Research Article

Lactobacillus gasseri NK109 Alleviates Lipopolysaccharide-Induced Cognitive Impairment in Mice by Up-Regulating Inflammation-Mediated BDNF Expression

Soo-Won Yun1https://orcid.org/0000-0002-8528-7586, Hee-Seo Park2https://orcid.org/0000-0001-7887-7900, Myung Joo Han1,*https://orcid.org/0000-0002-8028-145X, Dong-Hyun Kim2,*https://orcid.org/0000-0003-4783-7900
Author Information & Copyright
1Department of Food and Science, Kyung Hee University, Seoul 02447, Korea
2Neurobiota Research Center, College of Pharmacy, Kyung Hee University, Seoul 02447, Korea
*Corresponding author : Dong-Hyun Kim, Neurobiota Research Center, College of Pharmacy, Kyung Hee University, Seoul 02447, Korea, Tel: +82-2-961-0374, Fax: +82-2-957-5030, E-mail: dhkim@khu.ac.kr
*Corresponding author : Myung Joo Han, Department of Food and Science, Kyung Hee University, Seoul 02447, Korea, Tel: +82-2-961-0553, Fax: +82-2-961-0260, E-mail: mjhan@khu.ac.kr

Copyright © Korean Society for Lactic Acid Bacteria and Probiotics. This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Received: Feb 24, 2023; Revised: May 02, 2023; Accepted: May 04, 2023

Published Online: Jun 30, 2023

Abstract

Exposure to lipopolysaccharide (LPS) causes cognitive impairment (CI). In the preliminary study, Lactobacillus gasseri NK109 suppressed LPS-induced expression of proinflammatory cytokines in macrophages. Therefore, the effect of NK109 on LPS-increased CI was investigated in mice. Intraperitoneal injection of LPS caused CI-like behaviors and neuroinflammation. However, orally administered NK109 reduced LPS-increased CI-like behaviors and hippocampal IL-1β and TNF-α expression, whereas LPS-decreased BDNF expression increased. NK109 also reduced LPS-increased colonic myeloperoxidase, IL-1β, and TNF-α expression. The efficacy of NK109 was increased by the combination of soybean embryo ethanol extract (SE). These findings suggest that NK109 with SE can improve CI by alleviating inflammation-mediated BDNF expression, thereby being beneficial for dementia therapy.

Keywords: Lactobacillus gasseri NK109; cognitive impairment; lipopolysaccharide

INTRODUCTION

Alzheimer’s disease (AD) is the most common form of dementia, which is closely connected with systemic inflammation including neuroinflammation and neurotransmitter imbalance (Cao et al., 2020; Tarawneh and Holtzman, 2012). In particular, neuronal inflammation is known to be deteriorated by the alteration of gut microbiota and overexpression of bacterial toxins such as lipopolysaccharide (LPS) (Busche and Hyman, 2020; Cryan et al., 2020; Jang et al., 2018; Reitz and Mayeux, 2014). Exposure to gut bacteria LPS causes neuroinflammation and gut inflammation in rodents, resulting in cognitive impairment (CI) through the suppression of BDNF expression (Jang et al., 2018). BDNF regulates synapse in the many brain regions including hippocampus for maintaining memory storages (Afonso et al., 2019; Lu et al., 2014).

