3,765
Views
10
CrossRef citations to date
0
Altmetric
Review

Interlink between the gut microbiota and inflammation in the context of oxidative stress in Alzheimer’s disease progression

& ORCID Icon
Article: 2206504 | Received 20 Oct 2022, Accepted 18 Apr 2023, Published online: 01 May 2023

ABSTRACT

The microbiota-gut-brain axis is an important pathway of communication and may dynamically contribute to Alzheimer’s disease (AD) pathogenesis. Pathological commensal gut microbiota alterations, termed as dysbiosis, can influence intestinal permeability and break the blood–brain barrier which may trigger AD pathogenesis via redox signaling, neuronal, immune, and metabolic pathways. Dysbiosis increases the oxidative stress. Oxidants affect the innate immune system through recognizing microbial-derived pathogens by Toll-like receptors and initiating the inflammatory process. Most of the gut microbiome research work highlights the relationship between the gut microbiota and AD, but the contributory connection between precise bacteria and brain dysfunction in AD pathology cannot be fully demonstrated. Here, we summarize the current information of the fundamental connections between oxidative stress, inflammation, and gut dysbiosis in AD. This review emphasizes on the involvement of gut microbiota in the regulation of oxidative stress, inflammation, immune responses including central and peripheral cross-talk. It provides insights for novel preventative and therapeutic approaches in AD.

Introduction

Alzheimer’s disease (AD) is the most common cause of neurodegenerative disorders such as dementia. Cerebral extracellular amyloid β (Aβ) aggregation and intracellular neurofibrillary tangles (NFTs) formation are the primary histological hallmarks of AD. Citation1 Now a days, AD is the foremost global health affliction worldwide. The global prevalence of dementia rises exponentially with age. At 60–64 years it is around 0.7–1.8%, and over 90 years it goes up to 63.9% worldwide.Citation2 In 2021, about 6.2 million Americans were affected by AD; this number is projected to rise to nearly 13.8 million by 2060. Although the death rate by stroke, heart disease, and HIV were reduced between 2000 and 2019; unfortunately, AD death rate was increased by around 145%. During COVID-19 pandemic, AD death rate was increased by 6% in the USA.Citation3 In 2021, the total cost of healthcare-related dementia was estimated at around $355 billion in the USA.Citation3 Though the precise biological changes of AD, the different rates of progression among affected individuals and how AD can be prevented, slowed down, or stopped are still largely mysterious. Therefore, studying the mechanism of AD pathogenesis and finding new treatment strategies for the prevention and cure of AD is one of the most important challenges to be tackled in AD research.

Numerous evidences support that oxidative stress is one of the important cause of cell damage in ADCitation4,Citation5. The abundance of oxidative products which alter the major histopathology are increased in the AD brain.Citation6 Oxidative stress is mostly generated by reactive oxygen species (ROS) and reactive nitrogen species (RNS). Extreme accumulation of ROS induces neuronal damage.Citation4–6 Clearing or inhibiting the surplus ROS/RNS from the brain may be a fruitful treatment of AD. Oxidative stress will be discussed more in detail in the sections below.

Recent evidence suggested that age-related attenuation of gut microbiota biodiversity is considered as an important reason of AD pathogenesis.Citation7,Citation8 Gut microbiota can regulate the multiple neurochemical pathways through the “microbiota-gut-brain axis”. “microbiota-gut-brain axis” states to a bidirectional network communication between the central nervous system (CNS) and the gastrointestinal (GI) tract connecting various overlying pathways such as the autonomic, neuroendocrine, and immune systems including bacterial metabolites and neuromodulator molecules directly affecting the brain function (). Although the microbiota-gut-brain axis helps the appropriate function of the digestive tract, it also controls the biochemical signals between the sympathetic nervous system, endocrine glands, and specific brain regions such as the hypothalamus and the frontal cortex.Citation11

Figure 1. (a) Schematic representation of bidirectional communication between gut and brain through “microbiota-gut-brain axis”. The communications are mainly carried out by neural, endocrine, and immunological pathways. (b) in the lumen gut microbiota, microbial-derived metabolites such as short-chain fatty acids (SCFAs), neurotransmitters, amino acids, and bacterial amyloid interacts with the host immune system. These interactions affect the host metabolism and may activate the vagus nerve. Therefore these interactions are key to maintaining the overall health of the host. During dysbiosis, unfavorable conditions may cause the activation of corticotropin receptor, subsequently triggering adrenocorticotrophic hormone release and finally influencing cortisol release which leads to the loss of intestinal barrier integrity.Citation9,Citation10 As a result, intestinal and blood-brain barrier permeability is increased. Due to increased permeability, there is an increase in reactive oxygen species (ROS) in the neurons and microglia which may cause oxidative stress in neurodegenerative diseases such as Alzheimer’s disease.

Figure 1. (a) Schematic representation of bidirectional communication between gut and brain through “microbiota-gut-brain axis”. The communications are mainly carried out by neural, endocrine, and immunological pathways. (b) in the lumen gut microbiota, microbial-derived metabolites such as short-chain fatty acids (SCFAs), neurotransmitters, amino acids, and bacterial amyloid interacts with the host immune system. These interactions affect the host metabolism and may activate the vagus nerve. Therefore these interactions are key to maintaining the overall health of the host. During dysbiosis, unfavorable conditions may cause the activation of corticotropin receptor, subsequently triggering adrenocorticotrophic hormone release and finally influencing cortisol release which leads to the loss of intestinal barrier integrity.Citation9,Citation10 As a result, intestinal and blood-brain barrier permeability is increased. Due to increased permeability, there is an increase in reactive oxygen species (ROS) in the neurons and microglia which may cause oxidative stress in neurodegenerative diseases such as Alzheimer’s disease.

Gut microbiota communicates to the brain through four significant routes; first, vagus nerve activation which joins the muscular and mucosal layer of the GI tract to the brain stem; secondly, secretion of serotonin from enterochromaffin cells (EC) which are present in the gut epithelial lining; thirdly, dysfunction of microglia; and last direct transfer of the chemical signals (toxins, short-chain fatty acids, γ-aminobutyric acid, etc.) to the brain.Citation12–14 These four routes may work in combination to progress Aβ signaling cascade from the gut to the brain.

GI tract encloses trillions of commensal microorganisms and~1000 of its species which regulate a variety of metabolic functions and preserve membrane barrier functions mainly in the gut.Citation9 Within two years after birth, the community of microbes stabilizes the host GI tract, but depending upon peripheral factors such as age, diet, health, genetics, lifestyle, and environment differ their composition among individuals.Citation10 Furthermore, neurotransmitter γ-aminobutyric acid (GABA) can be produced by some beneficial gut microbiota like Bacteroides, Bifidobacterium, Parabacteroides, and Escherichia spp. Therefore, gut microbiota controls the level of neurotransmitters in the host organism.Citation15 With increasing age, pathological changes in the gut microbiota composition, termed as dysbiosis leads to inflammation, disrupts the blood–brain barrier (BBB), activates the immune system, and produces ROS and this is also seen in AD circumstancesCitation16(). Increasing abundance of Escherichia, Shigella spp., Bacteriodetes, and decreasing population of Bifidobacterium spp. with disturbed bacteriodetes versus firmicutes ratio might quicken inflammation and Aβ aggregation in AD.Citation17,Citation18 Therefore, a healthy well-balanced gut microbiome may reduce or prevent the detrimental effects of oxidative stress in AD. Here, we summarize the current research work on the association between oxidative stress and the gut microbiota in AD. In a nutshell, this review will provide evidence of the interlink between the gut microbiota and inflammation in the context of oxidative stress and the role of gut-microbial metabolites in AD.

Oxidative stress in AD

Oxidative stress is known as the imbalance between the ROS/RNS and antioxidant levels in cells. It disrupts the redox signaling pathway and contributes to microglial dysfunction in ADCitation19. ROS are mainly formed as the secondary product of the leaky electron transport chain (ETC) (complex I and III) in mitochondria.Citation4 In addition, monoamine oxidases (MAO) (present in the outer mitochondrial membrane),Citation20 an isoform of nitric oxide synthase (NOS) (present in neurons),Citation21 NADPH oxidases (NOX) (present in the plasma membrane, and phagosomes of polymorphonuclear neutrophils, abundance in cortex and hippocampus regions of the brain),Citation22 non-heme iron enzymes such as lipoxygenases (present in the cytoplasm),Citation23 xanthine oxidase, cytochrome P450 monooxygenase, cyclooxygenase and D-amino oxidase (present in the cytoplasm)Citation24 are important ROS producers. Peroxisomes also have a contribution to ROS formation.Citation25 Oxidative stress is augmented by the formation of superoxide (O2.−) by one electron from the molecular oxygen (O2), hydrogen peroxide (H2O2), peroxynitrite (ONOO−), and hydroxyl radicals (.OH) which are produced by the Fenton and Haber Weiss reaction in AD. The multi-valence of transition metals such as iron, copper, aluminum, and zinc help free radical formation causing oxidative stress. Lipid peroxidation, protein oxidation, nucleic acid damage, and advanced glycation end-products (AGEs) formation are the main four reactions which cause cellular damage in oxidative stress.Citation4 Oxidative stress biomarkers such as malondialdehyde, 4-hydroxynonenal, and F2-isoprostane (lipid oxidative damage); protein carbonyls and 3 nitrotyrosine (products of protein oxidation), 8-hydroxydeoxyguanosine (nucleic acid oxidation) are observed at high concentration in the blood and the cerebrospinal fluid (CSF) of AD patients.Citation26

In swift, dysfunction in cellular organelles like mitochondriaCitation27, endoplasmic reticulum due to unfolded protein response (UPR)Citation28, augmentation of metal ions in neuritic plaquesCitation29 and hyperactivation of microglia followed by the upregulation of NADPH oxidaseCitation30 are characterized by ROS/RNS productionCitation31 in AD. Oxidative stress can accelerate the aggregation of Aβ and vice versa.Citation4,Citation26 Additionally, hyperphosphorylated tau proteins may lead to reduced activity of NADH-ubiquinone reductase enzyme, increasing ROS production and mitochondrial dysfunction in AD.Citation32 Therefore, the relationship between oxidative stress and AD () is well documented in previous studies which indicate the importance of the antioxidant defense system in the brain.

Figure 2. Schematic representation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) formation in AD brain. Mitochondria is the main ROS producer. ROS are produced through electron leak from aerobic respiration in mitochondria. Superoxide (O2.−) is converted to hydrogen peroxide (H2O2) by superoxide dismutases (SOD). O2.− is also formed by the oxidation of NADPH by NADH oxidase enzymes (NOXs). Oxygen is produced by the reaction with H2O2 and catalase. In addition, H2O2 can produced hydroxyl radical (OH.) by Fenton and Haber–Weiss reaction in the presence of metal ions. On the other hand, reactive nitrogen species such as NO., CO3. are produced by the reaction of peroxide ions (ONOO.) and (O2.−). Exogenous source such as toxins or radiation may cause ROS/RNS production. ROS production may accelerate amyloid beta (Aβ) aggregation and tau hyperphosphorylation in Alzheimer’s disease., Streptococcus and Bacilli increases RNS production; whereas Salmonella and E. coli inhibit ATP production which accelerates oxidative stress, inflammation and thus may significantly influence amyloid-beta aggregation in AD.

