You are on page 1of 10

Biomedicine & Pharmacotherapy 153 (2022) 113430

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


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

Microbiota alteration and modulation in Alzheimer’s disease by


gerobiotics: The gut-health axis for a good mind
Duygu Ağagündüz a, Betül Kocaadam-Bozkurt b, Osman Bozkurt b, Heena Sharma c,
Renata Esposito d, Fatih Özoğul e, Raffaele Capasso f, *
a
Department of Nutrition and Dietetics, Gazi University, Emek, Ankara 06490, Turkey
b
Department of Nutrition and Dietetics, Erzurum Technical University, Yakutiye, Erzurum, Turkey
c
Dairy Technology Division, ICAR-National Dairy Research Institute, Karnal, Haryana 132001, India
d
Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L.Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy
e
Department of Seafood Processing Technology, Faculty of Fisheries, Cukurova University, 01330 Adana, Turkey
f
Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, NA, Italy

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

Keywords: The role of the gut microbiota in human health is one of the most important research topics. There is a strong
Gerobiotics relationship between the microbiota-gut-brain axis and cognitive functions. Since Alzheimer’s disease (AD) is a
Probiotics disease characterized by cognitive impairment, the influence of the gut microbiota in the development and
Alzheimer’s disease
treatment of the disease attracts considerable attention. Gerobiotics is a new concept that includes probiotics or
Gut-brain axis
Nutrition
derived postbiotics involved in delaying the aging process. Increasing evidence in the literature suggests that
gerobiotics has important roles in the pathogenesis and progression of AD, and even in its treatment, through
various mechanisms of action. Several researchers have established the linkage between ingestion of gerobiotics
and improved gut dysbiosis and cognitive functions, nevertheless the dose and duration of treatment differ based
on strain. Furthermore, oxidative-inflammatory pathways are mainly involved in the neuroprotective effects
caused by gerobiotics. This review provides the effects of gerobiotics on microbiota alteration and modulation in
AD.

1. Introduction the aging process and in addition to this, developing countries are wit­
nessing a huge propionate increase in the absolute numbers of elderly
Alzheimer’s disease (AD), a progressive neurological disorder, was persons, therefore, this disorder would be a challenging task to ensure
discovered more than a century ago by German psychiatrist Alois Alz­ public health systems in the future. Figures indicate that around 5–6
heimer [1]. Recently in the last few decades, researchers have focused million AD cases are reported every year and the population suffering
on exploring the epidemiology of AD and dementia, one of the most from dementia doubles after every 5 years among people aged more than
common clinical signs manifested by AD. Dementia is described as a 65 years [3].
clinical syndrome characterized by loss of memory, impaired language AD has a significant impact on both individuals and society as a
and related cognitive functions and day-to-day activities. It has been whole. It is often associated with reduced life expectancy, and physical-
reported that dementia affects more than 25 million people worldwide, functional disability and may lead to death in some cases. The popula­
and most these suffer from AD while total dementia cases are 50%− tion is more prone to stroke, musculoskeletal disorders and cardiovas­
70%. [2] All these cases have more serious implications due to the cular diseases due to AD [4]. Besides, decreased diversity and disturbed
globally increasing elderly population (>65 years). The US Centers for microbial ecology in microbiota are reported in AD and thus may be
Disease Control and Prevention (CDC) estimates an increase of 7–12% in associated with pathogenesis of AD and related metabolic outputs in AD
the elderly population from 2000 (420 million in numbers) to 2030 (1 [5,6]. Moreover, due to the disease involving multiple factors, AD de­
billion). Since the occurrence of AD is more positively correlated with mands certain interventions (protective factors) that might result in

* Corresponding author.
E-mail addresses: duyguturkozu@gazi.edu.tr (D. Ağagündüz), betulkocaadam@gmail.com (B. Kocaadam-Bozkurt), dytosmanbozkurt@gmail.com (O. Bozkurt), s.
heenavet@gmail.com (H. Sharma), renataesposito21@gmail.com (R. Esposito), fozogul@cu.edu.tr (F. Özoğul), rafcapas@unina.it (R. Capasso).

https://doi.org/10.1016/j.biopha.2022.113430
Received 22 June 2022; Received in revised form 14 July 2022; Accepted 14 July 2022
Available online 18 July 2022
0753-3322/© 2022 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
D. Ağagündüz et al. Biomedicine & Pharmacotherapy 153 (2022) 113430

stabilized cognitive functions, controlled neurological symptoms and of dysbiosis by gerobiotics and resulting in reduced pathogenesis of AD.
improved quality of life. One such intervention strategy is to target the
diet of people containing health-potentiating biotics, such as probiotics 2. Microbiota-gut-brain axis and Alzheimer’s disease
and their metabolites. International Scientific Association for Probiotic
and Prebiotic (ISAPP) are defined probiotics as “living microorganisms AD is a progressive neurodegenerative disease and extracellular
that create health benefits in the host when taken in sufficient amounts” amyloid-beta (Aß) plaques composed of amyloid and intracellular
and prebiotics as “inanimate food ingredients that support health in the neurofibrillary tangles composed of hyperphosphorylated tau play a role
host through microbiota modulation” [7,8]. On the other hand, ISAPP in its development [21]. It is thought that with aging, genetic and
has recently described postbiotics as “the preparation of non-viable environmental factors become influential in the development of AD.
microorganisms and/or components that provide health benefits to Especially recently, the idea that AD is a “systemic disease” has signif­
the host” and underlined that postbiotics must contain microbial cells or icantly become widespread [22].
cellular components that have been inactivated with or without me­ The role of the gut microbiota on human health is one of the most
tabolites, contributing to the observed health benefits [9]. Several re­ important research topics today [23]. Recent studies point to a signifi­
searchers have demonstrated the development and health beneficial cant relationship between the microbiota-gut-brain axis and cognitive
aspects of fermented foods using probiotics, prebiotics and/or the functions [19,24,25]. Since AD is a disease characterized by cognitive
combination of both (synbiotics) [10–13]. However, gerobiotics, a new impairment, the influence of the gut microbiota on the development of
concept, specifically includes those probiotics (and/or derived post­ the disease has become of a popular interest [26]. Simultaneously, many
biotics) which are mainly involved in delaying the aging process and recent studies have shown that AD can start in the gut and is closely
enhancing the health span of the host [14]. Several probiotic strains related to dysbiosis in the gut microbiota [27–29] (Fig. 1).
have conferred the status of gerobiotics, including B. longum BB68, The microbiota-gut-brain axis contains a complex, multi-directional
L. fermentum MBC2, L. gasseri SBT2055, B. infantis ATCC15697, interconnection system, through which the microbiota and brain have
L. paracasei PS23 L. brevis OW38, and others [15–18]. These bacterial bidirectional communication. This system includes neural (enteric ner­
strains modulate gut homeostasis and hamper the progression of AD, vous system, sympathetic and parasympathetic system), immune (in­
including inflammatory reactions and oxidative stress. Thus, the gut testinal and neural-immune system), neuroendocrine (hypothalamus-
microbiota composition plays a significant role in regulating the path­ pituitary-adrenal axis (HPA)) and metabolic systems. Metabolites, neu­
ways directly involved in the pathogenesis of AD and blood-brain barrier rotransmitters, and hormones produced by the microbiota via nervous
permeability [19]. Increased permeability of blood-brain barrier (BBB) and humoral pathways, cytokines and other immune signals reach the
might result in neuronal degradation, neuronal cell loss and, subse­ brain [30,31]. Microbiota produces some important metabolites asso­
quently, AD [20]. In this way, gerobiotics in the diet can be one of the ciated with changes in brain functions, including neurotransmitters such
most significant intervention to prevent the occurrence of AD by as serotonin, dopamine, γ-aminobutyric acid (GABA), and short-chain
modulating the microbiota composition and consequential response of fatty acids (SCFAs) [32,33]. It has also been stated that microbiota is
physiological processes in the brain. Therefore, this article focuses on effective in microglia maturation and functions [34].
the impact and significance of AD and the possible mechanisms by It is noted that oxidative stress and some changes that occur in the
which gerobiotics can modulate the gut-health axis. Additionally, it also microbiota influence amyloid plaque formation with neuro­
covers the recent animal and clinical studies supporting the counter-act inflammation activation in the cause of AD [35]. The effect of

Fig. 1. The relationship between the microbiota-gut-brain axis and Alzheimer’s Disease.

