banner



How To Increase E Coli In Gut

Introduction

Bidirectional networks between the brain and gut microbiota are maintained through the hypothalamus-pituitary-adrenal (HPA) axis and microbiota-gut-brain (MGB) centrality (1, 2). Exposure to external stressors, such as immobilization, stimulates the brain to secrete hormones such equally corticotrophin-releasing factor, via the HPA centrality, which stimulate the gut immune system and modify microbiota limerick and their byproduct product (iii–v). The overexpression of gut microbiota byproducts such as endotoxins disturbs gastrointestinal immune responses, which can crusade the secretion of neurotransmitters such as serotonin and catecholamines to fluctuate; this results in the occurrence of systemic inflammatory diseases such equally ulcerative colitis, obesity, and depression (half dozen–9). Oral administration of the commensal bacteria Escherichia coli, which is excessively proliferated by 2,4,half dozen-trinitrobenzenesulfonic acid (TNBS) or immobilization stress, and peritoneal injection of its lipopolysaccharide (LPS) cause colitis, hippocampal inflammation, cognitive pass up, and feet in mice via contradistinct microbiota (10, xi). LPS released from Bacteroides fragilis, which is abundant in the gut, have been suggested to crusade Alzheimer'south disease (AD) (12). A single peritoneal injection of LPS activates hippocampal astrocytes through interaction between the cells of the brain-immune interface and cytokine signals and its repeated injection activates microglia (13, xiv). The peritoneal injection of LPS also suppresses brain-derived neurotrophic factor (BDNF) and military camp response element bounden protein (CREB) expression by activating the NF-κB signaling pathway (13, 15). These results suggest that altered microbiota-induced endotoxemia may cause cognitive turn down and anxiety by inducing neuroinflammation in the brain.

The gut microbiota of healthy humans and animals consist of >ten11 bacteria per gram of gut contents (16, 17). They produce toxic compounds such as LPS and peptidoglycan (PG). LPS and PG are detected past macrophages, dendritic cells, and endothelial cells that are involved in the innate allowed system and then actuate the biosynthesis of inflammation mediators such as tumor necrosis factor (TNF)-α and interleukin (IL)-6, resulting in the inflammation (eighteen–xx). Excessive, chronic exposure to LPS in gut microbiota may cause systemic disorders via gut inflammation, such equally cognitive reject and low. However, the suppression of gut microbiota LPS production by the probiotic Lactobacillus plantarum C29 alleviates LPS- or TNBS-induced colitis and cognitive decline in mice (21, 22). Gut microbiota LPS production-inhibitory Lactobacillus brevis OW38 also increased cognitive function in anile mice (23). Oral administration of E. coli, which produces a big amount of LPS, significantly increases blood LPS levels in mice while treatment with Lactobacillus johnsonii significantly suppresses E. coli-induced cognitive decline and low (10, 11). These results suggest that regulating the balance between anti-inflammatory and inflammatory gut bacteria may be essential for the handling of neuropsychiatric disorders.

Therefore, we isolated inflammatory Eastward. coli K1 and anti-inflammatory Lactobacillus mucosae (formerly Lactobacillus reuteri) NK41 from good for you homo gut microbiota and examined whether K1 could cause altered microbiota, colitis, cognitive decline, and depression in mice and whether NK41 reduced K1-induced altered microbiota, cognitive turn down, and depression in mice.

Results

Furnishings of Escherichia coli K1 and Lactobacillus mucosae NK41 on the NF-κB Activation in Macrophages

To empathise how gut bacteria to regulate the occurrence of psychiatric disorders, we isolated gut bacteria and measured inflammatory and anti-inflammatory leaner from homo stools (Figure 1). Of these bacteria, K1 potently induced tumor necrosis cistron (TNF)-α expression and NF-κB activation in macrophages, like LPS, while NK41 did not affect them. NK41 potently suppressed LPS- or K1-induced TNF-α expression and NF-κB activation in activated macrophage. Furthermore, NK41 potently hindered LPS-induced TNF-α expression in BV-2 cells (Figure S1). K1 and NK41 were identified equally E. coli and 50. mucosae based on the results of Gram staining, API l CHL Kit (bioMerieux, Seoul, South korea), and 16S rDNA sequencing (ABI 3730XL Deoxyribonucleic acid analysis), respectively.

