Gut microbiota-derived metabolites mediate the neuroprotective effect of melatonin in cognitive impairment induced by sleep deprivation

Xintong Wang, Zixu Wang, Jing Cao, Yulan Dong, Yaoxing Chen, Xintong Wang, Zixu Wang, Jing Cao, Yulan Dong, Yaoxing Chen

Abstract

Sleep loss is a serious global health concern. Consequences include memory deficits and gastrointestinal dysfunction. Our previous research showed that melatonin can effectively improve cognitive impairment and intestinal microbiota disturbances caused by sleep deprivation (SD). The present study further explored the mechanism by which exogenous melatonin prevents SD-induced cognitive impairments. Here, we established fecal microbiota transplantation, Aeromonas colonization and LPS or butyrate supplementation tests to evaluate the role of the intestinal microbiota and its metabolites in melatonin in alleviating SD-induced memory impairment. RESULTS: Transplantation of the SD-gut microbiota into normal mice induced microglia overactivation and neuronal apoptosis in the hippocampus, cognitive decline, and colonic microbiota disorder, manifesting as increased levels of Aeromonas and LPS and decreased levels of Lachnospiraceae_NK4A136 and butyrate. All these events were reversed with the transplantation of SD + melatonin-gut microbiota. Colonization with Aeromonas and the addition of LPS produced an inflammatory response in the hippocampus and spatial memory impairment in mice. These changes were reversed by supplementation with melatonin, accompanied by decreased levels of Aeromonas and LPS. Butyrate administration to sleep-deprived mice restored inflammatory responses and memory impairment. In vitro, LPS supplementation caused an inflammatory response in BV2 cells, which was improved by butyrate supplementation. This ameliorative effect of butyrate was blocked by pretreatment with MCT1 inhibitor and HDAC3 agonist but was mimicked by TLR4 and p-P65 antagonists. CONCLUSIONS: Gut microbes and their metabolites mediate the ameliorative effects of melatonin on SD-induced cognitive impairment. A feasible mechanism is that melatonin downregulates the levels of Aeromonas and constituent LPS and upregulates the levels of Lachnospiraceae_NK4A136 and butyrate in the colon. These changes lessen the inflammatory response and neuronal apoptosis in the hippocampus through crosstalk between the TLR4/NF-κB and MCT1/ HDAC3 signaling pathways. Video Abstract.

Keywords: Cognitive impairment; Hippocampus; Melatonin; Microbial–gut–brain axis; Sleep deprivation.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2023. The Author(s).

Figures

Fig. 1
Fig. 1
Schematic and timeline of the experimental model. A Fecal microbiota transplantation (FMT) experiment. BAeromonas veronii colonization experiment. C LPS treatment experiment. D Butyrate treatment experiment. Mel: melatonin, MWM: Morris water maze, Vehicle: 2% ethanol sterile saline. SD: sleep deprivation, TAK-242: TLR4. inhibitor
Fig. 2
Fig. 2
The gut microbiota mediated the neuroprotective effect of melatonin in memory impairment induced by sleep deprivation. A Schematic illustration of experimental design. B Comparison of bacterial colony-forming unit (CFU) in feces from control- and Abs-treated mice (n = 10). C Track plot of spatial memory test (with hidden platform). D Track plot of spatial memory test (without hidden platform). E Latency to reach the platform (n = 8). F Path length to reach the platform (n = 8). G Path efficiency to reach the platform (n = 8). H Time spent in the target zone (n = 8). I Number of entries into the target zone (n = 8). J Images of the immunohistochemical microglia in the different experimental groups. The immunohistochemical results were processed using ImageJ. Bar = 50 μm. K IOD of Iba1-positive cells in the hippocampal cornu ammonis (CA)1, CA3, and dentate gyrus (DG) regions (n = 6). L–O The levels of cytokines (TNF-α, IL-6, IL-4, and IL-10) in the hippocampus (n = 5). CON-FMT: receiving control microbiota FMT mice, SD-FMT: receiving sleep deprivation microbiota FMT mice, SD + Mel-FMT: receiving SD + Mel (20 mg/kg) microbiota FMT mice, V-FMT: receiving vehicle microbiota FMT mice. The data represent the mean ± SEM, p < 0.05 was set as the threshold for significance by one-way ANOVA followed by post hoc comparisons using Tukey’s test for multiple groups’ comparisons, *p < 0.05, **p < 0.01, ***p < 0.001
