal elderly. The development of the next‐generation sequencing approach sheds a light on describing the composition of gut microbiota in the centenarians. Recently, numerous studies indicated that the composition of centenarians is different from the elderly and younger adults (Table 2). In a study from Italy, analysis of 21 centenarians revealed increased abundance of Bacilli, Bacteroidetes, and Proteobacteria compared with the elderly (Biagi et al., 2010). Meanwhile, microbiota sequencing of different ages (young adults, younger elderly, centenarians, and semisupercentenarians) indicated a decreased trend in the abundance of Coprococcus, Roseburia, and Faecalibacterium (E. Biagi et al., 2016). It is worth noting that the abundance of Akkermansia and Escherichia was significantly increased in the centenarians (E. Biagi et al., 2016). Akkermansia, known as the short‐chain fatty acids (SCFAs) producers, plays a critical role in anti‐inflammatory (Elena Biagi et al., 2016; Fanli Kong et al., 2016). This result was also confirmed by studies from China and Korea, which indicated the important role of these bacteria in the centenarians (B. S. Kim et al., 2019; F. Kong et al., 2016). An increased proportion of specific group of gut microbiota in centenarians might be associated with anti‐inflammatory effects and extended lifespan.
TABLE 2.
Taxon (Phylum/Genus) | Trend | Country | Subjects amount | Subjects composition | Method (Fecal Samples) | References | |
---|---|---|---|---|---|---|---|
Healthy Aging | |||||||
Actinobacteria | Bifidobacterium | ↓ | Italy | 9 samples | 3 centenarians (ages 99–102 years), 5 elderly (ages 59–75 years) and 1 adult (age 38 years) | Shotgun metagenomic sequencing | Rampelli et al. (2013) |
↑ | Italy | 65 subjects | 21 old (ages 99–107 years), 25 elderly (ages 68–88 years), 19 young (ages 21–33 years) subjects | DNA library construction/Shotgun metagenomic sequencing | Wu et al. (2019) | ||
↑ | Italy | 39 subjects, 39 samples | 24 semisupercentenarians (ages 105–109 years), 15 centenarians (ages 99–104 years), 15 elderly (ages 65–75 years), 15 young (ages 22–48 years) | 16S rRNA sequencing | E. Biagi et al. (2016) | ||
↓ | China | 1195 subjects | 86 elderly, 198 centenarians | 16S rRNA sequencing | Bian et al. (2017) | ||
↓ | Italy | 84 subjects | 21 centenarians (ages 99–104 years), 22 elderly (ages 63–76 years), 20 young (ages 25–40 years), 21 elderly (59–78 years, offspring of the centenarians) subjects | HITChip and quantitative PCR of 16S rRNA genes | Biagi et al. (2010) | ||
↓ | Japan | 367 subjects | ages between 0–104 years | 16S rRNA sequencing | Odamaki et al. (2016) | ||
Collinsella | ↑ | Korea | 56 subjects | 30 centenarians (ages 95–108 years), 17 elderly subjects (ages 67–79 years), 9 adults (ages 26–43 years) in longevity villages | 16S rRNA sequencing | B. S. Kim et al. (2019) | |
Eggerthella | ↑ | Italy | 84 subjects | 21 centenarians (ages 99–104 years), 22 elderly (ages 63–76 years), 20 young (ages 25–40 years), 21 elderly (59–78 years, offspring of the centenarians) subjects | HITChip and quantitative PCR of 16S rRNA genes | Biagi et al. (2010) | |
↑ | Italy | 62 samples | 11 young (ages 22–48 years), 13 elderly (ages 65–75 years), 15 centenarians (ages 99–104 years), and 23 semisupercentenarians (ages 105–109 years) subjects | Shotgun metagenomic sequencing | Rampelli et al. (2020) | ||
Bacteroidetes | Bacteroides | ↑ | Japan | 367 subjects | ages between 0 and 104 years | 16S rRNA sequencing | Odamaki et al. (2016) |
↑ | China | 1195 subjects | 86 elderly, 198 centenarians | 16S rRNA sequencing | Bian et al. (2017) | ||
↓ | Italy | 62 samples | 11 young (ages 22–48 years), 13 elderly (ages 65–75 years), 15 centenarians (ages 99–104 years), and 23 semisupercentenarians (ages 105–109 years) subjects | Shotgun metagenomic sequencing | Rampelli et al. (2020) | ||
Parabacteroides | ↑ | Italy | 65 subjects | 19 young (ages 19–33 years), 24 elderly (ages 68–88 years), 22 centenarians (ages >100 years) subjects | 16S rRNA sequencing/ITS1 sequencing | Wu et al. (2020) | |
Prevotella | ↓ | Italy | 65 subjects | 19 young (ages 19–33 years), 24 elderly (ages 68–88 years), 22 centenarians (ages >100 years) subjects | 16S rRNA sequencing/ITS1 sequencing | Wu et al. (2020) | |
↓ | Italy | 65 subjects | 21 old (ages 99–107 years), 25 elderly (ages 68–88 years), 19 young (ages 21–33 years) subjects | DNA library construction/shotgun metagenomic sequencing | Wu et al. (2019) | ||
↑ | China | 1195 subjects | 86 elderly, 198 centenarians | 16S rRNA sequencing | Bian et al. (2017) | ||
↓ | Korea | 56 subjects | 30 centenarians (ages 95–108 years), 17 elderly subjects (ages 67–79 years), 9 adults (ages 26–43 years) in longevity villages | 16S rRNA sequencing | B. S. Kim et al. (2019) | ||
Euryarchaeota | Methanobrevibacter | ↑ | Italy | 65 subjects | 21 old (ages 99–107 years), 25 elderly (ages 68–88 years), 19 young (ages 21–33 years) subjects | DNA library construction/shotgun metagenomic sequencing | Wu et al. (2019) |
↑ | Italy | 62 samples | 11 young (ages 22–48 years), 13 elderly (ages 65–75 years), 15 centenarians (ages 99–104 years), and 23 semisupercentenarians (ages 105–109 years) subjects | Shotgun metagenomic sequencing | Rampelli et al. (2020) | ||
Firmicutes | Bacillus | ↑ | Italy | 84 subjects | 21 centenarians (ages 99–104 years), 22 elderly (ages 63–76 years), 20 young (ages 25–40 years), 21 elderly (59–78 years, offspring of the centenarians) subjects | HITChip and quantitative PCR of 1S rRNA genes | Biagi et al. (2010) |
Clostridium | ↑ | China | 1195 subjects | 86 elderly, 198 centenarians | 16S rRNA sequencing | Bian et al. (2017) | |
↑ | Korea | 56 subjects | 30 centenarians (ages 95–108 years), 17 elderly subjects (ages 67–79 years), 9 adults (ages 26–43 years) in longevity villages | 16S rRNA sequencing | B. S. Kim et al. (2019) | ||
↓ | Italy | 84 subjects | 21 centenarians (ages 99–104 years), 22 elderly (ages 63–76 years), 20 young (ages 25–40 years), 21 elderly (59–78 years, offspring of the centenarians) subjects | HITChip and quantitative PCR of 16S rRNA genes | Biagi et al. (2010) | ||
Coprococcus | ↓ | Italy | 65 subjects | 21 old (ages 99–107 years), 25 elderly (ages 68–88 years), 19 young (ages 21–33 years) subjects | DNA library construction/shotgun metagenomic sequencing | Wu et al. (2019) | |
↓ | Italy | 62 samples | 11 young (ages 22–48 years), 13 elderly (ages 65–75 years), 15 centenarians (ages 99–104 years), and 23 semisupercentenarians (ages 105–109 years) subjects | Shotgun metagenomic sequencing | Rampelli et al. (2020) | ||
Dorea | ↑ | China | 1195 subjects | 86 elderly, 198 centenarians | 16S rRNA sequencing | Bian et al. (2017) | |
↓ | Italy | 65 subjects | 21 old (ages 99–107 years), 25 elderly (ages 68–88 years), 19 young (ages 21–33 years) subjects | DNA library construction/shotgun metagenomic sequencing | Wu et al. (2019) | ||
Roseburia | ↓ | Italy | 84 subjects | 21 centenarians (ages 99–104 years), 22 elderly (ages 63–76 years), 20 young (ages 25–40 years), 21 elderly (59–78 years, offspring of the centenarians) subjects | HITChip and quantitative PCR of 16S rRNA genes | Biagi et al. (2010) | |
Ruminococcus | ↓ | Italy | 65 subjects | 19 young (ages 19–33 years), 24 elderly (ages 68–88 years), 22 centenarians (ages >100 years) subjects | 16S rRNA sequencing/ITS1 sequencing | Wu et al. (2020) | |
↑ | Italy | 9 samples | 3 centenarians (ages 99–102 years), 5 elderly (ages 59–75 years) and 1 adult (age 38 years) | Shotgun metagenomic sequencing | Rampelli et al. (2013) | ||
↓ | Italy | 84 subjects | 21 centenarians (ages 99–104 years), 22 elderly (ages 63–76 years), 20 young (ages 25–40 years), 21 elderly (59–78 years, offspring of the centenarians) subjects | HITChip and quantitative PCR of 16S rRNA genes | Biagi et al. (2010) | ||
↓ | Italy | 65 subjects | 21 old (ages 99–107 years), 25 elderly (ages 68–88 years), 19 young (ages 21–33 years) subjects | DNA library construction/shotgun metagenomic sequencing | Wu et al. (2019) | ||
Eubacterium | ↓ | Italy | 9 samples | 3 centenarians (ages 99–102 years), 5 elderly (ages 59–75 years), and 1 adult (age 38 years) | Shotgun metagenomic sequencing | Rampelli et al. (2013) | |
↓ | Italy | 84 subjects | 21 centenarians (ages 99–104 years), 22 elderly (ages 63–76 years), 20 young (ages 25–40 years), 21 elderly (59–78 years, offspring of the centenarians) subjects | HITChip and quantitative PCR of 16S rRNA genes | Biagi et al. (2010) | ||
↓ | Italy | 65 subjects | 21 old (ages 99–107 years), 25 elderly (ages 68–88 years), 19 young (ages 21–33 years) subjects | DNA library construction/shotgun metagenomic sequencing | Wu et al. (2019) | ||
↓ | Italy | 62 samples | 11 young (ages 22–48 years), 13 elderly (ages 65–75 years), 15 centenarians (ages 99–104 years), and 23 semisupercentenarians (ages 105–109 years) subjects | Shotgun metagenomic sequencing | Rampelli et al. (2020) | ||
Faecalibacterium | ↓ | Italy | 9 samples | 3 centenarians (ages 99–102 years), 5 elderly (ages 59–75 years), and 1 adult (age 38 years) | Shotgun metagenomic sequencing | Rampelli et al. (2013) | |
↑ | China | 1195 subjects | 86 elderly, 198 centenarians | 16S rRNA sequencing | Bian et al. (2017) | ||
↓ | China | 67 subjects | centenarians and nonagenarians (ages ≥90 years) | 16S rRNA sequencing | Kong et al. (2019) | ||
↓ | Italy | 84 subjects | 21 centenarians (ages 99–104 years), 22 elderly (ages 63–76 years), 20 young (ages 25–40 years), 21 elderly (59–78 years, offspring of the centenarians) subjects | HITChip and quantitative PCR of 16S rRNA genes | Biagi et al. (2010) | ||
↓ | Italy | 65 subjects | 21 old (ages 99–107 years), 25 elderly (ages 68–88 years), 19 young (ages 21–33 years) subjects | DNA library construction/shotgun metagenomic sequencing | Wu et al. (2019) | ||
↓ | Italy | 62 samples | 11 young (ages 22–48 years), 13 elderly (ages 65–75 years), 15 centenarians (ages 99–104 years), and 23 semisupercentenarians (ages 105–109 years) subjects | Shotgun metagenomic sequencing | Rampelli et al. (2020) | ||
↓ | Korea | 56 subjects | 30 centenarians (ages 95–108 years), 17 elderly subjects (ages 67–79 years), 9 adults (ages 26–43 years) in longevity villages | 16S rRNA sequencing | B. S. Kim et al. (2019) | ||
Lachnospiraceae | ↓ | Japan | 367 subjects | ages between 0 and 104 years | 16S rRNA sequencing | Odamaki et al. (2016) | |
Lactobacillus | ↑ | Italy | 65 subjects | 19 young (ages 19–33 years), 24 elderly (ages 68–88 years), 22 centenarians (ages >100 years) subjects | 16S rRNA sequencing | Wu et al. (2020) | |
↑ | Spain | 153 subjects | 49 adults (ages <50 years), 58 elderly (ages 50–65 years), 46 old (ages 66–80 (n = 19), and >80 (n = 27) years) subjects | qPCR | Salazar et al. (2019) | ||
Marvinbryantia | ↑ | China | 1195 subjects | 86 elderly, 198 centenarians | 16S rRNA sequencing | Bian et al. (2017) | |
Proteobacteria | Desulfovibrio | ↑ | Italy | 65 subjects | 21 old (ages 99–107 years), 25 elderly (ages 68–88 years), 19 young (ages 21–33 years) subjects | DNA library construction/shotgun metagenomic sequencing | Wu et al. (2019) |
Escherichia/Shigella | ↑ | China | 67 subjects | centenarians and nonagenarians (ages ≥90 years) | 16S rRNA sequencing | Kong et al. (2019) | |
Escherichia | ↑ | Italy | 9 samples | 3 centenarians (ages 99–102 years), 5 elderly (ages 59–75 years) and 1 adult (age 38 years) | Shotgun metagenomic sequencing | Rampelli et al. (2013) | |
↑ | Italy | 62 samples | 11 young (ages 22–48 years), 13 elderly (ages 65–75 years), 15 centenarians (ages 99–104 years), and 23 semisupercentenarians (ages 105–109 years) subjects | Shotgun metagenomic sequencing | Rampelli et al. (2020) | ||
↑ | Korea | 56 subjects | 30 centenarians (ages 95–108 years), 17 elderly subjects (ages 67–79 years), 9 adults (ages 26–43 years) in longevity villages | 16S rRNA sequencing | B. S. Kim et al. (2019) | ||
Klebsiella | ↑ | Italy | 84 subjects | 21 centenarians (ages 99–104 years), 22 elderly (ages 63–76 years), 20 young (ages 25–40 years), 21 elderly (59–78 years, offspring of the centenarians) subjects | HITChip and quantitative PCR of 16S rRNA genes | Biagi et al. (2010) | |
Vibrio | ↑ | Italy | 84 subjects | 21 centenarians (ages 99–104 years), 22 elderly (ages 63–76 years), 20 young (ages 25–40 years), 21 elderly (59–78 years, offspring of the centenarians) subjects | HITChip and quantitative PCR of 16S rRNA genes | Biagi et al. (2010) | |
Verrucomicrobia | Akkermansia | ↑ | Italy | 84 subjects | 21 centenarians (ages 99–104 years), 22 elderly (ages 63–76 years), 20 young (ages 25–40 years), 21 elderly (59–78 years, offspring of the centenarians) subjects | HITChip and quantitative PCR of 16S rRNA genes | Biagi et al. (2010) |
↑ | Italy | 39 subjects, 39 samples | 24 semisupercentenarians (ages 105–109 years), 15 centenarians (ages 99–104 years), 15 elderly (ages 65–75 years), 15 young (ages 22–48 years) | 16S rRNA sequencing | E. Biagi et al. (2016) | ||
↑ | Italy | 62 samples | 11 young (ages 22–48 years), 13 elderly (ages 65–75 years), 15 centenarians (ages 99–104 years), and 23 semisupercentenarians (ages 105–109 years) subjects | Shotgun metagenomic sequencing | Rampelli et al. (2020) | ||
↑ | China | 168 subjects | 67 centenarians and nonagenarians, 54 elderly, and 47 young adults | 16S rRNA sequencing | F. Kong et al. (2016) | ||
↑ | Korea | 56 subjects | 30 centenarians (ages 95–108 years), 17 elderly subjects (ages 67–79 years), 9 adults (ages 26–43 years) in longevity villages | 16S rRNA sequencing | B. S. Kim et al. (2019) | ||
↑ | Spain | 153 subjects | 49 adults (ages <50 years), 58 elderly (ages 50–65 years), 46 old (ages 66–80 (n = 19), and >80 (n = 27) years) subjects | qPCR | Salazar et al. (2019) | ||
Synergistetes | Cloacibacillus | ↑ | Italy | 65 subjects | 19 young (ages 19–33 years), 24 elderly (ages 68–88 years), 22 centenarians (ages >100 years) subjects | 16S rRNA sequencing/ITS1 sequencing | Wu et al. (2020) |
Pyramidobacter | ↑ | Italy | 65 subjects | 21 old (ages 99–107 years), 25 elderly (ages 68–88 years), 19 young (ages 21–33 years) subjects | DNA library construction/shotgun metagenomic sequencing | Wu et al. (2019) |
3. ALTERATIONS IN MICROGLIA DURING AGE‐RELATED COGNITIVE DECLINE
Cognitive aging is an inevitable and natural process due to the physiological decline during aging (Harada et al., 2013). It should be distinguished from mild cognitive impairment, dementia, and other specific cognitive decline syndromes. However, not all mental abilities are declined during aging. Verbal ability, some numerical abilities, and semantic knowledge are stable until the late life (Hedden & Gabrieli, 2004). Conversely, mental functions related to memory, such as working memory, episodic memory, and problem‐solving, have a tendency to decline in the aged (Craik, 1994). Alterations in morphology and functions of microglia and followed neuroinflammation in the brain could lead to age‐related cognitive decline.