Figure 2. Schematic representation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) formation in AD brain. Mitochondria is the main ROS producer. ROS are produced through electron leak from aerobic respiration in mitochondria. Superoxide (O2.−) is converted to hydrogen peroxide (H2O2) by superoxide dismutases (SOD). O2.− is also formed by the oxidation of NADPH by NADH oxidase enzymes (NOXs). Oxygen is produced by the reaction with H2O2 and catalase. In addition, H2O2 can produced hydroxyl radical (OH.) by Fenton and Haber–Weiss reaction in the presence of metal ions. On the other hand, reactive nitrogen species such as NO., CO3. are produced by the reaction of peroxide ions (ONOO.) and (O2.−). Exogenous source such as toxins or radiation may cause ROS/RNS production. ROS production may accelerate amyloid beta (Aβ) aggregation and tau hyperphosphorylation in Alzheimer’s disease., Streptococcus and Bacilli increases RNS production; whereas Salmonella and E. coli inhibit ATP production which accelerates oxidative stress, inflammation and thus may significantly influence amyloid-beta aggregation in AD.

Oxidative stress: the role of gut microbiota

By the modulation of mitochondrial activity, commensal and pathogenic bacteria can change the cellular oxidative stress in the gutCitation33. Formylated peptides are produced by commensal bacteria and it binds with G protein-coupled receptors (GPCRs) on macrophages and neutrophils and trigger inflammationCitation34,Citation35. As a result, superoxide is produced by NOX-1 which increases cellular ROS.Citation36 In addition, nitrate and nitrites compounds can be converted to nitric oxide (NO) by the gut Lactobacilli and Bifidobacterium, creating a high abundance of NO in the gut epithelia. NO is also produced from L-arginine using NOS by Streptococcus and Bacilli. Citation37 Although the nanomolar concentration of NO is considered as a neuroprotective and neurotransmitter of noradrenergic, noncholinergic enteric neurons; the higher concentration of NO produces ROS/RNS which further forms hydroxyl radicals, resulting in detrimental effects to neuroinflammation, axonal degeneration, and neurodegenerative disorders.Citation38 Furthermore, Salmonella typhimurium, E. coli, Mycobacterium, and Streptococcus anginosus can produce hydrogen sulfide from sulfur-containing amino acids (e.g. Cysteine) in the GI tract by sulfur metabolism. Due to the high concentration of hydrogen sulfide, cyclooxygenase activity is inhibited which alters the metabolism toward glycolysis, resulting in the reduction of mitochondrial oxygen consumption, ATP production, and overexpression of pro‐inflammatory effects ().Citation39,Citation40 Additionally, dietary trimethylamine N‐oxide (TMAO) is accelerated oxidative stress by reducing superoxide dismutase levels, increasing malondialdehyde and glutathione peroxidase, and exacerbating inflammation by the production of proinflammatory cytokines such as IL-6, IL-10, IL-1β, TNF-∞ in plasma and liver of male Apolipoprotein E knock-out (ApoE−/−) mice.Citation41

Oxidative stress and inflammation in AD

Oxidative stress and inflammation are intimately connected in the pathophysiological conditions where redox homeostasis is interrupted.Citation42 Growing evidences indicate that inflammation/neuroinflammation is a significant contributor to AD development and exacerbation.Citation42,Citation43 Inflammation is mainly arbitrated by microglial and astroglial states in the brain. Microglia have an important role in the brain development and function. Deposition of Aβ increases neuronal injury and inflammation by triggering microglia activation in AD. Activated microglia release some pro-inflammatory cytokines such as IL-6, TNF-∞ and IL-1β which regulates the inflammation.Citation44 Interestingly, IL-1β regulates the amyloid precursor protein, parent protein of Aβ.Citation45 The CSF- IL-1β is higher in AD than in age-matched control patients.Citation46 By the activation of p38 mitogen-activated protein kinase and glycogen synthase kinase-3β pathway, highly concentrated IL-1β accelerates tau hyperphosphorylation and NFT formation in triple transgenic AD mouse model.Citation47

Additionally, microglia have a significant role in Aβ clearance and degradation.Citation48 During microglial phagocytosis over time, the efficiency of microglia gradually declines Aβ clearance, which reduces Aβ proteolysisCitation49 and increases Aβ deposition and aggregationCitation50. This incidence creates an overproduction of pro-inflammatory cytokines, which trigger more ROS production and AD onsetCitation19. Therefore, microglia is one of the important non-neuronal cells for plaque formation and oxidative stress initiation in the AD brain.

Microglial degeneration can be occurred by stimulating receptor expressed on myeloid cells 2 (TREM2), C×3C motif chemokine receptor 1 (CX3CR1), GABA, and other inflammatory cytokine mediators. The expression of TREM2 on the microglial cell surface increases the phagocytic activity and causes ROS production.Citation51 During the phagocytosis activity microglia produce ROS like O2.. Active microglia also generate O2−. via NOX-2 pathway. H2O2 and NO. also impose local inflammation by attracting microglia which are involved in oxidative stressCitation19 that may drive AD progression.

Accumulating evidences showed that the astrocytes are intensively involved in maintaining oxidative stress at physiological or pathological conditions.Citation52,Citation53 Astrocytes are playing a dual role in maintaining homeostasis of ROS/RNS regulation.Citation52 Under physiological conditions, astrocyte acts as a neuroprotector of CNS from oxidative injury. In this condition various antioxidants are secreted, endogenic antioxidative systems like nuclear factor E2-related factor 2 (Nrf2) is stimulated, the excitatory amino acids are removed, neurotransmitters are uptaken and metabolized, energy and neurotrophin are produced and finally ROS/RNS are degraded.Citation52,Citation54 Whereas, under pathophysiological conditions, astrocytes trigger by stimulation from microglial activation and neuronal degradation. As a result, excessive ROS/RNS are produced from impaired mitochondria and antioxidant production is reduced; and elevated pro-inflammatory cytokines which leads to a detrimental effect seen in AD. Aβ and pro-inflammatory cytokines can trigger the astrocytes, causing instigation of NF-kB pathway, subsequently more pro-inflammatory cytokines and chemokines are produced .Citation55 Astrocytes are the vital cells that control glutamate homeostasis which circuitously maintains oxidative stress.Citation56 In the pathological circumstance, excessive glutamate is secreted from the pre-synaptic membrane and collected in the synaptic cleft which may allow a large influx of Ca2+ by over-activating N-methyl-d-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. It starts with calcium overload and ROS production in mitochondria; and causes neurotoxicity.Citation57

Oligodendrocyte, the myelin-forming cells (myelinate neuronal axons) contribute to increased oxidative stress in the CNS. Oligodendrocytes have lower glutathione content and it increases the iron content in the brain, which makes them further susceptible to oxidative stress and damage.Citation58 Oligodendrocytes are also exposed to Aβ and lead to oxidative stress, resulting in demyelination.Citation59 Demyleination causes a reduction in neuronal action potential time with increased inflammation and oxidative stress, thereby contributing to cognitive impairment in AD.

Oxidative stress and inflammation: role of the gut microbiota

Gut microbiota plays numerous roles in the improvement of immune response, protection against pathogen colonizationCitation60 at the intestinal epithelial barrierCitation61, association with production/regulation of oxidative stress and inflammationCitation62 in the host. PreclinicalCitation63 and clinical studiesCitation64,Citation65 show that modifications in gut microbiota is connected with AD progression. Gram-negative bacteria such as E. coli and Shigella colonization leads to surge in the formation of bacterial amyloids and lipopolysaccharides (LPS) which causes peripheral systemic inflammation, resulting in dysfunctional GI permeability and impaired BBB function.Citation66,Citation67 Dysbiosis causes increase in pathogenic bacteria such as Escherichia, Shigella, Pseudomonas, Proteobacteria, and Verrucomicrobia and reduces beneficial bacteria e.g. Bifidobacterium, Bacteroides fragilis, Bacillus fragilis, Eubacterium hallii, Eubacterium rectale, and Faecalibacterium prausnitzii.Citation18 Gut microbial metabolites like indole-3-pyruvic acid and short-chain fatty acids (SCFAs) were gradually decreased from mild cognitive impairment (MCI) to AD patients.Citation68 The abundance of SCFA-producing bacteria such as Clostridia, Clostridiales Ruminococcaceae, Firmicutes, and Ruminococcus declined which indicates the AD progression by the host-microbe cross-talk signals.Citation68 Other studies revealed that Blautia, Desulfovibrio, EscherichiaShigella, and Akkermansia are markedly altered in the APP/PSI transgenic mice.Citation69,Citation70 High abundance of Enterotype I and III bacteria are connected with the incidence of dementia.Citation71 The level of Bifidobacterium, Blautia, Lactobacillus, and Sphingomonas were more than Anaerobacterium, Papillibacter, and Odoribacter in AD patients.Citation72 Additionally, the affluence of Firmicutes, Proteobacteria, Tenericutes Enterobacteriaceae, Coriobacteriaceae, Mogibacteriaceae, Phascolarcto-bacterium, and Coprococcus were higher in MCI patients than age-matched controls.Citation73 The abundances of Firmicutes, Actinobacteria, E. rectale, B. fragilis and Fusobacteriaceae were decreased whereas, the levels of Bacteroides, Tenericutes, E. coli, B. subtilis, Escherichia, shigella, Lactobacilli, Bifidobacteria, Prevotellaceae, Verrucomicrobia were increased in the feces when compared with respective control groups in both AD animal models and AD patientsCitation74. The higher abundance of fungi like, Phaffomyceteceae, Sclerotiniaceae, Cystofilobasidiace-ae, Togniniaceae, Trichocomaceae, Botrytis, Cladosporium, Kazachstania, Phaeoacremonium and lower affluence of Meyerozyma were found in MCI patients.Citation75 For example, dietary amines can be metabolized to trimethylamine by gut microbiota whereas liver upon nutrient absorption converts trimethylamine to TMAO. Trimethylamine availability can be influenced by the gut dysbiosis. Increased levels of TMAO can promote the upregulation of inflammatory cytokines that raise oxidative stress in AD.Citation76 TMAO were found to be elevated in the cerebrospinal fluid of AD and MCI patients.Citation77

Therefore, beneficially modifying the gut microbiota content can show an optimistic role in the declining ROS through SCFA such as butyrate production, whereas dysbiosis may accelerate the systematic inflammation, activation of microglia, and BBB damage by elevated TMAO and further promotes AD progression.Citation78

Interlink between oxidative stress, inflammation, and TLRs in AD

Toll-like receptors (TLRs) are type 1 transmembrane pattern recognition receptors that are composed of an extracytoplasmic leucine-rich repeat (LRR) domain, a single membrane-spanning helix, and a signaling Toll-interleukin-1 receptor (TIR) domain. Based on spatial distribution of TLRs, it is mainly divided into two groups. One group (TLR1, TLR2, TLR4, TLR5, TLR 6, and TLR11) are found on the plasma membranes and they can be activated by microbial products e.g. lipids, lipoproteins, and proteins. Other group (TLR3, TLR7, TLR8, and TLR9) is located in cytoplasmic compartments. It can be triggered by nucleic acid species.Citation79