www.frontiersin.org

Figure 1. Effects of gut bacteria K1 and NK41 on the TNF-α expression and NF-κB activation in macrophages. (A) Effects of NK41 and K1 on TNF-α expression and NF-κB activation in macrophages. (B) Effects of NK41 on TNF-α expression and NF-κB activation in LPS-stimulated macrophages. (C) Consequence of K1 on TNF-α expression and NF-κB activation in LPS-stimulated macrophages. (D) Issue of NK41 on NF-κB activation in K1-stimulated macrophages. Macrophage cells (one × 10half-dozen/mL) were incubated with K1 or NK41 (1 × tenthree or 1 × 105 CFU/mL) in the absence or presence of LPS for 2 h (for NF-κB) or 20 h (for TNF-α). p-p65 and p65 (NF-κB) were measured by immunoblotting. TNF-α was measured past ELISA kit. Data values are indicated every bit hateful ± SD (n = 4). # p < 0.05 vs. Con grouping treated with Vehicle lone. *p < 0.05 vs. grouping treated with K1 and LPS solitary.

Effects of Escherichia coli K1 and Lactobacillus mucosae NK41 on the Occurrence of Cerebral Decline and Depression in Mice

To empathize whether inflammatory and anti-inflammatory gut leaner were associated with the occurrence of psychiatric disorders, we examined the furnishings of K1 and NK41 on the occurrence of psychiatric disorders cerebral decline and depression in mice in the Y-maze, elevated plus maze (EPM), forced swimming (FS), tail suspension (TS), and Banes maze tasks (Figure 2). K1 at doses of 1 × ten8 and 1 × 109 colony-forming unit (CFU)/mouse/twenty-four hour period showed pregnant depressive behaviors in EPM and FS tasks (Figures 2B,C). Memory harm-similar behaviors were observed afterwards treatment with K1 at a dose of 1 × 109 CFU/mouse/day in the Y-maze task (Figure 2D). K1 at a dose of 1 × x9 CFU/mouse/day also increased the infiltration of Iba1+ cells into the hippocampus. Furthermore, K1 caused NF-κB activation in the hippocampus, while the BDNF expression and CREB phosphorylation were suppressed (Figures 2E,F and Figures S2A,B, S3). However, NK41 handling did non bear on the cerebral decline in the Y-maze and Banes maze tasks and depressive behaviors in the FS task, even at a dose of 1 × 109 CFU/mouse/day (Figures 2G–I). NK41 at a dose of ane × 109 CFU/mouse/twenty-four hour period did not affect Iba1+ prison cell population, NF-κB activation, and BDNF expression in the hippocampus (Figures 2J,K and Figures S2C,D, S3).

www.frontiersin.org

Effigy 2. Effects of K1 and NK41 on the occurrence of neuropsychiatric disorders in mice. (A) Experimental protocol. (B) Effect of K1 on the time spent in open arms (OT) in EPM task. (C) Effect of K1 on the immobility in the forced swimming task. (D) Effect of K1 on memory damage in Y-maze task. (E) Effect of K1 on the infiltration of Iba1+ cells into the hippocampus. (F) Effect of K1 on BDNF expression, CREB phosphorylation, and NF-κB activation in the hippocampus. (G) Upshot of NK41 on the depression in EPM chore. Effect of NK41 on the cogntive decline in the Y-maze (H) and Banes maze tasks (I). (J) Issue of NK41 on the infiltration of Iba1+ cells into the hippocampus. (One thousand) Consequence of NK41 on BDNF expression, CREB phosphorylation, and NF-κB activation in the hippocampus. Mice were exposed to K1 or NK41 (C, vehicle [1% maltose]; K7, 1 × 107 CFU/mouse/day of K1; K8, one × 108 CFU/mouse/twenty-four hours of K1; K9, ane × xix CFU/mouse/twenty-four hours of K1; NK8, one × 108 CFU/mouse/day of NK41; or NK9, ane × 109 CFU/mouse/twenty-four hours of NK41) daily for v days and thereafter treated with vehicle for 5 days. Normal command group (Con), not exposed to gut leaner, was treated with 1% maltose instead of gut bacteria. Data values were indicated as mean ± SD (northward = vii). *p < 0.05 vs. Con group.