3.1. Age‐related changes in morphology of microglia
Microglia, a group of resident immune cells in the brain, play a critical role in mediating neurogenesis, remodeling synapses, and regulating neuroinflammatory (Phi T. Nguyen et al., 2020; Pelvig et al., 2008; Wolfgang J. Streit et al., 2004). Recently, increasing studies indicated that microglia participated in several neurodegenerative diseases (V. Hugh Perry et al., 2010). At the stage of adult, microglia remain relatively homeostasis and the state of microglia is frequently mediated by factors existed in microenvironment of CNS (Van Rossum & Hanisch, 2004).
During the old age, obviously, morphological changes could be observed in microglia. Dystrophic microglia are a common group of microglia abundantly identified in aged human beings. A study including two nondemented individuals (38 and 68 years old) showed that dystrophic microglia presented with decreased fine branches, shortened tortuous processes, and/or formation of spheroids accompanied with significantly increased amount of dystrophic microglia in the aged brain (Wolfgang J. Streit et al., 2004). Similarly, Davies et al. observed reduced arborized area and length of processes in microglia during aging (Davies et al., 2017). With the progress of senescence, microglia show discontinuous processes and completely separate fragments of microglial cytoplasm under Iba‐1 staining in the AD cases (Davies et al., 2017; Tischer et al., 2016). Dystrophic microglia are also found to be close to Aβ plaques in postmortem AD patients (W. J. Streit et al., 2009). The markedly changed morphology of dystrophic microglia in the aged brain and AD supports the hypothesis that microglia experience a process of senescence during the course of aging, losing their abilities to respond to chronic inflammatory stimuli and no longer performing their neuroprotective duties (Bachstetter et al., 2015). Hypertrophic microglia or primed microglia are used to describe activated microglia in the brain, which could be observed in AD and accelerated aging (D. D. Raj et al., 2014; Walker & Lue, 2015). This type of microglia shows increased response to immune stimuli resulting in exaggerated inflammatory responses (V. H. Perry & Holmes, 2014). The morphological changes of hypertrophic microglia are different from that of dystrophic microglia. In an accelerated aging model of mice, microglia existed enlarged cell soma and thickened length of processes (D. D. Raj et al., 2014). No specific molecular markers are used to identify hypertrophic or primed microglia, while they express markers of activated macrophages, such as MHC‐II, CD11b, and IL‐1β, when stimulated by pathogens and high‐fat diet (Butler et al., 2020; Garner et al., 2018). The distribution of hypertrophic microglia is inhomogeneous in the aged brain. Hypertrophic microglia are frequently distributed in atrophied gray matter and left language regions compared with resting microglia (Ohm et al., 2021). Meanwhile, in the rhesus monkey, increased density of hypertrophic microglia in frontal white matter was associated with cognitive impairment (Shobin et al., 2017). It is shown that hypertrophic microglia were extremely close to amyloid‐beta (Aβ) plaques in several brain areas, especially cortex and hippocampus in aged human cases and transgenic mouse model of AD (Bouvier et al., 2016). In this situation, hypertrophic microglia showed upregulated CD33 expression, which is associated with decreased clearance and accumulation of Aβ plaques (Griciuc et al., 2013).
3.2. Age‐related changes in functions of microglia
Apart from the morphological change, the function of microglia is also changed during the process of priming. Accumulated pathogen‐associated molecular patterns (PAMPs) or danger‐associated molecular patterns (DAMPs), such as Aβ, tau, and α‐synuclein, in the aged brain induced microglial activation (Cai et al., 2014). Phagocytosis is an important function of microglia to clear dead cells, debris, and pathogens in the brain. Phagocytic receptors including CD36, TREM2b, and CD14 were found to upregulate in primed microglia (Raj et al., 2014). These microglia migrate to amyloid plaques and neurofibrillary tangles (NFTs) and attempt to sweep away them. However, in AD patients, microglia failed to effectively uptake amyloid plaque to prevent AD progression (Bamberger et al., 2003). Microglia adjacent to amyloid plaque showed impaired Ca2+ signaling characterized as increased frequency of spontaneous intracellular Ca2+ transients accompanied with smaller size (Brawek et al., 2014). Increased Ca2+ transients in vicinity of plaque may induce the release of pro‐inflammatory cytokines, such as TNF‐α and NO, which is relied on the concentration of Ca2+, deteriorate the neuroinflammation, and promote AD progression (Färber & Kettenmann, 2006; Hide et al., 2000). Toll‐like receptors (TLRs) are a group of family in pattern recognition, which is the crucial receptors in mediating phagocytosis of microglia. Upregulated TLR4 expression was observed in brain areas of AD patients, such as entorhinal cortex (Walter et al., 2007). On the one hand, TLR4‐mediated microglia activation, such as LPS (a classical TLR4 ligand) stimulation, induced clearance of Aβ plaques by microglial phagocytosis (Tahara et al., 2006). On the other hand, microglia activation mediated by TLR4 triggers the release of pro‐inflammatory cytokines (Papageorgiou et al., 2016). Besides, NLR family pyrin domain containing 3 (NLR3) inflammasome is aberrantly stimulated by PAMPs/DAMPs, which switches microglia into a pro‐inflammatory state (Ślusarczyk et al., 2018). Microglia also show increased proliferation as well as enhanced and uncontrolled inflammatory response to peripheral inflammatory factors, such as LPS, in a mouse model featured with accelerating aging (Raj et al., 2014). Single‐cell transcriptomic approaches indicated that iron metabolism‐related proteins, such as ferritin light chain (FTL), were differentially activated in dystrophic microglia in the aged brain (Shahidehpour et al., 2021). In a bioinformatic study based on HuMi_Aged gene, the proteome of aged human microglia showed that some signaling pathways, such as the ER‐phagosome pathway, endosomal pathway, and antigen processing‐cross presentation, were upregulated in aged human microglia (Olah et al., 2018). In general, morphological and functional changes of microglia during aging are likely to participate in the progression of cognitive aging.
3.3. Neuroinflammation in cognitive aging
Neuroinflammation is defined as the inflammation in the nervous tissue. Emerging evidence shows that neuroinflammation plays a vital role in the cognitive decline of older population. Normally, the CNS is isolated from inflammatory factors or pathogens by BBB, a specifically selective semipermeable structure composed of pericytes, endotheliocytes, and astrocytes. However, BBB leakage is observed in aged people, and 13.5% of the old population is found to have BBB dysfunction (Bowman et al., 2018). It is hard to define whether BBB breakdown happens before neuroinflammation in the aged. Factors from outside (periphery) and inside (CNS) might both be involved in BBB breakdown and neuroinflammation development (Figure 1).
Microbiota also involve in mediating the function of BBB, though the mechanism underlying microbiota‐BBB connections is still foggy. A previous study observed increased BBB leakage in germ‐free mice combined with decreased occludin and claudin‐5 expression, which indicated the role of gut microbiota in BBB formation (Braniste et al., 2014). During aging, dysbiosis of gut microbiota might lead to increased permeability of the gastrointestinal tract, which induces the increased level of circulating bacterial products, such as muramyl dipeptide (MDP) (Thevaranjan et al., 2017). In AD patients, the microbiota composition was characterized by decreased proportion of anti‐inflammatory bacteria (E.rectale) and increased abundance of pro‐inflammatory bacteria (Escherichia/Shigella), which was associated with the increased blood level of cytokines, such as IL‐6, NLRP3, and IL‐1β (Cattaneo et al., 2017). These elevated pro‐inflammatory cytokines accompanied bacterial products in the blood circulation might induce the production of NADPH and decrease tight junction proteins expression in BBB (Rochfort et al., 2014).
Vascular diseases, such as atherosclerosis, arteriolosclerosis, and stroke, promote BBB breakdown and make old patients more vulnerable to cognitive impairment (Chang et al., 2017; Skrobot et al., 2017; Wardlaw et al., 2017). Injured blood vessels, atheromatous plaque, and vascular occlusion induce the release of pro‐inflammatory cytokines and enroll immune cells to the damaged sites. In atherosclerosis, injured arteries are infiltrated with monocytes which lead to an increase of IL‐6 and activate macrophages (Tibolla et al., 2010). TNF‐α produced in vascular diseases induces the production of reactive oxygen species (ROS) and IL‐8, which further promotes vascular inflammation and cell infiltration (Hanrui Zhang et al., 2009). IL‐1 promotes the CNS‐specific inflammation through binding to interleukin‐1 receptor 1(IL‐1R1), a surface receptor on BBB endothelial cells. Activated IL‐1 signaling increases adhesion molecules expression, such as Vcam‐1 and Icam‐1 (Hauptmann et al., 2020). If the stimulated factors exist persistently, accumulated inflammatory factors release and sterile inflammation existence could damage the BBB integrity and increase its permeability, which facilitates the infiltration of pathogens and inflammatory cytokines in the brain and leads to neuroinflammation (Raj et al., 2015).
Intrinsic factors from CNS also induce the development of neuroinflammation, such as infection, death of neurons, Aβ peptide, and tau protein aggregation. Microglia take responsible for immune‐privileged and surveille the brain microenvironment, which could respond to the accumulated waste (Dukhinova et al., 2018). Besides, they also involve in regulating neuronal function and memory through mediating synaptic plasticity (Chu et al., 2019; C. Wang et al., 2020). The function of microglia is associated with its morphology (resting, hyperramified, amoeboid, and round) (Solano Fonseca et al., 2016). In the resting state, microglia release neurotransmitters, remodel synapses and perform as a surveillant to clear debris (Dá Mesquita et al., 2016). While in elderly human brains, microglia are activated by accumulated Aβ and tau protein in cortical tissues and are closely associated with cognitive decline (Felsky et al., 2019). In this condition, activated microglia show not only hyperramified and amoeboid phenotype but a shift to M1 phenotype (Chen et al., 2016). In the rodent model, though the composition of microglia clusters does not change during aging, special subpopulations of microglia are marked with inflammatory signals, such as Lgals3, cystatin F, and Ccl3, were significantly increased in aged brains (Hammond et al., 2019). Consistent with upregulated inflammatory markers, elevated inflammatory cytokines, and chemokines secreted by microglia are found in aged brain. In aged mice, IL‐6 mRNA expression level and supernatant IL‐6 from microglia are evidently increased compared with adult mice (Ye & Johnson, 1999). Increased IL‐6 binds to overexpressed IL‐6 receptor on microglia from aged mice and activates IL‐6 trans‐signaling pathway (Burton et al., 2013). Accordingly, microglia in such inflammatory state exacerbate neuron damage in aged brain (Hanisch & Kettenmann, 2007). Therefore, though microglia serve as “coordinator” between extracerebral and intracerebral environments, they will lose the role of protection with increased age and turn into an assistant of crimes.
4. MICROBIOTA, MICROGLIA, AND COGNITIVE DECLINE IN THE AGED
As microglia play a vital role in neurodegenerative diseases, abundant studies attempt to demonstrate the mechanism involved in it. In recent decades, scientists gradually realized that microglia and microbiota have amazing interplay. Microbiota influences the development, maturation, and function of microglia (Abdel‐Haq et al., 2019). In the aged, microbiota dysbiosis disturbs the microenvironment homeostasis and alters the state and function of microglia (Figure 2).
4.1. Microbiota, vagus nerve, and microglia
The vagus nerve connects the gut and brain through thousands of sensory and motor fibers. Accumulating evidence indicates that connections between gut microbiota and the vagus nerve appear to regulate the state of microglia and the level of inflammation in the CNS (Dinan & Cryan, 2017). To understand the communication between the specific bacteria in gut and the brain through the vagus nerve, we need to know how the bacteria can suppress or stimulate the signals transmitted through the vagus nerve. In the small and large intestine, vagal afferents terminate in the muscle layer as well as in the mucosal layer. In the muscle layer, vagal afferents end in longitudinal muscle and circular muscle to detect stretch and tension, thereby regulating intestinal movements. In the mucosal layer, vagal afferents are classified into two groups, vagal villi and vagal crypt afferents. Due to the close distance to the epithelial cell layer, gut peptides, metabolites, and chemicals absorbed by epithelial cells directly interact with vagus nerve endings.
A previous study showed that SCFAs secreted by gut bacteria, such as butyric acid, can activate vagal afferent in rat jejunum by a direct effect on afferent endings, while long‐chain fatty acids activate vagal afferents via a cholecystokinin‐dependent pathway (Lal et al., 2001). Consistent with this study, the release of bacterial byproducts induced by high‐fat diet leads to the response of vagal afferents terminals (Patterson et al., 2002). Besides, extracellular vehicles secreted by Paenalcaligenes hominis in GI of young mice could induce cognitive impairment, which can be inhibited by vagotomy (K.‐E. Lee et al., 2020). Similarly, oral administration of Lactobacillus rhamnosus induces a depression‐related behavior in adult mice through decreasing GABA receptor expression in the brain. This could also be attenuated by ablation of vagus nerve (Bravo et al., 2011; J. Zhang et al., 2020). Therefore, the gut microbiota might regulate signals of the vagus nerve by its metabolites.
Since changes caused by gut bacteria could suppress or stimulate vagal nerve terminals located in GI, signals transmitted by vagus nerve from GI to brain might influence the function of microglia (J. Kim et al., 2020). It is reported that non‐invasive vagus nerve stimulation causes a significant change of microglia branches number in mouse model of AD (Kaczmarczyk et al., 2018). If vagus nerve is damaged by vagotomy, increased microglia activation in hippocampus is observed in adult rats (Ronchi et al., 2012). Sen et al. revealed that obesity‐induced gut microbiota dysbiosis, especially an increase in Clostridia and Bacilli, downregulated IB4 binding and β3‐tubulin protein expression and therefore altered the vagal afferents from the gut and activated microglia (Sen et al., 2017). One possible pathway that contributed to microglia activation is acetylcholine (Ach) or norepinephrine (NE) pathway. In vitro incubation with Ach decreases IL‐1β and IL‐6 secretion and inhibits activation of JAK2/STAT3 and PI3K/AKT pathway in LPS‐induced microglia, therefore, switching microglia from M1 phenotype to M2 phenotype (Q. Zhang et al., 2017). Besides, activation of α7 nicotinic Ach receptor on microglia significantly prevents the release of inflammatory cytokines (De Simone et al., 2005). More studies are required to demonstrate the mechanisms that microbiota activates or suppresses microglia through which vagal afferents.