TLRs stimulate the downstream signaling transduction by identifying microbes-derived pathogens through the damage- and pathogen-associated molecular patterns (DAMPs and PAMPs) and start the inflammation. Firstly, TLRs change their confirmation based on PAMPs or DAMPs, and then dimerize to enhance the downstream signaling adaptors such as myeloid differentiation primary response protein 88 (MyD88), TIR domain-containing adaptor molecule (TIRAP), TIR domain-containing adaptor protein inducing interferon-b (TRIF) and TRIF-related adaptor molecule (TRAM) which stimulate the precise transcription factors and innate immune responses.Citation80 Except for TLR3, MyD88 is a common adaptor protein for all TLR-mediated signaling pathways.Citation81 Upon TLR involvement, the MyD88-dependent pathway can activate downstream signaling through the phosphorylation of IkB at ser 32 and 36 and it causes proteasomal degradation. As a result, free nuclear factor kappa B (NF-kb) translocates to the nucleus and activates targeted genes which produce proinflammatory cytokines such as TNF-α, IL-1, and IL-6 and oxidant enzymes such as NOX and iNOS which increases ROS levels. ROS accelerates NF-kB activation by enhancement of IKK phosphorylation by initiating protein kinase D (PKD) or obstructing protein phosphatase type 2A (PP2A).Citation82 In addition, TLRs also help ROS production within mitochondria and activate NOX by straight communication at the cell membrane or by increasing the phosphorylation of its p47phox subunit within the cytoplasm.Citation82 As a result, intracellular ROS level is increased and it helps to mobilize and dimerize TLRs and amplifies TLR responses (). Therefore, TLR-mediated inflammation plays a significant role in oxidative stress.

Figure 3. Schematic representation of TLR-mediated oxidative stress and inflammation in AD. TLRs help the formation of ROS through either direct interaction at the cell membrane or enhanced phosphorylation of its p47phox subunit within the cytoplasm. In the MyD88-dependent pathway, TLRs activate downstream signaling through the phosphorylation of IkB at Ser 32 and 36, and; it causes proteasomal degradation. Free nuclear factor kappa B (NF-Kb)) translocates to the nucleus and produces proinflammatory cytokines and oxidant enzymes which increases ROS levels. In addition, ROS accelerates the activation of NF-Kb by enhancement of IκB kinase (IKK) phosphorylation by activating PKD or inhibiting protein phosphatase type 2A (PP2A).

Figure 3. Schematic representation of TLR-mediated oxidative stress and inflammation in AD. TLRs help the formation of ROS through either direct interaction at the cell membrane or enhanced phosphorylation of its p47phox subunit within the cytoplasm. In the MyD88-dependent pathway, TLRs activate downstream signaling through the phosphorylation of IkB at Ser 32 and 36, and; it causes proteasomal degradation. Free nuclear factor kappa B (NF-Kb)) translocates to the nucleus and produces proinflammatory cytokines and oxidant enzymes which increases ROS levels. In addition, ROS accelerates the activation of NF-Kb by enhancement of IκB kinase (IKK) phosphorylation by activating PKD or inhibiting protein phosphatase type 2A (PP2A).

Although 11 human and 13 mouse TLRs are identified, recent evidence indicates TLR2, TLR4, and TLR9 are mainly involved in AD onset ().Citation80–82,Citation101 Although, the function of TLR2 in AD is controversial, some researchers believe that TLR2 can identify Aβ42 and trigger pro-inflammatory cytokines (e.g. TNF-α, IL-6, and IL-1) secretion.Citation83,Citation84 It was found that decreased levels of Aβ and higher toxicity of Aβ 1–42 can cause more cognitive decline in TLR2 knockout APP/PS1 mice.Citation85 It has been previously documented that uptake of Aβ42 by microglia is significantly increased by the activation of TLR2 expression.Citation86

Table 1. The effects of Toll-like receptors in Alzheimer’s disease.

Recently, we found that the integrity of the gut barrier was markedly decreased in Tg2576 AD mice which might increase bacterial amyloid (curli) burden in the gut before the appearance of Aβ deposition in the brain.Citation87 Curli stimulated TLR2 activation and co-localized with neuroendocrine marker PGP 9.5 within the epithelium and sub-mucosa of the gut in AD mice.Citation87 It indicated the vagus nerve activation by bacterial curli. Therefore, activation of TLR2 can stimulate AD pathogenesisCitation87,Citation88,Citation92,Citation102 It indicates that inhibition of TLR2 from periphery might be beneficial for AD. In-vivo and in-vitro studies also showed that phagocytosis and Aβ clearance in microglia were increased by the deficiency of TLR2Citation89. Secretion of pro-inflammatory cytokines and Aβ accumulation was reduced by the inhibition of TLR2 which improved the spatial learning performance in AD mouse models.Citation90,Citation91

TLR4 also plays a double role in AD pathogenesis. Jin et al. found that some cytokines such as IL-1β, IL-10, IL-17, and TNF-α were increased in TLR4 mutant Mo/Hu APPswe PS1dE9 transgenic mice (TLR4M AD mice), indicating TLR4 signaling may be involved in AD pathogenesis.Citation93 Song et al. also observed that TLR4 mutation decreased the microglial activation and accelerated Aβ deposition in AD mice, suggesting that microglial TLR4-mediated Aβ-induced neurotoxicity increases the clearance of Aβ deposition in the brain.Citation94 In addition, Qin et al. found that neuroinflammation could help neuronal autophagy, indicating mild TLR4 stimulation that attenuates AD-related tauopathy in tau-transgenic AD mice.Citation95 Go et al. found that TLR4 signaling in microglia was changed in the AD mouse model (TgAPP/PS1) and suggested the alteration of TLR4 signaling might help understand Aβ accumulation in the brain.Citation96

In addition, recent evidence indicated that activation of TLR9 signaling can defend neurons from stress;Citation97 Polymorphism of TLR9 may reduce the risk of ADCitation98 and TLR9 knockout mice showed impaired synaptic function.Citation99 Use of TLR9 agonists in AD mice cause the levels of Aβ aggregation and tau hyperphosphorylation declined which might improve cognitive deficits.Citation100 Therefore, immunomodulation through TLR9 may act as a probable therapeutic approach for AD but needs further investigation.

Oxidative stress and Nrf2-Keap 1 pathway in AD

Cellular central defense mechanism against oxidative stress is regulated by the nuclear factor E2-related factor 2 (Nrf2)-Kelch-like ECH-associated protein 1 (Keap1) signaling pathwayCitation103–105. Under homeostatic condition with low abundance of ROS, Keap1 binds with Nrf2 and then cullin-dependent E3 ubiquitin ligases the Nrf2 by proteasomal degradation. Under stimulation of elevated ROS levels, Nrf2 detaches from Keap1 and then Nrf2 transports into the nucleus, binds with antioxidant response elements (ARE) and finally promotes to increase downstream antioxidant enzyme genes (GCLC, GCLM, HO-1, and NQO1) which reduces the oxidative stress in ADCitation106. Numerous evidences reported that Nrf2 expression level are progressively reduced in the brain with the increasing of age. That leads to poorer clearance of resulting ROS immediately from the cytoplasm with ageCitation107–109. Consequently, oxidative damage and synaptic structural damage occurs in the neuronal cell, which is one of the primary reasons for AD. From current accumulating evidences, it is indicated that the activation of Nrf2-Keap 1 pathway ameliorates oxidative stress in AD ().

Figure 4. Schematic representation of the role of gut microbiota in Nrf2-keap 1 pathway and autophagy in AD. In homeostatic conditions, Nrf2 is degraded through proteasomal degradation. In oxidative stress conditions, Nrf2 dissociates from Keap1 and Nrf2 transport into the nucleus, binds with antioxidant response elements (ARE), and finally produces HO-1, NQo1, Catalase, and SOD. As a result, levels of anti-inflammatory cytokines are increased and pro-inflammatory cytokines levels are decreased. Lactobacilli can prompt their beneficial influence on host gut-epithelial tissue by the activation of lactobacilli-Nox-Nrf2 signaling. Lactobacilli and Bifidobacteria might help to decrease p62 levels and increase beclin-1 and LC3-II by activation of autophagic flux. Therefore, increased levels of Nrf2 and decreased levels of p62 can act as a neuroprotectant in AD.

Figure 4. Schematic representation of the role of gut microbiota in Nrf2-keap 1 pathway and autophagy in AD. In homeostatic conditions, Nrf2 is degraded through proteasomal degradation. In oxidative stress conditions, Nrf2 dissociates from Keap1 and Nrf2 transport into the nucleus, binds with antioxidant response elements (ARE), and finally produces HO-1, NQo1, Catalase, and SOD. As a result, levels of anti-inflammatory cytokines are increased and pro-inflammatory cytokines levels are decreased. Lactobacilli can prompt their beneficial influence on host gut-epithelial tissue by the activation of lactobacilli-Nox-Nrf2 signaling. Lactobacilli and Bifidobacteria might help to decrease p62 levels and increase beclin-1 and LC3-II by activation of autophagic flux. Therefore, increased levels of Nrf2 and decreased levels of p62 can act as a neuroprotectant in AD.

Oxidative stress and Nrf2-Keap 1 pathway: role of the gut microbiota

Numerous evidences suggest that cellular ROS is produced in the gut epithelial cells by the catalytic action of NADPH oxidases to the gut bacteria.Citation110,Citation111 Enzymatically ROS production by NADPH oxidase 1 (Nox1)Citation112 in epithelia is accelerated by pathogens, whereas symbiotic bacteria like Lactobacilli controls the intestinal epithelial cell proliferationCitation113, recompensation post-injuryCitation114, and modification of epithelial NF-kB signaling.Citation115 Jones et al. found that Nox1 is required to activate the Nrf2 pathway for the epithelial cytoprotection which is mediated by the gut bacteria Lactobacilli in the Drosophila and mice. Activated Nrf2 pathway boosts cytoprotective genes against environmental oxidative stress (). Therefore, Lactobacilli prompts their promising influence on host gut-epithelial tissue by the activation of lactobacilli dependent-Nox-Nrf2 signaling.Citation116 Therefore, Nrf2 signaling pathway initiates cytoprotection within the gut epithelial cells.Citation117

Crosstalk between autophagy and Nrf2 pathway in AD

Autophagy is a significant metabolic process which eliminates the misfolded proteins in the cells. Inception of autophagy and autophagosomes formation is other promising pathway for the treatment of AD.Citation107 Initiation of the Nrf2 pathway and autophagy are both helpful for the reduction of AD progression. There is a reciprocal relationship between autophagy and oxidative stress. After the elimination of Keap1 from Nrf2, keap1 binds with phosphorylated ubiquitin-binding protein p62 and finally triggers the autophagy by proteasome or lysosome pathway.Citation118 The degradation of p62 is controlled by autophagy at normal conditions, whereas oxidative stress activates p62 by the stimulation of p62-Keap1-Nrf2-ARE pathway.Citation119 Like Nrf2 signaling pathway, the released Nrf2 induces the expression of some autophagy genes such as genes of autophagy initiation (ULK1), substrate recognition (SQSTM1 and CALCOCO2), autophagosome forming (ATG4D, ATG7, and GABARAPL1), autophagosome extension (ATG2B and ATG5), lysosomal clearance (ATG4D), suggesting Nrf2 can stimulate the autophagy.Citation120,Citation121 In addition, p62 initiates tau protein degradation by selective autophagy,Citation122 and protect the neuronal homeostasisCitation123 . Hence, the p62-Keap1-Nrf2 positive feedback axis can act as a neuroprotective mechanismCitation124 and it connects the link between Nrf2 and autophagy pathways in AD pathogenesisCitation107 ().