Approximately 40% of patient with gut inflammation suffer from psychiatric diseases with gut dysbiosis (Almeida et al., 2020; Fousekis et al., 2021). The breaking of gut microbiota by antibiotics, pathogens, and stresses, causes the overgrowth of harmful bacteria such as Clostridioides difficile and toxic substances such as endotoxins (Dodiya et al., 2020; Konstantinidis et al., 2020). Chronic exposure to overproduced endotoxins occurs gut and systemic inflammation, resulting in psychiatric disorder (Jang et al., 2018; Marizzoni et al., 2020). Therefore, the alleviation of gut dysbiosis by probiotics lessens the endotoxin overexpression, leading to the amelioration of gut and systemic inflammation (Angelucci et al., 2019; Gomaa, 2020; Westfall et al., 2017). Gut bacteria lipopolysaccharide (LPS) production- inhibiting Lactobacillus plantarum NK151 alleviates CI with gut inflammation in mice (Lee et al., 2021b). Anti- inflammatory Bifidobacterium longum NK46 improves CI with colitis in rodents (Lee et al., 2019). IL-1β expression- inhibiting Lactobacillus gasseri NK109 also increases cognitive function in aged or Escherichia coli-treated mice (Yun et al., 2020; Yun et al., 2023). Anti- inflammatory Lactiplantibacillus plantarum C29 increases cognitive function in 5xFAD and aged mice by upregulating inflammation-mediated brain-derived neurotropic factor (BDNF) expression (Jeong et al., 2015; Lee et al., 2018). Based on these findings, up-regulating NF-κB activation- suppressed BDNF expression may be beneficial for the therapy of cognitive impairment. Furthermore, the cognitive impairment-ameliorating effects of probiotics are enhanced by the addition of natural products such as soybean (Glycine max), which also improves cognitive impairment (Hwang et al., 2019; Lee et al., 2021a; Lee et al., 2018). Nevertheless, the cognitive impairment- ameliorating action mechanism of NK109 with or without natural products remains elusive.

Therefore, to confirm whether the suppression of NF-κB signaling by probiotics could regulate CI, we examined the effects of Lactobacillus gasseri NK109 and its supplement combined with soybean embryo ethanol extract (SE) on LPS-induced CI in mice.

MATERIALS AND METHODS

Materials

LPS and MRS were purchased from Sigma (St Louis, MO) and BD (Franklin Lakes, NJ), respectively. A commercial soybean germ ethanol extract (SE, unprocessed, contained 0.2% soyasaponin Bb) was purchased from Mirae Biotech (Pocheon-shi, Korea).

Culture of NK109

NK109, which was isolated from human feces (female, 34 yrs, Korean) in the previous report (Yun et al., 2020), was cultured in MRS broth at 37°C for 24 h, centrifuged, and freeze-dried.

Culture of macrophages

Macrophage cells (peritoneal macrophages and BV2 cells) were cultured, as previously reported (Jeong et al., 2016; Lee et al., 2017). These cells (1 × 106 cells/mL) were incubated with LPS (100 ng/mL) in the absence or presence of NK109 (N4, 1 × 104 CFUs/mL; N6, 1 × 106 CFUs/mL) for 20 h. In the supernatant, proinflammatory cytokine levels were assayed.

Animals

C57BL/6 mice (male, 6 weeks old, 18–20 g) were purchased from Koatech (Pyeongtaek-shi, Korea), maintained under the controlled condition, as previously reported. All experiments were approved by the Committee for the Care and Use of Laboratory Animals at Kyung Hee University (IACC, KHUASP(SE)-21306) and were ethically carried out according to the University Guideline.

Mice with CI were prepared by intraperitoneally injecting LPS (10 μg/kg/day), as previously reported (Hong et al., 2014; Lee et al., 2011). (1) Mice were divided into four groups (NC, LP, NL, and NH), intraperitoneally injected with LPS daily for 5 days. Test agents (LP, vehicle alone; NL, 5 × 108 CFU/mouse of NK109; NH, 1 × 109 CFU/mouse of NK109) were orally gavaged daily for 5 days next day after LPS treatment. Each group consisted of 7 mice. (2) Mice were divided into five groups (NC, LP, NL, SENL, and SENH), intraperitoneally injected with LPS daily for 5 days. Thereafter, test agents (LP, vehicle alone; SENL, the mixture of 100 mg/kg/day of SE and 0.5 × 109 CFU/mouse/day of NK109; and SENH, the mixture of 100 mg/kg/day of SE and 1 × 109 CFU/mouse/day of NK109) were orally gavaged daily for 5 days. NC was treated with saline instead of LPS and test agents. Each group consisted of 7 mice.