4.2. Microbiota, blood circulation, and microglia
Microbial metabolites, such as SCFAs and dopamine, are key contributions that gut microbiota modulates host metabolic reactions and health (Macfarlane & Macfarlane, 2003; Rekdal et al., 2019). These metabolites are secreted into intestinal tract, absorbed across the gut, and circulated in the host circulation (Figure 2).
SCFAs, known as saturated fatty acids with less than six carbon atoms, are mainly derived from gut microbial fermentation of dietary fiber (Miller & Wolin, 1996). Currently, at least 8 kinds of SCFAs have been found in human gut. Among them, acetate, propionate, and butyrate are the predominant ingredients of SCFAs, which accounts for over 95% of total SCFAs (Cook & Sellin, 1998; Wong et al., 2006). Once produced in the colon, SCFAs are transported across the apical and basolateral membranes of colonocytes. For the apical membrane, SCFAs are mainly transported by monocarboxylate transporters (MCTs). MCT1 transports SCFAs in an H+ ‐dependent electroneutral manner, while sodium‐dependent monocarboxylate transporter (SMCT) 1 transports SCFA anion coupled with Na+ in a ratio of 1:2 or 1:3 (Al‐Mosauwi et al., 2016; Coady et al., 2004, 2007).
After crossing apical membrane of colonocytes, SCFAs could enter into the citric acid cycle to generate energy for colonocytes (Tan et al., 2014). SCFAs that are not consumed in the colonocytes could be transported across the basolateral membrane and enter into portal blood. MCT4 and MCT5 are the main transporters localized to the basolateral membranes, which are responsible for the transportation of SCFAs (Gill et al., 2005). Though SCFAs could be transported into the bloodstream, only a small amount of acetate (36%), propionate (9%), and butyrate (2%) could measure in the blood circulation (Boets et al., 2015).
When arriving at the BBB, SCFAs mediate the functions of microglia through direct or indirect contact so that they can influence cognition of the host. Accumulating studies have indicated the evidence of SCFAs crossing the BBB. In rat model, intracarotid injection of 14C labeled butyrate and pyruvate leads to increased 14C‐SCFAs concentration in the brain (Sarna et al., 1979). In human, 22 nmol/g of propionate and 15 nmol/g of butyrate are measured in brain by using gas chromatography (Bachmann et al., 1979). These part of SCFAs, that go through the BBB, directly regulates the maturation and activity of microglia in the brain. In germ‐free mice, SCFAs administration decreases the CSFR1 expression on immature microglia surface and promotes the maturation of microglia by binding to SCFA receptor‐free fatty acid receptor 2 (FFAR2) (Erny et al., 2015). Butyrate, a member of SCFAs, is shown to reduce LPS‐induced inflammation not only in rats but also in mice models (Huuskonen et al., 2004; Yamawaki et al., 2018). Manually increased uptake of butyrate from dietary attenuates the release of pro‐inflammatory cytokines from microglia in aged mice (Matt et al., 2018), which might be regulated by nuclear factor‐kappa B (NF‐kB) and extracellular signal‐regulated kinase (ERK) signaling pathways (Park et al., 2005). Similarly, administration of Clostridium butyricum in APP/PS1 mice attenuates microglial activation, reduces TNF‐α and IL‐1β production, and improves cognitive impairment by suppressing the phosphorylation of NF‐kB p65 in microglia (J. Sun et al., 2020). Consistent with this, additional supplement of propionate by prebiotics reverses age‐related percentage of activated microglia in mice (Boehme et al., 2018). Aside from SCFAs that going across the BBB, SCFAs could also interact with BBB and might indirectly regulate the activation of microglia in the brain. In germ‐free adult mice, administration of Clostridium tyrobutyricum or Bacteroides thetaiotaomicron, which mainly produce SCFAs, decreases the permeability of BBB and increases the expression of occludin. Oral gavage of butyrate in germ‐free adult mice for 3 days receives the same effect on BBB (Braniste et al., 2014). In aged mice, application of Lactobacillus and sodium butyrate reverses cognitive impairment after operation by increasing the expression of tight junction proteins in BBB (Wen et al., 2020). During aging, increased permeability of BBB allows circulating pro‐inflammatory cytokines and pathogens (MDP and LPS) penetration (Thevaranjan et al., 2017; Zhan et al., 2018). SCFAs induced BBB tight junctions repair might prevent the microglia activation by contacting with circulating inflammatory cytokines. However, there is still a lack of direct evidence to illustrate this axis. More studies are required to demonstrate the mechanism involved in SCFAs mediated BBB permeability regulation.
4.3. Microbiota, immune cells, and microglia
Communications between microbiota and immune cells in the host have been recognized for decades. Growing evidence indicates that gut microbiota might interact with the peripheral immune system to indirectly modulate the function of microglia in the brain (Figure 2).
In the periphery, microbiota‐originated molecular patterns, including lipopolysaccharide (LPS), bacterial flagellum, and CpG DNA, are also known as microbial‐associated molecular patterns (MAMPs), which could mediate the functions of immune cells including macrophages, dendritic cells, and neutrophils (Timothy R Sampson & Mazmanian, 2015). Age‐related gut microbiota dysbiosis increases the paracellular permeability of colons and allows LPS to leak into blood circulation. This induces TNF and IL‐6 release and deteriorates the ability of bone marrow‐derived macrophages (BMDMs) to extinguish Streptococcus pneumoniae (Thevaranjan et al., 2017). Additionally, deficiency of pro‐inflammatory cytokines secretion in macrophages lowers the ability of macrophages to support B cells response to LPS in aged mice (Chelvarajan et al., 2006). Dysfunction of BMDMs in aged rats increased the responsiveness to LPS in vitro by increasing the expression of IFNγ receptor and toll‐like receptor 4 (TLR4) (Barrett et al., 2015). Inflammatory signals produced by dysfunctional peripheral macrophages in old age transduce to microglia not only via leaked BBB in old brain but also meningeal cells (Liu et al., 2013). After receiving these inflammatory signals, microglia displays a hyperactive response in aged rodents, which presents as upregulation of major histocompatibility complex class II (MHC‐II), IL‐1β, IL‐10, and TLR‐2 (Henry et al., 2009). Meanwhile, IL‐4‐induced M2 promoting effects are less sensitive in aged microglia (Fenn et al., 2012). Some signaling pathways in microglia are downregulated, such as transforming growth factor β (TGF‐β1), mothers against decapentaplegic homolog 3 (Smad3) pathway (Tichauer et al., 2014). These changes in microglia might lead to enhanced neuroinflammation and synapse remodeling in aged mice (Kondo et al., 2011).
T cells also involve in the gut microbiota‐induced microglia function regulation. Groups of specific T cells diffuse almost everywhere in the human body to surveille the peripheral environment, discriminate antigens, and extinguish pathogens. In the gut, T cells are not only accumulated in Peyer's patches to recognize the luminal antigens sampled by M cells in the follicle‐associated epithelium, but resided in the lamina propria as CD4+ T cells to mediate the immune response (MacDonald & Monteleone, 2005). During the aging process, the integrity of gut barrier is decreased as a result of passive factors, including increased epithelial apoptosis, overactivated c‐Jun N‐terminal kinase (JNK) signaling pathway in intestinal epithelial stem cells and decreased expression of tight junctions proteins (Branca et al., 2019; Farhadi et al., 2003). Besides, aging‐associated gut microbiota dysbiosis also contributes to increased intestinal permeability (Thevaranjan et al., 2017). In this situation, metabolites from gut microbiota, pro‐inflammatory cytokines, and even bacteria translocate into lamina propria through leaky gut epithelial barrier, which induces T cells to produce IL‐6 and TNF‐α (Heumann et al., 1994). Results from rhesus macaque models indicate that lower levels of Firmicutes and higher levels of archaeal and proteobacterial species in aged animals are associated with chronic systemic inflammation and increased memory CD4 T cells in the brain (Pallikuth et al., 2021).
However, in which way do activated T cells in the gut influence the functions of microglia in the brain? Inflammation induced by T cells in the gut could lead to chronic systemic inflammation as a result of immune homeostasis damage in aged individuals. Age‐driven changes in regulatory T cells (Tregs) weaken their ability to suppress immune reactions, such as IL‐17 and IL‐2 secretion by T cells, which deteriorates the imbalance of the immune homeostasis and promotes inflammation in the body (Churov et al., 2020). These reactive T cells activated in the peripheral immune system infiltrate in the brain partly as a result of gut antigens activation (Mora et al., 2003). Growing evidence indicates that inflammatory activities increased the number of dendritic cells and T cells in 12 months old mice (Stichel & Luebbert, 2007). Though the mechanism of T cells recruitment in the brain is not clear, single‐cell analysis in old brains reveals that T cells infiltrate in old brain neurogenic niches and express interferon‐γ, which inhibits the growth of neural stem cells (Dulken et al., 2019; Moreno‐Valladares et al., 2020). As microglia are the immune cells resided in the brain, infiltrated T cells might have a crosstalk with microglia to modulate the functions of the aged brain. Previously, isolated MHC‐II+ microglia are proved to effectively support the activation of CD4+ T cells, which is stimulated by an encephalitogenic myelin basic protein, to produce IFN‐γ and TNF‐α not IL‐2 (Ford et al., 1996). Similarly, under specific in vitro conditions, microglia expressing MHC‐II induces the activity of CD4 regulatory T cells by low‐dose IFN‐γ (Ebner et al., 2013). In AD mice model and human AD specimen, Iba‐1+ and CD68+ microglia are found to be colocalized with CD8+ T cells at the site of Aβ plaques, which indicates the interaction between microglia and T cells in the brain (M. S. Unger et al., 2018). C. Laurent et al. reveals that chemokine (C‐C motif) ligand 3 (CCL3) exclusively produced by microglia in hippocampus of AD mice model facilitates CD8+ T cells infiltration, and elimination of these infiltrated T cells restores the cognitive function of AD mice (Laurent et al., 2017). In this study, unfortunately, it is unknown whether microglia depletion could attenuate CD8+ T cells infiltration in the hippocampus. Unexpectedly, microglia ablation by PLX5622, a colony‐stimulating factor 1 receptor (CSF1R) inhibitor, increases the number of CD3+/ CD8+ T cells in the brain of APP/PS1 mice, while this treatment has no improvement on cognition of APP/PS1 mice (M. Unger et al., 2018). Increased CD8+ T cells in the brain might neutralize the influence of microglia ablation on neuroinflammation, which makes PLX5622 fail to restore the cognitive functions. Furthermore, microglia in aged brain partly switch to phagocytic phenotype as a response of accumulating myelin pathology, which also leads to cognitive impairment (Shobin et al., 2017). These data suggest that neuroinflammation induced by crosstalk between microglia and infiltrated T cells in the aged brain plays a vital role in cognitive deficits.
5. MICROBIAL DYSBIOSIS AND MICROGLIAL DYSFUNCTION IN NEURODEGENERATIVE DISEASES
Considering the wide range of physiological functions of microglia in brain development and homeostasis maintenance, microglia are believed to participate in the pathogenesis of several neurodegenerative diseases. As we discussed above, the morphology and functions of microglia are affected by factors and signals originated from brain or the periphery. Therefore, it has been recognized that several neurodegenerative diseases are not only originated in the brain but affected by periphery factors. Here, we briefly discuss the potential role of microbiota‐microglia connections in AD and PD.
5.1. AD
AD is characterized as a neurodegenerative disease with slowly but progressively loss of memory, cognition, and motor functions (Burns & Iliffe, 2009). Currently, the recognized pathological hallmarks of AD are amyloid plaques deposition and tau protein aggregation (Savelieff et al., 2013). Factors in the brain and peripheral environment involved in the pathogenesis of AD, eventually leading to severe neuroinflammation and neurons loss.
Microglia, as the resident phagocytes in the brain, play an important role in Aβ clearance contributing to homeostasis of Aβ production and removal (C. Y. D. Lee & Landreth, 2010). Continuous exposure to Aβ species, microglia switch to an activated state with increased proliferation and expression of CD36, iNOS, and CD14 (Martin et al., 2017; Olmos‐Alonso et al., 2016). Activated microglia, on the one hand, lead to Aβ clearance by increased phagocytosis. Pattern recognition receptors (PRPs) expressed on the surface of microglia drive microglia binding to different species of Aβ, and microglia rapidly transport these plaques into endolysosomal compartments to degradate (Mandrekar et al., 2009; C. Venegas & Heneka, 2017). On the other hand, chronic activation of microglia results in chronic neuroinflammation and neurotoxicity by pro‐inflammatory factors release. Long‐term binding of Aβ peptides to PRPs induces enhanced release of pro‐inflammatory cytokines in microglia, such as IL‐1β and TNF‐α, which could promote the progression of AD (Q. Li et al., 2018). Therefore, it seems that the function of microglia is dependent on the environment around microglia.
Considering the potential interplay between the gut microbiota and microglia, bacteria in the gastrointestinal tract might regulate the pathogenesis of AD. Changed composition of gut microbiota is observed several mice models of AD and AD patients. The decreased abundance of Firmicutes, Verrucomicrobia, Proteobacteria, and Actinobacteria is observed in old age of AD mice models (Harach et al., 2017). Additionally, abundant phyla proportion of Firmicutes and Bacteroidetes and families of Lachnospiraceae, Ruminococcaceae, and Bacteroidaceae are observed in AD patients (Vogt et al., 2017). Support for the hypothesis that microbiota‐microglia connections involve in AD development, and progression was that gut metabolites such as SCFAs could affect the development of AD by mediating the function of microglia and neuroinflammation (J. Sun et al., 2020). SCFAs level was found to be altered in AD patients compared with healthy controls (Nagpal et al., 2019). A recent study indicated that SCFAs supplementation in germ‐free AD mice increases microglia recruitment to Aβ plaques and promotes Aβ deposition likely by mediating the phenotype of microglia (Colombo et al., 2021). Previously, Venegas et al. suggested that microglia promote Aβ plaques deposition in AD mice model by secreting ASC specks, which are pro‐inflammatory protein complexes (Carmen Venegas et al., 2017). However, loss of Trem2 accelerates amyloid seeds due to reduced Aβ clearance, accompanied with reduced plaque‐associated ApoE (Parhizkar et al., 2019). Therefore, it seems that the function of microglia is largely depended on the peripheral environment.
5.2. PD
PD is defined as a long‐term degenerative disease occurred in the CNS that mainly affects the motor system. The common symptoms of PD are resting tremor, rigidity, and slowness of movement (Politis et al., 2010). And the main pathological changes in PD are cell death in basal ganglia which mainly leads to death of dopaminergic neurons (Davie, 2008). Complicated interactions between genetic and environmental factors are believed to cause the pathogenesis of PD (Kalia & Lang, 2015).
The evidence that microglia involve in the pathogenesis of PD is revealed by several studies. Sruti et al. found that p.R47H variant in Trem2 of microglia is a risk factor for PD (Rayaprolu et al., 2013). Besides, positron emission tomography (PET) revealed that the activation of microglia is associated with the early pathogenesis of PD possibly by releasing cytokines (Gerhard et al., 2006). The distribution of microglia is specific in PD, which predominantly distributed in substantia nigra and striatum (Grabert et al., 2016). This leads to region‐specific release of cytokines, such as TNF‐α and IL‐6, affecting the death of dopaminergic neurons (Nagatsu & Sawada, 2005). Microglia are also stimulated by α‐synuclein (a misfolded protein in Lewy bodies) and promote the release of pro‐inflammatory cytokines in turn (George et al., 2019). In summary, these results support that microglia play a pivotal role in progression of PD.