Crosstalk between autophagy and Nrf2 pathway: role of the gut microbiota

Recent evidence has demonstrated that modulation of the gut microbiota induces the potential beneficial effects on neurochemical pathways which can slow down AD progression.Citation125,Citation126 Bonfili et al. observed that four months of treatment in 8-week-old male 3×-Tg AD mice, the probiotic mixture (SLAB51) containing nine live bacterial strains (Streptococcus thermophilus, Bifidobacteria (B. longum, B. breve, B. infantis), Lactobacilli (L. acidophilus, L. plantarum, L. paracasei, L. delbrueckii subsp. bulgaricus, L. brevis) have neuroprotective effect on the initial stage of AD. In the treatment with SLAB51 in AD mice, autophagic markers such as beclin-1 and LC3-II were increased and the level of p62 was decreased, indicating the autophagic flux activation. SLAB51 treatment ameliorated brain damage with improved cognition and reduced levels of Aβ through the fractional recovery of ubiquitin-proteasome system and autophagy pathways.Citation125 Te´gla´s et al. also found that using probiotic supplementation (with Bifidobacterium longum and Lactobacillus acidophilus) for twenty weeks, delayed the progression of AD in 3-month old, male APP/PS1 transgenic mice. They found that Nrf2 levels were elevated in the probiotic supplemented mice. Probiotics were found to stimulate the antioxidants such as superoxide dismutase (SOD)Citation126 and finally it promotes cellular defense mechanism against oxidative stress in AD. Through the microbiota-gut-brain axis Lactobacillus reuteri can promote the production and absorption of indole-3-aldehyde and indole-3-propionic acid in to the brainCitation127 that results in suppressed neuroinflammation and improved astrocyte activation. (). Overall, it is clear that intervention with probiotics has a promising effect on suppressing oxidative stress and improving cognition in AD.

Neuroprotection: role of the gut metabolites

The rising line of evidence expose that the gut microbiota and its metabolites such as polyphenols, SCFAs, antioxidants, vitamins etc. regulate many biosynthetic pathways which may have favorable or unfavorable effects on the host system ().Citation12 Gut microbiota can change neurotransmitter function of Brain-Derived Neurotrophic Factor (BDNF) by either kynurenine pathway or the action and availability of SCFAs in the brain.Citation144 Hence, potent gut microbiota is of utmost importance and can maintain neuronal health through antioxidative or anti-inflammatory pathways. The gut microbiome also regulates the penetrability of metabolites to BBB and improves intestinal barrier integrity; a hindrance to the intestinal gut microbiota colonization with pathogens, can reverse these protective effects.

Table 2. Common gut microbiota and their metabolites which act as neuroprotection in AD.

Recent studies revealed that Vit B and Vit K can improve neuronal health in the brain development and function.Citation139,Citation145 Deficiency in Vit B and Vit K correlated with decline in the memory functions of AD patients. Vit K2 (menaquinone-4) have antioxidative properties and markedly inhibits the rotenone-induced p38 activation, ROS production, and caspase-1 activity and finally reestablished mitochondrial membrane potential.Citation146 Vit K is produced by Escherichia coli, Klebsiella pneumoniae, Propionibacterium, and Eubacterium; Bacillus subtilis and E. coli produces B2 (riboflavin); Bifidobacterium, Lactococcus lactis, and Streptococcus thermophilus produces B9 (folic acid); Lactobacillus reuteri and Propionibacterium freudenreichii produces B12 (cobalamin).Citation139 There might be a great benefit in exploring these bacterial interaction with the host in the context of AD.

With the help of the gut bacteria, dietary amino acids (e.g. tyrosine, tryptophan, and phenylalanine) are metabolized into SCFAs, indole derivatives, neurotransmitters, organic acids, amines, and ammonia.Citation147 The products of tryptophan metabolism such as tryptamine, tryptophan derivatives act as neuroactive molecule.Citation148 Indole propanoic acid (an indole derivative) acts as antioxidant by beneficially influencing decline in neuroinflammation.Citation149 Metabolized products of arginine and agmatine, ameliorates ROS production and indicates the therapeutic effects in CNS disorders.Citation150 In-vitro and in-vivo studies also showed that agmatine defends astrocytes and microglia from the detrimental effect of oxidative stress.Citation151 Useful metabolites like SCFAs formed by intestinal bacteria support to decrease ROS by controlling the activity of mitochondria.Citation12 SCFAs decrease oxidative stress by declining microglial activation in the brain.Citation152 In addition, SCFAs prevent neurotoxic Aβ aggregation in AD by obstracting Aβ40/42 assembly.Citation153

The abundances of Bifidobacterium spp. decreases with increase in Proteobacteria spp. with growing age that might contribute to AD pathogenesis. Bifidobacterium have an important role in maintaining hippocampal plasticity and memory functions through the regulation of cholesterol levels by accumulating the serum leptin levels.Citation154,Citation155 Taken together, metabolites produced by the gut microbiota prevent and ameliorate oxidative stress associated with CNS depending upon the gut health of an individual.

Metabolic and neuroactive metabolites produced by gut microbiota have beneficial effects on the host health. Gut bacteria like Lactobacilli and Bifidobacterium can produce neurotransmitter such as γ-Aminobutyric acid (GABA) which can control glucose homeostasis and change the behavioral activity in the host.Citation156 Tiwari V. et al. showed that synaptic plasticity in the hippocampus was changed by the reduced level of GABA with augmentation of glutamatergic neurotransmission in the AβPPswe-PS1dE9 mice model of AD.Citation157 GABA-producing bacteria like Lactobacilli may recover the metabolic and depressive-like behavioral abnormalities in mice.Citation158 In addition, Cyanobacteria produces β-methylamino-L-alanine (BMAA), neurotoxins causing cognitive impairment.Citation159 Therefore, gut microbiota plays the main role in modulating ROS production in the CNS. The role of gut microbiota in modulating the host oxidative stress both peripherally and centrally looks very promising and needs further attention with more evidences to treat AD patients before symptoms occur in the brain.

Conclusion

There are enough evidences from our own research and other research works as highlighted in this review that the interrelationship of the gut microbiota and the brain has controlled to transformative advances in neuroscience research. Gut dysbiosis acts as an important player in modulating the microbiota-gut-brain axis that may contribute to increased inflammation and accelerate amyloid β aggregation by TLR associate signaling cascade upon AD onset. Gut microbiota can influence AD pathogenesis via increased neuroinflammation elevated oxidative stress, dysregulated neurotransmitters, reduced SCFAs, elevated TLRs and increased toxins. All these pathways are interlinked. In this review, we primarily explored the role of dysbiotic gut microbiota on triggering oxidative stress and inflammation. In addition, we have also focused on studies stating maintenance of eubiosis by inhibition of pro-inflammatory cytokines, increasing anti-inflammatory cytokines, preventing oxidative stress and inflammation, and improving bacterial metabolites with beneficial function to improve host health. To date, most of the gut microbiome research work mainly emphasize the connections between the gut microbiota and AD, although the contributory relationship between specific bacteria and the brain dysfunction in AD are not demonstrated functionally. Therefore, the specific function of the precise gut microbiota in AD patients persist as indefinable. If we could understand these interactions and molecular mechanisms in detail then the commensal gut bacteria can be utilized as targets for novel noninvasive diagnosis and future treatment strategies of AD. Furthermore, increasing the usage of germ-free, specific gene knockout and humanized sporadic AD animal models for the characterization of the gut microbiota and its cross-communication with the host in AD pathophysiology is needed. Targeting the potential beneficial bacteria (as described in this review) as a novel intervention strategy can prohibit or slow AD onset or counteract its development. In addition, new treatment strategies such as fecal microbiota transfer, probiotics (beneficial bacterial cocktails), beneficial metabolite supplementations to improve the brain and the gut health in pre-symptomatic AD patients may have translational value. Therefore, efficacy of the gut microbiota and its effective metabolites in neurodegenerative and neuroprotective mechanisms is continued to be open for further investigation. Therefore, it may provide new suggestions in the pathology and treatment to AD.

Author Contributions

T.K.D. and B.P.G. constructed the design and outlined the content. T.K.D. reviewed and analyzed the literatures, drafted the manuscript, and prepared the figures; B.P.G. obtained the funding, editing and supervised the writing of the review. All authors critically revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Acknowledgments

All diagrams are generated using BioRender.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

This was supported by the National Institutes of Health (NIA 5R01AG070934-03 to B.P.G)