2
D. Ağagündüz et al. Biomedicine & Pharmacotherapy 153 (2022) 113430

microbiota on the pathogenesis of AD is associated with the capacity of benefits for AD [44]. Another study on mice with AD revealed
the microorganisms. It hosts secrete neurotransmitters, metabolites, and improvement in memory loss and learning abilities in the group
high amounts of amyloid and lipopolysaccharides (LPS), which trigger receiving probiotics (B. lactis, B. longum, L.fermentum, L. acidophilus)
the central nervous system (CNS) inflammation and cerebrovascular compared with the control group. Additionally, the improvement in
degeneration [36]. It was reported that patients with AD have three malondialdehyde (MDA) and superoxide dismutase levels in the pro­
times higher plasma LPS levels compared with healthy controls [26]. biotic group reduced the increased oxidative stress resulting from AD.
Besides, it was revealed that plasma LPS concentration was negatively Consequently, this study revealed that probiotics have effects on the
correlated with the viable count of intestinal Bifidobacterium, which pathogenesis of AD by improving memory impairment and regulating
retains the integrity of the intestinal barrier and shows the microbiota [47].
anti-inflammatory effects by preventing bacterial/endotoxin trans­ A biochemical pathway seen in AD focused on acetylcholine, a
location [37]. neurotransmitter with an important role in the hippocampus. Normally,
It was further stated that there was a relationship between amyloid the enzyme acetylcholinesterase inactivates acetylcholine. In AD,
plaque formation and pro-inflammatory gut bacteria (e.g., Proteobac­ metabolically low acetylcholine levels and high acetylcholinesterase
teria). These bacterial species induce the secretion of proinflammatory levels are frequently reported. Considering this information, in a study,
cytokines by producing high amounts of amyloid, which may lead to the rodents were given scopolamine (1 mg/kg/day) for 10 days to cause
deterioration of cognitive functions [38]. The brain-derived neuro­ forgetfulness (to create an AD-like condition), L. rhamnosus and curcu­
trophic factor (BDNF) regulated by the microbiota plays a neuro­ min affected the acetylcholine level by regulating the acetylcholines­
protective role. In patients with AD, intestinal homeostasis changes with terase level and improving memory loss [46]. In another study, mice
oxidative stress, and a significant decrease is observed in BDNF levels were injected D-Galactose intraperitoneally and were induced AD. It was
[39]. It is believed that this may contribute to the decrease in cognitive seen that the administration of L. plantarum MTCC1325 had a positive
function and progressive neurodegeneration of hippocampal neurons. effect on the entire ATPase system in the mouse brain and delayed
For these reasons, studies on the relationship between the neurodegeneration as it stabilizes the structural and functional integrity
microbiota-gut-brain axis and the progression of AD have become a of the biological membranes by regulating the ionic concentration
popular interest [23]. gradient, and thus protects neurons [48]. Moreover, some studies have
been conducted in the mouse model to improve LPS-induced cognitive
3. Recent evidence for gerobiotics and Alzheimer’s disease impairment-like behaviors. For example, one study examined the effects
of L. plantarum NK151 and B. longum NK173 on cognitive impairment
In this section, animal and human studies with gerobiotic strains that caused by LPS. L. plantarum NK151 and B.longum NK173 prevent LPS
are considered important in the development of AD are discussed and production by E. coli in mice. The study revealed that upregulating BDNF
some current shreds of evidence are displayed in Table 1 and Table 2. expression through Nuclear Factor kappa B (NF-κB) and suppressing
fecal and blood bacterial LPS levels reduced IL-1β, IL-6, and TNF-α ex­
3.1. Animal studies pressions. In this way, an improvement was achieved in cognitive
impairment-like behavior caused by E. coli K1 or LPS and colitis. Thus, it
The effects of different bacterial strains on the pathogenesis and has been reported that these probiotic strains may be effective in the
prognosis of AD have been shown in different animal models using development and treatment of AD and dementia [41].
different strain types and dose interventions. Thus, in a study conducted In a recent study on aged mice conducted with Akkermansia muci­
on Wistar rats with Alzheimer’s, moderate-intensity interval training niphila strain which is named as a gerobiotic, it was observed that this
was applied daily for 8 weeks with L. plantarum and B. bifidum. It was strain increased the phagocytic activities of macrophages and the ac­
found that choline acetyltransferase (ChAT) and BDNF increased tivities of neutral killer (NK) cells as another mechanism. It was also
significantly in the Hippocampus in the group that received exercise found to improve oxidative stress parameters (glutathione peroxidase,
together with probiotics. It has been reported that the increase in ChAT reductase activity, lipid peroxidation level). It was stated that Akker­
levels also increases probiotic-induced cholinergic transmission. Exer­ mansia muciniphila strain could transform into short-chain fatty acids,
cise and these probiotic strains significantly repair tissue damage and reach the hypothalamus through the blood-brain barrier and affect
improve short-term memory in AD [40]. neurotransmitters. In this way, it positively affects the central nervous
Song et al. (2021) reported that the L. plantarum DP189 strain was system and immune system [45]. Similarly, in a different study, aged
applied to mice with AD for 10 weeks, and it was seen that this inter­ mice (18 months) were administered L. brevis OW38 strain (a gerobiotic)
vention improved dysbiosis by modulating the gut microbiota; increased orally for 8 weeks (1 ×109 cfu). It was reported that the expression of
serotonin, dopamine, and GABA levels; and improved neuronal damage, inflammatory markers such as Tumor Necrosis Factor (TNF), NF-kB,
Aß deposition, and tau pathology and thus influenced the pathological interleukin (IL)− 1β, and LPS was inhibited, which can improve colitis
processes positively [42]. Additionally, it was revealed that L. plantarum and memory loss
DP189 regulated the PI3 K/Akt/GSK-3β pathway and prevented tau BDNF, which is among the neurotrophic factors, plays an important
hyperphosphorylation. Therefore, L. plantarum DP189 may be a thera­ role in neuronal growth, differentiation, survival and plasticity. It is used
peutic strategy in AD. as a marker for a decrease observed in BDNF levels because of deterio­
Huang et al. (2022) worked on mice with streptozotocin-accelerated ration in cognitive functions with aging. Long-term supplementation (43
cognitive dysfunction and reported that L. plantarum PS128 supplements weeks) of L. paracasei K71 strain in aged rodents was observed to in­
regulated gliosis, propionic acid levels, and glycogen synthase kinase 3 ß crease c-AMP response element binding protein (CREB) and BDNF
activity, and thus prevented damage caused by intracerebroventricular expression in the hippocampus, thereby improving cognitive function
streptozotocin injection. The authors showed that they proposed PS128 [51].
supplementation as a potential strategy for preventing and/or delaying Nuclear factor erythroid 2-related factor 2 (Nrf-2), which is a tran­
the progression of AD [43]. scription factor regulating antioxidant and detoxifying enzymes, pro­
Supplementing Bifidobacterium bifidum TMC3115 (5 ×108 colony- tects against oxidative stress. A study conducted on aged rodents
forming unit [cfu]) and L. plantarum 45 (LP45) (5 × 108 cfu) on mice revealed that the L. plantarum AR501 strain increased the expression of
with AD for 22 weeks reversed intestinal dysbiosis by reducing Bacter­ antioxidant genes (GCLm: Glutamate-cysteine-ligase regulatory sub-
oides species and increasing the density of Acetatifactor and Millionella. unit, GCLc: Glutamate cysteine-ligase catalytic subunit, GST: Gluta­
These results suggest that these strains repair spatial memory impair­ thione S-Transferase) by affecting the activation pathways of Nrf-2 [53].
ment and modify the gut microbiome, which may have potential TNF-α, which is a cytokine that indicates inflammation, may increase

3
D. Ağagündüz et al. Biomedicine & Pharmacotherapy 153 (2022) 113430

Table 1
Animal studies evaluating the relationship between gerobiotics and Alzheimer’s disease (a summary).
Strains Subjects and Procedures Physiological effects Mechanism of Actions References