Though PD is a brain disorder mainly affected the function of the brain, recent evidence suggests that factors from gastrointestinal tract might play a role in pathogenesis of PD. The composition of gut microbiota in PD patients is significantly changed compared with healthy controls. Lin et al. found that the diversity of gut microbiota in PD patients is less than that of healthy controls (Lin et al., 2019). Increased abundance of Verrucomicrobia, Prevotella, Mucispirillum, Porphyromonas, Lactobacillus, and Parabacteroides was observed in PD patients, and dysbiosis of gut microbiota was associated with elevated inflammatory response (Lin et al., 2019). Meanwhile, metabolites derived from gut microbiota are altered in PD patients, which presents as markedly reduced SCFAs (M. M. Unger et al., 2016). It should be noticed that SCFA‐producing KEGG pathways are upregulated in mice who received fecal transplants from PD donors (T. R. Sampson et al., 2016). The indications that microbiota‐microglia connections might drive the pathogenesis of PD are based on the following observations. Germ‐free mice that overexpressed human α‐synuclein display motor dysfunction, GI dysfunction, and microglial activation (T. R. Sampson et al., 2016). Conducting oral gavage for these mice with mixture of SCFAs induces α‐synuclein aggregation and morphological changes of microglia, which promotes motor deficits (T. R. Sampson et al., 2016). Another study also observed reduced microbial dysbiosis, reduced SCFAs level and microglia activation, and alleviated motor deficits in PD mice when received fecal transplants from healthy mice (M.‐F. Sun et al., 2018). Though these findings reveal part role of microbiota‐microglia connections in PD, the precise mechanism that gut microbiota affects motor deficits and neuroinflammation is still unclear.
6. FUTURE PERSPECTIVES
Aging is a major risk of neurodegenerative diseases, such as AD. In recent years, microglia, the resident immune cells in the CNS, are revealed to involve in the development of neuroinflammation and be mediated by gut microbiota due to the involvement of microbiota in microglia maturation and function. To date, however, the studies likely only give a glimpse into the tip of the iceberg that the interplay between the gut microbiota and microglia during aging. The emergence of new tools, such as single‐cell analysis and spatial transcriptomic sequencing (ST‐seq) used to feature the microbiota and microglia, will accelerate the further recognition on the crosstalk between gut microbiota and microglia (Masuda et al., 2020). Recently, a multifunctional technology, named high‐phylogenetic‐resolution microbiota mapping by fluorescence in situ hybridization (HiPR‐FISH), achieves identification of taxa in situ only with a single round of confocal imaging and reveals the spatial network disruption of gut microbiota in mice treated with antibiotics (Shi et al., 2020). Apart from in vitro studies, noninvasively in vivo imaging and tracking of gut microbiota could greatly promote the knowledge in microbiota‐microglia interactions. Previously, metabolic oligosaccharide engineering and biorthogonal click chemistry were used to label commensal bacteria in the gut and demonstrated the distribution and colonization of labeled bacteria in vivo by two‐photon microscopy and non‐invasive whole‐body imaging (Geva‐Zatorsky et al., 2015). Radiolabeled probes (18F‐FDS and 64Cu‐NOTA‐DBCO) could also achieve in vivo imaging of gut microbiota by using PET/CT (Y. Wang et al., 2020; Z. Zhang et al., 2021). Meanwhile, the developed microscopy techniques, such as two‐photon microscopy, multiphoton microscopy and allows to observe the activity of microglia in the living brain (Hierro‐Bujalance et al., 2018; Zhao et al., 2018). These studies shed light on the characterization of comprehensive changes in gut microbiota and microglia of aged individuals, promoting the screening of specific markers in cognitive aging and speeding the development of microbiota or microglia‐targeted therapies.
One of the priority questions remains to be clarified is what the certain mechanism involved in the phenotypic and functional changes of microglia by altered gut microbiota. It seems that gut microbiota changes and microglia alterations may interact as both cause and effect. The observed changes of gut microbiota might be a cause or result of neuroinflammation or microglia alterations. Metabolites or neurotransmitters derived from gut commensal microbiota have the potential to attenuate and even restore the age‐induced alterations of microglia and finally improve cognitive impairment. Therefore, further investigations are called to reveal the amazing interplay between gut microbiota and microglia so that the microbiota or microglia‐targeted therapies come true and benefit the patients. Additionally, technologies development in noninvasively and in vivo monitoring the composition of gut microbiota and functions of microglia will greatly accelerate the clinical practice of these therapeutic strategies.
CONFLICT OF INTEREST
The authors declare no conflict of interest.
AUTHOR CONTRIBUTIONS
MT, LC, HZ, and WC conceived the manuscript. RZ finished the manuscript and figures. SQ summarized the tables. JZ contributed to the manuscript revision and Figure 2 preparation. All authors contributed to the manuscript revision.
ACKNOWLEDGMENTS
We thank the support of National Natural Science Foundation of China (81725009, 21788102 and 82030049) and National Key R&D Program of China (2021YFE0108300).
Zhou, R. , Qian, S. , Cho, W. C. S. , Zhou, J. , Jin, C. , Zhong, Y. , Wang, J. , Zhang, X. , Xu, Z. , Tian, M. , Chan, L. W. C. , & Zhang, H. (2022). Microbiota‐microglia connections in age‐related cognition decline. Aging Cell, 21, e13599. 10.1111/acel.13599
Contributor Information
Mei Tian, Email: meitian@zju.edu.cn.
Lawrence W. C. Chan, Email: wing.chi.chan@polyu.edu.hk.
Hong Zhang, Email: hzhang21@zju.edu.cn.
REFERENCES
- Abdel‐Haq, R. , Schlachetzki, J. C. , Glass, C. K. , & Mazmanian, S. K. (2019). Microbiome–microglia connections via the gut–brain axis. Journal of Experimental Medicine, 216(1), 41–59. 10.1084/jem.20180794 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Al‐mosauwi, H. , Ryan, E. , McGrane, A. , Riveros‐Beltran, S. , Walpole, C. , Dempsey, E. , Courtney, D. , Fearon, N. , Winter, D. , Baird, A. , & Stewart, G. (2016). Differential protein abundance of a basolateral MCT1 transporter in the human gastrointestinal tract. Cell Biology International, 40(12), 1303–1312. 10.1002/cbin.10684 [DOI] [PubMed] [Google Scholar]
- Amarya, S. , Singh, K. , & Sabharwal, M. (2015). Changes during aging and their association with malnutrition. Journal of Clinical Gerontology and Geriatrics, 6(3), 78–84. 10.1016/j.jcgg.2015.05.003 [DOI] [Google Scholar]
- Bachmann, C. , Colombo, J.‐P. , & Berüter, J. (1979). Short chain fatty acids in plasma and brain: quantitative determination by gas chromatography. Clinica Chimica Acta, 92(2), 153–159. 10.1016/0009-8981(79)90109-8 [DOI] [Google Scholar]
- Bachstetter, A. D. , Van Eldik, L. J. , Schmitt, F. A. , Neltner, J. H. , Ighodaro, E. T. , Webster, S. J. , Patel, E. , Abner, E. L. , Kryscio, R. J,. , & Nelson, P. T. (2015). Disease‐related microglia heterogeneity in the hippocampus of Alzheimer’s disease, dementia with Lewy bodies, and hippocampal sclerosis of aging. Acta Neuropathologica Communications, 3(1), 32. 10.1186/s40478-015-0209-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baldwin, C. L. , & Greenwood, P. M. (2020). Editorial: cognitive and brain aging: interventions to promote well‐being in old age. Frontiers in Aging Neuroscience, 11. 10.3389/fnagi.2019.00353 [DOI] [Google Scholar]
- Bamberger, M. E. , Harris, M. E. , McDonald, D. R. , Husemann, J. , & Landreth, G. E. (2003). A cell surface receptor complex for fibrillar β‐amyloid mediates microglial activation. The Journal of Neuroscience, 23(7), 2665. 10.1523/JNEUROSCI.23-07-02665.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barrett, J. P. , Costello, D. A. , O’Sullivan, J. , Cowley, T. R. , & Lynch, M. A. (2015). Bone marrow‐derived macrophages from aged rats are more responsive to inflammatory stimuli. Journal of Neuroinflammation, 12(1), 1–11. 10.1186/s12974-015-0287-7 [DOI] [Google Scholar]
- Biagi, E. , Franceschi, C. , Rampelli, S. , Severgnini, M. , Ostan, R. , Turroni, S. , Consolandi, C. , Quercia, S. , Scurti, M. , Monti, D. , Capri, M. , Brigidi, P. , & Candela, M. (2016). Gut microbiota and extreme longevity. Current Biology, 26(11), 1480–1485. 10.1016/j.cub.2016.04.016 [DOI] [PubMed] [Google Scholar]
- Biagi, E. , Nylund, L. , Candela, M. , Ostan, R. , Bucci, L. , Pini, E. , Nikkïla, J. , Monti, D. , Satokari, R. , Franceschi, C. , Brigidi, P. , & De Vos, W. (2010). Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PLoS One, 5(5), e10667. 10.1371/journal.pone.0010667 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bian, G. , Gloor, G. B. , Gong, A. , Jia, C. , Zhang, W. , Hu, J. , & Li, J. (2017). The gut microbiota of healthy aged chinese is similar to that of the healthy young. mSphere, 2(5), e00327. 10.1128/mSphere.00327-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bisanz, J. E. , Upadhyay, V. , Turnbaugh, J. A. , Ly, K. , & Turnbaugh, P. J. (2019). Meta‐analysis reveals reproducible gut microbiome alterations in response to a high‐fat diet. Cell Host & Microbe, 26(2), 265–272. e264. 10.1016/j.chom.2019.06.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boehme, M. , Van de Wouw, M. , Van Sandhu, K. , Lyons, K. , Fouhy, F. , Olavarria Ramirez, L. , Van Leuven, L. , Golubeva, A. , Scott, K. A. , Stanton, C. , Dinan, T. G. , Schellekens, H. , & Cryan, J. F. (2018). Targeting the gut microbiome to reverse microglia activation and stress‐induced immune priming in ageing. European Neuropsychopharmacology, 28, S18–S19. 10.1016/j.euroneuro.2017.12.038 [DOI] [Google Scholar]
- Boets, E. , Deroover, L. , Houben, E. , Vermeulen, K. , Gomand, S. V. , Delcour, J. A. , & Verbeke, K. (2015). Quantification of in vivo colonic short chain fatty acid production from inulin. Nutrients, 7(11), 8916–8929. 10.3390/nu7115440 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bouvier, D. S. , Jones, E. V. , Quesseveur, G. , Davoli, M. A. , A. Ferreira, T. , Quirion, R. , Mechawar, N. , & Murai, K. K. (2016). High resolution dissection of reactive glial nets in alzheimer's disease. Scientific Reports, 6, 24544. 10.1038/srep24544 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bowman, G. L. , Dayon, L. , Kirkland, R. , Wojcik, J. , Peyratout, G. , Severin, I. C. , Henry, H. , Oikonomidi, A. , Migliavacca, E. , Bacher, M. , & Popp, J. (2018). Blood‐brain barrier breakdown, neuroinflammation, and cognitive decline in older adults. Alzheimer's & Dementia, 14(12), 1640–1650. 10.1016/j.jalz.2018.06.2857 [DOI] [Google Scholar]
- Branca, J. J. , Gulisano, M. , & Nicoletti, C. (2019). Intestinal epithelial barrier functions in ageing. Ageing Research Reviews, 54, 100938. 10.1016/j.arr.2019.100938 [DOI] [PubMed] [Google Scholar]
- Braniste, V. , Al‐Asmakh, M. , Kowal, C. , Anuar, F. , Abbaspour, A. , Tóth, M. , Korecka, A. , Bakocevic, N. , Ng, L. G. , Kundu, P. , Gulyás, B. , Halldin, C. , Hultenby, K. , Nilsson, H. , Hebert, H. , Volpe, B. T. , Diamond, B. , & Pettersson, S. (2014). The gut microbiota influences blood‐brain barrier permeability in mice. Science Translational Medicine, 6(263), 263ra158‐263ra158. 10.1126/scitranslmed.3009759 [DOI] [Google Scholar]
- Bravo, J. A. , Forsythe, P. , Chew, M. V. , Escaravage, E. , Savignac, H. M. , Dinan, T. G. , & Cryan, J. F. (2011). Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proceedings of the National Academy of Sciences, 108(38), 16050–16055. [Google Scholar]
- Brawek, B. , Schwendele, B. , Riester, K. , Kohsaka, S. , Lerdkrai, C. , Liang, Y. , & Garaschuk, O. (2014). Impairment of in vivo calcium signaling in amyloid plaque‐associated microglia. Acta Neuropathologica, 127(4), 495–505. 10.1007/s00401-013-1242-2 [DOI] [PubMed] [Google Scholar]
- Burns, A. , & Iliffe, S. (2009). Alzheimer's disease. BMJ, 338, b158. 10.1136/bmj.b158 [DOI] [PubMed] [Google Scholar]
- Burton, M. D. , Rytych, J. L. , Freund, G. G. , & Johnson, R. W. (2013). Central inhibition of interleukin‐6 trans‐signaling during peripheral infection reduced neuroinflammation and sickness in aged mice. Brain, Behavior, and Immunity, 30, 66–72. 10.1016/j.bbi.2013.01.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Butler, M. J. , Cole, R. M. , Deems, N. P. , Belury, M. A. , & Barrientos, R. M. (2020). Fatty food, fatty acids, and microglial priming in the adult and aged hippocampus and amygdala. Brain, Behavior, and Immunity, 89, 145–158. 10.1016/j.bbi.2020.06.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai, Z. , Hussain, M. D. , & Yan, L.‐J. (2014). Microglia, neuroinflammation, and beta‐amyloid protein in Alzheimer's disease. International Journal of Neuroscience, 124(5), 307–321. 10.3109/00207454.2013.833510 [DOI] [PubMed] [Google Scholar]
- Camilleri, M. , Lee, J. S. , Viramontes, B. , Bharucha, A. E. , & Tangalos, E. G. (2000). Insights into the pathophysiology and mechanisms of constipation, irritable bowel syndrome, and diverticulosis in older people. Journal of the American Geriatrics Society, 48(9), 1142–1150. 10.1111/j.1532-5415.2000.tb04793.x [DOI] [PubMed] [Google Scholar]