Fig. 3
Fig. 3
Composition of the colonic microbiota in FMT-treated mice. A,B Alpha diversity evaluation of colon microbial richness and evenness by measuring Chao and Ace diversity indexes. C Principal component analysis (PCA). D PCoA score plot. E Nonmetric multidimensional scaling (NMDS) score plot based on the binary_jaccard distance plot based on the OTU of the gut microbe. F Shannon curves. G OTU rank curves. H Rarefaction curves. I Rank abundance curve. J Unweighted pair-group method with arithmetic mean (UPGMA) analysis (at the phylum level). K Unweighted pair-group method with arithmetic mean (UPGMA) analysis (at the genus level) in the mice cecum of the CON-FMT, SD-FMT, and SD + Mel-FMT groups. CON-FMT: receiving control microbiota FMT mice, SD-FMT: receiving sleep deprivation microbiota FMT mice, SD + Mel-FMT: receiving SD + Mel (20 mg/kg) microbiota FMT mice. The data represent the mean ± SEM, p < 0.05 was set as the threshold for significance by one-way ANOVA followed by post hoc comparisons using Tukey’s test for multiple groups’ comparisons, *p < 0.05, **p < 0.01, ***p < 0.001
Fig. 4
Fig. 4
Composition and the key microflora of the colonic microbiota in FMT-treated mice. A Relative abundances of colonic microbiota at the phylum level in the 3 groups. B Relative abundances of gut microbiota at the genus level in the 3 groups. C Linear discriminant analysis effect size (LEfSe) was performed to identify the bacteria that are differentially represented between the different groups. D–K Relative abundance of p_Bacteroidota, p_Proteobacteria, c_Gammaproteobacteria, g_Lachnospiraceae_NK4A136, g_Eubacterium_xylanophilum, g_Ruminococcus_1, g_ Lachnospiraceae_A2, and g_Turicionas in the colon microbiota based on the LefSe results. Solid and dashed lines indicate the mean and median, respectively. CON-FMT: receiving control microbiota FMT mice, SD-FMT: receiving sleep deprivation microbiota FMT mice, SD + Mel-FMT: receiving SD + Mel (20 mg/kg) microbiota FMT mice
Fig. 5
Fig. 5
Composition of the colonic microbiota metabolites in FMT-treated mice. A Venn based on the microbiota metabolites. B β-diversity of principal component analysis (PCA). C Orthogonal projections to latent structures-discriminate analysis (OPLS-DA) score plot was performed on colon. D In the permutation validation plot the Y-axis intercepts of R2 and Q2 are 0.587 and 0.273, respectively, indicating that the model is valid. E Volcano plot based on the differential metabolite screening compared with the CON-FMT and SD-FMT groups. F volcano plot based on the differential metabolite screening compared with the CON-FMT and SD + Mel-FMT groups. G volcano plot based on the differential metabolite screening compared with the SD-FMT and SD-FMT groups. H The relative abundance of butyric acid. I The relative abundance of L-Tryptophan. J Heatmap showing the relative abundance of the key identified 41 metabolites (p < 0.05, VIP > 1). CON-FMT: receiving control microbiota FMT mice, SD-FMT: receiving sleep deprivation microbiota FMT mice, SD + Mel-FMT: receiving SD + Mel (20 mg/kg) microbiota FMT mice. The data represent the mean ± SEM, p < 0.05 was set as the threshold for significance by one-way ANOVA followed by post hoc comparisons using Tukey’s test for multiple groups’ comparisons, *p < 0.05, **p < 0.01, ***p < 0.001
Fig. 6
Fig. 6