Cognitive behaviors were measured in the Y-maze task (YMT), novel object recognition test (NORT), and Barnes maze task (BMT) (Yun et al., 2021; Yun et al., 2023). Mice was killed by exposure to CO2 in a chamber. Bloods, brains, and colons were collected and stored at – 80°C. The biomarkers were assayed using enzyme-linked immunosorbent assay (ELISA). For the immunostaining, mice were perfused and brain and colon tissues were collected, post-fixed with paraformaldehyde, cytoprotected, freezed, immunostained, and observed using a confocal microscope.

Statistics

Experimental data are described as mean ± S.D. The significance was analyzed using one-way ANOVA followed by Duncan’s multiple range test (p < 0.05).

RESUTLS

Effect of NK109 on the expression of proinflammatory cytokines in LPS-stimulated macrophages

To understand whether NK109 could mitigate inflammation-involved cognitive decline, we first investigated the effect of NK109 on proinflammatory cytokine expression in LPS-stimulated macrophages (Fig. 1). LPS treatment significantly up-regulated TNF-α and IL-1β expression in peritoneal macrophages and BV2 cells. However, NK109 inhibited aLPS-induced TNF-α and IL-1β expression.

labp-9-1-1-g1
Fig. 1. The effects of NK109 on the TNF-α and IL-1β expression in LPS-stimulated macrophages. Effect on the TNF-α (A) and IL-1β expression (B) in peritoneal macrophages. Effect on the TNF-α (C) and IL-1β expression (D) in BV2 cells. Cells were incubated with NK109 (N4, 1 ’ 104 CFU/mL; N6, 1 ’ 106 CFU mg/mL). NC was treated with vehicle (PBS). Data values were described as mean ± S.D. (n = 4). #p<0.05 vs NC group. *p<0.05 vs LPS group.
Download Original Figure
Effect of NK109 on LPS-induced CI and colitis in mice

Next, we investigated the effect of NK109 on LPS-induced CI in mice (Fig. 2). Intraperitoneally injected LPS impaired cognitive behaviors in the YMT to 65.2% of normal control (NC) mice. However, oral administrations of NK109 improved LPS-impaired cognitive function. NK109 at a dose of 50 mg/kg (NL) restored LPS-impaired cognitive function to 89.8% of NC mice. However, LPS or NK109 did not significantly affect average values of the arm entry numbers in the YMT. LPS treatment also reduced exploration in the NORT to 28.9% of NC mice and increased the escape time (on the third day) in the BMT to 192.8% of NC mice. However, NK109 alleviated exploration and latency. NL treatment increased LPS-impaired exploration time to 58.9% of NC mice and decreased LPS-induced escape time to 115.1% of NC mice. The CI-ameliorating effect of NH was higher without significance than that of NL. Furthermore, NK109 suppressed IL-1β and TNF-α expression in the brain, whereas BDNF expression increased. NK109 also decreased LPS-induced myeloperoxidase, IL-1β, and TNF-α expression in the colon (Fig. 3).

labp-9-1-1-g2
Fig. 2. The effect of NK109 on the LPS-induced CI in mice. Effects on CI-like behaviors in the YMT (A), NORT (B), and BMT (C). (B) Effects on hippocampal BDNF (D), IL-1β (E), and TNF-α (F) levles. NC was treated with vehicle (saline) instead of LPS and test agents. Test agents (LP, vehicle alone; NL, 0.5 × 108 CFU/mouse of NK109; NH, 1 × 109 CFU/mouse of NK109) were orally gavaged in mice daily for 5 days. Data values are as mean ± SD (n = 7). #p<0.05 vs NC group. *p<0.05 vs LP group.
Download Original Figure
labp-9-1-1-g3
Fig. 3. The effect of NK109 on the LPS-induced colitis in mice. Effects on myeloperoxidase (MPO) (A), IL-1β (B), and TNF-α (C) levels in the colon. NC was treated with vehicle (saline) instead of LPS and test agents. Test agents (LP, vehicle alone; NL, 0.5×108 CFU/mouse of NK109; NH, 1×109 CFU/mouse of NK109) were orally gavaged in mice daily for 5 days. Data values are as mean ± SD (n = 7). #p<0.05 vs NC group. *p<0.05 vs LP group.
Download Original Figure
The combined effects of NK109 with SE on LPS-induced CI and colitis in mice