Lactiplantibacillus Forty male Wistar rats Short-term memory was improved in Aβ (Aβ + MIIT + Probiotics group) [40]
plantarum Five groups (n = 8 in + MIIT + Probiotics group Increase in ChAT↑
Bifidobacterium bifidum each) BDNF level↑
1. Control,
2. AD models receiving Aβ
3. Aβ + MIIT (AD rats undergoing
MIIT)
4. Aβ + Probiotics (AD rats fed
Lactobacillus plantarum and
Bifidobacterium bifidum),
5. Aβ + MIIT + Probiotics (AD rats
receiving both treatments).
8 weeks
Bifidobacterium longum C57BL (n = 6) mice (6-week old) Alleviated Escherichia coli K1- or LPS-induced NF-κB-mediated BDNF expression was [41]
NK173 Bifidobacterium longum NK173 and cognitive upregulated by suppressing the fecal and
Lactiplantibacillus Lactobacillus plantarum NK151 from a impairment-like behaviors and colitis blood bacterial LPS levels.
plantarum NK151 human fecal bacteria IL-1β↓
IL-6↓
TNF-α↓
Lactiplantibacillus AD mice Prevented cognitive dysfunction Serotonin↑ [42]
plantarum DP189 10 weeks Neuronal damage, tau pathology, and Dopamine↑
amyloid-beta deposition were recovered. GABA↑
Gut dysbiosis was improved. Tau hyperphosphorylation was prevented
with the regulation of PI3 K/Akt/GSK-3β
Lactiplantibacillus PS128 (1010 (cfu/mL) was Prevents cognitive dysfunction caused by Regulates glycogen synthase kinase 3 beta [43]
plantarum PS128 administered via oral gavage to 6- intracerebroventricular streptozotocin activity, propionic acid levels, and glucose
month-old male AD mice that infection
received an intracerebroventricular
injection of streptozotocin (3 mg/kg)
Lactiplantibacillus The APP/PS1 mice Combined TMC3115 and LP45 Abundance of Bacteroides↓ [44]
plantarum 45 (LP45) Four groups: supplementation alleviated gut dysbiosis The abundance of Acetatifactor↑
Bifidobacterium bifidum 1. Alzheimer’s disease (AD) İmproved spatial memory impairment The abundance of Millionella.↑
TMC311 2. TMC3115 group (1 × 109 cfu),
3. LP45 group (1 × 109 cfu) and
4. A mixture group of TMC3115 (5 ×
108 cfu) and LP45 (5 × 108 cfu)
22 weeks
Akkermansia muciniphila Old rodent group (Control) Akkermansia muciniphila can reach the Provides positive effects on anxiety. [45]
(AKK) Old rodent group (Experiment) hypothalamus by passing through the blood- Phagocytic activities of macrophages↑
2 × 108 cfu brain barrier by being reduced to short-chain NK cells activities↑
4 weeks fatty acids. It affects behavior by acting on Provides improvement in oxidative stress
neurotransmitters in the hypothalamus. parameters (glutathione peroxidase,
Its positive effects on the central nervous reductase activity, lipid peroxidation level)
system and immune system are also thought
to be effective in improving life expectancy.
Lacticaseibacillus Rodents were separated into 5 groups There was improvement in memory loss in the Lactobacillus rhamnosus and curcumin affect [46]
rhamnosus UBLR-58 for 10 days: group that received Lactobacillus rhamnosus the acetylcholine level by regulating the
Group 1: Saline (1 mL/kg/day) and curcumin. acetylcholinesterase level.
Group 2: Scopolamine (1 mL/kg/ Oxidative stress↓
day)
Group 3: Scopolamine (1 mL/kg/
day) +Donepezil (2 mg/kg/day)
Group 4: Curcumin (205 mg/kg/day)
+ Scopolamine (1 mL/kg/day)
Group 5: Curcumin (205 mg/kg/day)
+ Scopolamine (1 mL/kg/day)
+ Lactobacillus rhamnosus (1 ×106
cfu/day)
Lactobacillus acidophilus Mice were separated into 5 groups for Probiotic intake has been shown to provide Oxidative stress↓ [47]
Bifidobacterium longum 8 weeks: improvement in memory loss and learning in Improvement in MDA and SOD levels
Bifidobacterium lactis Group 1: Control group mice with AD. In addition, it has been
Limosilactobacillus Group 2: Control-Probiotic group observed that probiotics reduce neurologic
fermentum Group 3: Saline inflammation by reducing increased
Group 4: AH Group oxidative stress.
Group 5: AH- Probiotic group (2 g
(1 × 1010 cfu /day))
Lactiplantibacillus 48 healthy Wistar rats: Rats induced by AD were treated with The activities of the membrane transport [48]
plantarum MTCC1325 1. Control group, Lactobacillus plantarum MTCC1325 and all ATPases system were improved.
2. AD model ATPase enzyme components turned to near Lactobacillus plantarum MTCC1325 stabilizes
3. Normal control rats+ Lactobacillus normal levels within 30 days. the structural and functional integrity of
plantarum MTCC1325 biological membranes and regulates the
4. AD-induced rats+ Lactobacillus ionic concentration gradient, thereby
plantarum MTCC1325 (10 mL/kg protecting the neurons.
body weight, 12 × 108 cfu /mL)
(continued on next page)

4
D. Ağagündüz et al. Biomedicine & Pharmacotherapy 153 (2022) 113430

Table 1 (continued )
Strains Subjects and Procedures Physiological effects Mechanism of Actions References

8 weeks
Levilactobacillus brevis Old mice (18 months old) Colitis formation↓ TNF↓ [49]
OW38 8 weeks (1 × 109 cfu/rodent, orally) Memory impairment↓ Interleukin (IL) − 1β↓
Nuclear factor Kappa β (NF-K β)↓
Lipopolysaccharide↓
Lacticaseibacillus paracasei Group of non-aged mice The decrease in muscle mass and muscle Mitochondrial Function↑ [50]
PS23 Old mouse group (control) strength decreased in the mouse group IL10↑
Old mouse group (LPPS23) receiving LPPS23 compared to the control ROS↓
orally for 12 weeks group.
(1 × 109 cfu/ rodent) It has been found to slow the development of
sarcopenia.
Lacticaseibacillus paracasei Old group (LPK71) Cognitive function↑ BDNF↑ [51]
K71 Old group (control) Memory impairment↓ CREB↑
for 43 weeks
Lactobacillus helveticus Old control group It has been found to reduce aging-related liver NRF-2↑ [52]
KLDS1.8701 Old group (LHKLDS1.8701) damage by decreasing oxidative stress in the
liver.
Lactiplantibacillus Old rodents Antioxidant level↑ Regulates the activity of SOD, CAT and GPX [53]
plantarum AR501 6 weeks It has been found to reduce aging-related liver Regulates the expression of antioxidant
1010 cfu /mL damage by decreasing oxidative stress in the genes (GST, GCLc, GCLm)
liver. NRF-2↑
Lactiplantibacillus pentosus Memory Impairment↓ Oxidative stress↓ [54]
var. plantarum C29 Age-related inflammation↓ BDNF level↑
CREB↑
M1 macrophages↓
(TNF-α, Arginine II)
Lactiplantibacillus Old mice were separated into 5 Learning ability↑ Mitochondrial Functions↑ [55]
plantarum NDC 75017 groups of 10. Memory ability↑
7 weeks
(1 × 108, 1 × 109, 1 × 1010 cfu /mL)