Correlation between the microbiome composition and phenotypic variables. A The correlation of top focus metabolites and the signature microbiota (Spearman correlation test, FDR < 0.05). B The contents of acetate in the feces (n = 8). C The contents of propionate in the feces (n = 8). D The contents of butyrate in the feces (n = 8). E The levels of LPS in the hippocampus (n = 7). F Relative abundance of colonic Aeromonas (n = 7). G Plots of correlation analysis between the fecal level of butyrate and colonic Aeromonas. X-axis represented the Relative abundance of colonic Aeromonas; Y-axis represented the fecal butyrate levels. CON-FMT: receiving control microbiota FMT mice, SD-FMT: receiving sleep deprivation microbiota FMT mice, SD + Mel-FMT: receiving SD + Mel (20 mg/kg) microbiota FMT mice. The data represent the mean ± SEM, p < 0.05 was set as the threshold for significance by one-way ANOVA followed by post hoc comparisons using Tukey’s test for multiple groups’ comparisons, *p < 0.05, **p < 0.01, ***p < 0.001
Fig. 7
Fig. 7
Melatonin ameliorates the occurrence of neuroinflammation and memory impairment in mice induced by Aeromonas colonization. A Schematic illustration of experimental design. B Track plot of spatial memory test (with hidden platform). C Track plot of spatial memory test (without hidden platform). D Latency to reach the platform (n = 8). E Path length to reach the platform (n = 8). F Time spent in the target zone (n = 8). G Number of entries into the target zone (n = 8). H Path efficiency to reach the platform (n = 8). I The levels of LPS in the hippocampus (n = 7). J–M The levels of cytokines (TNF-α, IL-6, IL-4, and IL-10) in the hippocampus (n = 5). N Images of the immunohistochemical microglia in the different experimental groups. The immunohistochemical results were processed using ImageJ. Bar = 50 μm. O IOD of Iba1-positive cells in the hippocampal cornu ammonis (CA)1, CA3, and dentate gyrus (DG) regions (n = 6). P–T Relative protein levels of TLR4, HDAC3, p-IκB, p-P65, and cleaved caspase-3 in the hippocampus (n = 6). CON: control group, Aero: Aeromonas colonization group, A + Mel: Aeromonas + melatonin (20 mg/kg) group. The data represent the mean ± SEM, p < 0.05 was set as the threshold for significance by one-way ANOVA followed by post hoc comparisons using Tukey’s test for multiple groups’ comparisons, *p < 0.05, **p < 0.01, ***p < 0.001
Fig. 8
Fig. 8
Melatonin ameliorates LPS-induced neuroinflammation and memory impairment in mice. A Schematic illustration of experimental design. B Track plot of spatial memory test (with hidden platform). C Latency to reach the platform (n = 8). D Path length to reach the platform (n = 8). E Path efficiency to reach the platform (n = 8). F Track plot of spatial memory test (without hidden platform). G Time spent in the target zone (n = 8). H Number of entries into the target zone (n = 8). I Images of the immunohistochemical microglia in the different experimental groups. The immunohistochemical results were processed using ImageJ. Bar = 50 μm. J The levels of LPS in the hippocampus (n = 7). K IOD of Iba1-positive cells in the hippocampal cornu ammonis (CA)1, CA3, and dentate gyrus (DG) regions (n = 6). L–O The levels of cytokines (TNF-α, IL-6, IL-4, and IL-10) in the hippocampus (n = 5). P–T Relative protein levels of TLR4, HDAC3, p-IκB, p-P65, and cleaved caspase-3 in the hippocampus (n = 6). CON: control group, LPS: lipopolysaccharides (2 mg/kg) group, LPS + Mel: LPS + melatonin (20 mg/kg) group, LPS + TAK-242: LPS + TAK-242 (TLR4 inhibitor, 150 mg/kg) group. The data represent the mean ± SEM, p < 0.05 was set as the threshold for significance by one-way ANOVA followed by post hoc comparisons using Tukey’s test for multiple groups’ comparisons, *p < 0.05, **p < 0.01, ***p < 0.001
Fig. 9
Fig. 9