Lactobacillus mucosae NK41 Alleviated Escherichia coli K1-Induced Altered Microbiota in Mice

To empathise whether K1 and NK41 could shift gut microbiota limerick, we examined their effects on the gut microbiota composition in mice (Figures 3A–C). The estimated operational taxonomic unit (OTU) richness and Shannon's diversity index were decreased in mice treated with K1 or NK41 compared to those in control mice. To friction match the length and position of fecal bacterial 16S rRNA gene sequences, nosotros performed principal coordinate analysis. The bacterial customs of control mouse carrion was different from that of mouse ones treated with K1 or NK41. NK41 treatment similarly shifted K1-treated mouse gut microbiota to command mouse ones. At the phylum level, Proteobacteria and Actinobacteria populations showed a higher abundance in the K1-treated group compared to those in the command mouse ones, while the Bacteroidetes and Verrucomicrobioa populations showed a lower affluence. The Proteobacteria population showed a lower abundance and the Verrucomicrobia population showed a higher abundance in the NK41-treated group. Desulfovibrionaceae, Coriobacteriaceae, and Lactobacillaceae populations showed a higher abundance in the K1-treated grouping, while the Bacteroidaceae, AC160630_f, Helicobacteriaceae, Odoribacteriaceae, Prevotellaceae, and Rikenellaceae populations showed a lower abundance. Akkermansiaceae, Bacteroidaceae, and Lactobacillaceae populations showed a higher abundance in the NK1-treated group, while the Hellicobacteriaceae, Lachnospiraceae, Odoribacteriace, Prevotellaceae, and Rikenellaceae, Runinococcaceae populations showed a lower abundance (Table S1). At the genus level, Desulfovibrio, PAC001512_g, HM123997_g, Akkermansia, and PAC001472_g populations showed a higher abundance in the K1-treated grouping, while the PAC001074_g, PAC001692_g, and Oscillibacter populations showed a lower abundance. PAC001485_g, PAC000198_g, Akkermansia, and PAC001472_g populations showed a higher abundance in the NK1-treated group, while the Alistipes, PAC001692_g, Oscillibacter, Muribaculum, and PAC001112_g populations showed a lower abundance (Table S2). Furthermore, NK41 treatment shifted K1-induced gut microbiota composition to those in the control mouse i: the Proteobacteria and Bacteroidetes populations showed a lower abundance, while the Verrucomicrobia population showed a higher abundance in the K1-treated group. Linear discriminant assay (LDA) issue size (LefSe) analysis was likewise performed to confirm the different furnishings of K1 and NK41 on gut microbiota (Figure 3D and Figures S4, S5). K1-treated mice had a college abundance of Coridobacteriaceae, Bacillaceae, Gemella_f, and Clostridiaceae populations, while NK41 handling resulted in a college abundance of Bacteroidaceae, Lactobacillaceae, Eubacteriaceae, and Acholeplasmataceae populations in command mice and Staphylococcaceae, Carnobacteriaceae, and Gammaproteobacteria populations in K1-treated mice. When the fecal E. coli and L. mucosae were analyzed in the mouse feces treated with K1 and/or NK41 by using qPCR, K1 treatment significantly increased the E. coli population and decreased the Fifty. mucosae population (Figure 3E). However, NK41 treatment significantly decreased K1-induced E. coli population. Furthermore, NK41 treatment suppressed K1-induced LPS production (Figure 3F).

www.frontiersin.org

Figure iii. NK41 suppressed K1-induced altered microbiota in the feces mice. Furnishings on the composition of gut microbiota, analyzed by the pyrosequencing: phylum (A), chief coordinate assay (PCoA) plot based on weighted pairwise Fast UniFrac analysis (B), and OTUs and Shannon (C). (D) Cladogram generated by LEfSE indicating meaning differences in gut microbial (family) abundances among Con (blue), NK (purple), Grand (ruby), and KN (greenish) groups. Yellowish nodes represent species with no significant difference. The threshold logarithmic score set at 2.0 in the family level and ranked. (Due east) Effects on fecal Escherichia coli and Lactobacillus mucosae, assessed by qPCR. (F) Effects on the fecal LPS level. LPS levels were assayed past ELISA kits. (G) The affluence of bacterial genes predicted using the method of PICRUSt. The difference was analyzed using the Kruskal-Wallis H test. (H) Effects on the MUC1 and MUC2 expression in the colon. (I) Histological examination of colons, stained with alcian bluish. NK and Thou groups were exposed to Lactobacillus mucosae NK41 (1 × ten9 CFU/mouse/day of NK41) and Escherichia coli K1 (ane × 109 CFU/mouse/day) daily for v days, respectively, and thereafter treated with vehicle (1% maltose) daily for 5 days. KN group was exposed to Escherichia coli K1 (i × 10ix CFU/mouse/day) daily for v days and thereafter treated with Lactobacillus mucosae NK41 (1 × 10nine CFU/mouse/day of NK41) daily for 5 days. Con group was treated with vehicle instead of gut bacteria. Data values were indicated as mean ± SD (n = 5). # p < 0.05 vs. Con grouping. *p < 0.05 vs. One thousand group.