Oral administration of NK109 and heat-processed SE mix increased cognitive function in aged mice (Yun et al., 2023). Therefore, to investigate whether SE could increase the CI-ameliorating effect of NK109, we investigated the effects of NL, SENL, and SENH on LPS-induced CI in mice (Fig. 4). NL, SENL, and SENH all alleviated LPS-induced CI-like behaviors (Fig. 4A-C). They suppressed IL-1β and TNF-α expression and NF-κB+Iba1+ cell number in the brain, whereas BDNF expression and BDNF+NeuN+ cell number increased (Fig. 4D-H). They also decreased LPS-induced myeloperoxidase, IL-1β, and TNF-α expression and NF-κB+CD11c+ cell number in the colon (Fig. 5). Of these, SENH most potently alleviated CI-like behaviors, down-regulated proinflammatory cytokine expression in the brain and colon, and up-regulated BDNF expression in the hippocampus, followed by SENL and NL. However, there were not significant differences between them.

labp-9-1-1-g4
Fig. 4. The effects of SENL and SENH on LPS-induced CI in mice. Effects on CI-like behaviors in the YMT (A), NORT (B), and BMT (C). Effects on hippocampal BDNF (D), IL-1β (E), and TNF-α (F) levels and BDNF+NeuN+ (G) and NF-κB+Iba1+ cell numbers (H) NC was treated with vehicle (saline) instead of LPS and test agents. Test agents (LP, vehicle alone; NL, 0.5 × 109 CFU/mouse of NK109; SENL, [SE (100 mg/kg/day of SE) plus NL (0.5 × 109 CFU/mouse of NK109)]; and SENH, [SE (100 mg/kg/day of SE) plus NH (1 × 109 CFU/mouse of NK109)]) were orally gavaged in mice daily for 5 days. Data values are as mean ± SD (n = 7). #p<0.05 vs NC group. *p<0.05 vs LP group.
Download Original Figure
labp-9-1-1-g5
Fig. 5. The effects of SENL and SENH on LPS-induced colitis in mice. Effects on myeloperoxidase (MPO) (A), IL-1β (B), and TNF-α (C) levels and NF-κB+CD11c+ cell number (D) in the colon. NC was treated with vehicle (saline) instead of LPS and test agents. Test agents (LP, vehicle alone; NL, 0.5 × 109 CFU/mouse of NK109; SENL, [SE (100 mg/kg/day of SE) plus NL (0.5 × 109 CFU/mouse of NK109)]; and SENH, [SE (100 mg/kg/day of SE) plus NH (1 × 109 CFU/mouse of NK109)]) were orally gavaged in mice daily for 5 days. Data values are as mean ± SD (n = 7). #p<0.05 vs NC group. *p<0.05 vs LP group.
Download Original Figure

DISCUSSION

Brain and gut are at the bidirectional cross-talks through neural, immune, and endocrine networks (Cryan et al., 2019; Maqsood and Stone, 2016). Gut inflammation significantly increased the occurrence of dementia (Jang et al., 2018). The attenuation of gut inflammation by probiotics or drugs alleviates CI and depression (Park et al., 2020; Sakurai et al., 2022; Yang et al., 2020; Yun et al., 2021). Anti-inflammatory Limosilactobacillus mucosae NK41 mitigated LPS-impaired cognitive function and gut inflammation in mice (Lee et al., 2019). Lactiplantibacillus plantarum C29 and DW2009, a C29-fermented soybean, increased cognitive function and alleviated colitis in 5xFAD mice (Lee et al., 2018). They also alleviated CI and gut inflammation in mice treated with LPS or D-galactose (Lee et al., 2021a; Woo et al., 2014). Furthermore, DW2009 improved cognitive function in patients with mild CI (Hwang et al., 2019). These results suggest that anti-inflammatory probiotics may alleviate CI as well as gut inflammation in vivo through the activation of gut-brain axis.