Effect of butyrate on Mel improved memory impairment in sleep-deprived mice. A Schematic illustration of experimental design. B Track plot of spatial memory test (with hidden platform). C Latency to reach the platform (n = 8). D Path length to reach the platform (n = 8). E Path efficiency to reach the platform (n = 8). F Track plot of spatial memory test (without hidden platform). G Number of entries into the target zone (n = 8). H Time spent in the target zone (n = 8). I Images of the immunohistochemical microglia in the different experimental groups. The immunohistochemical results were processed using ImageJ. Bar = 50 μm. J–L IOD of Iba1-positive cells in the hippocampal cornu ammonis (CA)1, CA3, and dentate gyrus (DG) regions (n = 6). M–P The levels of cytokines (TNF-α, IL-6, IL-4, and IL-10) in the hippocampus (n = 5). Q–T Relative protein levels of HDAC3, p-IκB, p-P65, and cleaved caspase-3 in the hippocampus (n = 6). CON: control group, SD: sleep deprivation group, SD + Mel: SD + melatonin (20 mg/kg) group, SD + Abs: SD + antibiotics group, SD + Abs + Mel: SD + antibiotics + Mel group, SD + Abs + Butyrate: SD + antibiotics + Butyrate (40 mM) group. The data represent the mean ± SEM, p < 0.05 was set as the threshold for significance by one-way ANOVA followed by post hoc comparisons using Tukey’s test for multiple groups’ comparisons, *p < 0.05, **p < 0.01, ***p < 0.001
Fig. 10
Fig. 10
Effect of butyrate on inflammatory response and cell neurotoxicity in BV2 cells induced by LPS. A Schematic illustration of experimental design. B–E The levels of cytokines (TNF-α, IL-6, IL-4, and IL-10) in the BV2 cells (n = 5). F–H Relative protein levels of HDAC3, p-IκB and p-P65 in the BV2 cells (n = 6). I Relative protein levels of cleaved caspase-3 in the HT22 cells (n = 6). TAK-242: TLR4 inhibitor, PDTC:NF-κB antagonists, AZD3965:MCT1 inhibitor, ITSA-1: HDAC3 agonist. The data represent the mean ± SEM, p < 0.05 was set as the threshold for significance by one-way ANOVA followed by post hoc comparisons using Tukey’s test for multiple groups’ comparisons. *p < 0.05, **p < 0.01, ***p < 0.001 compared to the control group. # p < 0.05, ## p < 0.01, ### p < 0.001 compared to LPS group. + p < 0.05, +  + p < 0.01, +  +  + p < 0.001 compared to LPS + Mel group
Fig. 11
Fig. 11
Schematic diagram of the protective effects of melatonin on cognitive impairment caused by sleep deprivation through the microbiota–gut–brain axis. Briefly, gut microbes and their metabolites mediate the ameliorative effect of melatonin on SD-induced cognitive impairment. A feasible mechanism is that Mel downregulates the Aeromonas population and production of the constituent LPS production and upregulates the Lachnospiraceae_NK4A136 population and the production of the butyrate metabolite by remodeling gut microbiota homeostasis. These events inhibit the TLR4/HDAC3/NF-κB signaling pathway, thereby preventing neuroinflammation and ultimately alleviating neuronal apoptosis and memory impairment in sleep-deprived mice. HDAC3: histone deacetylase3, LPS: lipopolysaccharide, Mel: melatonin, NF-κB: nuclear factor-κB, SD: sleep deprivation, TLR4: Toll-like receptor 4

References

    1. Troynikov O, Watson CG, Nawaz N. Sleep environments and sleep physiology: a review. J Therm Biol. 2018;78:192–203.
    1. Wang X, Wang Z, Cao J, Dong Y, Chen Y. Melatonin alleviates acute sleep deprivation-induced memory loss in mice by suppressing hippocampal ferroptosis. Front Pharmacol. 2021;12:708645.
    1. Shahrbabaki SS, Linz D, Hartmann S, Redline S, Baumert M. Sleep arousal burden is associated with long-term all-cause and cardiovascular mortality in 8001 community-dwelling older men and women. Eur Heart J. 2021;42(21):2088–2099.
    1. McAlpine CS, Kiss MG, Rattik S, He S, Vassalli A, Valet C, et al. Sleep modulates haematopoiesis and protects against atherosclerosis. Nature. 2019;566(7744):383–387.
    1. Liu X, Chen B, Huang Z, Duan R, Li H, Xie L, et al. Effects of poor sleep on the immune cell landscape as assessed by single-cell analysis. Commun Biol. 2021;4(1):1325.
    1. Jameson KG, Olson CA, Kazmi SA, Hsiao EY. Toward understanding microbiome-neuronal signaling. Mol Cell. 2020;78(4):577–583.