Oral administration of NK41 and/or K1 modified the gut bacterial cistron abundance related to the catabolism and anabolism of polysaccharides and fatty acids in the gut microbiota (Figure 3G). K1 treatment suppressed the abundance of gut bacterial genes related to iv-α-glucanotransferase, α-fucosidase 2, glucuronyl transferase, glycogen phosphorylase, UDP-2,3-diacylglucosamine hydrolase, and heparinheparan sulfate lyase while that related to β-glucosidase and pectin lyase was increased. All the same, NK41 treatment increased the K1-suppressed the abundance of gut bacterial genes related to 4-α-glucanotransferase, α-fucosidase, β-glucosidase, glycogen phosphorylase, and UDP-2,3-diacylglucosamine acyltransferase, while the β-glucosidase, pectin lyase, and urea cycle-related gut bacterial gene affluence was suppressed.

To empathize whether NK41 and K1 could touch the biosynthesis of mucins such equally MUC1 and MUC2 in the intestine, we examined their effects on the mucin expression in the colon (Figure 3H). K1 handling significantly induced the expression of MUC2, non MUC1, while NK41 treatment did not affect the expression of MUC1 and MUC2. Furthermore, NK41 treatment significantly suppressed MUC1 and MUC2 expression. When the colon of mice was stained with alcian blue, the colon of K1-treated mice was strongly stained, manifested past disrupted and shortened epithelia (Figure 3I).

Lactobacillus mucosae NK41 Alleviated Escherichia coli K1-Induced Colitis in Mice

Oral gavage of K1 treatment acquired colitis in mice (Figure 4). Thus, K1 treatment caused colon shortening and induced myeloperoxidase activity, IL-6 and TNF-α expression, and NF-κB activation in the colon (Figures 4A–Due east). Furthermore, K1 treatment increased the infiltration of NF-κB+/CD11b+ and CD11b+/CD11c+ cells (activated dendritic cells [DCs] and macrophages) into the colon (Figures 4F,K). NK41 treatment significantly reduced K1-induced colon shortening, macroscopic score, myeloperoxidase activity, IL-half dozen, TNF-α, and MUC2 expression, NF-κB activation, and infiltration of CD11b+ and/or CD11c+ cells, while MUC1 was affected. NK41 treatment likewise increased the K1-suppressed claudin-ane and occludin expression (Effigy 4E). Furthermore, NK41 alleviated K1-induced mucin layer damage in the colon.

www.frontiersin.org

Effigy 4. NK41 signiticantly suppressed K1-induced gut inflammation in mice. Furnishings on colon length (A), myeloperoxidase (MPO) activity (B), and TNF-α (C) and IL-half dozen (D) expression in the colon. (E) Effects on occludin and claudin-1 expression and NF-kB activation (Eastward) in the colon. Furnishings on CD11b+/CD11c+ (F) and NF-κB+/CD11c+ cell populations (G) in the colon. NK and Chiliad groups were exposed to Lactobacillus mucosae NK41 (1 × x9 CFU/mouse/day of NK41) and Escherichia coli K1 (1 × xix CFU/mouse/day) daily for 5 days, respectively, and thereafter treated with vehicle (1% maltose) daily for 5 days. KN group was exposed to Escherichia coli K1 (i × 10nine CFU/mouse/twenty-four hours) daily for 5 days and thereafter treated with Lactobacillus mucosae NK41 (1 × 109 CFU/mouse/solar day of NK41) daily for v days. Con group was treated with vehicle instead of gut bacteria. Colonic p65, p-p65, and β-actin were analyzed by immunoblotting. TNF-α and IL-6 levels were assayed past ELISA kits. NF-κB+, CD11b+, and CD11c+ cells were measured using a confocal microscope. Arrows indicate postive cells. Information values were indicated as mean ± SD (n = seven). # p < 0.05 vs. Con grouping. *p < 0.05 vs. K group.