- Cattaneo, A. , Cattane, N. , Galluzzi, S. , Provasi, S. , Lopizzo, N. , Festari, C. , Ferrari, C. , Guerra, U. P. , Paghera, B. , Muscio, C. , Bianchetti, A. , Volta, G. D. , Turla, M. , Cotelli, M. S. , Gennuso, M. , Prelle, A. , Zanetti, O. , Lussignoli, G. , Mirabile, D. , … Frisoni, G. B. (2017). Association of brain amyloidosis with pro‐inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiology of Aging, 49, 60–68. 10.1016/j.neurobiolaging.2016.08.019 [DOI] [PubMed] [Google Scholar]
- Chang, J. , Mancuso, M. R. , Maier, C. , Liang, X. , Yuki, K. , Yang, L. U. , Kwong, J. W. , Wang, J. , Rao, V. , Vallon, M. , Kosinski, C. , Zhang, J. J. H. , Mah, A. T. , Xu, L. , Li, L. E. , Gholamin, S. , Reyes, T. F. , Li, R. , Kuhnert, F. , … Kuo, C. J. (2017). Gpr124 is essential for blood–brain barrier integrity in central nervous system disease. Nature Medicine, 23(4), 450–460. 10.1038/nm.4309 [DOI] [Google Scholar]
- Chaudhari, D. S. , Dhotre, D. P. , Agarwal, D. M. , Gaike, A. H. , Bhalerao, D. , Jadhav, P. , Mongad, D. , Lubree, H. , Sinkar, V. P. , Patil, U. K. , Salvi, S. , Bavdekar, A. , Juvekar, S. K. , & Shouche, Y. S. (2020). Gut, oral and skin microbiome of Indian patrilineal families reveal perceptible association with age. Scientific Reports, 10(1), 5685. 10.1038/s41598-020-62195-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chelvarajan, R. L. , Liu, Y. , Popa, D. , Getchell, M. L. , Getchell, T. V. , Stromberg, A. J. , & Bondada, S. (2006). Molecular basis of age‐associated cytokine dysregulation in LPS‐stimulated macrophages. Journal of Leukocyte Biology, 79(6), 1314–1327. 10.1189/jlb.0106024 [DOI] [PubMed] [Google Scholar]
- Chen, W. W. , Zhang, X. , & Huang, W. J. (2016). Role of neuroinflammation in neurodegenerative diseases. Molecular Medicine Reports, 13(4), 3391–3396. 10.3892/mmr.2016.4948 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheung, S. G. , Goldenthal, A. R. , Uhlemann, A.‐C. , Mann, J. J. , Miller, J. M. , Sublette, M. E. (2019). Systematic review of gut microbiota and major depression. Frontiers in Psychiatry, 10(34), 34. 10.3389/fpsyt.2019.00034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chu, C. , Murdock, M. H. , Jing, D. , Won, T. H. , Chung, H. , Kressel, A. M. , & Zhou, L. (2019). The microbiota regulate neuronal function and fear extinction learning. Nature, 574(7779), 543–548. 10.1038/s41586-019-1644-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Churov, A. V. , Mamashov, K. Y. , & Novitskaia, A. V. (2020). Homeostasis and the functional roles of CD4+ Treg cells in aging. Immunology Letters. 226, 83–89. 10.1016/j.imlet.2020.07.004 [DOI] [PubMed] [Google Scholar]
- Claesson, M. J. , Cusack, S. , O'Sullivan, O. , Greene‐Diniz, R. , de Weerd, H. , Flannery, E. , Marchesi, J. R. , Falush, D. , Dinan, T. , Fitzgerald, G. , Stanton, C. , van Sinderen, D. , O'Connor, M. , Harnedy, N. , O'Connor, K. , Henry, C. , O'Mahony, D. , Fitzgerald, A. P. , Shanahan, F. , … O'Toole, P. W. (2011). Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proceedings of the National Academy of Sciences, 108(supplement_1), 4586–4591. 10.1073/pnas.1000097107 [DOI] [Google Scholar]
- Claesson, M. J. , Jeffery, I. B. , Conde, S. , Power, S. E. , O’Connor, E. M. , Cusack, S. , Harris, H. M. B. , Coakley, M. , Lakshminarayanan, B. , O’Sullivan, O. , Fitzgerald, G. F. , Deane, J. , O’Connor, M. , Harnedy, N. , O’Connor, K. , O’Mahony, D. , van Sinderen, D. , Wallace, M. , Brennan, L. , … O’Toole, P. W. (2012). Gut microbiota composition correlates with diet and health in the elderly. Nature, 488(7410), 178–184. 10.1038/nature11319 [DOI] [PubMed] [Google Scholar]
- Coady, M. J. , Chang, M. H. , Charron, F. M. , Plata, C. , Wallendorff, B. , Sah, J. F. , & Lapointe, J. Y. (2004). The human tumour suppressor gene SLC5A8 expresses a Na+‐monocarboxylate cotransporter. Journal of Physiology, 557(Pt 3), 719–731. 10.1113/jphysiol.2004.063859 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coady, M. J. , Wallendorff, B. , Bourgeois, F. , Charron, F. , & Lapointe, J. Y. (2007). Establishing a definitive stoichiometry for the Na+/monocarboxylate cotransporter SMCT1. Biophysical Journal, 93(7), 2325–2331. 10.1529/biophysj.107.108555 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colombo, A. V. , Sadler, R. K. , Llovera, G. , Singh, V. , Roth, S. , Heindl, S. , Sebastian Monasor, L. , Verhoeven, A. , Peters, F. , Parhizkar, S. , Kamp, F. , Gomez de Aguero, M. , MacPherson, A. J. , Winkler, E. , Herms, J. , Benakis, C. , Dichgans, M. , Steiner, H. , Giera, M. , … Liesz, A. (2021). Microbiota‐derived short chain fatty acids modulate microglia and promote Aβ plaque deposition. eLife, 10, e59826. 10.7554/eLife.59826 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cook, S. , & Sellin, J. (1998). Short chain fatty acids in health and disease. Alimentary Pharmacology & Therapeutics, 12(6), 499–507. 10.1046/j.1365-2036.1998.00337.x [DOI] [PubMed] [Google Scholar]
- Craik, F. I. (1994). Memory changes in normal aging. Current Directions in Psychological Science, 3(5), 155–158. 10.1111/1467-8721.ep10770653 [DOI] [Google Scholar]
- Dá Mesquita, S. , Ferreira, A. C. , Sousa, J. C. , Correia‐Neves, M. , Sousa, N. , & Marques, F. (2016). Insights on the pathophysiology of Alzheimer's disease: The crosstalk between amyloid pathology, neuroinflammation and the peripheral immune system. Neuroscience & Biobehavioral Reviews, 68, 547–562. 10.1016/j.neubiorev.2016.06.014 [DOI] [PubMed] [Google Scholar]
- Daria, A. , Colombo, A. , Llovera, G. , Hampel, H. , Willem, M. , Liesz, A. , Haass, C. , & Tahirovic, S. (2017). Young microglia restore amyloid plaque clearance of aged microglia. EMBO Journal, 36(5), 583–603. 10.15252/embj.201694591 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davie, C. A. (2008). A review of Parkinson's disease. British Medical Bulletin, 86, 109–127. 10.1093/bmb/ldn013 [DOI] [PubMed] [Google Scholar]
- Davies, D. S. , Ma, J. , Jegathees, T. , & Goldsbury, C. (2017). Microglia show altered morphology and reduced arborization in human brain during aging and Alzheimer's disease. Brain Pathology, 27(6), 795–808. 10.1111/bpa.12456 [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Simone, R. , Ajmone‐Cat, M. A. , Carnevale, D. , & Minghetti, L. (2005). Activation of α7 nicotinic acetylcholine receptor by nicotine selectively up‐regulates cyclooxygenase‐2 and prostaglandin E 2 in rat microglial cultures. Journal of Neuroinflammation, 2(1), 1–10. 10.1186/1742-2094-2-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dinan, T. G. , & Cryan, J. F. (2017). Brain–gut–microbiota axis—mood, metabolism and behaviour. Nature Reviews Gastroenterology & Hepatology, 14(2), 69–70. 10.1038/nrgastro.2016.200 [DOI] [PubMed] [Google Scholar]
- Dukhinova, M. , Kopeikina, E. , & Ponomarev, E. D. (2018). Usage of multiparameter flow cytometry to study microglia and macrophage heterogeneity in the central nervous system during neuroinflammation and neurodegeneration. In Cellular Heterogeneity (pp. 167–177). Springer. 10.1007/978-1-4939-7680-5_10 [DOI] [Google Scholar]
- Dulken, B. W. , Buckley, M. T. , Negredo, P. N. , Saligrama, N. , Cayrol, R. , Leeman, D. S. , & Pluvinage, J. V. (2019). Single‐cell analysis reveals T cell infiltration in old neurogenic niches. Nature, 571(7764), 205–210. 10.1038/s41586-019-1362-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ebner, F. , Brandt, C. , Thiele, P. , Richter, D. , Schliesser, U. , Siffrin, V. , Schueler, J. , Stubbe, T. , Ellinghaus, A. , Meisel, C. , Sawitzki, B. , & Nitsch, R. (2013). Microglial activation milieu controls regulatory T cell responses. The Journal of Immunology, 191(11), 5594–5602. 10.4049/jimmunol.1203331 [DOI] [PubMed] [Google Scholar]
- Erny, D. , Hrabě de Angelis, A. L. , Jaitin, D. , Wieghofer, P. , Staszewski, O. , David, E. , Keren‐Shaul, H. , Mahlakoiv, T. , Jakobshagen, K. , Buch, T. , Schwierzeck, V. , Utermöhlen, O. , Chun, E. , Garrett, W. S. , McCoy, K. D. , Diefenbach, A. , Staeheli, P. , Stecher, B. , Amit, I. , & Prinz, M. (2015). Host microbiota constantly control maturation and function of microglia in the CNS. Nature Neuroscience, 18(7), 965–977. 10.1038/nn.4030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Färber, K. , & Kettenmann, H. (2006). Functional role of calcium signals for microglial function. Glia, 54(7), 656–665. 10.1002/glia.20412 [DOI] [PubMed] [Google Scholar]
- Farhadi, A. , Banan, A. , Fields, J. , & Keshavarzian, A. (2003). Intestinal barrier: an interface between health and disease. Journal of Gastroenterology and Hepatology, 18(5), 479–497. 10.1046/j.1440-1746.2003.03032.x [DOI] [PubMed] [Google Scholar]
- Felsky, D. , Roostaei, T. , Nho, K. , Risacher, S. L. , Bradshaw, E. M. , Petyuk, V. , Schneider, J. A. , Saykin, A. , Bennett, D. A. , & De Jager, P. L. (2019). Neuropathological correlates and genetic architecture of microglial activation in elderly human brain. Nature Communications, 10(1), 1–12. 10.1038/s41467-018-08279-3 [DOI] [Google Scholar]
- Fenn, A. M. , Henry, C. J. , Huang, Y. , Dugan, A. , & Godbout, J. P. (2012). Lipopolysaccharide‐induced interleukin (IL)‐4 receptor‐α expression and corresponding sensitivity to the M2 promoting effects of IL‐4 are impaired in microglia of aged mice. Brain, Behavior, and Immunity, 26(5), 766–777. 10.1016/j.bbi.2011.10.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fjell, A. M. , & Walhovd, K. B. (2010). Structural brain changes in aging: courses, causes and cognitive consequences. Reviews in the Neurosciences, 21(3), 187–221. 10.1515/REVNEURO.2010.21.3.187 [DOI] [PubMed] [Google Scholar]
- Ford, A. L. , Foulcher, E. , Lemckert, F. A. , & Sedgwick, J. D. (1996). Microglia induce CD4 T lymphocyte final effector function and death. Journal of Experimental Medicine, 184(5), 1737–1745. 10.1084/jem.184.5.1737 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fujii, Y. , Nguyen, T. T. T. , Fujimura, Y. , Kameya, N. , Nakamura, S. , Arakawa, K. , & Morita, H. (2019). Fecal metabolite of a gnotobiotic mouse transplanted with gut microbiota from a patient with Alzheimer’s disease. Bioscience, Biotechnology, and Biochemistry, 83(11), 2144–2152. 10.1080/09168451.2019.1644149 [DOI] [PubMed] [Google Scholar]
- Fung, T. C. , Olson, C. A. , & Hsiao, E. Y. (2017). Interactions between the microbiota, immune and nervous systems in health and disease. Nature Neuroscience, 20(2), 145. 10.1038/nn.4476 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garner, K. M. , Amin, R. , Johnson, R. W. , Scarlett, E. J. , & Burton, M. D. (2018). Microglia priming by interleukin‐6 signaling is enhanced in aged mice. Journal of Neuroimmunology, 324, 90–99. 10.1016/j.jneuroim.2018.09.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- George, S. , Rey, N. L. , Tyson, T. , Esquibel, C. , Meyerdirk, L. , Schulz, E. , Pierce, S. , Burmeister, A. R. , Madaj, Z. , Steiner, J. A. , Escobar Galvis, M. L. , Brundin, L. , & Brundin, P. (2019). Microglia affect α‐synuclein cell‐to‐cell transfer in a mouse model of Parkinson's disease. Molecular Neurodegeneration, 14(1), 34. 10.1186/s13024-019-0335-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gerhard, A. , Pavese, N. , Hotton, G. , Turkheimer, F. , Es, M. , Hammers, A. , Eggert, K. , Oertel, W. , Banati, R. B. , & Brooks, D. J. (2006). In vivo imaging of microglial activation with [11C](R)‐PK11195 PET in idiopathic Parkinson's disease. Neurobiology of Disease, 21(2), 404–412. 10.1016/j.nbd.2005.08.002 [DOI] [PubMed] [Google Scholar]
- Geva‐Zatorsky, N. , Alvarez, D. , Hudak, J. E. , Reading, N. C. , Erturk‐Hasdemir, D. , Dasgupta, S. , von Andrian, U. H. , & Kasper, D. L. (2015). In vivo imaging and tracking of host–microbiota interactions via metabolic labeling of gut anaerobic bacteria. Nature Medicine, 21(9), 1091–1100. 10.1038/nm.3929 [DOI] [Google Scholar]
- Gill, R. K. , Saksena, S. , Alrefai, W. A. , Sarwar, Z. , Goldstein, J. L. , Carroll, R. E. , Ramaswamy, K. , & Dudeja, P. K. (2005). Expression and membrane localization of MCT isoforms along the length of the human intestine. American Journal of Physiology Cell Physiology, 289(4), C846–852. 10.1152/ajpcell.00112.2005 [DOI] [PubMed] [Google Scholar]
- Goldberg, J. , Currais, A. , Prior, M. , Fischer, W. , Chiruta, C. , Ratliff, E. , Daugherty, D. , Dargusch, R. , Finley, K. , Esparza‐Moltó, P. B. , Cuezva, J. M. , Maher, P. , Petrascheck, M. , & Schubert, D. (2018). The mitochondrial ATP synthase is a shared drug target for aging and dementia. Aging Cell, 17(2), e12715. 10.1111/acel.12715 [DOI] [Google Scholar]
- Grabert, K. , Michoel, T. , Karavolos, M. H. , Clohisey, S. , Baillie, J. K. , Stevens, M. P. , Freeman, T. C. , Summers, K. M. , & McColl, B. W. (2016). Microglial brain region−dependent diversity and selective regional sensitivities to aging. Nature Neuroscience, 19(3), 504–516. 10.1038/nn.4222 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Green, D. R. , Galluzzi, L. , & Kroemer, G. (2011). Mitochondria and the autophagy–inflammation–cell death axis in organismal aging. Science, 333(6046), 1109–1112. 10.1126/science.1201940 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Griciuc, A. , Serrano‐Pozo, A. , Parrado, A. R. , Lesinski, A. N. , Asselin, C. N. , Mullin, K. , Hooli, B. , Choi, S. H. , Hyman, B. T. , & Tanzi, R. E. (2013). Alzheimer’s disease risk gene CD33 inhibits microglial uptake of amyloid beta. Neuron, 78(4), 631–643. 10.1016/j.neuron.2013.04.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hammond, T. R. , Dufort, C. , Dissing‐Olesen, L. , Giera, S. , Young, A. , Wysoker, A. , Walker, A. J. , Gergits, F. , Segel, M. , Nemesh, J. , Marsh, S. E. , Saunders, A. , Macosko, E. , Ginhoux, F. , Chen, J. , Franklin, R. J. M. , Piao, X. , McCarroll, S. A. , & Stevens, B. (2019). Single‐cell RNA sequencing of microglia throughout the mouse lifespan and in the injured brain reveals complex cell‐state changes. Immunity, 50(1), 253–271. 10.1016/j.immuni.2018.11.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hanisch, U.‐K. , & Kettenmann, H. (2007). Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nature Neuroscience, 10(11), 1387–1394. 10.1038/nn1997 [DOI] [PubMed] [Google Scholar]