References

  • DeTure MA, Dickson DW. The neuropathological diagnosis of Alzheimer’s disease. Mol Neurodegener. 2019;14(1):32. doi:10.1186/s13024-019-0333-5.
  • Prince M, Bryce R, Albanese E, Wimo A, Ribeiro W, Ferri CP. The global prevalence of dementia: a systematic review and metaanalysis. Alzheimer’s & Dementia. 2013;9(1):63. doi:10.1016/j.jalz.2012.11.007.
  • Alzheimer’s disease facts and figures. Alzheimer’s & Dementia. 2021;17(3):327–19. doi:10.1002/alz.12328.
  • Kanti Das T, Wati MR, Fatima-Shad K. Oxidative stress gated by Fenton and Haber Weiss reactions and its association with Alzheimer’s disease. Arch Neurosci. 2014;2(3). doi:10.5812/archneurosci.20078.
  • Singh A, Kukreti R, Saso L, Kukreti S. Oxidative stress: a key modulator in neurodegenerative diseases. Molecules. 2019;24(8):1583. doi:10.3390/molecules24081583.
  • Christen Y. Oxidative stress and Alzheimer disease. Am J Clin Nutr. 2000;71(2):621S–629S. doi:10.1093/ajcn/71.2.621s.
  • Zheng D, Liwinski T, Elinav E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020;30(6):492–506. doi:10.1038/s41422-020-0332-7.
  • Santoro A, Zhao J, Wu L, Carru C, Biagi E, Franceschi C. Microbiomes other than the gut: inflammaging and age-related diseases. Semin Immunopathol. 2020;42(5):589–605. doi:10.1007/s00281-020-00814-z.
  • Morais LH, Schreiber HL, Mazmanian SK. The gut microbiota–brain axis in behaviour and brain disorders. Nat Rev Microbiol. 2021;19(4):241–255. doi:10.1038/s41579-020-00460-0.
  • Cho I, Blaser MJ. The human microbiome: at the interface of health and disease. Nat Rev Genet. 2012;13(4):260–270. doi:10.1038/nrg3182.
  • Bonfili L, Cecarini V, Gogoi O, Gong C, Cuccioloni M, Angeletti M, Rossi G, Eleuteri AM. Microbiota modulation as preventative and therapeutic approach in Alzheimer’s disease. FEBS J. 2021;288(9):2836–2855. doi:10.1111/febs.15571.
  • Shandilya S, Kumar S, Kumar Jha N, Kumar Kesari K, Ruokolainen J. Interplay of gut microbiota and oxidative stress: perspective on neurodegeneration and neuroprotection. J Adv Res. 2022;38:223–244. doi:10.1016/j.jare.2021.09.005.
  • Ganesh BP, Versalovic J. Luminal conversion and immunoregulation by probiotics. Front Pharmacol. 2015;6. doi:10.3389/fphar.2015.00269.
  • Ganesh BP, Fultz R, Ayyaswamy S, Versalovic J. Microbial interactions with the intestinal epithelium and beyond: focusing on immune cell maturation and homeostasis. Curr Pathobiol Rep. 2018;6(1):47–54. doi:10.1007/s40139-018-0165-y.
  • Strandwitz P, Kim KH, Terekhova D, Liu JK, Sharma A, Levering J, McDonald D, Dietrich D, Ramadhar TR, Lekbua A, et al. GABA-modulating bacteria of the human gut microbiota. Nat Microbiol. 2018;4(3):396–403. doi:10.1038/s41564-018-0307-3.
  • Liu S, Gao J, Zhu M, Liu K, Zhang HL. Gut microbiota and dysbiosis in Alzheimer’s disease: implications for pathogenesis and treatment. Mol Neurobiol. 2020;57(12):5026–5043. doi:10.1007/s12035-020-02073-3.
  • Zhang Y, Geng R, Tu Q. Gut microbial involvement in Alzheimer’s disease pathogenesis. Aging. 2021;13(9):13359–13371. doi:10.18632/aging.202994.
  • Cattaneo A, Cattane N, Galluzzi S, Provasi S, Lopizzo N, Festari C, Ferrari C, Guerra UP, Paghera B, Muscio C, et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol Aging. 2017;49:60–68. doi:10.1016/j.neurobiolaging.2016.08.019.
  • Simpson DSA, Oliver PL. ROS generation in microglia: understanding oxidative stress and inflammation in neurodegenerative disease. Antioxidants. 2020;9(8):743. doi:10.3390/antiox9080743.
  • Nagatsu T. Progress in Monoamine Oxidase (MAO) research in relation to genetic engineering. Neurotoxicology. 2004;25(1–2):11–20. doi:10.1016/S0161-813X(03)00085-8.
  • Forstermann U, Sessa WC. Nitric oxide synthases: regulation and function. Eur Heart J. 2012;33(7):829–837. doi:10.1093/eurheartj/ehr304.
  • Tejada-Simon MV, Serrano F, Villasana LE, Kanterewicz BI, Wu G-Y, Quinn MT, Klann E. Synaptic localization of a functional NADPH oxidase in the mouse hippocampus. Mol Cell Neurosci. 2005;29(1):97–106. doi:10.1016/j.mcn.2005.01.007.
  • Nanda B, Nataraju A, Rajesh R, Rangappa K, Shekar M, Vishwanath B. PLA2 mediated arachidonate free radicals: pLA2 inhibition and neutralization of free radicals by anti-oxidants – a new role as anti-inflammatory molecule. Curr Top Med Chem. 2007;7(8):765–777. doi:10.2174/156802607780487623.
  • Angelova PR, Abramov AY. Role of mitochondrial ROS in the brain: from physiology to neurodegeneration. FEBS Lett. 2018;592(5):692–702. doi:10.1002/1873-3468.12964.
  • Sun Y, Lu Y, Saredy J, Wang X, Drummer IV C, Shao Y, Saaoud F, Xu K, Liu M, Yang WY, et al. ROS systems are a new integrated network for sensing homeostasis and alarming stresses in organelle metabolic processes. Redox Biol. 2020;37:101696. doi:10.1016/j.redox.2020.101696.
  • Michalska P, León R. When it comes to an end: oxidative stress crosstalk with protein aggregation and neuroinflammation induce neurodegeneration. Antioxidants. 2020;9(8):740. doi:10.3390/antiox9080740.
  • Wang W, Zhao F, Ma X, Perry G, Zhu X. Mitochondria dysfunction in the pathogenesis of Alzheimer’s disease: recent advances. Mol Neurodegener. 2020;15(1):30. doi:10.1186/s13024-020-00376-6.
  • Ajoolabady A, Lindholm D, Ren J, Pratico D. ER stress and UPR in Alzheimer’s disease: mechanisms, pathogenesis, treatments. Cell Death Dis. 2022;13(8):706. doi:10.1038/s41419-022-05153-5.
  • Liu F, Zhang Z, Zhang L, Meng R-N, Gao J, Jin M, Li M, Wang X-P. Effect of metal ions on Alzheimer’s disease. Brain Behav. 2022;12(3). doi:10.1002/brb3.2527.
  • Wilkinson BL, Landreth GE. The microglial NADPH oxidase complex as a source of oxidative stress in Alzheimer’s disease. J Neuroinflammation. 2006;3(1):30. doi:10.1186/1742-2094-3-30.
  • di Meo S, Reed TT, Venditti P, Victor VM. Role of ROS and RNS sources in physiological and pathological conditions. Oxid Med Cell Longev. 2016;2016:1–44. doi:10.1155/2016/1245049.
  • Abramov AY, Potapova EV, Dremin VV, Dunaev AV. Interaction of oxidative stress and misfolded proteins in the mechanism of neurodegeneration. Life. 2020;10(7):101. doi:10.3390/life10070101.
  • Saint-Georges-Chaumet Y, Edeas M, Carbonetti N. Microbiota–mitochondria inter-talk: consequence for microbiota–host interaction. Pathog Dis. 2016;74(1):ftv096. doi:10.1093/femspd/ftv096.
  • Wentworth CC, Jones RM, Kwon YM, Nusrat A, Neish AS. Commensal-epithelial signaling mediated via formyl peptide receptors. Am J Pathol. 2010;177(6):2782–2790. doi:10.2353/ajpath.2010.100529.
  • Dorward DA, Lucas CD, Chapman GB, Haslett C, Dhaliwal K, Rossi AG. The role of formylated peptides and formyl peptide receptor 1 in governing neutrophil function during acute inflammation. Am J Pathol. 2015;185(5):1172–1184. doi:10.1016/j.ajpath.2015.01.020.
  • MIGEOTTE I, COMMUNI D, PARMENTIER M. Formyl peptide receptors: a promiscuous subfamily of G protein-coupled receptors controlling immune responses. Cytokine Growth Factor Rev. 2006;17(6):501–519. doi:10.1016/j.cytogfr.2006.09.009.
  • Tiso M, Schechter AN, Jourd’heuil D. Nitrate reduction to nitrite, nitric oxide and ammonia by gut bacteria under physiological conditions. Plos One. 2015;10(3):e0119712. doi:10.1371/journal.pone.0119712.
  • Wang B, Yao M, Lv L, Ling Z, Li L. The human microbiota in health and disease. Engineering. 2017;3(1):71–82. doi:10.1016/J.ENG.2017.01.008.
  • Leschelle X, Goubern M, Andriamihaja M, Blottière HM, Couplan E, Gonzalez-Barroso MDM, Petit C, Pagniez A, Chaumontet C, Mignotte B, et al. Adaptative metabolic response of human colonic epithelial cells to the adverse effects of the luminal compound sulfide. Biochimica Et Biophysica Acta (BBA) - General Subjects. 2005;1725(2):201–212. doi:10.1016/j.bbagen.2005.06.002.
  • Beaumont M, Andriamihaja M, Lan A, Khodorova N, Audebert M, Blouin J-M, Grauso M, Lancha L, Benetti P-H, Benamouzig R, et al. Detrimental effects for colonocytes of an increased exposure to luminal hydrogen sulfide: the adaptive response. Free Radical Biol Med. 2016;93:155–164. doi:10.1016/j.freeradbiomed.2016.01.028.
  • He Z, Kwek E, Hao W, Zhu H, Liu J, Ma KY, Chen Z-Y. Hawthorn fruit extract reduced trimethylamine-N-oxide (TMAO)-exacerbated atherogenesis in mice via anti-inflammation and anti-oxidation. Nutr Metab. 2021;18(1):6. doi:10.1186/s12986-020-00535-y.
  • Bhatt S, Nagappa AN, Patil CR. Role of oxidative stress in depression. Drug Discov Today. 2020;25(7):1270–1276. doi:10.1016/j.drudis.2020.05.001.
  • Łuc M, Misiak B, Pawłowski M, Stańczykiewicz B, Zabłocka A, Szcześniak D, Pałęga A, Rymaszewska J. Gut microbiota in dementia critical review of novel findings and their potential application. Prog Neuro-Psychopharmacol Biol Psychiatry. 2021;104:110039. doi:10.1016/j.pnpbp.2020.110039.
  • Wang WY, Tan MS, Yu JT, Tan L. Role of pro-inflammatory cytokines released from microglia in Alzheimer’s disease. Ann Transl Med. 2015;3(10). doi:10.3978/j.issn.2305-5839.2015.03.49.