In the present study, NK109 improved LPS-impaired CI, neuroinflammation, and colitis in mice. In particular, NK109 suppressed proinflammatory cytokine expression and NF-κB-positive immune cells in the colon and brain. In the previous study, NK109 attenuated Escherichia coli-induced CI and depression in mice by regulating NF-κB activation and gut microbiota dysbiosis and cognitive function in aged mice (Yun et al., 2020). These findings suggest that NK109 may attenuate CI by suppressing inflammatory response.

When NK109 was combined with SE, the combinations also alleviated LPS-impaired cognitive behaviors, suppressed NF-κB-positive cells and proinflammatory cytokine in the brain and colon, whereas BDNF level and BDNF-positive cells increased. Moreover, SENH more potently alleviated CI than SENL or NL. Although the efficacy of SENH against cognitive impairment was not significantly different to that of NK109 or SENL, the combination of NK109 and SE additively alleviated CI and colitis. In addition, NF-κB activation-induced neuroinflammation suppresses BDNF expression in the brain (Carreño et al., 2011; Kairisalo et al., 2009; Lee et al., 2021a; Marini et al., 2004; Ying et al., 2002). These results suggest that the combination of NK109 and SE may strongly alleviate CI by suppressing inflammatory responses and inducing BDNF expression. Moreover, the combination of probiotics with natural products may be beneficial for improving their efficacies.

In conclusion, heat-processing may increase the efficacy of SE on CI. The combined treatment of SE and NK109 may additively mitigate CI by the regulation of NF-κB- mediated BDNF expression.

Funding

This research was supported by the Commercialization Promotion Agency for R&D Outcomes (COMPA) funded by the Ministry of Science and ICT (COMPA 2021-0102).

Conflicts of Interest

The authors declare no conflict of interest.

References

1.

Almeida C, Oliveira R, Soares R, and Barata P (2020) Influence of gut microbiota dysbiosis on brain function: a systematic review. Porto Biomed J,5.

2.

Angelucci F, Cechova K, Amlerova J, and Hort J (2019) Antibiotics, gut microbiota, and Alzheimer’s disease. J. Neuroinflammation. 16, 108.

3.

Busche MA and Hyman BT (2020) Synergy between amyloid-β and tau in Alzheimer’s disease. Nat. Neurosci. 23, 1183-1193.

4.

Cao Q, Tan CC, Xu W, Hu H, Cao XP, Dong Q, Tan L, and Yu JT (2020) The prevalence of dementia: A systematic review and meta-analysis. J. Alzheimers Dis. 73, 1157-1166.

5.

Carreño FR, Walch JD, Dutta M, Nedungadi TP, and Cunningham JT (2011) Brain-derived neurotrophic factor-tyrosine kinase B pathway mediates NMDA receptor NR2B subunit phosphorylation in the supraoptic nuclei following progressive dehydration. J. Neuroendocrinol. 23, 894-905.

6.

Cryan JF, O’Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, Codagnone MG, Cussotto S, Fulling C, Golubeva AV, Guzzetta KE, Jaggar M, Long-Smith CM, Lyte JM, Martin JA, Molinero-Perez A, Moloney G, Morelli E, Morillas E, O’Connor R, Cruz-Pereira JS, Peterson VL, Rea K, Ritz NL, Sherwin E, Spichak S, Teichman EM, van de Wouw M, Ventura-Silva AP, Wallace-Fitzsimons SE, Hyland N, Clarke G, and Dinan TG (2019) The microbiota-gut-brain axis. Physiol. Rev. 99, 1877-2013.

7.

Cryan JF, O’Riordan KJ, Sandhu K, Peterson V, and Dinan TG (2020) The gut microbiome in neurological disorders. Lancet Neurol. 19, 179-194.

8.

Dodiya HB, Forsyth CB, Voigt RM, Engen PA, Patel J, Shaikh M, Green SJ, Naqib A, Roy A, Kordower JH, Pahan K, Shannon KM, and Keshavarzian A (2020) Chronic stress-induced gut dysfunction exacerbates Parkinson’s disease phenotype and pathology in a rotenone-induced mouse model of Parkinson’s disease. Neurobiol Dis. 135, 104352.

9.