- Harach, T. , Marungruang, N. , Duthilleul, N. , Cheatham, V. , Mc Coy, K. D. , Frisoni, G. , Neher, J. J. , Fåk, F. , Jucker, M. , Lasser, T. , & Bolmont, T. (2017). Reduction of Abeta amyloid pathology in APPPS1 transgenic mice in the absence of gut microbiota. Scientific Reports, 7, 41802. 10.1038/srep41802 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harada, C. N. , Love, M. C. N. , & Triebel, K. L. (2013). Normal cognitive aging. Clinics in Geriatric Medicine, 29(4), 737–752. 10.1016/j.cger.2013.07.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hauptmann, J. , Johann, L. , Marini, F. , Kitic, M. , Colombo, E. , Mufazalov, I. A. , Krueger, M. , Karram, K. , Moos, S. , Wanke, F. , Kurschus, F. C. , Klein, M. , Cardoso, S. , Strauß, J. , Bolisetty, S. , Lühder, F. , Schwaninger, M. , Binder, H. , Bechman, I. , … Waisman, A. (2020). Interleukin‐1 promotes autoimmune neuroinflammation by suppressing endothelial heme oxygenase‐1 at the blood–brain barrier. Acta Neuropathologica, 140(4), 549–567. 10.1007/s00401-020-02187-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hedden, T. , & Gabrieli, J. D. (2004). Insights into the ageing mind: a view from cognitive neuroscience. Nature Reviews Neuroscience, 5(2), 87–96. 10.1038/nrn1323 [DOI] [PubMed] [Google Scholar]
- Henry, C. J. , Huang, Y. , Wynne, A. M. , & Godbout, J. P. (2009). Peripheral lipopolysaccharide (LPS) challenge promotes microglial hyperactivity in aged mice that is associated with exaggerated induction of both pro‐inflammatory IL‐1β and anti‐inflammatory IL‐10 cytokines. Brain, Behavior, and Immunity, 23(3), 309–317. 10.1016/j.bbi.2008.09.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heumann, D. , Barras, C. , Severin, A. , Glauser, M. , & Tomasz, A. (1994). Gram‐positive cell walls stimulate synthesis of tumor necrosis factor alpha and interleukin‐6 by human monocytes. Infection and Immunity, 62(7), 2715–2721. 10.1128/iai.62.7.2715-2721.1994 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hide, I. , Tanaka, M. , Inoue, A. , Nakajima, K. , Kohsaka, S. , Inoue, K. , & Nakata, Y. (2000). Extracellular ATP triggers tumor necrosis factor‐alpha release from rat microglia. Journal of Neurochemistry, 75(3), 965–972. 10.1046/j.1471-4159.2000.0750965.x [DOI] [PubMed] [Google Scholar]
- Hierro‐Bujalance, C. , Bacskai, B. J. , & Garcia‐Alloza, M. (2018). In Vivo Imaging of Microglia With Multiphoton Microscopy. Frotiers in Aging Neuroscience, 10, 10.3389/fnagi.2018.00218 [DOI] [Google Scholar]
- Hugon, P. , Dufour, J.‐C. , Colson, P. , Fournier, P.‐E. , Sallah, K. , & Raoult, D. (2015). A comprehensive repertoire of prokaryotic species identified in human beings. The Lancet Infectious Diseases, 15(10), 1211–1219. 10.1016/S1473-3099(15)00293-5 [DOI] [PubMed] [Google Scholar]
- Huuskonen, J. , Suuronen, T. , Nuutinen, T. , Kyrylenko, S. , & Salminen, A. (2004). Regulation of microglial inflammatory response by sodium butyrate and short‐chain fatty acids. British Journal of Pharmacology, 141(5), 874–880. 10.1038/sj.bjp.0705682 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jeffery, I. B. , Lynch, D. B. , & O'Toole, P. W. (2016). Composition and temporal stability of the gut microbiota in older persons. The ISME Journal, 10(1), 170–182. 10.1038/ismej.2015.88 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang, H. , Ling, Z. , Zhang, Y. , Mao, H. , Ma, Z. , Yin, Y. , Wang, W. , Tang, W. , Tan, Z. , Shi, J. , Li, L. , & Ruan, B. (2015). Altered fecal microbiota composition in patients with major depressive disorder. Brain, Behavior, and Immunity, 48, 186–194. 10.1016/j.bbi.2015.03.016 [DOI] [PubMed] [Google Scholar]
- Johnson, E. C. B. , Dammer, E. B. , Duong, D. M. , Ping, L. , Zhou, M. , Yin, L. , Higginbotham, L. A. , Guajardo, A. , White, B. , Troncoso, J. C. , Thambisetty, M. , Montine, T. J. , Lee, E. B. , Trojanowski, J. Q. , Beach, T. G. , Reiman, E. M. , Haroutunian, V. , Wang, M. , Schadt, E. , … Seyfried, N. T. (2020). Large‐scale proteomic analysis of Alzheimer’s disease brain and cerebrospinal fluid reveals early changes in energy metabolism associated with microglia and astrocyte activation. Nature Medicine, 26(5), 769–780. 10.1038/s41591-020-0815-6 [DOI] [Google Scholar]
- Kaczmarczyk, R. , Tejera, D. , Simon, B. J. , & Heneka, M. T. (2018). Microglia modulation through external vagus nerve stimulation in a murine model of Alzheimer's disease. Journal of Neurochemistry, 146(1), 76–85. 10.1111/jnc.14284 [DOI] [Google Scholar]
- Kalia, L. V. , & Lang, A. E. (2015). Parkinson's disease. Lancet, 386(9996), 896–912. 10.1016/s0140-6736(14)61393-3 [DOI] [PubMed] [Google Scholar]
- Kim, B.‐S. , Choi, C. W. , Shin, H. , Jin, S.‐P. , Bae, J.‐S. , Han, M. , Seo, E. Y. , Chun, J. , & Chung, J. H. (2019). Comparison of the gut microbiota of centenarians in longevity villages of south korea with those of other age groups. Journal of Microbiology and Biotechnology, 29(3), 429–440. 10.4014/jmb.1811.11023 [DOI] [PubMed] [Google Scholar]
- Kim, J. S. , Kirkland, R. A. , Lee, S. H. , Cawthon, C. R. , Rzepka, K. W. , Minaya, D. M. , de Lartigue, G. , Czaja, K. , & de La Serre, C. B. (2020). Gut microbiota composition modulates inflammation and structure of the vagal afferent pathway. Physiology & Behavior, 225, 113082. 10.1016/j.physbeh.2020.113082 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kondo, S. , Kohsaka, S. , & Okabe, S. (2011). Long‐term changes of spine dynamics and microglia after transient peripheral immune response triggered by LPS in vivo. Molecular Brain, 4(1), 1–16. 10.1186/1756-6606-4-27 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong, F. , Deng, F. , Li, Y. , & Zhao, J. (2019). Identification of gut microbiome signatures associated with longevity provides a promising modulation target for healthy aging. Gut Microbes, 10(2), 210–215. 10.1080/19490976.2018.1494102 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong, F. , Hua, Y. , Zeng, B. , Ning, R. , Li, Y. , & Zhao, J. (2016). Gut microbiota signatures of longevity. Current Biology, 26(18), R832–r833. 10.1016/j.cub.2016.08.015 [DOI] [PubMed] [Google Scholar]
- Kumar, M. , Babaei, P. , Ji, B. , & Nielsen, J. (2016). Human gut microbiota and healthy aging: Recent developments and future prospective. Nutr Healthy Aging, 4(1), 3–16. 10.3233/nha-150002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lal, S. , Kirkup, A. J. , Brunsden, A. M. , Thompson, D. G. , & Grundy, D. (2001). Vagal afferent responses to fatty acids of different chain length in the rat. American Journal of Physiology‐Gastrointestinal and Liver Physiology, 281(4), G907–G915. 10.1152/ajpgi.2001.281.4.G907 [DOI] [PubMed] [Google Scholar]
- La‐Ongkham, O. , Nakphaichit, M. , Nakayama, J. , Keawsompong, S. , & Nitisinprasert, S. (2020). Age‐related changes in the gut microbiota and the core gut microbiome of healthy Thai humans. 3 Biotech, 10(6), 276. 10.1007/s13205-020-02265-7 [DOI] [Google Scholar]
- Laurent, C. , Dorothée, G. , Hunot, S. , Martin, E. , Monnet, Y. , Duchamp, M. , Dong, Y. , Légeron, F.‐P. , Leboucher, A. , Burnouf, S. , Faivre, E. , Carvalho, K. , Caillierez, R. , Zommer, N. , Demeyer, D. , Jouy, N. , Sazdovitch, V. , Schraen‐Maschke, S. , Delarasse, C. , … Blum, D. (2017). Hippocampal T cell infiltration promotes neuroinflammation and cognitive decline in a mouse model of tauopathy. Brain, 140(1), 184–200. 10.1093/brain/aww270 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee, C. Y. D. , & Landreth, G. E. (2010). The role of microglia in amyloid clearance from the AD brain. Journal of Neural Transmission, 117(8), 949–960. 10.1007/s00702-010-0433-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee, K.‐E. , Kim, J.‐K. , Han, S.‐K. , Lee, D. Y. , Lee, H.‐J. , Yim, S.‐V. , & Kim, D.‐H. (2020). The extracellular vesicle of gut microbial Paenalcaligenes hominis is a risk factor for vagus nerve‐mediated cognitive impairment. Microbiome, 8(1), 1–18. 10.1186/s40168-020-00881-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li, D. , Lang, W. , Zhou, C. , Wu, C. , Zhang, F. , Liu, Q. , Yang, S. , & Hao, J. (2018). Upregulation of microglial ZEB1 ameliorates brain damage after acute ischemic stroke. Cell Reports, 22(13), 3574–3586. 10.1016/j.celrep.2018.03.011 [DOI] [PubMed] [Google Scholar]
- Li, Q. , Chen, L. , Liu, X. , Li, X. , Cao, Y. , Bai, Y. , & Qi, F. (2018). Pterostilbene inhibits amyloid‐β‐induced neuroinflammation in a microglia cell line by inactivating the NLRP3/caspase‐1 inflammasome pathway. Journal of Cellular Biochemistry, 119(8), 7053–7062. 10.1002/jcb.27023 [DOI] [PubMed] [Google Scholar]
- Lin, C.‐H. , Chen, C.‐C. , Chiang, H.‐L. , Liou, J.‐M. , Chang, C.‐M. , Lu, T.‐P. , Chuang, E. Y. , Tai, Y.‐C. , Cheng, C. , Lin, H.‐Y. , & Wu, M.‐S. (2019). Altered gut microbiota and inflammatory cytokine responses in patients with Parkinson's disease. Journal of Neuroinflammation, 16(1), 129. 10.1186/s12974-019-1528-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu, Y. , Wu, Z. , Zhang, X. , Ni, J. , Yu, W. , Zhou, Y. , & Nakanishi, H. (2013). Leptomeningeal cells transduce peripheral macrophages inflammatory signal to microglia in reponse to Porphyromonas gingivalis LPS. Mediators of Inflammation, 2013, 407562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lynch, S. V. , & Pedersen, O. (2016). The human intestinal microbiome in health and disease. New England Journal of Medicine, 375(24), 2369–2379. 10.1056/NEJMra1600266 [DOI] [PubMed] [Google Scholar]
- MacDonald, T. T. , & Monteleone, G. (2005). Immunity, inflammation, and allergy in the gut. Science, 307(5717), 1920–1925. 10.1126/science.1106442 [DOI] [PubMed] [Google Scholar]
- Macfarlane, S. , & Macfarlane, G. T. (2003). Regulation of short‐chain fatty acid production. Proceedings of the Nutrition Society, 62(1), 67–72. 10.1079/PNS2002207 [DOI] [PubMed] [Google Scholar]
- Mäkivuokko, H. , Tiihonen, K. , Tynkkynen, S. , Paulin, L. , & Rautonen, N. (2010). The effect of age and non‐steroidal anti‐inflammatory drugs on human intestinal microbiota composition. British Journal of Nutrition, 103(2), 227–234. 10.1017/s0007114509991553 [DOI] [PubMed] [Google Scholar]
- Mandrekar, S. , Jiang, Q. , Lee, C. Y. D. , Koenigsknecht‐Talboo, J. , Holtzman, D. M. , & Landreth, G. E. (2009). Microglia Mediate the Clearance of Soluble Aβ through Fluid Phase Macropinocytosis. The Journal of Neuroscience, 29(13), 4252. 10.1523/JNEUROSCI.5572-08.2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martin, E. , Boucher, C. , Fontaine, B. , & Delarasse, C. (2017). Distinct inflammatory phenotypes of microglia and monocyte‐derived macrophages in Alzheimer's disease models: effects of aging and amyloid pathology. Aging Cell, 16(1), 27–38. 10.1111/acel.12522 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Masuda, T. , Sankowski, R. , Staszewski, O. , & Prinz, M. (2020). Microglia heterogeneity in the single‐cell era. Cell Reports, 30(5), 1271–1281. 10.1016/j.celrep.2020.01.010 [DOI] [PubMed] [Google Scholar]
- Matt, S. M. , Allen, J. M. , Lawson, M. A. , Mailing, L. J. , Woods, J. A. , & Johnson, R. W. (2018). Butyrate and dietary soluble fiber improve neuroinflammation associated with aging in mice. Frontiers in Immunology, 9, 1832. 10.3389/fimmu.2018.01832 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mattson, M. P. , & Arumugam, T. V. (2018). Hallmarks of brain aging: adaptive and pathological modification by metabolic states. Cell Metabolism, 27(6), 1176–1199. 10.1016/j.cmet.2018.05.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller, T. L. , & Wolin, M. J. (1996). Pathways of acetate, propionate, and butyrate formation by the human fecal microbial flora. Applied and Environmental Microbiology, 62(5), 1589–1592. 10.1128/aem.62.5.1589-1592.1996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mora, J. R. , Bono, M. R. , Manjunath, N. , Weninger, W. , Cavanagh, L. L. , Rosemblatt, M. , & Von Andrian, U. H. (2003). Selective imprinting of gut‐homing T cells by Peyer's patch dendritic cells. Nature, 424(6944), 88–93. 10.1038/nature01726 [DOI] [PubMed] [Google Scholar]
- Moreno‐Valladares, M. , Moreno‐Cugnon, L. , Silva, T. M. , Garcés, J. P. , Saenz‐Antoñanzas, A. , Álvarez‐Satta, M. , & Matheu, A. (2020). CD8+ T cells are increased in the subventricular zone with physiological and pathological aging. Aging Cell, 19(9), e13198. 10.1111/acel.13198 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morrison, K. E. , Jašarević, E. , Howard, C. D. , & Bale, T. L. (2020). It's the fiber, not the fat: significant effects of dietary challenge on the gut microbiome. Microbiome, 8(1), 15. 10.1186/s40168-020-0791-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moskalev, A. A. , Shaposhnikov, M. V. , Plyusnina, E. N. , Zhavoronkov, A. , Budovsky, A. , Yanai, H. , & Fraifeld, V. E. (2013). The role of DNA damage and repair in aging through the prism of Koch‐like criteria. Ageing Research Reviews, 12(2), 661–684. 10.1016/j.arr.2012.02.001 [DOI] [PubMed] [Google Scholar]
- Nagatsu, T. , & Sawada, M. (2005). Inflammatory process in parkinsons disease: role for cytokines. Current Pharmaceutical Design, 11(8), 999–1016. 10.2174/1381612053381620 [DOI] [PubMed] [Google Scholar]