    1. Chang L, Wei Y, Hashimoto K. Brain-gut-microbiota axis in depression: a historical overview and future directions. Brain Res Bull. 2022;182:44–56.
    1. Shi N, Li N, Duan X, Niu H. Interaction between the gut microbiome and mucosal immune system. Mil Med Res. 2017;4:14.
    1. Diaz Heijtz R, Wang S, Anuar F, Qian Y, Björkholm B, Samuelsson A, et al. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A. 2011;108(7):3047–3052.
    1. Matenchuk BA, Mandhane PJ, Kozyrskyj AL. Sleep, circadian rhythm, and gut microbiota. Sleep Med Rev. 2020;53:101340.
    1. Gao T, Wang Z, Dong Y, Cao J, Lin R, Wang X, et al. Role of melatonin in sleep deprivation-induced intestinal barrier dysfunction in mice. J Pineal Res. 2019;67(1):e12574.
    1. Wang Z, Chen WH, Li SX, He ZM, Zhu WL, Ji YB, et al. Gut microbiota modulates the inflammatory response and cognitive impairment induced by sleep deprivation. Mol Psychiatry. 2021;26:6277–6292.
    1. de Theije CG, Wopereis H, Ramadan M, van Eijndthoven T, Lambert J, Knol J, et al. Altered gut microbiota and activity in a murine model of autism spectrum disorders. Brain Behav Immun. 2014;37:197–206.
    1. Sun MF, Shen YQ. Dysbiosis of gut microbiota and microbial metabolites in Parkinson's Disease. Ageing Res Rev. 2018;45:53–61.
    1. Carloni S, Bertocchi A, Mancinelli S, Bellini M, Erreni M, Borreca A, et al. Identification of a choroid plexus vascular barrier closing during intestinal inflammation. Science. 2021;374:439–448.
    1. Obrenovich MEM. Leaky gut, leaky brain? Microorganisms. 2018;6(4):107.
    1. Tena D, González-Praetorius A, Gimeno C, Pérez-Pomata MT. Bisquert J [Extraintestinal infection due to Aeromonas spp.: review of 38 cases] Enferm Infecc Microbiol Clin. 2007;25:235–241.
    1. Zhang W, Li Z, Yang H, Wang G, Liu G, Wang Y, et al. Aeromonas sobria Induces proinflammatory cytokines production in mouse macrophages via activating NLRP3 inflammasome signaling pathways. Front Cell Infect Microbiol. 2021;11:691445.
    1. Zhao Y, Jaber V, Lukiw WJ. Secretory products of the human GI tract microbiome and their potential impact on Alzheimer’s disease (AD): detection of lipopolysaccharide (LPS) in AD hHippocampus. Front Cell Infect Microbiol. 2017;7:318.
    1. Elahy M, Jackaman C, Mamo JC, Lam V, Dhaliwal SS, Giles C, et al. Blood-brain barrier dysfunction developed during normal aging is associated with inflammation and loss of tight junctions but not with leukocyte recruitment. Immun Ageing. 2015;12:2.
    1. Zhang J, Zheng Y, Luo Y, Du Y, Zhang X, Fu J. Curcumin inhibits LPS-induced neuroinflammation by promoting microglial M2 polarization via TREM2/TLR4/NF-κB pathways in BV2 cells. Mol Immunol. 2019;116:29–37.
    1. Muhammad T, Ali T, Ikram M, Khan A, Alam SI, Kim MO. Melatonin rescue oxidative stress-mediated neuroinflammation/ neurodegeneration and memory impairment in scopolamine-induced amnesia mice model. J Neuroimmune Pharmacol. 2019;14:278–294.
    1. Yawoot N, Govitrapong P, Tocharus C, Tocharus J. Ischemic stroke, obesity, and the anti-inflammatory role of melatonin. BioFactors. 2021;47:41–58.
    1. Zhang B, Chen T, Cao M, Yuan C, Reiter RJ, Zhao Z, et al. Gut microbiota dysbiosis induced by decreasing endogenous melatonin mediates the pathogenesis of Alzheimer’s disease and obesity. Front Immunol. 2022;13:900132.