Lactobacillus mucosae NK41 Suppressed Escherichia coli K1-Induced Cognitive Pass up and Low in Mice

Adjacent, we examined whether NK41 could regulate the occurrence of K1-induced psychiatric disorders in mice (Figure v). Oral gavage of K1 caused cognitive refuse in mice: its handling significantly decreased spontaneous amending in the Y-maze job, the interaction time in NOR task, and latency time in the Barnes maze chore (Figures 5B–D). Withal, NK41 treatment significantly reduced K1-induced cerebral turn down in Y-maze, novel object recognition (NOR) maze, and Barnes maze tasks to 98.1, 98.3, and 98.nine% for the control mice, respectively. Oral gavage of K1 also caused anxiety/depression: its treatment increased immobility in the FS chore to 186.1% for the command mice (Figure 5E). Furthermore, K1 treatment significantly decreased the fourth dimension spent in open arms and light compartment during the EPM and light/dark transition (LDT) tasks, respectively (Figures 5F–I). However, oral assistants of NK41 significantly reduced the fourth dimension spent in open arms (OT) in the EPM and LDT tasks to 109.5 and 98.4% for the command mice, respectively, and immobility in the FS task to 94.half dozen% for the control mice (Figure 5D).

www.frontiersin.org

Effigy v. NK41 signiticantly suppressed K1-induced neuropsychiatric disorders in mice. (A) Experimental protocol. Result on the noesis office in Y-maze (B), NOR (C), and Banes maze (D). Issue on the depressive behaviors in the forced swimming (East), EPM (F: OT, fourth dimension spent in open up arms; G: OE, open up arm entries), and light-dark transition tasks (H: TL, time spent in the light night compartment; I: NT, number of transitions into the light dark compartment). Effect on the infitration of NF-κB+/Iba1+ (J), LPS+/Iba1+ (K), and BDNF+/NeuN+ cells (L) into the hippocampus. Effects on IL-six (M), TNF-α (N), and BDNF expression, CREB phosphorylation, and NF-κB activation (O). Furnishings on the LPS (P), IL-6 (Q), and TNF-α levels (R) in the blood. NK and K groups were exposed to Lactobacillus mucosae NK41 (1 × 109 CFU/mouse/day of NK41) and Escherichia coli K1 (1 × ten9 CFU/mouse/twenty-four hour period) daily for v days, respectively, and thereafter treated with vehicle (1% maltose) daily for v days. KN grouping was exposed to Escherichia coli K1 (i × 10ix CFU/mouse/day) daily for v days and thereafter treated with Lactobacillus mucosae NK41 (1 × ten9 CFU/mouse/day of NK41) daily for v days. Con group was treated with vehicle instead of gut bacteria. TNF-α, IL-6, and LPS were assayed past ELISA. p65, p-p65, CREB, p-CREB, BDNF, and β-actin were analyzed by immunoblotting. Iba1+, NF-κB+, LPS+ and NeuN+ cells were measured using a confocal microscope). # p < 0.05 vs. Con group. *p < 0.05 vs. K group.

K1 treatment increased the infiltration of activated/phagocytic microglial (NF-κB+/Iba1+, LPS+/Iba1+) cells into the hippocampus while the BDNF+/NeuN+ cell population was reduced (Figures 5J–L). Furthermore, K1 treatment induced NF-κB activation in the hippocampus (Figure 5O). However, handling with NK41 suppressed K1-induced activation of NF-κB and infiltration of activated microglial cells and induced the K1-suppressed BDNF+/NeuN+ cells population in the hippocampus. NK41 treatment also suppressed K1-induced LPS, IL-half-dozen, and TNF-α levels in the blood (Figures 5P–R).