- Nagpal, R. , Neth, B. J. , Wang, S. , Craft, S. , & Yadav, H. (2019). Modified Mediterranean‐ketogenic diet modulates gut microbiome and short‐chain fatty acids in association with Alzheimer's disease markers in subjects with mild cognitive impairment. EBioMedicine, 47, 529–542. 10.1016/j.ebiom.2019.08.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nguyen, P. T. , Dorman, L. C. , Pan, S. , Vainchtein, I. D. , Han, R. T. , Nakao‐Inoue, H. , Taloma, S. E. , Barron, J. J. , Molofsky, A. B. , Kheirbek, M. A. , & Molofsky, A. V. (2020). Microglial remodeling of the extracellular matrix promotes synapse plasticity. Cell, 182(2), 388–403.e315. 10.1016/j.cell.2020.05.050 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nicoletti, C. (2015). Age‐associated changes of the intestinal epithelial barrier: local and systemic implications. Expert Review of Gastroenterology & Hepatology, 9(12), 1467–1469. 10.1586/17474124.2015.1092872 [DOI] [PubMed] [Google Scholar]
- Odamaki, T. , Kato, K. , Sugahara, H. , Hashikura, N. , Takahashi, S. , Xiao, J.‐Z. , Abe, F. , & Osawa, R. O. (2016). Age‐related changes in gut microbiota composition from newborn to centenarian: a cross‐sectional study. BMC Microbiology, 16, 90. 10.1186/s12866-016-0708-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ohm, D. T. , Fought, A. J. , Martersteck, A. , Coventry, C. , Sridhar, J. , Gefen, T. , Weintraub, S. , Bigio, E. , Mesulam, M.‐M. , Rogalski, E. , & Geula, C. (2021). Accumulation of neurofibrillary tangles and activated microglia is associated with lower neuron densities in the aphasic variant of Alzheimer’s disease. Brain Pathology, 31(1), 189–204. 10.1111/bpa.12902 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olah, M. , Patrick, E. , Villani, A.‐C. , Xu, J. , White, C. C. , Ryan, K. J. , Piehowski, P. , Kapasi, A. , Nejad, P. , Cimpean, M. , Connor, S. , Yung, C. J. , Frangieh, M. , McHenry, A. , Elyaman, W. , Petyuk, V. , Schneider, J. A. , Bennett, D. A. , De Jager, P. L. , & Bradshaw, E. M. (2018). A transcriptomic atlas of aged human microglia. Nature Communications, 9(1), 1–8. 10.1038/s41467-018-02926-5 [DOI] [Google Scholar]
- Olmos‐Alonso, A. , Schetters, S. T. T. , Sri, S. , Askew, K. , Mancuso, R. , Vargas‐Caballero, M. , Holscher, C. , Perry, V. H. , & Gomez‐Nicola, D. (2016). Pharmacological targeting of CSF1R inhibits microglial proliferation and prevents the progression of Alzheimer’s‐like pathology. Brain, 139(3), 891–907. 10.1093/brain/awv379 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Organization, W. H. (2015). World report on ageing and health. World Health Organization. [Google Scholar]
- Pallikuth, S. , Russell, K. B. , Sirupangi, T. , Mendez, R. , Kvistad, D. K. , Pahwa, R. , & Pahwa, S. (2021). Age associated microbiome modulation and its association with systemic inflammation in a rhesus macaque model. Frontiers in Immunology, 12. 10.3389/fimmu.2021.748397 [DOI] [Google Scholar]
- Papageorgiou, I. E. , Lewen, A. , Galow, L. V. , Cesetti, T. , Scheffel, J. , Regen, T. , Hanisch, U.‐K. , & Kann, O. (2016). TLR4‐activated microglia require IFN‐γ to induce severe neuronal dysfunction and death in situ. Proceedings of the National Academy of Sciences, 113(1), 212. 10.1073/pnas.1513853113 [DOI] [Google Scholar]
- Parhizkar, S. , Arzberger, T. , Brendel, M. , Kleinberger, G. , Deussing, M. , Focke, C. , Nuscher, B. , Xiong, M. , Ghasemigharagoz, A. , Katzmarski, N. , Krasemann, S. , Lichtenthaler, S. F. , Müller, S. A. , Colombo, A. , Monasor, L. S. , Tahirovic, S. , Herms, J. , Willem, M. , Pettkus, N. , … Haass, C. (2019). Loss of TREM2 function increases amyloid seeding but reduces plaque‐associated ApoE. Nature Neuroscience, 22(2), 191–204. 10.1038/s41593-018-0296-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park, J.‐S. , Woo, M.‐S. , Kim, S.‐Y. , Kim, W.‐K. , & Kim, H.‐S. (2005). Repression of interferon‐γ‐induced inducible nitric oxide synthase (iNOS) gene expression in microglia by sodium butyrate is mediated through specific inhibition of ERK signaling pathways. Journal of Neuroimmunology, 168(1–2), 56–64. 10.1016/j.jneuroim.2005.07.003 [DOI] [PubMed] [Google Scholar]
- Patterson, L. M. , Zheng, H. , & Berthoud, H. R. (2002). Vagal afferents innervating the gastrointestinal tract and CCKA‐receptor immunoreactivity. The Anatomical Record, 266(1), 10–20. 10.1002/ar.10026 [DOI] [PubMed] [Google Scholar]
- Pelvig, D. P. , Pakkenberg, H. , Stark, A. K. , & Pakkenberg, B. (2008). Neocortical glial cell numbers in human brains. Neurobiology of Aging, 29(11), 1754–1762. 10.1016/j.neurobiolaging.2007.04.013 [DOI] [PubMed] [Google Scholar]
- Perry, V. H. , & Holmes, C. (2014). Microglial priming in neurodegenerative disease. Nature Reviews Neurology, 10(4), 217–224. 10.1038/nrneurol.2014.38 [DOI] [PubMed] [Google Scholar]
- Perry, V. H. , Nicoll, J. A. R. , & Holmes, C. (2010). Microglia in neurodegenerative disease. Nature Reviews Neurology, 6(4), 193–201. 10.1038/nrneurol.2010.17 [DOI] [PubMed] [Google Scholar]
- Politis, M. , Wu, K. , Molloy, S. , G. Bain, P. , Chaudhuri, K. R. , & Piccini, P. (2010). Parkinson's disease symptoms: The patient's perspective. Movement Disorders, 25(11), 1646–1651. 10.1002/mds.23135 [DOI] [PubMed] [Google Scholar]
- Raj, D. D. A. , Jaarsma, D. , Holtman, I. R. , Olah, M. , Ferreira, F. M. , Schaafsma, W. , Brouwer, N. , Meijer, M. M. , de Waard, M. C. , van der Pluijm, I. , Brandt, R. , Kreft, K. L. , Laman, J. D. , de Haan, G. , Biber, K. P. H. , Hoeijmakers, J. H. J. , Eggen, B. J. L. , & Boddeke, H. W. G. M. (2014). Priming of microglia in a DNA‐repair deficient model of accelerated aging. Neurobiology of Aging, 35(9), 2147–2160. 10.1016/j.neurobiolaging.2014.03.025 [DOI] [PubMed] [Google Scholar]
- Raj, D. D. A. , Moser, J. , Pol, S. M. A. , Os, R. P. , Holtman, I. R. , Brouwer, N. , Oeseburg, H. , Schaafsma, W. , Wesseling, E. M. , Dunnen, W. , Biber, K. P. H. , Vries, H. E. , Eggen, B. J. L. , & Boddeke, H. W. G. M. (2015). Enhanced microglial pro‐inflammatory response to lipopolysaccharide correlates with brain infiltration and blood–brain barrier dysregulation in a mouse model of telomere shortening. Aging Cell, 14(6), 1003–1013. 10.1111/acel.12370 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rampelli, S. , Candela, M. , Turroni, S. , Biagi, E. , Collino, S. , Franceschi, C. , O'Toole, P. W. , & Brigidi, P. (2013). Functional metagenomic profiling of intestinal microbiome in extreme ageing. Aging (Albany NY), 5(12), 902–912. 10.18632/aging.100623 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rampelli, S. , Soverini, M. , D'Amico, F. , Barone, M. , Tavella, T. , Monti, D. , & Candela, M. (2020). Shotgun metagenomics of gut microbiota in humans with up to extreme longevity and the increasing role of xenobiotic degradation. mSystems, 5(2), 20. 10.1128/mSystems.00124-20 [DOI] [Google Scholar]
- Rayaprolu, S. , Mullen, B. , Baker, M. , Lynch, T. , Finger, E. , Seeley, W. W. , & Ross, O. A. (2013). TREM2 in neurodegeneration: evidence for association of the p.R47H variant with frontotemporal dementia and Parkinson’s disease. Molecular Neurodegeneration, 8(1), 19. 10.1186/1750-1326-8-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rekdal, V. M. , Bess, E. N. , Bisanz, J. E. , Turnbaugh, P. J. , & Balskus, E. P. (2019). Discovery and inhibition of an interspecies gut bacterial pathway for Levodopa metabolism. Science, 364, 6445. 10.1126/science.aau6323 [DOI] [Google Scholar]
- Rochfort, K. D. , Collins, L. E. , Murphy, R. P. , & Cummins, P. M. (2014). Downregulation of Blood‐Brain Barrier Phenotype by Proinflammatory Cytokines Involves NADPH Oxidase‐Dependent ROS Generation: Consequences for Interendothelial Adherens and Tight Junctions. PLoS One, 9(7), e101815. 10.1371/journal.pone.0101815 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ronchi, G. , Ryu, V. , Fornaro, M. , & Czaja, K. (2012). Hippocampal plasticity after a vagus nerve injury in the rat. Neural Regeneration Research, 7(14), 1055. 10.3969/j.issn.1673-5374.2012.14.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salazar, N. , Arboleya, S. , Fernández‐Navarro, T. , de Los Reyes‐Gavilán, C. G. , Gonzalez, S. , & Gueimonde, M. (2019). Age‐associated changes in gut microbiota and dietary components related with the immune system in adulthood and old age: A cross‐sectional study. Nutrients, 11(8), 1765. 10.3390/nu11081765 [DOI] [Google Scholar]
- Salazar, N. , López, P. , Valdés, L. , Margolles, A. , Suárez, A. , Patterson, Á. M. , Cuervo, A. , Reyes‐Gavilán, C. G. D. L. , Ruas‐Madiedo, P. , Gonzalez, S. , & Gueimonde, M. (2013). Microbial targets for the development of functional foods accordingly with nutritional and immune parameters altered in the elderly. Journal of the American College of Nutrition, 32(6), 399–406. 10.1080/07315724.2013.827047 [DOI] [PubMed] [Google Scholar]
- Sampson, T. R. , Debelius, J. W. , Thron, T. , Janssen, S. , Shastri, G. G. , Ilhan, Z. E. , Challis, C. , Schretter, C. E. , Rocha, S. , Gradinaru, V. , Chesselet, M.‐F. , Keshavarzian, A. , Shannon, K. M. , Krajmalnik‐Brown, R. , Wittung‐Stafshede, P. , Knight, R. , & Mazmanian, S. K. (2016). Gut microbiota regulate motor deficits and neuroinflammation in a model of parkinson's disease. Cell, 167(6), 1469–1480.e1412. 10.1016/j.cell.2016.11.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sampson, T. R. , & Mazmanian, S. K. (2015). Control of brain development, function, and behavior by the microbiome. Cell Host & Microbe, 17(5), 565–576. 10.1016/j.chom.2015.04.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sarna, G. , Bradbury, M. , Cremer, J. E. , Lai, J. , & Teal, H. M. (1979). Brain metabolism and specific transport at the blood‐brain barrier after portocaval anastomosis in the rat. Brain Research, 160(1), 69–83. 10.1016/0006-8993(79)90601-2 [DOI] [PubMed] [Google Scholar]
- Savelieff, M. G. , Lee, S. , Liu, Y. , & Lim, M. H. (2013). Untangling amyloid‐β, tau, and metals in alzheimer’s disease. ACS Chemical Biology, 8(5), 856–865. 10.1021/cb400080f [DOI] [PubMed] [Google Scholar]
- Schiffrin, E. J. , Morley, J. E. , Donnet‐Hughes, A. , & Guigoz, Y. (2010). The inflammatory status of the elderly: the intestinal contribution. Mutation Research, 690(1–2), 50–56. 10.1016/j.mrfmmm.2009.07.011 [DOI] [PubMed] [Google Scholar]
- Schmidtner, A. K. , Slattery, D. A. , Gläsner, J. , Hiergeist, A. , Gryksa, K. , Malik, V. A. , Hellmann‐Regen, J. , Heuser, I. , Baghai, T. C. , Gessner, A. , Rupprecht, R. , Di Benedetto, B. , & Neumann, I. D. (2019). Minocycline alters behavior, microglia and the gut microbiome in a trait‐anxiety‐dependent manner. Translational Psychiatry, 9(1), 1–12. 10.1038/s41398-019-0556-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sen, T. , Cawthon, C. R. , Ihde, B. T. , Hajnal, A. , DiLorenzo, P. M. , Claire, B. , & Czaja, K. (2017). Diet‐driven microbiota dysbiosis is associated with vagal remodeling and obesity. Physiology & Behavior, 173, 305–317. 10.1016/j.physbeh.2017.02.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shahidehpour, R. K. , Higdon, R. E. , Crawford, N. G. , Neltner, J. H. , Ighodaro, E. T. , Patel, E. , Price, D. , Nelson, P. T. , & Bachstetter, A. D. (2021). Dystrophic microglia are associated with neurodegenerative disease and not healthy aging in the human brain. Neurobiology of Aging, 99, 19–27. 10.1016/j.neurobiolaging.2020.12.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi, H. , Shi, Q. , Grodner, B. , Lenz, J. S. , Zipfel, W. R. , Brito, I. L. , & De Vlaminck, I. (2020). Highly multiplexed spatial mapping of microbial communities. Nature, 1–6. 10.1038/s41586-020-2983-4 [DOI] [Google Scholar]
- Shobin, E. , Bowley, M. P. , Estrada, L. I. , Heyworth, N. C. , Orczykowski, M. E. , Eldridge, S. A. , Calderazzo, S. M. , Mortazavi, F. , Moore, T. L. , & Rosene, D. L. (2017). Microglia activation and phagocytosis: relationship with aging and cognitive impairment in the rhesus monkey. Geroscience, 39(2), 199–220. 10.1007/s11357-017-9965-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Skrobot, O. A. , O'Brien, J. , Black, S. , Chen, C. , DeCarli, C. , Erkinjuntti, T. , Ford, G. A. , Kalaria, R. N. , Pantoni, L. , Pasquier, F. , Roman, G. C. , Wallin, A. , Sachdev, P. , Skoog, I. , Taragano, F. E. , Kril, J. , Cavalieri, M. , Jellinger, K. A. , Kovacs, G. G. , … Kehoe, P. G. (2017). The vascular impairment of cognition classification consensus study. Alzheimer's & Dementia, 13(6), 624–633. 10.1016/j.jalz.2016.10.007 [DOI] [Google Scholar]
- Ślusarczyk, J. , Trojan, E. , Głombik, K. , Piotrowska, A. , Budziszewska, B. , Kubera, M. , Popiołek‐Barczyk, K. , Lasoń, W. , Mika, J. , & Basta‐Kaim, A. (2018). Targeting the NLRP3 inflammasome‐related pathways via tianeptine treatment‐suppressed microglia polarization to the M1 phenotype in lipopolysaccharide‐stimulated cultures. International Journal of Molecular Sciences, 19(7), 1965. 10.3390/ijms19071965 [DOI] [Google Scholar]
- Solano Fonseca, R. , Mahesula, S. , Apple, D. M. , Raghunathan, R. , Dugan, A. , Cardona, A. , O'Connor, J. , & Kokovay, E. (2016). Neurogenic niche microglia undergo positional remodeling and progressive activation contributing to age‐associated reductions in neurogenesis. Stem Cells and Development, 25(7), 542–555. 10.1089/scd.2015.0319 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stevens, B. R. , Goel, R. , Seungbum, K. , Richards, E. M. , Holbert, R. C. , Pepine, C. J. , & Raizada, M. K. (2018). Increased human intestinal barrier permeability plasma biomarkers zonulin and FABP2 correlated with plasma LPS and altered gut microbiome in anxiety or depression. Gut, 67(8), 1555–1557. 10.1136/gutjnl-2017-314759 [DOI] [Google Scholar]