Fousekis FS, Katsanos AH, Kourtis G, Saridi M, Albani E, Katsanos KH, and Christodoulou DK (2021) Inflammatory bowel disease and patients with mental disorders: What do we know?. J. Clin. Med. Res. 13, 466-473.

10.

Gomaa EZ (2020) Human gut microbiota/microbiome in health and diseases: a review. Antonie Van Leeuwenhoek. 113, 2019-2040.

11.

Hong SW, Yoo DH, Woo JY, Jeong JJ, Yang JH and Kim DH (2014) Soyasaponins Ab and Bb prevent scopolamine-induced memory impairment in mice without the inhibition of acetylcholinesterase. J. Agric. Food Chem. 62, 2062-2068.

12.

Hwang YH, Park S, Paik JW, Chae SW, Kim DH., Jeong DG, Ha E, Kim M, Hong G, Park SH, Jung SJ, Lee SM, Na KH, Kim J and Chung YC (2019) Efficacy and Safety of Lactobacillus plantarum C29-fermented soybean (DW2009) in individuals with mild cognitive impairment: A 12-week, multi-center, randomized, double-blind, placebo-controlled clinical trial. Nutrients. 11, 305.

13.

Jang SE, Lim SM, Jeong JJ, Jang HM, Lee HJ, Han MJ and Kim DH (2018) Gastrointestinal inflammation by gut microbiota disturbance induces memory impairment in mice. Mucosal. Immunol. 11, 369-379.

14.

Jeong JJ, Kim KA, Hwang YJ, Han MJ and Kim DH (2016) Anti-inflammaging effects of Lactobacillus brevis OW38 in aged mice. Benef. Microbes. 7, 707-718.

15.

Jeong JJ, Woo JY, Kim KA, Han MJ and Kim DH (2015) Lactobacillus pentosus var. plantarum C29 ameliorates age-dependent memory impairment in Fischer 344 rats. Lett. Appl. Microbiol. 60, 307-314.

16.

Kairisalo M, Korhonen L, Sepp M, Pruunsild P, Kukkonen JP, Kivinen J, Timmusk T, Blomgren K and Lindholm D (2009) NF-kappaB-dependent regulation of brain-derived neurotrophic factor in hippocampal neurons by X-linked inhibitor of apoptosis protein. Eur. J. Neurosci. 30, 958-966.

17.

Konstantinidis T, Tsigalou C, Karvelas A, Stavropoulou E, Voidarou C, Bezirtzoglou E (2020) Effects of antibiotics upon the gut microbiome: A review of the literature. Biomedicines. 8, 502.

18.

Lee DY, Kim JK, Yun SW, Han MJ and Kim DH (2021a) DW2009 Elevates the efficacy of donepezil against cognitive impairment in Mice. Nutrients. 13, 3273.

19.

Lee DY, Shin YJ, Kim JK, Jang HM, Joo MK and Kim DH (2021b) Alleviation of cognitive impairment by gut microbiota lipopolysaccharide production-suppressing Lactobacillus plantarum and Bifidobacterium longum in mice. Food Funct. 12, 10750-10763.

20.

Lee HJ, Hwang YH and Kim DH (2018) Lactobacillus plantarum C29-fermented soybean (DW2009) Alleviates memory impairment in 5XFAD transgenic mice by regulating microglia activation and gut microbiota composition. Mol. Nutr. Food Res. 62, e1800359.

21.

Lee HJ, Lee KE, Kim JK, and Kim DH (2019) Suppression of gut dysbiosis by Bifidobacterium longum alleviates cognitive decline in 5XFAD transgenic and aged mice. Sci. Rep. 9, 11814.

22.

Lee HJ, Lim SM, Ko DB, Jeong JJ, Hwang YH and Kim DH (2017) Soyasapogenol B and genistein attenuate lipopolysaccharide-induced memory impairment in mice by the modulation of NF-κB-mediated BDNF expression. J. Agric. Food Chem. 65, 6877-6885.

23.

Lee IA, Park YJ, Joh EH, and Kim DH (2011) Soyasaponin Ab ameliorates colitis by inhibiting the binding of lipopolysaccharide (LPS) to toll-like receptor (TLR)4 on macrophages. J. Agric. Food Chem. 59, 13165-13172.