AD: Alzheimer’s disease; MIIT: Moderate-intensity interval training; Aβ: Amyloid beta-peptide; ChAT: choline acetyltransferase; BDNF: Brain derived neurotrophic
factor; LPS: Lipopolysaccharides; CFU: Colony forming unit; NF-kB: Nuclear Factor kappa B; IL-1B: Interleukin 1 beta; IL- 6: Interleukin 6; TNF- α: Tumor Necrosis
Factor-alfa; GABA: Gamma-aminobutyric acid; PI3K: Phosphoinositide 3-kinase; GSK-3β: Glycogen synthase kinase-3- beta; NK cells: Natural Killer Cells; SOD: Su­
peroxide dismutase; CAT: Catalase; GPX: Glutathione Peroxidase; GST: Glutathione S-Transferase; GCLc: Glutamate cysteine ligase catalytic subunit; GCLm:
Glutamate-cysteine ligase regulatory subunit; NRF-2: Nuclear factor erythroid-2; CREB: c-AMP response element binding protein. RBANS: Repeatable Battery for the
Assessment of Neuropsychological Status.

with aging. Woo et al. (2014) reported that TNF-α levels increase due to an increase in cognitive functions were observed because of selenium
aging in old rodents. These old rodents were administered the L. pentosus and probiotic co-supplementation (B. longum, B.bifidum, L. acidophilus)
var. plantarum C29 strain for 5 weeks. At the end of the intervention, it for 12 weeks [59]. In a different study on B. longum BB536, B. infantis
was found that TNF-α expression and inflammation due to aging M-63, B. breve M-16 V, and B. breve B-3 strains, it was reported that
decreased [54]. Additionally, it was reported that BDNF and CREB levels BDNF levels increased, cognitive function improved, and
increased, memory impairment decreased, and cognitive function depression-anxiety level decreased even in healthy elderly individuals
improved. Similarly, in a study conducted with the L. plantarum NDC [58]. In another randomized controlled study with B. longum BORI and
75017 strain, an increase was found in memory ability [55]. B. bifidum BGN4 strains, it was found that the gut microbiota signifi­
cantly changed, mental flexibility was promoted, and stress was allevi­
ated in the group receiving probiotics during a 12-week intervention
3.2. Human studies [66].
In another randomized controlled study, probiotic milk containing
Similar to animal models, the potential effects of gerobiotics in AD (200 mL/day) B. bifidum, L. casei, L. acidophilus, and L. fermentum
have been investigated in human clinical studies, although compara­ (2 ×109 cfu/g for each) was administered to patients with AD for 12
tively limited. For example, in a study, L. plantarum C29 strain was given weeks, and similarly, it was found that cognitive functions increased and
to the elderly with mild cognitive impairment for 12 weeks and it was CRP levels decreased. Additionally, it was seen that the MDA levels
observed that serum BDNF level increased, and cognitive function and decreased [60]. A study conducted with probiotic-fermented milk with
attention level improved (Hwang et al., 2019). In similar studies using kefir grains revealed that oxidative stress, systemic inflammation, and
the Bifidobacterium breve A1 strain in elderly individuals with mild blood cell damage decreased in patients with AD because of the 90-day
cognitive impairment, it was reported that cognitive performance intervention, and cognitive performance increased [65]. These studies
improved at the end of the intervention although the intervention times show that some special probiotic strains, which are not only in their
varied (12–24 weeks) [62–64]. nutraceutical form but also in the food matrix, may have a gerobiotic
The effects of gerobiotics vary depending on the strain used, the effect and lead to improvements in patients with AD.
dose, and the severity of AD and show their effects through different A 12-week randomized controlled study with Bifidobacterium and
mechanisms. A study conducted with patients with AD revealed that Lactobacillus strains revealed that probiotic supplementation was inef­
microbiota dysbiosis and the immune system improved when various fective against cognitive and biochemical parameters in severe AD. It
Bifidobacterium and Lactobacillus strains were applied for 4 weeks [61]. was shown that not only the formulation and dosage of probiotic bac­
Consistent with this finding, it was found that IL-10 levels and the ac­ teria but also the severity of the disease and the duration of adminis­
tivity of natural killer (NK) cells increased when the L. casei shirota strain tration might influence the outcomes of treatment [67].
was given to elderly individuals for 4 weeks [56]. In a randomized
controlled study conducted with individuals with AD (n = 79), a
decrease in CRP levels, an increase in the total antioxidant capacity, and

5
D. Ağagündüz et al. Biomedicine & Pharmacotherapy 153 (2022) 113430

Table 2
Human studies evaluating the relationship between gerobiotics and Alzheimer’s disease (a summary).
Strains Subjects and procedure Physiological Effect Mechanism of Actions References

Lacticaseibacillus casei Shirota Older adults (n = 30) Improve immune system The activity of neutral killer cells↑ [56]
Every day (1.3 × 1010 cfu beverage Level of Serum interleukin (IL)− 10↑
containing probiotic/placebo group
milk)
4 weeks
Lactiplantibacillus plantarum C29 Older adults (n = 100) Cognitive function↑ BDNF level↑ [57]
(800 mg supplementation/ Attention level↑
placebo)
800 mg a day
12 weeks
Bifidobacterium infantis M-63 Older adults (n = 38) Cognitive function↑ BDNF level↑ [58]
Bifidobacterium longum BB536 20 supplement Level of depression-anxiety↓ Gastrointestinal System Function↑
Bifidobacterium breve B-3 Every day (1.25 × 1010 cfu/day) Body Mass Index↓
Bifidobacterium breve M-16V placebo (n = 18) Frequency of defecation↑
12 weeks
Selenium and probiotic co- Patients with AD (n = 79) Cognitive function↑ hs-CRP↓ [59]
supplementation 12 weeks total antioxidant capacity↑
Bifidobacterium longum (2 × 109 cfu/day each) TNF-α↑
Lactobacillus acidophilus PPAR-γ↑
Bifidobacterium bifidum
Lactobacillus acidophilus Patients with AD (n = 60) Cognitive function↑ MDA↓ [60]
Limosilactobacillus fermentum 12 weeks CRP↓
Lacticaseibacillus casei (2 × 109 cfu/g)
Bifidobacterium bifidum
Lactococcus lactis W19 AD patients (n = 20) Modulate gut microbiota↑ Modulate tryptophan metabolism in serum [61]
Lacticaseibacillus casei W56 4 weeks Immune system and the activation of macrophages and/or
Lactobacillus acidophilus W22 dendritic cells
Lacticaseibacillus paracasei W20
Lactiplantibacillus plantarum W62
Bifidobacterium lactis W52
Bifidobacterium lactis W51
Bifidobacterium bifidum W23
Ligilactobacillus salivarius W24
Bifidobacterium breve A1 Older adults (n = 27) with mild Cognitive function ↑ MMSE scores↑ [62]
cognitive impairment
24 weeks
Bifidobacterium breve A1 Probiotic group (n = 59) elderly Cognitive function↑ RBANS score↑ [63]
with memory complaints and control MMSE score
group (n = 58)
12 weeks
Bifidobacterium breve A1 Probiotic group (n = 40) and control Memory functions↑ Increased Repeatable Battery for the [64]
group (n = 40) Assessment of Neuropsychological Status
16 weeks scores
Probiotic-fermented milk with kefir AD patients (n = 13) Improve cognitive deficits Inflammatory cytokine markers↓ [65]
grains 12 weeks Improvement of serum protein oxidation,
mitochondrial dysfunction, DNA damage/
repair, and apoptosis
Oxidative stress markers↓
NO↑
Probiotic containing Bifidobacterium Probiotic group (n = 27) and control Mental flexibility↑ BDNF level↑ [66]
longum BORI and Bifidobacterium group (n = 26) Alleviate stress The gut microbiome significantly shifted
bifidum BGN4 12 weeks (Eubacterium and Clostridiales)
Multi-strain probiotics containing Probiotic group (n = 25) and 23 in In severe AD patients, cognitive and Not defined [67]
three bacteria including either control group (n = 23) biochemical functions were not
Lactiplantibacillus plantarum, 12 weeks influenced by probiotics
Limosilactobacillus fermentum, and
Bifidobacterium lactis or
Bifidobacterium longum,
Lactobacillus acidophilus, and
Bifidobacterium bifidum

AD: Alzheimer’s disease; BDNF: Brain-derived neurotrophic factor; hs-CRP: high sensitivity C-reactive protein; TNF-α: Tumor necrosis factor - alfa; PPAR-γ: Peroxisome
proliferator-activated receptor-gamma; NO: Nitric oxide; CFU: Colony forming unit; MDA: Malondialdehyde; NO: Nitric oxide; MMSE: Mini Mental State Examination