Discussion

Excessive exposure to stressors such as immobilization, loftier-fat diet, and pathogen infection disrupts the gut immune system and microbiota composition through the activation of the HPA and/or MGB axis, resulting in the occurrence of altered microbiota and neuropsychiatric disorders (24–26). Long-term feeding with a high-fatty nutrition causes obesity, colitis, and psychiatric disorders including cognitive refuse and feet in mice by increasing the gut Proteobacteria population (27). Intrarectal injection of TNBS causes colitis and cognitive reject by increasing the gut Proteobacteria population including E. coli and decreasing L. mucosae population (x). Exposure to immobilization stress causes colitis and anxiety/depression in mice by increasing Enterobacteriaceae including Eastward. coli and decreasing the populations of L. johnsonii and Fifty. plantarum (11). Furthermore, the oral gavage of E. coli causes colitis, cerebral reject, and depression in mice by increasing fecal and blood LPS levels (10, 11). However, handling with L. mucosae, isolated from mice, significantly alleviated E. coli-induced cerebral refuse in mice (10). Treatment with L. johnsonii, isolated from mouse feces, significantly mitigated E. coli-induced anxiety-like behaviors in mice (11). These results suggest that gut microbiota consist of a variety of bacteria including potential causative and protective bacteria regarding neuropsychiatric disorders. Withal, what kinds of gut bacteria tin cause and reduce cognitive refuse and anxiety/depression remain unclear.

In the present study, we isolated inflammatory E. coli K1, which caused NF-κB activation in macrophages, and anti-inflammatory L. mucosae NK41, which hindered K1 lysate- or LPS-induced NF-κB activation in macrophages. K1 significantly induced TNF-α expression in macrophages, while NK41 suppressed TNF-α expression. Furthermore, oral gavage of K1 caused colitis and hippocampal inflammation via alteration of gut microbiota in mice, resulting in cognitive pass up and depression/anxiety. Exposure to K1 caused feet/depression likewise every bit gut microbiota amending that had a higher abundance of Proteobacteria and Actinobacteria populations and a lower affluence of Bacteroidetes and Verrucomicrobioa populations, while these bacterial alterations and cognitive reject and anxiety/low were alleviated by NK41 treatment. Treatment with NK41 showed a higher abundance of Lactobacillaceae and Eubacteriaceae, and Bacteroidaceae populations. These results advise that the overgrowth of Proteobacteria including East. coli K1 in the intestine past exposure to endogenous and exogenous stressors may induce cognitive decline and anxiety/depression. Oral administration of NK41 showed a lower affluence of K1-induced Proteobacteria and Enterobacteriaceae populations and LPS production in the gut microbiota of mice. These results suggest that the induction of Lactobacillaceae and Bacteroidaceae growth including L. mucosae NK41 can alleviate gut microbiota-mediated cognitive pass up and anxiety/depression. Furthermore, NK41 treatment significantly inhibited K1-induced colon shortening, colonic myeloperoxidase activity, and infiltration of DCs and macrophages into the colon. Furthermore, NK41 suppressed the K1-induced expression of TNF-α and IL-6 and activation of NF-κB in the colon and increased the expression of tight junction proteins claudin-1 and occludin. NK41 treatment lowered LPS levels in the carrion and blood. Jang et al. reported that fecal transplantation of IS-treated mouse feces, oral gavage of the gram-negative E. coli independent in information technology, and peritoneal injection of its LPS caused colitis: they induced myeloperoxidase activity and suppressed tight junction poly peptide expression in the colon (11). They also found that E. coli treatment increased the absorption of orally administered fluorescein isothiocyanate-labeled dextran into the blood of mice. These results suggest that Due east. coli K1 tin induce the excessive LPS production in gut microbiota, which leads to gut inflammation, resulting in the elevation of claret LPS by increasing gut membrane permeability. We too institute that NK41 restrained K1-induced blood TNF-α and IL-6 levels in mice. NK41 as well reduced K1-induced activated/phagocytic microglial (Iba1+) jail cell populations in the hippocampus. Furthermore, NK41 suppressed K1-induced TNF-α, IL-6, and MUC2 expression and NF-κB activation in the colon and increased K1-suppressed BDNF expression and CREB phosphorylation in the hippocampus. Moreover, NK41 treatment simultaneously improved K1-induced cognitive refuse and depression in mice.