24.

Maqsood R and Stone TW (2016) The gut-brain axis, BDNF, NMDA and CNS disorders. Neurochem. Res. 41, 2819-2835.

25.

Marini AM, Jiang X, Wu X, Tian F, Zhu D, Okagaki P, and Lipsky RH (2004) Role of brain-derived neurotrophic factor and NF-kappaB in neuronal plasticity and survival: From genes to phenotype. Restor. Neurol. Neurosci. 22, 121-130.

26.

Marizzoni M, Cattaneo A, Mirabelli P, Festari C, Lopizzo N, Nicolosi V, Mombelli E, Mazzelli M, Luongo D, Naviglio D, Coppola L, Salvatore M and Frisoni GB (2020) short-chain fatty acids and lipopolysaccharide as mediators between gut dysbiosis and amyloid pathology in Alzheimer’s disease. J. Alzheimers. Dis. 78, 683-697.

27.

Park MR, Shin M, Mun D, Jeong SY, Jeong DY, Song M, Ko G, Unno T, Kim Y and Oh S (2020) Probiotic Lactobacillus fermentum strain JDFM216 improves cognitive behavior and modulates immune response with gut microbiota. Sci. Rep. 10, 21701.

28.

Reitz C and Mayeux R (2014) Alzheimer disease: epidemiology, diagnostic criteria, risk factors and biomarkers. Biochem. Pharmacol. 88, 640-651.

29.

Sakurai K, Toshimitsu T, Okada E, Anzai S, Shiraishi I, Inamura N, Kobayashi S, Sashihara T and Hisatsune T (2022) Effects of Lactiplantibacillus plantarum OLL2712 on memory function in older adults with declining memory: A randomized placebo-controlled trial. Nutrients. 14, 4300.

30.

Tarawneh R and Holtzman DM (2012) The clinical problem of symptomatic Alzheimer disease and mild cognitive impairment. Cold Spring Harb. Perspect. Med. 2, a006148.

31.

Westfall S, Lomis N, Kahouli I, Dia SY, Singh SP, and Prakash S (2017) Microbiome, probiotics and neurodegenerative diseases: deciphering the gut brain axis. Cell. Mol. Life Sci. 74, 3769-3787.

32.

Woo JY, Gu W, Kim KA, Jang SE, Han MJ, and Kim DH (2014) Lactobacillus pentosus var. plantarum C29 ameliorates memory impairment and inflammaging in a D-galactose-induced accelerated aging mouse model. Anaerobe. 27, 22-26.

33.

Yang X, Yu D, Xue L, Li H and Du J (2020) Probiotics modulate the microbiota-gut-brain axis and improve memory deficits in aged SAMP8 mice. Acta Pharm. Sin. B. 10, 475-487.

34.

Ying SW, Futter M, Rosenblum K, Webber MJ, Hunt SP, Bliss TV, and Bramham CR (2002) Brain-derived neurotrophic factor induces long-term potentiation in intact adult hippocampus: requirement for ERK activation coupled to CREB and upregulation of Arc synthesis. J. Neurosci. 22, 1532-1540.

35.

Yun SW, Kim JK, Han MJ, and Kim DH (2021) Lacticaseibacillus paracasei NK112 mitigates Escherichiacoli-induced depression and cognitive impairment in mice by regulating IL-6 expression and gut microbiota. Benef. Microbes. 12, 541-551.

36.

Yun SW, Kim JK, Lee KE, Oh YJ, Choi HJ, Han MJ and Kim DH (2020) A probiotic Lactobacillus gasseri alleviates Escherichia coli-induced cognitive impairment and depression in mice by regulating IL-1β expression and gut microbiota. Nutrients. 12, 3441.

37.

Yun SW, Park HS, Shin YJ, Ma X, Han MJ, Kim DH (2023) Lactobacillus gasseri NK109 and its supplement alleviate cognitive impairment in mice by modulating NF-κB activation, BDNF expression, and gut microbiota composition. Nutrients. 15, 790.