4. The mechanisms of action of gerobiotics on Alzheimer’s probiotics are considered both protective and promising therapeutic
disease agents in AD due to their effects on the gut microbiota [23].
Probiotics are known as “live microorganisms that affect the health
Studies on the significance of the microbiota-gut-brain axis in the of the host positively when taken in sufficient quantities” [69]. Gero­
pathogenesis of AD show this axis as an important target in preventing biotics include probiotics or derived postbiotics involved in delaying the
and slowing the development of the disease [31,35]. Recent evidence aging process [14]. However, this concept is usually used to meet the
has revealed that gut microbiota is important in gut-brain interactions concept of probiotics than postbiotics in the literature. Increasing evi­
[68]. Changes in the gut microbiota composition may lead to the dence in the literature suggests that gerobiotics has important roles in
development of various neurological disorders such as AD. Thus, the development and progression of AD, and even in the treatment,

6
D. Ağagündüz et al. Biomedicine & Pharmacotherapy 153 (2022) 113430

through various mechanisms of action [68]. bacterial LPS levels and with the NF-kB mediated upregulation of BDNF
expression, gerobiotics decrease TNF-α, IL-1β, and IL-6 expressions. In
4.1. Mechanisms of action on the central nervous system this way, it contributes to an improvement in cognitive impairment-like
behavior and colitis [41,49].
The microbiome plays a major role in synaptic transmission at the With the above-mentioned mechanisms of action, probiotics are
molecular mechanism levels. Numerous studies have revealed that thought to play a significant role in the gut-brain axis and modulate
probiotics can produce neuromodulators and neurotransmitters, brain functions (Fig. 2). Although the aim is to explain how probiotics
including norepinephrine, GABA, serotonin, dopamine, acetylcholine, modulate the gut-brain axis, the precise and concrete mechanisms have
and BDNF [42,48,70]. It has also been stated that probiotics improve not yet been fully elucidated [23].
neuronal damage, Aß accumulation, and tau pathology in AD, thereby
improving pathological processes and cognitive functions [42,71]. 5. Potential therapeutic effects of probiotics on Alzheimer’s
Gerobiotics affect the acetylcholine level by regulating the acetyl disease
cholinesterase level and improving memory and cognitive functions [40,
46]. It was revealed that memory loss can be improved by inhibiting the There is a debate on whether the current treatments applied to AD
expression of inflammatory markers such as TNF, IL-1β, NF-kB and LPS cause positive or negative influences on the gut microbiota due to a few
[49]. Additionally, the inhibition of the expression of inflammatory interesting findings in the literature [74]. For example, similar to AD
markers increased the expression of c-AMP response element binding drugs, it has been reported that some pesticides have inhibitory effects
protein (CREB) and BDNF in the hippocampus, thereby improving on acetylcholinesterase and can modulate the gut microbiota composi­
cognitive function [51,54,66]. It was also reported that it had glycogen tion in animals [75]. The treatment of rats with high doses of tacrine led
synthase kinase 3 β activity and gliosis regulatory effects [43]. to some negative effects on the gut microbiota [76]. In this respect, AD
Human studies revealed that with gerobiotic supplementation, drugs may worsen the course of the disease in the long term due to their
serum BDNF levels increase and cognitive function and attention negative effects on the gut microbiota. Accordingly, intestinal dysbiosis
improved [57,58]. can be prevented and/or improved when treated with pre/probiotics
and AD drugs. A paper reviewing the therapeutic effects of probiotics in
4.2. Antioxidant and anti-inflammatory mechanisms of action AD reported that probiotic supplementation could produce neuro­
protective effects, increase cognitive function and modulate gut micro­
Antioxidant and anti-inflammatory effects have been mentioned biota dysbiosis, which is associated with oxidative-inflammatory
among the positive roles of probiotics. Oxidative stress is a significant pathways [77].
factor in the development of AD, which suggests that probiotics may Supporting this view, probiotics affect normal brain activity as well
have important protective effects against neurodegenerative diseases as improve the cognitive functions of patients with AD [60]. It was also
[35]. found that mice with AD treated with pre-and probiotics had higher
It was reported that some Lactobacillus and Bifidobacterium strains cognitive performance and fewer Aβ plaques in their hippocampus [78].
reduce the level of pro-inflammatory cytokines and neutralized ROS In conclusion, the latest data show that pre/probiotic therapy in AD is an
[68,72,73]. It has also been argued that the structural and functional important area of research considering its relationship with conven­
integrity of biological membranes could be stabilized by gerobiotics tional drug therapies. It must be investigated whether administering
through the regulation of the ionic concentration gradient [48]. It was pre/probiotics and currently available drugs at the same time produces
further shown that gerobiotics reduced the elevated oxidative stress due synergistic beneficial effects.
to AD by improving glutathione peroxidase, superoxide dismutase and
lipid peroxidation levels such as MDA levels [45,47,53]. 6. Conclusion and future directions
Human studies revealed that gerobiotics increase the total antioxi­
dant capacity with a decrease in C-reactive protein (CRP) and MDA AD is a neurological disorder manifested by clinical symptoms and
levels, thereby regulating cognitive functions [59,60]. It has also been affects the elderly population. Clinical evidence suggests that dietary
reported that gerobiotics reduce various cytokine markers effective in interventions can be a strategic measure to prevent the occurrence of
inflammation, increase NO bioavailability, improve serum protein such disorders. Probiotics and/or postbiotics, noticeably gerobiotics,
oxidation, DNA damage/apoptosis and mitochondrial dysfunction, and significantly influence the fundamental aging process by modulating the
thus improve cognitive functions [65]. interaction between gut microbiota and neuronal network via the
microbiota-gut-brain axis. Several researchers have established the
4.3. Mechanisms of action on microbiota and immune system linkage between ingestion of gerobiotics and improved gut dysbiosis and
cognitive functions, nevertheless the dose and duration of treatment
The gut microbiota can influence brain activity via humoral activity differ based on strain. However, we may comment that a minimum of 4
or the vagus nerve. Therefore, the deterioration in the gut microbiota weeks of some gerobiotics ingestion appears to be beneficial for AD and
may cause brain dysfunction. At this point, it is stated that gerobiotics related symptoms in some human studies according to literature, but the
increase the phagocytic activities of macrophages and the activities of range of intervention time of gerobiotics to observe AD prognosis in
NK cells and thus have positive effects on the immune system [45,56]. It animal studies is quite wide (4–24 weeks) and it is not easy to reach a
has also been reported that with the modulation of tryptophan meta­ general judgment. Besides, oxidative-inflammatory pathways are
bolism in serum and the activation of macrophages and/or dendritic mainly involved in the neuroprotective effects caused by gerobiotics.
cells, probiotics modulate the microbiota and immune system [61]. Since the pathogenesis of AD is quite complex, other pathways that
Intestinal dysbiosis causes increased inflammation, activation of the evaluate the effect of probiotics need to be unveiled. Moreover, most of
HPA axis, and changes in neurotransmitter and bacterial metabolite the investigations have been conducted on animal models and clinical
levels, which may lead to abnormal signaling through the vagus nerve. studies are limited to a small sample size. Therefore, it becomes
Bacterial migration (leaky gut) and inflammation occur as the integrity imperative to conduct longitudinal human studies with a larger sample
of the gastrointestinal barrier decreases. Inflammation disrupts the for stringent validation of the results. It is worth noting that a single
integrity of the blood-brain barrier. Probiotics may normalize all of mechanism or pathway would never yield promising results, thus the
these processes. Studies have shown that some probiotics, called gero­ interactive pathways may be deduced to a reasonable explanation
biotics, also modulate dysbiosis by modulating gut microbiota [42,47]. behind the role of gerobiotics and their bioactive components in the
Consistent with this, it was found that by suppressing blood and fecal improvement of AD.