  • Anderson P. Differential effects of interleukin-1β and S100B on amyloid precursor protein in rat retinal neurons. Clin Ophthalmol. 2009;235:235. doi:10.2147/OPTH.S2684. Published online February.
  • Hesse R, Wahler A, Gummert P, Kirschmer S, Otto M, Tumani H, Lewerenz J, Schnack C, von Arnim CAF. Decreased IL-8 levels in CSF and serum of AD patients and negative correlation of MMSE and IL-1β. BMC Neurol. 2016;16(1):185. doi:10.1186/s12883-016-0707-z.
  • Ghosh S, Wu MD, Shaftel SS, Kyrkanides S, LaFerla FM, Olschowka JA, O’Banion MK. Sustained interleukin-1β overexpression exacerbates tau pathology despite reduced amyloid burden in an Alzheimer’s mouse model. J Neurosci. 2013;33(11):5053–5064. doi:10.1523/JNEUROSCI.4361-12.2013.
  • Ries M, Sastre M. Mechanisms of Aβ clearance and degradation by glial cells. Front Aging Neurosci. 2016;8:8. doi:10.3389/fnagi.2016.00160.
  • Lee CYD, Landreth GE. The role of microglia in amyloid clearance from the AD brain. J Neural Transm. 2010;117(8):949–960. doi:10.1007/s00702-010-0433-4.
  • Lai AY, McLaurin J. Clearance of amyloid-β peptides by microglia and macrophages: the issue of what, when and where. Future Neurol. 2012;7(2):165–176. doi:10.2217/fnl.12.6.
  • Liu W, Taso O, Wang R, Bayram S, Graham AC, Garcia-Reitboeck P, Mallach A, Andrews WD, Piers TM, Botia JA, et al. Trem2 promotes anti-inflammatory responses in microglia and is suppressed under pro-inflammatory conditions. Hum Mol Genet. 2020;29(19):3224–3248. doi:10.1093/hmg/ddaa209.
  • Chen Y, Qin C, Huang J, Tang X, Liu C, Huang K, Xu J, Guo G, Tong A, Zhou L. The role of astrocytes in oxidative stress of central nervous system: a mixed blessing. Cell Prolif. 2020;53(3). doi:10.1111/cpr.12781.
  • Lee KH, Cha M, Lee BH. Crosstalk between neuron and glial cells in oxidative injury and neuroprotection. Int J Mol Sci. 2021;22(24):13315. doi:10.3390/ijms222413315.
  • Bylicky MA, Mueller GP, Day RM. Mechanisms of endogenous neuroprotective effects of astrocytes in brain injury. Oxid Med Cell Longev. 2018;2018:1–16. doi:10.1155/2018/6501031.
  • Yang S, Magnutzki A, Alami NO. IKK2/NF-κB Activation in astrocytes reduces amyloid β deposition: a process associated with specific microglia polarization. Cells. 2021;10(10):2669. doi:10.3390/cells10102669.
  • Kim Y, Park J, Choi YK. The role of astrocytes in the central nervous system focused on BK channel and heme oxygenase metabolites: a review. Antioxidants. 2019;8(5):121. doi:10.3390/antiox8050121.
  • Belov Kirdajova D, Kriska J, Tureckova J, Anderova M. Ischemia-triggered glutamate excitotoxicity from the perspective of glial cells. Front Cell Neurosci. 2020;14:14. doi:10.3389/fncel.2020.00051.
  • Spaas J, van Veggel L, Schepers M, Tiane A, van Horssen J, Wilson DM, Moya PR, Piccart E, Hellings N, Eijnde BO, et al. Oxidative stress and impaired oligodendrocyte precursor cell differentiation in neurological disorders. Cell Mol Life Sci. 2021;78(10):4615–4637. doi:10.1007/s00018-021-03802-0.
  • Lloret A, Esteve D, Lloret MA, Monllor P, López B, León JL, Cervera-Ferri A. Is oxidative stress the link between cerebral small vessel disease, sleep disruption, and oligodendrocyte dysfunction in the onset of Alzheimer’s disease? Front Physiol. 2021;12:12. doi:10.3389/fphys.2021.708061.
  • Pickard JM, Zeng MY, Caruso R, Núñez G. Gut microbiota: role in pathogen colonization, immune responses, and inflammatory disease. Immunol Rev. 2017;279(1):70–89. doi:10.1111/imr.12567.
  • Ghosh S, Whitley CS, Haribabu B, Jala VR. Regulation of intestinal barrier function by microbial metabolites. Cell Mol Gastroenterol Hepatol. 2021;11(5):1463–1482. doi:10.1016/j.jcmgh.2021.02.007.
  • Dumitrescu L, Popescu-Olaru I, Cozma L, Tulbă D, Hinescu ME, Ceafalan LC, Gherghiceanu M, Popescu BO. Oxidative stress and the microbiota-gut-brain axis. Oxid Med Cell Longev. 2018;2018:1–12. doi:10.1155/2018/2406594.
  • Jung JH, Kim G, Byun MS, Lee JH, Yi D, Park H, Lee DY. Gut microbiome alterations in preclinical Alzheimer’s disease. Plos One. 2022;17(11):e0278276. doi:10.1371/journal.pone.0278276.
  • Zhuang ZQ, Shen LL, Li WW, Fu X, Zeng F, Gui L, Lü Y, Cai M, Zhu C, Tan Y-L, et al. Gut microbiota is altered in patients with Alzheimer’s disease. J Alzheimer’s Dis. 2018;63(4):1337–1346. doi:10.3233/JAD-180176.
  • Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Merluzzi AP, Johnson SC, Carlsson CM, Asthana S, Zetterberg H, Blennow K, et al. Gut microbiome alterations in Alzheimer’s disease. Sci Rep. 2017;7(1):13537. doi:10.1038/s41598-017-13601-y.
  • Loffredo L, Ettorre E, Zicari AM, Inghilleri M, Nocella C, Perri L, Spalice A, Fossati C, De Lucia MC, Pigozzi F, et al. Oxidative stress and gut-derived lipopolysaccharides in neurodegenerative disease: role of NOX2. Oxid Med Cell Longev. 2020;2020:1–7. doi:10.1155/2020/8630275.
  • Kesika P, Suganthy N, Sivamaruthi BS, Chaiyasut C. Role of gut-brain axis, gut microbial composition, and probiotic intervention in Alzheimer’s disease. Life Sci. 2021;264:118627. doi:10.1016/j.lfs.2020.118627.
  • Wu L, Han Y, Zheng Z, Peng G, Liu P, Yue S, Zhu S, Chen J, Lv H, Shao L, et al. Altered gut microbial metabolites in amnestic mild cognitive impairment and Alzheimer’s disease: signals in host–Microbe interplay. Nutrients. 2021;13(1):228. doi:10.3390/nu13010228.
  • Chen Y, Fang L, Chen S, Zhou H, Fan Y, Lin L, Li J, Xu J, Chen Y, Ma Y, et al. Gut microbiome alterations precede cerebral amyloidosis and microglial pathology in a mouse model of Alzheimer’s disease. Biomed Res Int. 2020;2020:1–15. doi:10.1155/2020/8456596.
  • Zhang A, Ma Z, Kong L. High‐throughput lipidomics analysis to discover lipid biomarkers and profiles as potential targets for evaluating efficacy of Kai‐Xin‐San against APP/PS1 transgenic mice based on UPLC–Q/TOF–MS. Biomed Chromatogr. 2020;34(2). doi:10.1002/bmc.4724
  • Saji N, Murotani K, Hisada T, Kunihiro T, Tsuduki T, Sugimoto T, Kimura A, Niida S, Toba K, Sakurai T. Relationship between dementia and gut microbiome-associated metabolites: a cross-sectional study in Japan. Sci Rep. 2020;10(1):8088. doi:10.1038/s41598-020-65196-6.
  • Zhou Y, Wang Y, Quan M, Zhao H, Jia J. Gut microbiota changes and their correlation with cognitive and neuropsychiatric symptoms in Alzheimer’s disease. J Alzheimer’s Dis. 2021;81(2):583–595. doi:10.3233/JAD-201497.
  • Nagpal R, Neth BJ, Wang S, Craft S, Yadav H. 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. 2019;47:529–542. doi:10.1016/j.ebiom.2019.08.032.
  • Megur A, Baltriukienė D, Bukelskienė V, Burokas A. The microbiota–gut–brain axis and Alzheimer’s disease: neuroinflammation is to blame? Nutrients. 2020;13(1):37. doi:10.3390/nu13010037.
  • Nagpal R, Neth BJ, Wang S, Mishra SP, Craft S, Yadav H. Gut microbiome and its interaction with diet, gut bacteria and Alzheimer’s disease markers in subjects with mild cognitive impairment: a pilot study. EBioMedicine. 2020;59:102950. doi:10.1016/j.ebiom.2020.102950.
  • Janeiro M, Ramírez M, Milagro F, Martínez J, Solas M. Implication of Trimethylamine N-Oxide (TMAO) in disease: potential biomarker or new therapeutic target. Nutrients. 2018;10(10):1398. doi:10.3390/nu10101398.
  • Vogt NM, Romano KA, Darst BF, Engelman CD, Johnson SC, Carlsson CM, Asthana S, Blennow K, Zetterberg H, Bendlin BB, et al. The gut microbiota-derived metabolite trimethylamine N-oxide is elevated in Alzheimer’s disease. Alzheimers Res Ther. 2018;10(1):124. doi:10.1186/s13195-018-0451-2.
  • Shabbir U, Tyagi A, Elahi F, Aloo SO, Oh DH. The potential role of polyphenols in oxidative stress and inflammation induced by gut microbiota in Alzheimer’s disease. Antioxidants. 2021;10(9):1370. doi:10.3390/antiox10091370.
  • Moresco EMY, LaVine D, Beutler B. Toll-like receptors. Curr Biol. 2011;21(13):R488–493. doi:10.1016/j.cub.2011.05.039.
  • Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol. 2004;4(7):499–511. doi:10.1038/nri1391.
  • Kawai T, Akira S. TLR signaling. Semin Immunol. 2007;19(1):24–32. doi:10.1016/j.smim.2006.12.004.
  • Lugrin J, Rosenblatt-Velin N, Parapanov R, Liaudet L. The role of oxidative stress during inflammatory processes. Biol Chem. 2014;395(2):203–230. doi:10.1515/hsz-2013-0241.
  • Jana M, Palencia CA, Pahan K. Fibrillar amyloid-β peptides activate microglia via TLR2: implications for Alzheimer’s disease. J Immunol. 2008;181(10):7254–7262. doi:10.4049/jimmunol.181.10.7254.
  • Lin W, Ding M, Xue J, Leng W. The role of TLR2/JNK/NF-κB pathway in amyloid β peptide-induced inflammatory response in mouse NG108-15 neural cells. Int Immunopharmacol. 2013;17(3):880–884. doi:10.1016/j.intimp.2013.09.016.
  • Richard KL, Filali M, Préfontaine P, Rivest S. Toll-like receptor 2 acts as a natural innate immune receptor to clear amyloid β1-42 and delay the cognitive decline in a mouse model of Alzheimer’s disease. J Neurosci. 2008;28(22):5784–5793. doi:10.1523/JNEUROSCI.1146-08.2008.
  • Chen K, Iribarren P, Hu J, Chen J, Gong W, Cho EH, Lockett S, Dunlop NM, Wang JM. Activation of toll-like receptor 2 on microglia promotes cell uptake of Alzheimer disease-associated amyloid β peptide. J Biol Chem. 2006;281(6):3651–3659. doi:10.1074/jbc.M508125200.