    1. Messner M, Huether G, Lorf T, Ramadori G, Schwörer H. Presence of melatonin in the human hepatobiliary-gastrointestinal tract. Life Sci. 2001;69:543–551.
    1. Wu Y, He F, Zhang C, Zhang Q, Su X, Zhu X, et al. Melatonin alleviates titanium nanoparticles induced osteolysis via activation of butyrate/GPR109A signaling pathway. J Nanobiotechnology. 2021;19(1):170.
    1. He J, Zhang P, Shen L, Niu L, Tan Y, Chen L, et al. Short-chain fatty acids and their association with signalling pathways in inflammation, glucose and lipid metabolism. Int J Mol Sci. 2020;21(17):6356.
    1. Stilling RM, van de Wouw M, Clarke G, Stanton C, Dinan TG, Cryan JF. The neuropharmacology of butyrate: the bread and butter of the microbiota-gut-brain axis? Neurochem Int. 2016;99:110–132.
    1. Dalile B, Van Oudenhove L, Vervliet B, Verbeke K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nat Rev Gastroenterol Hepatol. 2019;16(8):461–478.
    1. Wang H, Zhang M, Li J, Liang J, Yang M, Xia G, et al. Gut microbiota is causally associated with poststroke cognitive impairment through lipopolysaccharide and butyrate. J Neuroinflammation. 2022;19(1):76.
    1. Hou YF, Shan C, Zhuang SY, Zhuang QQ, Ghosh A, Zhu KC, et al. Gut microbiota-derived propionate mediates the neuroprotective effect of osteocalcin in a mouse model of Parkinson’s disease. Microbiome. 2021;9(1):34.
    1. Sun MF, Zhu YL, Zhou ZL, Jia XB, Xu YD, Yang Q, et al. Neuroprotective effects of fecal microbiota transplantation on MPTP-induced Parkinson’s disease mice: gut microbiota, glial reaction and TLR4/TNF-α signaling pathway. Brain Behav Immun. 2018;70:48–60.
    1. Rao J, Xie R, Lin L, Jiang J, Du L, Zeng X, et al. Fecal microbiota transplantation ameliorates gut microbiota imbalance and intestinal barrier damage in rats with stress-induced depressive-like behavior. Eur J Neurosci. 2021;53:3598–3611.
    1. Suh SH, Choe K, Hong SP, Jeong SH, Mäkinen T, Kim KS, et al. Gut microbiota regulates lacteal integrity by inducing VEGF-C in intestinal villus macrophages. EMBO Rep. 2019;20(4):e46927.
    1. Quast C, Pruesse E, Yilmaz P, Gerken J, Schweer T, Yarza P, et al. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 2013;41:D590–596.
    1. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, et al. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12(6):R60.
    1. Liu Z, Dai X, Zhang H, Shi R, Hui Y, Jin X, et al. Gut microbiota mediates intermittent-fasting alleviation of diabetes-induced cognitive impairment. Nat Commun. 2020;11(1):855.
    1. Kim N, Jeon SH, Ju IG, Gee MS, Do J, Oh MS, et al. Transplantation of gut microbiota derived from Alzheimer’s disease mouse model impairs memory function and neurogenesis in C57BL/6 mice. Brain Behav Immun. 2021;98:357–365.
    1. Jing Y, Yu Y, Bai F, Wang L, Yang D, Zhang C, et al. Effect of fecal microbiota transplantation on neurological restoration in a spinal cord injury mouse model: involvement of brain-gut axis. Microbiome. 2021;9(1):59.
    1. Zhao Z, Ning J, Bao XQ, Shang M, Ma J, Li G, et al. Fecal microbiota transplantation protects rotenone-induced Parkinson’s disease mice via suppressing inflammation mediated by the lipopolysaccharide-TLR4 signaling pathway through the microbiota-gut-brain axis. Microbiome. 2021;9(1):226.
    1. Dicker AJ, Huang JTJ, Lonergan M, Keir HR, Fong CJ, Tan B, et al. The sputum microbiome, airway inflammation, and mortality in chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2021;147(1):158–167.
    1. Galea I. The blood-brain barrier in systemic infection and inflammation. Cell Mol Immunol. 2021;18(11):2489–2501.
    1. Fernández-Bravo A, Figueras MJ. An update on the genus aeromonas: Taxonomy, epidemiology, and pathogenicity. Microorganisms. 2020;8(1):129.