7
D. Ağagündüz et al. Biomedicine & Pharmacotherapy 153 (2022) 113430

Fig. 2. The mechanism of action of gerobiotics on Alzheimer’s Disease. TNF: Tumor Necrosis Factor; NF-kB: Nuclear Factor kappa B; LPS: Lipopolysaccharides; ROS:
Reactive Oxygen Species; CRP: C-reactive protein; MDA: Malondialdehyde; NK cells: Natural Killer cells; GABA: γ-aminobutyric acid; BDNF: brain-derived neuro­
trophic factor; CREB: c-AMP Response Element Binding Protein; NO: Nitric oxide.

CRediT authorship contribution statement [2] M. Prince, A. Comas-Herrera, M. Knapp, M. Guerchet, M. Karagiannidou, World
Alzheimer report 2016: improving healthcare for people living with dementia:
coverage, quality and costs now and in the future, 2016.
Duygu Ağagündüz: Conceptualization, Writing – review & editing. [3] C. Qiu, M. Kivipelto, E. von Strauss, Epidemiology of Alzheimer’s disease:
Heena Sharma, Betül Kocaadam-Bozkurt, Renata Esposito, Osman occurrence, determinants, and strategies toward intervention, Dialog. Clin.
Bozkurt: Writing – original draft. Fatih Özoğul, Raffaele Capasso: Neurosci. 11 (2) (2009) 111–128.
[4] C.B. Wong, Y. Kobayashi, J. Xiao, Probiotics for preventing cognitive impairment
Supervision. All authors read and approved the final manuscript. in Alzheimer’s disease, Gut Microbiota Brain Axis (2018) 85–104.
[5] T.G. Dinan, J.F. Cryan, Gut instincts: microbiota as a key regulator of brain
development, ageing and neurodegeneration, J. Physiol. 595 (2) (2017) 489–503.
[6] J. Xi, D. Ding, H. Zhu, R. Wang, F. Su, W. Wu, Z. Xiao, X. Liang, Q. Zhao, Z. Hong,
Conflict of interest statement Disturbed microbial ecology in Alzheimer’s disease: evidence from the gut
microbiota and fecal metabolome, BMC Microbiol. 21 (1) (2021) 1–13.
The authors declare that they have no known competing financial [7] C. Hill, F. Guarner, G. Reid, G.R. Gibson, D.J. Merenstein, B. Pot, L. Morelli, R.B.
Canani, H.J. Flint, S. Salminen, Expert consensus document: the International
interests or personal relationships that could have appeared to influence Scientific Association for Probiotics and Prebiotics consensus statement on the
the work reported in this paper. scope and appropriate use of the term probiotic, Nat. Rev. Gastroenterol. Hepatol.
(2014).
[8] G.R. Gibson, R. Hutkins, M.E. Sanders, S.L. Prescott, R.A. Reimer, S.J. Salminen,
Data availability
K. Scott, C. Stanton, K.S. Swanson, P.D. Cani, Expert consensus document: the
International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus
No data were used in the research described in this article. statement on the definition and scope of prebiotics, Nat. Rev. Gastroenterol.
Hepatol. 14 (8) (2017) 491–502.
[9] S. Salminen, M.C. Collado, A. Endo, C. Hill, S. Lebeer, E.M. Quigley, M.E. Sanders,
Acknowledgments R. Shamir, J.R. Swann, H. Szajewska, The International Scientific Association of
Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope
of postbiotics, Nat. Rev. Gastroenterol. Hepatol. 18 (9) (2021) 649–667.
Fig. 1 and Fig. 2 were created with BioRender.com. [10] P. Chand, M.D. Kumar, A.K. Singh, G.K. Deshwal, P.S. Rao, S.K. Tomar, H. Sharma,
Low-calorie synbiotic yoghurt from indigenous probiotic culture and combination
of inulin and oligofructose: improved sensory, rheological, and textural attributes,
References J. Food Process. Preserv. 45 (4) (2021), e15322.

[1] H. Hippius, G. Neundörfer, The discovery of Alzheimer’s disease, Dialogues in


clinical neuroscience, 2022.