  • Das TK, Blasco-Conesa MP, Korf J, Honarpisheh P, Chapman MR, Ganesh BP. Bacterial amyloid curli associated gut epithelial neuroendocrine activation predominantly observed in Alzheimer’s disease mice with central amyloid-β pathology. J Alzheimer’s Dis. 2022;88(1):191–205. doi:10.3233/JAD-220106.
  • Letiembre M, Liu Y, Walter S, Hao W, Pfander T, Wrede A, Schulz-Schaeffer W, Fassbender K. Screening of innate immune receptors in neurodegenerative diseases: a similar pattern. Neurobiol Aging. 2009;30(5):759–768. doi:10.1016/j.neurobiolaging.2007.08.018.
  • Liu S, Liu Y, Hao W, Wolf L, Kiliaan AJ, Penke B, Rübe CE, Walter J, Heneka MT, Hartmann T, et al. TLR2 is a primary receptor for Alzheimer’s amyloid β peptide to trigger neuroinflammatory activation. J Immunol. 2012;188(3):1098–1107. doi:10.4049/jimmunol.1101121.
  • McDonald CL, Hennessy E, Rubio-Araiz A, Keogh B, McCormack W, McGuirk P, Reilly M, Lynch MA. Inhibiting TLR2 activation attenuates amyloid accumulation and glial activation in a mouse model of Alzheimer’s disease. Brain Behav Immun. 2016;58:191–200. doi:10.1016/j.bbi.2016.07.143.
  • Costello DA, Carney DG, Lynch MA. α-TLR2 antibody attenuates the Aβ-mediated inflammatory response in microglia through enhanced expression of SIGIRR. Brain Behav Immun. 2015;46:70–79. doi:10.1016/j.bbi.2015.01.005.
  • Walter S, Letiembre M, Liu Y, Heine H, Penke B, Hao W, Bode B, Manietta N, Walter J, Schulz-Schüffer W, et al. Role of the Toll-like receptor 4 in neuroinflammation in Alzheimer’s disease. Cell Physiol Biochem. 2007;20(6):947–956. doi:10.1159/000110455.
  • Jin JJ, Kim HD, Maxwell JA, Li L, Fukuchi KI. Toll-like receptor 4-dependent upregulation of cytokines in a transgenic mouse model of Alzheimer’s disease. J Neuroinflammation. 2008;5(1):23. doi:10.1186/1742-2094-5-23.
  • Song M, Jin J, Lim JE, Kou J, Pattanayak A, Rehman JA, Kim H-D, Tahara K, Lalonde R, Fukuchi K-I. TLR4 mutation reduces microglial activation, increases Aβ deposits and exacerbates cognitive deficits in a mouse model of Alzheimer’s disease. J Neuroinflammation. 2011;8(1):92. doi:10.1186/1742-2094-8-92.
  • Qin Y, Liu Y, Hao W, Decker Y, Tomic I, Menger MD, Liu C, Fassbender K. Stimulation of TLR4 attenuates Alzheimer’s disease–related symptoms and pathology in tau-transgenic mice. J Immunol. 2016;197(8):3281–3292. doi:10.4049/jimmunol.1600873.
  • Go M, Kou J, Lim JE, Yang J, Fukuchi KI. Microglial response to LPS increases in wild-type mice during aging but diminishes in an Alzheimer’s mouse model: implication of TLR4 signaling in disease progression. Biochem Biophys Res Commun. 2016;479(2):331–337. doi:10.1016/j.bbrc.2016.09.073.
  • Shintani Y, Drexler HC, Kioka H, Terracciano CMN, Coppen SR, Imamura H, Akao M, Nakai J, Wheeler AP, Higo S, et al. Toll-like receptor 9 protects non-immune cells from stress by modulating mitochondrial ATP synthesis through the inhibition of SERCA2. EMBO Rep. 2014;15(4):438–445. doi:10.1002/embr.201337945.
  • Wang YL, Tan MS, Yu JT, Zhang W, Hu N, Wang H-F, Jiang T, Tan L. Toll-like receptor 9 promoter polymorphism is associated with decreased risk of Alzheimer’s disease in han Chinese. J Neuroinflammation. 2013;10(1):886. doi:10.1186/1742-2094-10-101.
  • Scholtzova H, Do E, Dhakal S, Sun Y, Liu S, Mehta PD, Wisniewski T. Innate immunity stimulation via toll-like receptor 9 ameliorates vascular amyloid pathology in Tg-SwDI mice with associated cognitive benefits. J Neurosci. 2017;37(4):936–959. doi:10.1523/JNEUROSCI.1967-16.2016.
  • Scholtzova H, Chianchiano P, Pan J, Sun Y, Goñi F, Mehta PD, Wisniewski T. Amyloid ß and Tau Alzheimer¿s disease related pathology is reduced by toll-like receptor 9 stimulation. Acta Neuropathol Commun. 2014;2(1):101. doi:10.1186/s40478-014-0101-2.
  • Lin C, Zhao S, Zhu Y, Fan Z, Wang J, Zhang B, Chen Y. Microbiota-gut-brain axis and toll-like receptors in Alzheimer’s disease. Comput Struct Biotechnol J. 2019;17:1309–1317. doi:10.1016/j.csbj.2019.09.008.
  • Bilkei-Gorzo A. Genetic mouse models of brain ageing and Alzheimer’s disease. Pharmacol Ther. 2014;142(2):244–257. doi:10.1016/j.pharmthera.2013.12.009.
  • Fão L, Mota SI, Rego AC. Shaping the Nrf2-ARE-related pathways in Alzheimer’s and parkinson’s diseases. Ageing Res Rev. 2019;54:100942. doi:10.1016/j.arr.2019.100942.
  • Saha S, Buttari B, Profumo E, Tucci P, Saso L. A perspective on Nrf2 signaling pathway for neuroinflammation: a potential therapeutic target in Alzheimer’s and parkinson’s diseases. Front Cell Neurosci. 2022;15:15. doi:10.3389/fncel.2021.787258.
  • Kim S, Indu Viswanath AN, Park JH, Lee HE, Park AY, Choi JW, Kim HJ, Londhe AM, Jang BK, Lee J, et al. Nrf2 activator via interference of Nrf2-Keap1 interaction has antioxidant and anti-inflammatory properties in Parkinson’s disease animal model. Neuropharmacology. 2020;167:107989. doi:10.1016/j.neuropharm.2020.107989.
  • Zhang W, Feng C, Jiang H. Novel target for treating Alzheimer’s diseases: crosstalk between the Nrf2 pathway and autophagy. Ageing Res Rev. 2021;65:101207. doi:10.1016/j.arr.2020.101207.
  • Zhang H, Davies KJA, Forman HJ. Oxidative stress response and Nrf2 signaling in aging. Free Radical Biol Med. 2015;88:314–336. doi:10.1016/j.freeradbiomed.2015.05.036.
  • Ramsey CP, Glass CA, Montgomery MB, Lindl KA, Ritson GP, Chia LA, Hamilton RL, Chu CT, Jordan-Sciutto KL. Expression of Nrf2 in neurodegenerative diseases. J Neuropathol Exp Neurol. 2007;66(1):75–85. doi:10.1097/nen.0b013e31802d6da9.
  • Rojo AI, Pajares M, Rada P, Nuñez A, Nevado-Holgado AJ, Killik R, Van Leuven F, Ribe E, Lovestone S, Yamamoto M, et al. NRF2 deficiency replicates transcriptomic changes in Alzheimer’s patients and worsens APP and TAU pathology. Redox Biol. 2017;13:444–451. doi:10.1016/j.redox.2017.07.006.
  • Alam A, Leoni G, Wentworth CC, Kwal JM, Wu H, Ardita CS, Swanson PA, Lambeth JD, Jones RM, Nusrat A, et al. Redox signaling regulates commensal-mediated mucosal homeostasis and restitution and requires formyl peptide receptor 1. Mucosal Immunol. 2014;7(3):645–655. doi:10.1038/mi.2013.84.
  • Jones RM, Luo L, Ardita CS, Richardson AN, Kwon YM, Mercante JW, Alam A, Gates CL, Wu H, Swanson PA, et al. Symbiotic lactobacilli stimulate gut epithelial proliferation via Nox-mediated generation of reactive oxygen species. Embo J. 2013;32(23):3017–3028. doi:10.1038/emboj.2013.224.
  • Lambeth JD, Neish AS. Nox enzymes and new thinking on reactive oxygen: a double-edged sword revisited. Annu Rev Pathol Mech Dis. 2014;9(1):119–145. doi:10.1146/annurev-pathol-012513-104651.
  • Wentworth CC, Alam A, Jones RM, Nusrat A, Neish AS. Enteric commensal bacteria induce extracellular signal-regulated kinase pathway signaling via formyl peptide receptor-dependent redox modulation of dual specific phosphatase 3. J Biol Chem. 2011;286(44):38448–38455. doi:10.1074/jbc.M111.268938.
  • Swanson PA, Kumar A, Samarin S, Vijay-Kumar M, Kundu K, Murthy N, Hansen J, Nusrat A, Neish AS. Enteric commensal bacteria potentiate epithelial restitution via reactive oxygen species-mediated inactivation of focal adhesion kinase phosphatases. Proc Natl Acad Sci. 2011;108(21):8803–8808. doi:10.1073/pnas.1010042108.
  • Kumar A, Wu H, Collier-Hyams LS, Hansen JM, Li T, Yamoah K, Pan Z-Q, Jones DP, Neish AS. Commensal bacteria modulate cullin-dependent signaling via generation of reactive oxygen species. Embo J. 2007;26(21):4457–4466. doi:10.1038/sj.emboj.7601867.
  • Jones RM, Desai C, Darby TM, Luo L, Wolfarth A, Scharer C, Ardita C, Reedy A, Keebaugh E, Neish A. Lactobacilli modulate epithelial cytoprotection through the Nrf2 pathway. Cell Rep. 2015;12(8):1217–1225. doi:10.1016/j.celrep.2015.07.042.
  • Jones RM, Neish AS. Redox signaling mediated by the gut microbiota. Free Radical Biol Med. 2017;105:41–47. doi:10.1016/j.freeradbiomed.2016.10.495.
  • Fan W, Tang Z, Chen D, Moughon D, Ding X, Chen S, Zhu M, Zhong Q. Keap1 facilitates p62-mediated ubiquitin aggregate clearance via autophagy. Autophagy. 2010;6(5):614–621. doi:10.4161/auto.6.5.12189.
  • Shah SZA, Zhao D, Hussain T, Sabir N, Mangi MH, Yang L. P62-Keap1-NRF2-ARE pathway: a contentious player for selective targeting of autophagy, oxidative stress and mitochondrial dysfunction in prion diseases. Front Mol Neurosci. 2018;11:11. doi:10.3389/fnmol.2018.00310.
  • Joshi G, Gan KA, Johnson DA, Johnson JA. Increased Alzheimer’s disease–like pathology in the APP/PS1ΔE9 mouse model lacking Nrf2 through modulation of autophagy. Neurobiol Aging. 2015;36(2):664–679. doi:10.1016/j.neurobiolaging.2014.09.004.
  • Pajares M, Jiménez-Moreno N, García-Yagüe ÁJ, Escoll M, de Ceballos ML, Van Leuven F, Rábano A, Yamamoto M, Rojo AI, Cuadrado A. Transcription factor NFE2L2/NRF2 is a regulator of macroautophagy genes. Autophagy. 2016;12(10):1902–1916. doi:10.1080/15548627.2016.1208889.