- Stevens, B. R. , Roesch, L. , Thiago, P. , Russell, J. T. , Pepine, C. J. , Holbert, R. C. , Raizada, M. K. , & Triplett, E. W. (2020). Depression phenotype identified by using single nucleotide exact amplicon sequence variants of the human gut microbiome. Molecular Psychiatry, 26(8), 4277–4287, 10.1038/s41380-020-0652-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stichel, C. , & Luebbert, H. (2007). Inflammatory processes in the aging mouse brain: participation of dendritic cells and T‐cells. Neurobiology of Aging, 28(10), 1507–1521. 10.1016/j.neurobiolaging.2006.07.022 [DOI] [PubMed] [Google Scholar]
- Stones, M. J. (2019). Age differences, age changes and their generalizability in marathon running by master athletes. Frontiers in Psychology, 10, 2161. 10.3389/fpsyg.2019.02161 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Streit, W. J. , Braak, H. , Xue, Q. S. , & Bechmann, I. (2009). Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer's disease. Acta Neuropathologica, 118(4), 475–485. 10.1007/s00401-009-0556-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Streit, W. J. , Mrak, R. E. , & Griffin, W. S. T. (2004). Microglia and neuroinflammation: a pathological perspective. Journal of Neuroinflammation, 1(1), 14. 10.1186/1742-2094-1-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Streit, W. J. , Sammons, N. W. , Kuhns, A. J. , & Sparks, D. L. (2004). Dystrophic microglia in the aging human brain. Glia, 45(2), 208–212. 10.1002/glia.10319 [DOI] [PubMed] [Google Scholar]
- Sun, J. , Xu, J. , Yang, B. , Chen, K. , Kong, Y. , Fang, N. , & Liu, J. (2020). Effect of Clostridium butyricum against Microglia‐Mediated Neuroinflammation in Alzheimer's Disease via Regulating Gut Microbiota and Metabolites Butyrate. Molecular Nutrition & Food Research, 64(2), 1900636. 10.1002/mnfr.201900636 [DOI] [Google Scholar]
- Sun, M.‐F. , Zhu, Y.‐L. , Zhou, Z.‐L. , Jia, X.‐B. , Xu, Y.‐D. , Yang, Q. , Cui, C. , & Shen, Y.‐Q. (2018). Neuroprotective effects of fecal microbiota transplantation on MPTP‐induced Parkinson’s disease mice: Gut microbiota, glial reaction and TLR4/TNF‐α signaling pathway. Brain, Behavior, and Immunity, 70, 48–60. 10.1016/j.bbi.2018.02.005 [DOI] [PubMed] [Google Scholar]
- Tahara, K. , Kim, H.‐D. , Jin, J.‐J. , Maxwell, J. A. , Li, L. , & Fukuchi, K.‐I. (2006). Role of toll‐like receptor signalling in Aβ uptake and clearance. Brain, 129(11), 3006–3019. 10.1093/brain/awl249 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tan, J. , McKenzie, C. , Potamitis, M. , Thorburn, A. N. , Mackay, C. R. , & Macia, L. (2014). The role of short‐chain fatty acids in health and disease. Advances in Immunology, 121, 91–119. 10.1016/b978-0-12-800100-4.00003-9 [DOI] [PubMed] [Google Scholar]
- Thevaranjan, N. , Puchta, A. , Schulz, C. , Naidoo, A. , Szamosi, J. C. , Verschoor, C. P. , Loukov, D. , Schenck, L. P. , Jury, J. , Foley, K. P. , Schertzer, J. D. , Larché, M. J. , Davidson, D. J. , Verdú, E. F. , Surette, M. G. , & Bowdish, D. M. E. (2017). Age‐associated microbial dysbiosis promotes intestinal permeability, systemic inflammation, and macrophage dysfunction. Cell Host & Microbe, 21(4), 455–466. e454. 10.1016/j.chom.2017.03.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thursby, E. , & Juge, N. (2017). Introduction to the human gut microbiota. Biochemical Journal, 474(11), 1823–1836. 10.1042/BCJ20160510 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tibolla, G. , Norata, G. D. , Meda, C. , Arnaboldi, L. , Uboldi, P. , Piazza, F. , Ferrarese, C. , Corsini, A. , Maggi, A. , Vegeto, E. , & Catapano, A. L. (2010). Increased atherosclerosis and vascular inflammation in APP transgenic mice with apolipoprotein E deficiency. Atherosclerosis, 210(1), 78–87. 10.1016/j.atherosclerosis.2009.10.040 [DOI] [PubMed] [Google Scholar]
- Tichauer, J. E. , Flores, B. , Soler, B. , Eugenín‐von Bernhardi, L. , Ramírez, G. , & von Bernhardi, R. (2014). Age‐dependent changes on TGFβ1 Smad3 pathway modify the pattern of microglial cell activation. Brain, Behavior, and Immunity, 37, 187–196. 10.1016/j.bbi.2013.12.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tischer, J. , Krueger, M. , Mueller, W. , Staszewski, O. , Prinz, M. , Streit, W. J. , & Bechmann, I. (2016). Inhomogeneous distribution of Iba‐1 characterizes microglial pathology in Alzheimer's disease. Glia, 64(9), 1562–1572. 10.1002/glia.23024 [DOI] [PubMed] [Google Scholar]
- Unger, M. S. , Marschallinger, J. , Kaindl, J. , Klein, B. , Johnson, M. , Khundakar, A. A. , Roßner, S. , Heneka, M. T. , Couillard‐Despres, S. , Rockenstein, E. , Masliah, E. , Attems, J. , & Aigner, L. (2018). Doublecortin expression in CD8+ T‐cells and microglia at sites of amyloid‐β plaques: A potential role in shaping plaque pathology? Alzheimer's & Dementia, 14(8), 1022–1037. 10.1016/j.jalz.2018.02.017 [DOI] [Google Scholar]
- Unger, M. , Schernthaner, P. , Marschallinger, J. , Mrowetz, H. , & Aigner, L. (2018). Microglia prevent peripheral immune cell invasion and promote an anti‐inflammatory environment in the brain of APP‐PS1 transgenic mice. Journal of Neuroinflammation, 15(1), 1–23. 10.1186/s12974-018-1304-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Unger, M. M. , Spiegel, J. , Dillmann, K.‐U. , Grundmann, D. , Philippeit, H. , Bürmann, J. , Faßbender, K. , Schwiertz, A. , & Schäfer, K.‐H. (2016). Short chain fatty acids and gut microbiota differ between patients with Parkinson's disease and age‐matched controls. Parkinsonism & Related Disorders, 32, 66–72. 10.1016/j.parkreldis.2016.08.019 [DOI] [PubMed] [Google Scholar]
- Valles‐Colomer, M. , Falony, G. , Darzi, Y. , Tigchelaar, E. F. , Wang, J. , Tito, R. Y. , Schiweck, C. , Kurilshikov, A. , Joossens, M. , Wijmenga, C. , Claes, S. , Van Oudenhove, L. , Zhernakova, A. , Vieira‐Silva, S. , & Raes, J. (2019). The neuroactive potential of the human gut microbiota in quality of life and depression. Nature Microbiology, 4(4), 623–632. 10.1038/s41564-018-0337-x [DOI] [Google Scholar]
- Van Rossum, D. , & Hanisch, U. K. (2004). Microglia. Metabolic Brain Disease, 19(3–4), 393–411. 10.1023/B:MEBR.0000043984.73063.d8 [DOI] [PubMed] [Google Scholar]
- Velin, Å. K. , Ericson, A. C. , Braaf, Y. , Wallon, C. , & Söderholm, J. D. (2004). Increased antigen and bacterial uptake in follicle associated epithelium induced by chronic psychological stress in rats. Gut, 53(4), 494. 10.1136/gut.2003.028506 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Venegas, C. , & Heneka, M. T. (2017). Danger‐associated molecular patterns in Alzheimer's disease. Journal of Leukocyte Biology, 101(1), 87–98. 10.1189/jlb.3MR0416-204R [DOI] [PubMed] [Google Scholar]
- Venegas, C. , Kumar, S. , Franklin, B. S. , Dierkes, T. , Brinkschulte, R. , Tejera, D. , Vieira‐Saecker, A. , Schwartz, S. , Santarelli, F. , Kummer, M. P. , Griep, A. , Gelpi, E. , Beilharz, M. , Riedel, D. , Golenbock, D. T. , Geyer, M. , Walter, J. , Latz, E. , & Heneka, M. T. (2017). Microglia‐derived ASC specks cross‐seed amyloid‐β in Alzheimer’s disease. Nature, 552(7685), 355–361. 10.1038/nature25158 [DOI] [PubMed] [Google Scholar]
- Vijay‐Kumar, M. , Aitken, J. D. , Carvalho, F. A. , Cullender, T. C. , Mwangi, S. , Srinivasan, S. , Sitaraman, S. V. , Knight, R. , Ley, R. E. , & Gewirtz, A. T. (2010). Metabolic syndrome and altered gut microbiota in mice lacking Toll‐like receptor 5. Science, 328(5975), 228–231. 10.1126/science.1179721 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vogt, N. M. , Kerby, R. L. , Dill‐McFarland, K. A. , Harding, S. J. , Merluzzi, A. P. , Johnson, S. C. , Carlsson, C. M. , Asthana, S. , Zetterberg, H. , Blennow, K. , Bendlin, B. B. , & Rey, F. E. (2017). Gut microbiome alterations in Alzheimer's disease. Scientific Reports, 7(1), 13537. 10.1038/s41598-017-13601-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walker, D. G. , & Lue, L. F. (2015). Immune phenotypes of microglia in human neurodegenerative disease: challenges to detecting microglial polarization in human brains. Alzheimers Res Ther, 7(1), 56. 10.1186/s13195-015-0139-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walter, S. , Letiembre, M. , Liu, Y. , Heine, H. , Penke, B. , Hao, W. , Bode, B. , Manietta, N. , Walter, J. , Schulz‐Schüffer, W. , & Fassbender, K. (2007). Role of the Toll‐Like Receptor 4 in Neuroinflammation in Alzheimer’s Disease. Cellular Physiology and Biochemistry, 20(6), 947–956. 10.1159/000110455 [DOI] [PubMed] [Google Scholar]
- Wang, C. , Yue, H. , Hu, Z. , Shen, Y. , Ma, J. , Li, J. , & Shi, P. (2020). Microglia mediate forgetting via complement‐dependent synaptic elimination. Science, 367(6478), 688–694. 10.1126/science.aaz2288 [DOI] [PubMed] [Google Scholar]
- Wang, Y. , Zhang, C. , Lai, J. , Zhao, Y. , Lu, D. , Bao, R. , Feng, X. , Zhang, T. , & Liu, Z. (2020). Noninvasive PET tracking of post‐transplant gut microbiota in living mice. European Journal of Nuclear Medicine and Molecular Imaging, 47(4), 991–1002. 10.1007/s00259-019-04639-3 [DOI] [PubMed] [Google Scholar]
- Wardlaw, J. M. , Makin, S. J. , Valdés Hernández, M. C. , Armitage, P. A. , Heye, A. K. , Chappell, F. M. , Muñoz‐Maniega, S. , Sakka, E. , Shuler, K. , Dennis, M. S. , & Thrippleton, M. J. (2017). Blood‐brain barrier failure as a core mechanism in cerebral small vessel disease and dementia: evidence from a cohort study. Alzheimer's & Dementia, 13(6), 634–643. 10.1016/j.jalz.2016.09.006 [DOI] [Google Scholar]
- Wen, J. , Ding, Y. , Wang, L. , & Xiao, Y. (2020). Gut microbiome improves postoperative cognitive function by decreasing permeability of the blood‐brain barrier in aged mice. Brain Research Bulletin, 164, 249–256. 10.1016/j.brainresbull.2020.08.017 [DOI] [PubMed] [Google Scholar]
- Wong, J. M. , De Souza, R. , Kendall, C. W. , Emam, A. , & Jenkins, D. J. (2006). Colonic health: fermentation and short chain fatty acids. Journal of Clinical Gastroenterology, 40(3), 235–243. 10.1097/00004836-200603000-00015 [DOI] [PubMed] [Google Scholar]
- Wu, L. U. , Zeng, T. , Deligios, M. , Milanesi, L. , Langille, M. G. I. , Zinellu, A. , Rubino, S. , Carru, C. , & Kelvin, D. J. (2020). Age‐related variation of bacterial and fungal communities in different body habitats across the young. Elderly, and Centenarians in Sardinia. Msphere, 5(1), 10.1128/mSphere.00558-19 [DOI] [Google Scholar]
- Wu, L. , Zeng, T. , Zinellu, A. , Rubino, S. , Kelvin, D. J. , & Carru, C. (2019). A cross‐sectional study of compositional and functional profiles of gut microbiota in sardinian centenarians. mSystems, 4(4). 10.1128/mSystems.00325-19 [DOI] [Google Scholar]
- Xu, C. , Lee, S. K. , Zhang, D. , & Frenette, P. S. (2020). The gut microbiome regulates psychological‐stress‐induced inflammation Immunity, 53(2), 417–428. 10.1016/j.immuni.2020.06.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamawaki, Y. , Yoshioka, N. , Nozaki, K. , Ito, H. , Oda, K. , Harada, K. , Shirawachi, S. , Asano, S. , Aizawa, H. , Yamawaki, S. , Kanematsu, T. , & Akagi, H. (2018). Sodium butyrate abolishes lipopolysaccharide‐induced depression‐like behaviors and hippocampal microglial activation in mice. Brain Research, 1680, 13–38. 10.1016/j.brainres.2017.12.004 [DOI] [PubMed] [Google Scholar]
- Ye, S.‐M. , & Johnson, R. W. (1999). Increased interleukin‐6 expression by microglia from brain of aged mice. Journal of Neuroimmunology, 93(1–2), 139–148. 10.1016/S0165-5728(98)00217-3 [DOI] [PubMed] [Google Scholar]
- Yousef, H. , Czupalla, C. J. , Lee, D. , Chen, M. B. , Burke, A. N. , Zera, K. A. , Zandstra, J. , Berber, E. , Lehallier, B. , Mathur, V. , Nair, R. V. , Bonanno, L. N. , Yang, A. C. , Peterson, T. , Hadeiba, H. , Merkel, T. , Körbelin, J. , Schwaninger, M. , Buckwalter, M. S. , … Wyss‐Coray, T. (2019). Aged blood impairs hippocampal neural precursor activity and activates microglia via brain endothelial cell VCAM1. Nature Medicine, 25(6), 988–1000. 10.1038/s41591-019-0440-4 [DOI] [Google Scholar]
- Zhan, X. , Stamova, B. , & Sharp, F. R. (2018). Lipopolysaccharide associates with amyloid plaques, neurons and oligodendrocytes in alzheimer’s disease brain: A review. Frontiers in Aging Neuroscience, 10(42), 10.3389/fnagi.2018.00042 [DOI] [Google Scholar]
- Zhang, H. , Park, Y. , Wu, J. , Chen, X. , Lee, S. , Yang, J. , Dellsperger, K. C. , & Zhang, C. (2009). Role of TNF‐α in vascular dysfunction. Clinical Science, 116(3), 219–230. 10.1042/CS20080196 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang, H. , Sparks, J. B. , Karyala, S. V. , Settlage, R. , & Luo, X. M. (2015). Host adaptive immunity alters gut microbiota. ISME Journal, 9(3), 770–781. 10.1038/ismej.2014.165 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang, J. , Ma, L. , Chang, L. , Pu, Y. , Qu, Y. , & Hashimoto, K. (2020). A key role of the subdiaphragmatic vagus nerve in the depression‐like phenotype and abnormal composition of gut microbiota in mice after lipopolysaccharide administration. Translational Psychiatry, 10(1), 1–13. 10.1038/s41398-020-00878-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang, Q. , Lu, Y. , Bian, H. , Guo, L. , & Zhu, H. (2017). Activation of the α7 nicotinic receptor promotes lipopolysaccharide‐induced conversion of M1 microglia to M2. American Journal of Translational Research, 9(3), 971. [PMC free article] [PubMed] [Google Scholar]
- Zhang, Z. , Xu, D. , Fang, J. , Wang, D. , Zeng, J. , Liu, X. , & Zhao, X. (2021). In situ live imaging of gut microbiota. mSphere, 6(5), e0054521. 10.1128/mSphere.00545-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao, Y.‐J. , Yu, T.‐T. , Zhang, C. , Li, Z. , Luo, Q.‐M. , Xu, T.‐H. , & Zhu, D. (2018). Skull optical clearing window for in vivo imaging of the mouse cortex at synaptic resolution. Light: Science & Applications, 7(2), 17153. 10.1038/lsa.2017.153 [DOI] [Google Scholar]