IL-6, TNF-α, and corticosterone are highly expressed in patients with anxiety and depression (28, 29). Excessive IL-vi and corticostrone expression was increased by stressors such as immobilization and pathogen infection via the activation of the HPA axis. Treatment with therapeutic drugs for psychiatric disorders reduces blood IL-six and corticosterone levels, increases BDNF expression, and alleviates neuropsychiatric disorders (10, 11, 30). Treatment with corticosterone suppresses BDNF expression in SH-SY5Y cells in vitro and in mice. BDNF induces de novo synthesis of proteins such as synaptophysin and drebrin, which are involved in neural and synaptic plasticity (31, 32). Additionally, systemic exposure to LPS activates microglia and increased expression of pro-inflammatory cytokines in the brain of mice (x, 11). We also found that oral gavage of E. coli in mice caused endotoxemia and hippocampal inflammation. Therefore, E. coli-induced endotoxemia may cause inflammation in the brain including the hippocampus. LPS causes systemic neuroinflammation, resulting in retentiveness impairment by the modulation of NF-κB-mediated BDNF/CREB expression (xv, 33). Therefore, regulating endotoxemia-mediated corticosterone and BDNF expression can exist useful for the handling of psychiatric disorders.

We too found that Due east. coli K1 treatment significantly increased the expression of MUC2, not MUC1, and mucin layer in the colon while the colonic epithelia were disrupted. K1 treatment likewise increased the gut bacterial factor abundance related to polysaccharide breaking and biosynthesis. MUC1 and MUC2 are increased in patients and mice with inflammation (34, 35). LPS from gram-negative Pseudomonas aeruginosa upregulates MUC2 transcription through activation of NF-κB (34). E. coli K1 treatment caused NF-κB activation in the gut and encephalon and increased blood LPS levels, as previously reported in mice treated with IS or LPS (10, xi). These results suggest that, although the biosynthesis of mucins such as MUC2 is accelerated, the overgrowth of Due east. coli can increase gut bacterial LPS product and crusade colitis that the speed of mucosal repair does not be overcome. Gut bacteria-induced inflammation can cause systemic inflammation including hippocampal inflammation due to the increase of LPS in the claret via increased gut membrane permeability. L. mucosae NK41 restrained altered microbiota-induced bacterial LPS product, blood LPS levels, and hippocampal inflammation and brought well-nigh BDNF expression and CREB expression. Neuro-inflammation decreases the expression of BDNF in the mouse hippocampus, and reduced hippocampal BDNF is associated with memory and learning deficiencies (33, 36). BDNF is influenced past the gut microbiota (33). However, oral administration of L. plantarum and Bifidobacterium infantis reduced anxiety-like beliefs past restoring noradrenaline levels and protecting gut microbiota alteration, respectively (37, 38). Bifidobacterium adolescentis IM38, a human gut bacterium, inhibited IS-induced feet by regulating the GABAA receptor (39). L. johnsonii, a commensal gut bacterium of mice, suppressed IS-induced anxiety in mice past hindering gut microbiota LPS production (11). Bifidobacterium breve strain A1 prevents cognitive impairment in Alzheimer's disease (40). L. plantarum C29 improved cerebral function in mice and patient with mild cognitive pass up by regulating NF-κB activation and inducing BDNF expression (22, 41). L. plantarum 299v improves cognitive functions in patients with major depression by decreasing kynurenine concentration (42). Lactobacillus rhamnosus (JB-one) regulates emotional behavior by the consecration of GABA(Aα2) expression in the hippocampus (43). Moreover, the induction of altered microbiota by stressors such as antibiotics caused cognitive function via the MGB axis (2, 11). These results propose that NK41 can reduce cognitive decline and low by suppressing gastrointestinal and hippocampal inflammation via the regulation of contradistinct microbiota and bacterial LPS production. Furthermore, the regulation of K1 and NK41 on the gut inflammation and altered microbiota should be closely associated with the occurrence of neuropsychiatric disorders. Nevertheless, further studies on the detailed memory impairment-ameliorating mechanism of NK41 and pathogenic mechanism of K1 are necessary.

In Decision, gut microbiota, which consist of inflammatory and anti-inflammatory bacteria in humans and animals, are bidirectionally continued to the brain: the overgrowth of inflammatory leaner such as E. coli in the alimentary canal tin cause psychiatric disorders with gut inflammation and the superiority of anti-inflammatory bacteria such as L. mucosae tin can convalesce psychiatric disorders with the attenuation of altered microbiota (Figure 6).

www.frontiersin.org

Effigy half dozen. Interplay between Escherichia coli K1 and Lactobacillus mucosae NK41 on the occurrence of neuropsychotric disorders and contradistinct microbiota.