    1. Mendoza-Barberá E, Merino S, Tomás J. Surface glucan structures in Aeromonas spp. Mar Drugs. 2021;19(11):649.
    1. Ahmad M. Mercuric resistant bacteria Aeromonas exhibits neurologic toxic effects on the developmental motor reflexes, and brain oxidative stress in mice offspring. Microb Pathog. 2018;114:169–175.
    1. Banks WA, Gray AM, Erickson MA, Salameh TS, Damodarasamy M, Sheibani N, et al. Lipopolysaccharide-induced blood-brain barrier disruption: roles of cyclooxygenase, oxidative stress, neuroinflammation, and elements of the neurovascular unit. J Neuroinflammation. 2015;12:223.
    1. Gogolla N. The brain remembers where and how inflammation struck. Cell. 2021;184:5851–5853.
    1. Peng X, Luo Z, He S, Zhang L, Li Y. Blood-brain barrier disruption by lipopolysaccharide and sepsis-associated encephalopathy. Front Cell Infect Microbiol. 2021;11:768108.
    1. Wang P, Gao J, Ke W, Wang J, Li D, Liu R, et al. Resveratrol reduces obesity in high-fat diet-fed mice via modulating the composition and metabolic function of the gut microbiota. Free Radic Biol Med. 2020;156:83–98.
    1. Sheng K, Yang J, Xu Y, Kong X, Wang J, Wang Y. Alleviation effects of grape seed proanthocyanidin extract on inflammation and oxidative stress in a D-galactose-induced aging mouse model by modulating the gut microbiota. Food Funct. 2022;13:1348–1359.
    1. Mukherjee A, Lordan C, Ross RP, Cotter PD. Gut microbes from the phylogenetically diverse genus Eubacterium and their various contributions to gut health. Gut Microbes. 2020;12(1):1802866.
    1. Lukić I, Getselter D, Ziv O, Oron O, Reuveni E, Koren O, et al. Antidepressants affect gut microbiota and Ruminococcus flavefaciens is able to abolish their effects on depressive-like behavior. Transl Psychiatry. 2019;9(1):133.
    1. Siddiqui MT, Cresci GAM. The immunomodulatory functions of butyrate. J Inflamm Res. 2021;14:6025–6041.
    1. Wang C, Zheng D, Weng F, Jin Y, He L. Sodium butyrate ameliorates the cognitive impairment of Alzheimer's disease by regulating the metabolism of astrocytes. Psychopharmacology. 2022;239:215–227.
    1. Gao T, Wang Z, Dong Y, Cao J, Chen Y. Melatonin-mediated colonic microbiota metabolite butyrate prevents acute sleep deprivation-induced colitis in mice. Int J Mol Sci. 2021;22(21):11894.
    1. Borst K, Dumas AA, Prinz M. Microglia: immune and non-immune functions. Immunity. 2021;54(10):2194–2208.
    1. Abdel-Haq R, Schlachetzki JCM, Glass CK, Mazmanian SK. Microbiome-microglia connections via the gut-brain axis. J Exp Med. 2019;216:41–59.
    1. Wei P, Keller C, Li L. Neuropeptides in gut-brain axis and their influence on host immunity and stress. Comput Struct Biotechnol J. 2020;18:843–851.
    1. Lee SJ, Lee S. Toll-like receptors and inflammation in the CNS. Curr Drug Targets Inflamm Allergy. 2002;1:181–191.
    1. Needham BD, Kaddurah-Daouk R, Mazmanian SK. Gut microbial molecules in behavioural and neurodegenerative conditions. Nat Rev Neurosci. 2020;21(12):717–731.
    1. Ikeda T, Nishida A, Yamano M, Kimura I. Short-chain fatty acid receptors and gut microbiota as therapeutic targets in metabolic, immune, and neurological diseases. Pharmacol Ther. 2022;239:108273.
    1. Wenzel TJ, Gates EJ, Ranger AL, Klegeris A. Short-chain fatty acids (SCFAs) alone or in combination regulate select immune functions of microglia-like cells. Mol Cell Neurosci. 2020;105:103493.

Source: PubMed

3
Prenumerera