8
D. Ağagündüz et al. Biomedicine & Pharmacotherapy 153 (2022) 113430

[11] H. Sharma, F. Ozogul, E. Bartkiene, J.M. Rocha, Impact of lactic acid bacteria and the hippocampus of rats with Alzheimer’s disease, Neurosci. Lett. 756 (2021),
their metabolites on the techno-functional properties and health benefits of 135949.
fermented dairy products, Crit. Rev. Food Sci. Nutr. (2021) 1–23. [41] D.-Y. Lee, Y.-J. Shin, J.-K. Kim, H.-M. Jang, M.-K. Joo, D.-H. Kim, Alleviation of
[12] E. Dimidi, S.R. Cox, M. Rossi, K. Whelan, Fermented foods: definitions and cognitive impairment by gut microbiota lipopolysaccharide production-
characteristics, impact on the gut microbiota and effects on gastrointestinal health suppressing Lactobacillus plantarum and Bifidobacterium longum in mice, Food
and disease, Nutrients 11 (8) (2019). Funct. 12 (21) (2021) 10750–10763.
[13] B. Yilmaz, H. Sharma, E. Melekoglu, F. Ozogul, Recent developments in dairy kefir- [42] X. Song, Z. Zhao, Y. Zhao, Z. Wang, C. Wang, G. Yang, S. Li, Lactobacillus
derived lactic acid bacteria and their health benefits, Food Biosci. 46 (2022), plantarum DP189 prevents cognitive dysfunction in D-galactose/AlCl3 induced
101592. mouse model of Alzheimer’s disease via modulating gut microbiota and PI3K/Akt/
[14] Y.C. Tsai, L.H. Cheng, Y.W. Liu, O.J. Jeng, Y.K. Lee, Gerobiotics: probiotics GSK-3β signaling pathway, Nutr. Neurosci. (2021) 1–13.
targeting fundamental aging processes, Biosci. Microbiota Food Health 40 (1) [43] H.-J. Huang, J.-L. Chen, J.-F. Liao, Y.-H. Chen, M.-W. Chieu, Y.-Y. Ke, C.-C. Hsu, Y.-
(2021) 1–11. C. Tsai, H.M. Hsieh-Li, Correction: Lactobacillus plantarum PS128 prevents
[15] R. Sharma, Y. Padwad, Probiotic bacteria as modulators of cellular senescence: cognitive dysfunction in alzheimer’s disease mice by modulating propionic acid
emerging concepts and opportunities, Gut Microbes 11 (3) (2020) 335–349. levels, glycogen synthase kinase 3 beta activity, and gliosis, BMC Complement.
[16] K. Jayanama, O. Theou, Effects of probiotics and prebiotics on frailty and ageing: a Med. Ther. 22 (1) (2022) 1–3.
narrative review, Curr. Clin. Pharmacol. 15 (3) (2020) 183–192. [44] F. Wang, T. Xu, Y. Zhang, T. Zheng, Y. He, F. He, Y. Jiang, Long-term combined
[17] E. Setbo, K. Campbell, P. O’Cuiv, R. Hubbard, Utility of probiotics for maintenance administration of Bifidobacterium bifidum TMC3115 and Lactobacillus plantarum
or improvement of health status in older people—a scoping review, J. Nutr. Health 45 alleviates spatial memory impairment and gut dysbiosis in APP/PS1 mice, FEMS
Aging 23 (4) (2019) 364–372. Microbiol. Lett. 367 (7) (2020) fnaa048.
[18] H.S. Lye, Y.T. Lee, S.Y. Ooi, L.K. Teh, L.N. Lim, L.K. Wei, Modifying progression of [45] E.D.-D. Cerro, M. Lambea, J. Félix, N. Salazar, M. Gueimonde, M. De la Fuente,
aging and reducing the risk of neurodegenerative diseases by probiotics and Daily ingestion of Akkermansia mucciniphila for one month promotes healthy
synbiotics, Front. Biosci. 10 (2018) 344–351. aging and increases lifespan in old female mice, Biogerontology 23 (1) (2022)
[19] N. Saji, K. Murotani, N. Sato, T. Tsuduki, T. Hisada, M. Shinohara, T. Sugimoto, 35–52.
S. Niida, K. Toba, T. Sakurai, Relationship between plasma neurofilament light [46] C. Patel, S. Pande, S. Acharya, Potentiation of anti-Alzheimer activity of curcumin
chain, gut microbiota, and dementia: a cross-sectional study, J. Alzheimer’s Dis. by probiotic Lactobacillus rhamnosus UBLR-58 against scopolamine-induced
JAD 86 (3) (2022) 1323–1335. memory impairment in mice, Naunyn-Schmiede’s Arch. Pharmacol. 393 (10)
[20] A. Rutsch, J.B. Kantsjö, F. Ronchi, The gut-brain axis: how microbiota and host (2020) 1955–1962.
inflammasome influence brain physiology and pathology, Front. Immunol. (2020) [47] S. Athari Nik Azm, A. Djazayeri, M. Safa, K. Azami, B. Ahmadvand,
3237. F. Sabbaghziarani, M. Sharifzadeh, M. Vafa, Lactobacilli and bifidobacteria
[21] S. Tiwari, V. Atluri, A. Kaushik, A. Yndart, M. Nair, Alzheimer’s disease: ameliorate memory and learning deficits and oxidative stress in β-amyloid (1–42)
pathogenesis, diagnostics, and therapeutics, Int. J. Nanomed. 14 (2019) 5541. injected rats, Appl. Physiol. Nutr. Metab. 43 (7) (2018) 718–726.
[22] J.-H. Hou, Y.-N. Ou, W. Xu, P.-F. Zhang, L. Tan, J.-T. Yu, Association of peripheral [48] N. Mallikarjuna, K. Praveen, K. Yellamma, Role of Lactobacillus plantarum
immunity with cognition, neuroimaging, and Alzheimer’s pathology, Alzheimer’s MTCC1325 in membrane-bound transport ATPases system in Alzheimer’s disease-
Res. Ther. 14 (1) (2022) 1–11. induced rat brain, BioImpacts BI 6 (4) (2016) 203.
[23] L. Guo, J. Xu, Y. Du, W. Wu, W. Nie, D. Zhang, Y. Luo, H. Lu, M. Lei, S. Xiao, Effects [49] J.-J. Jeong, K. Kim, Y.-J. Hwang, M. Han, D.-H. Kim, Anti-inflammaging effects of
of gut microbiota and probiotics on Alzheimer’s disease, Transl. Neurosci. 12 (1) Lactobacillus brevis OW38 in aged mice, Benef. Microbes 7 (5) (2016) 707–718.
(2021) 573–580. [50] L.-H. Chen, S.-Y. Huang, K.-C. Huang, C.-C. Hsu, K.-C. Yang, L.-A. Li, C.-H. Chan,
[24] C. Proctor, P. Thiennimitr, N. Chattipakorn, S.C. Chattipakorn, Diet, gut microbiota H.-Y. Huang, Lactobacillus paracasei PS23 decelerated age-related muscle loss by
and cognition, Metab. Brain Dis. 32 (1) (2017) 1–17. ensuring mitochondrial function in SAMP8 mice, Aging 11 (2) (2019) 756.
[25] K. Meyer, A. Lulla, K. Debroy, J.M. Shikany, K. Yaffe, O. Meirelles, L.J. Launer, [51] H.M. Corpuz, S. Ichikawa, M. Arimura, T. Mihara, T. Kumagai, T. Mitani,
Association of the gut microbiota with cognitive function in midlife, JAMA Netw. S. Nakamura, S. Katayama, Long-term diet supplementation with Lactobacillus
Open 5 (2) (2022) e2143941-e2143941. paracasei K71 prevents age-related cognitive decline in senescence-accelerated
[26] C. Jiang, G. Li, P. Huang, Z. Liu, B. Zhao, The gut microbiota and Alzheimer’s mouse prone 8, Nutrients 10 (6) (2018) 762.
disease, J. Alzheimer’s Dis. 58 (1) (2017) 1–15. [52] B. Li, S.E. Evivie, J. Lu, Y. Jiao, C. Wang, Z. Li, F. Liu, G. Huo, Lactobacillus
[27] N.M. Vogt, R.L. Kerby, K.A. Dill-McFarland, S.J. Harding, A.P. Merluzzi, S. helveticus KLDS1. 8701 alleviates d-galactose-induced aging by regulating Nrf-2
C. Johnson, C.M. Carlsson, S. Asthana, H. Zetterberg, K. Blennow, Gut microbiome and gut microbiota in mice, Food Funct. 9 (12) (2018) 6586–6598.
alterations in Alzheimer’s disease, Sci. Rep. 7 (1) (2017) 1–11. [53] X. Lin, Y. Xia, G. Wang, Z. Xiong, H. Zhang, F. Lai, L. Ai, Lactobacillus plantarum
[28] H. Choi, D. Lee, I. Mook-Jung, Gut microbiota as a hidden player in the AR501 alleviates the oxidative stress of D-galactose-induced aging mice liver by
pathogenesis of Alzheimer’s disease, J. Alzheimer’s Dis. (2022) 1–26. upregulation of Nrf2–mediated antioxidant enzyme expression, J. Food Sci. 83 (7)
[29] C. Sheng, K. Yang, B. He, W. Du, Y. Cai, Y. Han, Combination of gut microbiota and (2018) 1990–1998.
plasma amyloid-β as a potential index for identifying preclinical Alzheimer’s [54] J.-Y. Woo, W. Gu, K.-A. Kim, S.-E. Jang, M.J. Han, D.-H. Kim, Lactobacillus
disease: a cross-sectional analysis from the SILCODE study, Alzheimer’s Res. Ther. pentosus var. plantarum C29 ameliorates memory impairment and inflammaging
14 (1) (2022) 1–15. in a D-galactose-induced accelerated aging mouse model, Anaerobe 27 (2014)
[30] T.C. Fung, The microbiota-immune axis as a central mediator of gut-brain 22–26.
communication, Neurobiol. Dis. 136 (2020), 104714. [55] X. Peng, J. Meng, T. Chi, P. Liu, C. Man, S. Liu, Y. Guo, Y. Jiang, Lactobacillus
[31] M. De la Fuente, The role of the microbiota-gut-brain axis in the health and illness plantarum NDC 75017 alleviates the learning and memory ability in aging rats by
condition: a focus on Alzheimer’s disease, J. Alzheimer’s Dis. 81 (4) (2021) reducing mitochondrial dysfunction, Exp. Ther. Med. 8 (6) (2014) 1841–1846.
1345–1360. [56] H. Dong, I. Rowland, L.V. Thomas, P. Yaqoob, Immunomodulatory effects of a
[32] C. González-Arancibia, J. Urrutia-Piñones, J. Illanes-González, J. Martinez-Pinto, probiotic drink containing Lactobacillus casei Shirota in healthy older volunteers,
R. Sotomayor-Zárate, M. Julio-Pieper, J.A. Bravo, Do your gut microbes affect your Eur. J. Nutr. 52 (8) (2013) 1853–1863.
brain dopamine? Psychopharmacology 236 (5) (2019) 1611–1622. [57] Y.-H. Hwang, S. Park, J.-W. Paik, S.-W. Chae, D.-H. Kim, D.-G. Jeong, E. Ha,
[33] T.G. Dinan, J.F. Cryan, Brain–gut–microbiota axis—mood, metabolism and M. Kim, G. Hong, S.-H. Park, Efficacy and safety of Lactobacillus plantarum C29-
behaviour, Nat. Rev. Gastroenterol. Hepatol. 14 (2) (2017) 69–70. fermented soybean (DW2009) in individuals with mild cognitive impairment: a 12-
[34] D. Erny, A.L.H. de Angelis, D. Jaitin, P. Wieghofer, O. Staszewski, E. David, week, multi-center, randomized, double-blind, placebo-controlled clinical trial,
H. Keren-Shaul, T. Mahlakoiv, K. Jakobshagen, T. Buch, Host microbiota Nutrients 11 (2) (2019) 305.
constantly control maturation and function of microglia in the CNS, Nat. Neurosci. [58] T. Inoue, Y. Kobayashi, N. Mori, M. Sakagawa, J.-Z. Xiao, T. Moritani, N. Sakane,
18 (7) (2015) 965–977. N. Nagai, Effect of combined bifidobacteria supplementation and resistance
[35] P. Kesika, N. Suganthy, B.S. Sivamaruthi, C. Chaiyasut, Role of gut-brain axis, gut training on cognitive function, body composition and bowel habits of healthy
microbial composition, and probiotic intervention in Alzheimer’s disease, Life Sci. elderly subjects, Benef. Microbes 9 (6) (2018) 843–853.
264 (2021), 118627. [59] O.R. Tamtaji, R. Heidari-Soureshjani, N. Mirhosseini, E. Kouchaki, F. Bahmani,
[36] M. Bostanciklioğlu, The role of gut microbiota in pathogenesis of Alzheimer’s E. Aghadavod, M. Tajabadi-Ebrahimi, Z. Asemi, Probiotic and selenium co-
disease, J. Appl. Microbiol. 127 (4) (2019) 954–967. supplementation, and the effects on clinical, metabolic and genetic status in
[37] P.D. Cani, A.M. Neyrinck, F. Fava, C. Knauf, R.G. Burcelin, K.M. Tuohy, G. Gibson, Alzheimer’s disease: a randomized, double-blind, controlled trial, Clin. Nutr. 38 (6)
N.M. Delzenne, Selective increases of bifidobacteria in gut microflora improve (2019) 2569–2575.
high-fat-diet-induced diabetes in mice through a mechanism associated with [60] E. Akbari, Z. Asemi, R. Daneshvar Kakhaki, F. Bahmani, E. Kouchaki, O.R. Tamtaji,
endotoxaemia, Diabetologia 50 (11) (2007) 2374–2383. G.A. Hamidi, M. Salami, Effect of probiotic supplementation on cognitive function
[38] A. Cattaneo, N. Cattane, S. Galluzzi, S. Provasi, N. Lopizzo, C. Festari, C. Ferrari, U. and metabolic status in Alzheimer’s disease: a randomized, double-blind and
P. Guerra, B. Paghera, C. Muscio, Association of brain amyloidosis with pro- controlled trial, Front. Aging Neurosci. 8 (2016) 256.
inflammatory gut bacterial taxa and peripheral inflammation markers in [61] F. Leblhuber, K. Steiner, B. Schuetz, D. Fuchs, J.M. Gostner, Probiotic
cognitively impaired elderly, Neurobiol. Aging 49 (2017) 60–68. supplementation in patients with Alzheimer’s dementia-an explorative
[39] M. Kaushik, P. Kaushik, S. Parvez, Memory related molecular signatures: the pivots intervention study, Curr. Alzheimer Res. 15 (12) (2018) 1106–1113.
for memory consolidation and Alzheimer’s related memory decline, Ageing Res. [62] Y. Kobayashi, T. Kinoshita, A. Matsumoto, K. Yoshino, I. Saito, J.-Z. Xiao,
Rev. (2022), 101577. Bifidobacterium breve A1 supplementation improved cognitive decline in older
[40] S. Shamsipour, G. Sharifi, F. Taghian, Impact of interval training with probiotic (L. adults with mild cognitive impairment: an open-label, single-arm study, J. Prev.
plantarum/Bifidobacterium bifidum) on passive avoidance test, ChAT and BDNF in Alzheimer’s Dis. 6 (1) (2019) 70–75.