  • Lau A, Wang XJ, Zhao F, Villeneuve NF, Wu T, Jiang T, Sun Z, White E, Zhang DD. A noncanonical mechanism of Nrf2 activation by autophagy deficiency: direct Interaction between Keap1 and p62. Mol Cell Biol. 2010;30(13):3275–3285. doi:10.1128/MCB.00248-10.
  • Salminen A, Kaarniranta K, Haapasalo A, Hiltunen M, Soininen H, Alafuzoff I. Emerging role of p62/sequestosome-1 in the pathogenesis of Alzheimer’s disease. Prog Neurobiol. 2012;96(1):87–95. doi:10.1016/j.pneurobio.2011.11.005.
  • Gureev AP, Sadovnikova IS, Starkov NN, Starkov AA, Popov VN. p62-Nrf2–p62 Mitophagy Regulatory Loop as a Target for Preventive Therapy of Neurodegenerative Diseases. Brain Sci. 2020;10(11):847. doi:10.3390/brainsci10110847.
  • Bonfili L, Cecarini V, Berardi S, Scarpona S, Suchodolski JS, Nasuti C, Fiorini D, Boarelli MC, Rossi G, Eleuteri AM. Microbiota modulation counteracts Alzheimer’s disease progression influencing neuronal proteolysis and gut hormones plasma levels. Sci Rep. 2017;7(1):2426. doi:10.1038/s41598-017-02587-2.
  • Téglás T, Ábrahám D, Jókai M, Kondo S, Mohammadi R, Fehér J, Szabó D, Wilhelm M, Radák Z. Exercise combined with a probiotics treatment alters the microbiome, but moderately affects signalling pathways in the liver of male APP/PS1 transgenic mice. Biogerontology. 2020;21(6):807–815. doi:10.1007/s10522-020-09895-7.
  • Li Z, Zhu H, Zhang L, Qin C. The intestinal microbiome and Alzheimer’s disease: a review. Animal Model Exp Med. 2018;1(3):180–188. doi:10.1002/ame2.12033.
  • Barrett E, Ross RP, O’Toole PW, Fitzgerald GF, Stanton C. γ-Aminobutyric acid production by culturable bacteria from the human intestine. J Appl Microbiol. 2012;113(2):411–417. doi:10.1111/j.1365-2672.2012.05344.x.
  • Pokusaeva K, Johnson C, Luk B, Uribe G, Fu Y, Oezguen N, Matsunami RK, Lugo M, Major A, Mori-Akiyama Y, et al. GABA-producing bifidobacterium dentium modulates visceral sensitivity in the intestine. Neurogastroenterology & Motility. 2017;29(1):e12904. doi:10.1111/nmo.12904.
  • Siragusa S, de Angelis M, di Cagno R, Rizzello CG, Coda R, Gobbetti M. Synthesis of γ-aminobutyric acid by lactic acid bacteria isolated from a variety of Italian cheeses. Appl Environ Microbiol. 2007;73(22):7283–7290. doi:10.1128/AEM.01064-07.
  • Shishov VA, Kirovskaya TA, Kudrin VS, Oleskin AV. Oleskin a v. Amine neuromediators, their precursors, and oxidation products in the culture of Escherichia coli K-12. Appl Biochem Microbiol. 2009;45(5):494–497. doi:10.1134/S0003683809050068.
  • Hamamah S, Aghazarian A, Nazaryan A, Hajnal A, Covasa M. Role of microbiota-gut-brain axis in regulating dopaminergic signaling. Biomedicines. 2022;10(2):436. doi:10.3390/biomedicines10020436.
  • Stanaszek PM, Snell JF, O’Neill JJ. Isolation, extraction, and measurement of acetylcholine from lactobacillus plantarum. Appl Environ Microbiol. 1977;34(2):237–239. doi:10.1128/aem.34.2.237-239.1977.
  • Horiuchi Y, Kimura R, Kato N, Fujii T, Seki M, Endo T, Kato T, Kawashima K. Evolutional study on acetylcholine expression. Life Sci. 2003;72(15):1745–1756. doi:10.1016/S0024-3205(02)02478-5.
  • Russell WR, Hoyles L, Flint HJ, Dumas ME. Colonic bacterial metabolites and human health. Curr Opin Microbiol. 2013;16(3):246–254. doi:10.1016/j.mib.2013.07.002.
  • Özoğul F. Production of biogenic amines by morganella morganii, Klebsiella pneumoniae and Hafnia alvei using a rapid HPLC method. Eur Food Res Technol. 2004;219(5):465–469. doi:10.1007/s00217-004-0988-0.
  • Ganesh BP, Hall A, Ayyaswamy S, Nelson JW, Fultz R, Major A, Haag A, Esparza M, Lugo M, Venable S, et al. Diacylglycerol kinase synthesized by commensal lactobacillus reuteri diminishes protein kinase C phosphorylation and histamine-mediated signaling in the mammalian intestinal epithelium. Mucosal Immunol. 2018;11(2):380–393. doi:10.1038/mi.2017.58.
  • Landete JM, Ferrer S, Pardo I. Biogenic amine production by lactic acid bacteria, acetic bacteria and yeast isolated from wine. Food Control. 2007;18(12):1569–1574. doi:10.1016/j.foodcont.2006.12.008.
  • LeBlanc JG, Milani C, de Giori GS, Sesma F, van Sinderen D, Ventura M. Bacteria as vitamin suppliers to their host: a gut microbiota perspective. Curr Opin Biotechnol. 2013;24(2):160–168. doi:10.1016/j.copbio.2012.08.005.
  • Roche HM, Terres AM, Black IB, Gibney MJ, Kelleher D. Fatty acids and epithelial permeability: effect of conjugated linoleic acid in Caco-2 cells. Gut. 2001;48(6):797–802. doi:10.1136/gut.48.6.797.
  • Chen Y, Yang B, Ross RP, Jin Y, Stanton C, Zhao J, Zhang H, Chen W. Orally administered CLA ameliorates DSS-induced colitis in mice via intestinal barrier improvement, oxidative stress reduction, and inflammatory cytokine and gut microbiota modulation. J Agric Food Chem. 2019;67(48):13282–13298. doi:10.1021/acs.jafc.9b05744.
  • Ren Q, Yang B, Zhang H, Ross RP, Stanton C, Chen H, Chen W. C9, t11, c15-CLNA and t9, t11, c15-CLNA from Lactobacillus plantarum ZS2058 ameliorate dextran sodium sulfate-induced colitis in mice. J Agric Food Chem. 2020;68(12):3758–3769. doi:10.1021/acs.jafc.0c00573.
  • Zelante T, Iannitti RG, Cunha C, De Luca A, Giovannini G, Pieraccini G, Zecchi R, D’Angelo C, Massi-Benedetti C, Fallarino F, et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity. 2013;39(2):372–385. doi:10.1016/j.immuni.2013.08.003.
  • Maqsood R, Stone TW. The Gut-Brain Axis, BDNF, NMDA and CNS disorders. Neurochem Res. 2016;41(11):2819–2835. doi:10.1007/s11064-016-2039-1.
  • Alisi L, Cao R, de Angelis C, Cafolla A, Caramia F, Cartocci G, Librando A, Fiorelli M. The relationships between vitamin k and cognition: a review of current evidence. Front Neurol. 2019;10:10. doi:10.3389/fneur.2019.00239.
  • Xia YY, Pei LY, Gao F, Hu Q-S, Zhang Y, Chen D, Wang G-H. Vitamin K2 suppresses rotenone-induced microglial activation in vitro. Acta Pharmacol Sin. 2016;37(9):1178–1189. doi:10.1038/aps.2016.68.
  • Dodd D, Spitzer MH, van Treuren W, Merrill BD, Hryckowian AJ, Higginbottom SK, Le A, Cowan TM, Nolan GP, Fischbach MA, et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature. 2017;551(7682):648–652. doi:10.1038/nature24661.
  • Jaglin M, Rhimi M, Philippe C, Pons N, Bruneau A, Goustard B, Daugé V, Maguin E, Naudon L, Rabot S. Indole, a signaling molecule produced by the gut microbiota, negatively impacts emotional behaviors in rats. Front Neurosci. 2018;12(APR). doi:10.3389/fnins.2018.00216.
  • Yanovsky I, Finkin-Groner E, Zaikin A, Lerman L, Shalom H, Zeeli S, Weill T, Ginsburg I, Nudelman A, Weinstock M. Carbamate derivatives of indolines as cholinesterase inhibitors and antioxidants for the treatment of Alzheimer’s disease. J Med Chem. 2012;55(23):10700–10715. doi:10.1021/jm301411g.
  • Chai J, Luo L, Hou F, Fan X, Yu J, Ma W, Tang W, Yang X, Zhu J, Kang W, et al. Agmatine Reduces lipopolysaccharide-mediated oxidant response via activating PI3K/Akt pathway and up-regulating Nrf2 and HO-1 expression in macrophages. Plos One. 2016;11(9):e0163634. doi:10.1371/journal.pone.0163634.
  • Ahn SK, Hong S, Park YM, Lee WT, Park KA, Lee JE. Effects of agmatine on hypoxic microglia and activity of nitric oxide synthase. Brain Res. 2011;1373:48–54. doi:10.1016/j.brainres.2010.12.002.
  • Erny D, Hrabě de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, Keren-Shaul H, Mahlakoiv T, Jakobshagen K, Buch T, et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci. 2015;18(7):965–977. doi:10.1038/nn.4030.
  • Ho L, Ono K, Tsuji M, Mazzola P, Singh R, Pasinetti GM. Protective roles of intestinal microbiota derived short chain fatty acids in Alzheimer’s disease-type beta-amyloid neuropathological mechanisms. Expert Rev Neurother. 2018;18(1):83–90. doi:10.1080/14737175.2018.1400909.
  • Yin YN, Yu QF, Fu N, Liu XW, Lu FG. Effects of four Bifidobacteria on obesity in high-fat diet induced rats. World J Gastroenterol. 2010;16(27):3394–3401. doi:10.3748/wjg.v16.i27.3394.
  • Bo TB, Wen J, Zhao YC, Tian SJ, Zhang XY, Wang DH. Bifidobacterium pseudolongum reduces triglycerides by modulating gut microbiota in mice fed high-fat food. J Steroid Biochem Mol Biol. 2020;198:105602. doi:10.1016/j.jsbmb.2020.105602.
  • Yunes RA, Poluektova EU, Dyachkova MS, Klimina KM, Kovtun AS, Averina OV, Orlova VS, Danilenko VN. GABA production and structure of gadB/gadC genes in lactobacillus and bifidobacterium strains from human microbiota. Anaerobe. 2016;42:197–204. doi:10.1016/j.anaerobe.2016.10.011.
  • Tiwari V, Patel AB. Impaired glutamatergic and gabaergic function at early age in aβppswe-ps1de9 mice: implications for Alzheimer’s disease. J Alzheimer’s Dis. 2012;28(4):765–769. doi:10.3233/JAD-2011-111502.
  • Patterson E, Ryan PM, Wiley N, Carafa I, Sherwin E, Moloney G, Franciosi E, Mandal R, Wishart DS, Tuohy K, et al. Gamma-aminobutyric acid-producing lactobacilli positively affect metabolism and depressive-like behaviour in a mouse model of metabolic syndrome. Sci Rep. 2019;9(1):16323. doi:10.1038/s41598-019-51781-x.
  • Rodgers KJ, Main BJ, Samardzic K. Cyanobacterial neurotoxins: their occurrence and mechanisms of toxicity. Neurotox Res. 2018;33(1):168–177. doi:10.1007/s12640-017-9757-2.