9
D. Ağagündüz et al. Biomedicine & Pharmacotherapy 153 (2022) 113430

[63] Y. Kobayashi, T. Kuhara, M. Oki, J.-Z. Xiao, Effects of Bifidobacterium breve A1 on [70] J.S. Generoso, V.V. Giridharan, J. Lee, D. Macedo, T. Barichello, The role of the
the cognitive function of older adults with memory complaints: a randomised, microbiota-gut-brain axis in neuropsychiatric disorders, Braz. J. Psychiatry 43
double-blind, placebo-controlled trial, Benef. Microbes 10 (5) (2019) 511–520. (2020) 293–305.
[64] J. Xiao, N. Katsumata, F. Bernier, K. Ohno, Y. Yamauchi, T. Odamaki, [71] M. Grochowska, T. Laskus, M. Radkowski, Gut microbiota in neurological
K. Yoshikawa, K. Ito, T. Kaneko, Probiotic Bifidobacterium breve in improving disorders, Arch. Immunol. Ther. Exp. 67 (6) (2019) 375–383.
cognitive functions of older adults with suspected mild cognitive impairment: a [72] M. Azad, A. Kalam, M. Sarker, D. Wan, Immunomodulatory effects of probiotics on
randomized, double-blind, placebo-controlled trial, J. Alzheimer’s Dis. 77 (1) cytokine profiles, BioMed Res. Int. 2018 (2018).
(2020) 139–147. [73] P.S. Begum, G. Madhavi, S. Rajagopal, B. Viswanath, M.A. Razak,
[65] A.M.M. Ton, B.P. Campagnaro, G.A. Alves, R. Aires, L.Z. Côco, C.M. Arpini, V. Venkataratnamma, Probiotics as functional foods: potential effects on human
T. Guerra e Oliveira, M. Campos-Toimil, S.S. Meyrelles, T.M.C. Pereira, Oxidative health and its impact on neurological diseases, Int. J. Nutr. Pharmacol. Neurol. Dis.
stress and dementia in Alzheimer’s patients: effects of synbiotic supplementation, 7 (2) (2017) 23.
Oxid. Med. Cell. Longev. 2020 (2020). [74] J. Hort, M. Valis, F. Angelucci, Administration of pre/probiotics with conventional
[66] C.-S. Kim, L. Cha, M. Sim, S. Jung, W.Y. Chun, H.W. Baik, D.-M. Shin, Probiotic drug treatment in Alzheimer’s disease, Neural Regen. Res. 15 (3) (2020) 448.
supplementation improves cognitive function and mood with changes in gut [75] Y. Liang, J. Zhan, D. Liu, M. Luo, J. Han, X. Liu, C. Liu, Z. Cheng, Z. Zhou, P. Wang,
microbiota in community-dwelling older adults: a randomized, double-blind, Organophosphorus pesticide chlorpyrifos intake promotes obesity and insulin
placebo-controlled, multicenter trial, J. Gerontol. Ser. A 76 (1) (2021) 32–40. resistance through impacting gut and gut microbiota, Microbiome 7 (1) (2019)
[67] A. Agahi, G.A. Hamidi, R. Daneshvar, M. Hamdieh, M. Soheili, A. Alinaghipour, S. 1–15.
M. Esmaeili Taba, M. Salami, Does severity of Alzheimer’s disease contribute to its [76] L.Y. Yip, C.C. Aw, S.H. Lee, Y.S. Hong, H.C. Ku, W.H. Xu, J.M.X. Chan, E.J.
responsiveness to modifying gut microbiota? A double blind clinical trial, Front. Y. Cheong, K.R. Chng, A.H.Q. Ng, The liver–gut microbiota axis modulates
Neurol. (2018) 662. hepatotoxicity of tacrine in the rat, Hepatology 67 (1) (2018) 282–295.
[68] N. Kim, M. Yun, Y.J. Oh, H.J. Choi, Mind-altering with the gut: modulation of the [77] H.-F. Ji, L. Shen, Probiotics as potential therapeutic options for Alzheimer’s
gut-brain axis with probiotics, J. Microbiol. 56 (3) (2018) 172–182. disease, Appl. Microbiol. Biotechnol. 105 (20) (2021) 7721–7730.
[69] C. Hill, F. Guarner, G. Reid, G.R. Gibson, D.J. Merenstein, B. Pot, L. Morelli, R. [78] D. Abraham, J. Feher, G.L. Scuderi, D. Szabo, A. Dobolyi, M. Cservenak, J. Juhasz,
B. Canani, H.J. Flint, S. Salminen, P.C. Calder, M.E. Sanders, The International B. Ligeti, S. Pongor, M.C. Gomez-Cabrera, Exercise and probiotics attenuate the
Scientific Association for Probiotics and Prebiotics consensus statement on the development of Alzheimer’s disease in transgenic mice: role of microbiome, Exp.
scope and appropriate use of the term probiotic, Nat. Rev. Gastroenterol. Hepatol. Gerontol. 115 (2019) 122–131.
11 (8) (2014) 506–514.

10

You might also like