Methods

Civilisation of Escherichia coli K1 and Lactobacillus mucosae NK41

K1 and NK41 were selected from good for you human being gut microbiota according to the method of Jang et al. (10). These bacteria were cultured in general media such as general anaerobic medium (GAM) and MRS (BD, Franklin Lakes, NJ). Cultured bacteria were collected by centrifugation (v,000 yard, xx min, 4°C) and done with saline. The collected cells were suspended in saline (for in vitro experiments) or 1% maltose (for in vivo experiments).

To decide the dosage of these bacteria in the in vivo experiment, K1 at doses of 1 × x7, i × x8, and 1 × tenix CFU/mouse/day was orally gavaged for 5 days in mice and depression-similar behaviors were measured in the EPM chore, equally the previously reported (44). NK41 at doses of 1 × 108 and 1 × 109 CFU/mouse/solar day was orally gavaged for 5 days in K1-treated mice and behaviors were measured in the Y-maze and elevated plus maze (EPM) tasks, as previously reported (10).

Isolation and Culture of Peritoneal Macrophages

Macrophages were isolated from the peritoneal crenel of mice co-ordinate to the method of Jang et al. (45). Nerveless cells were suspended in RPMI 1640 containing 10% FBS and ane% antibiotics (RFA), seeded in vi-well plate, incubated at 37°C for a 24-hour interval, and washed with RFA. Attached cells (1 × 10half-dozen cells/well) were used every bit macrophages. To measure anti-inflammatory effect of gut leaner, macrophage was treated with LPS (100 ng/mL) in the absenteeism or presence of gut bacteria (ane × 10iii or ane × 105 CFU/mL) for 90 min (for p65 and p-p65) or twenty h (for TNF-α).

Animals

Specific pathogen-free male C57BL/6J mice (nineteen–21 chiliad, 5 weeks-old) were purchased from Orient Inc. (Seoul, South Korea). All mice were housed in wire cages (3–four mice per cage) at 20–22°C, fifty ± ten% humidity, and 12-h light/dark cycle (lights on from 07:30 to 19:xxx), fed standard laboratory chow and h2o ad libitum. Mice were used in the experiments later the acclimation for 1 week. All animate being experiments were canonical by The Committee for the Care and Utilize of Laboratory Animals in Kyung Hee University and performed in accordance with The Kyung Hee Academy Guidelines for Laboratory Animals Care and Usage (IACUC No. KHUASP(SE)-18089).

To examine the effects of K1 and NK41 on the occurrence and evolution of psychiatric disorders cerebral turn down and anxiety/low, mice were randomly divided into iii groups (Control, EC, and NK groups). Each group consisted of vii mice. Mice (EC and NK groups) were orally gavaged with the K1 intermission (1 × ten9 CFU, suspended in 100 μL of ane% maltose) once a day for 5 days according to the method of Jang et al. (x). The command group was treated with 1% maltose instead of the K1 break and NK41 (for the NK group, 1 × 10nine CFU/mouse/day) or vehicle (for the command and EC groups) was orally administered once a day for five days from 24 h afterwards treatment with K1 pause. Behavioral tasks were performed 24 h later on NK41 treatment. Mice were then anesthetized by CO2 asphyxiation, followed by blood depict for biochemical assays. The colon and hippocampus were removed. The specimens were stored at −eighty°C until use in an ELISA assay, immunoblotting, and enzyme activity assay.

Cognitive and Depressive Behavioral Tasks

To evaluate the anti-depressive effects of gut leaner in mice, the EPM job was performed in the plus-maze apparatus (consisting of two open [30 × 7 cm] and two enclosed arms [30 × 7 cm] with 20-cm-high walls extending from a central platform [seven × 7 cm] on a single central support to a acme of 60 cm above the floor) for 5 min according to the method of Jang et al. (44). The tail suspension (TS) job was measured according to the method of Dunn and Swiergiel (46). Mice were suspended on the edge of a tabular array 30 cm above the floor by taping ane cm from the tail tip. Immobility time was measured for five min. Mice were judged to be immobile, when they did not motility and hanged passively. The forced swimming (FS) chore was performed in a circular transparent plastic jar (twenty × xl cm3) containing fresh water (25°C) to a height of 25 cm. Immobility time was measured during v min. Mice were judged to be immobile, when they remained floating in the water without struggling.

To evaluate the cognitive effects of gut bacteria in mice, get-go Y-maze was performed in a three-arm horizontal maze (40 cm long and 3 cm wide with 12-cm-high walls) (10). A mouse was initially placed within one arm and the sequence and number of arm entries were manually recorded for viii min. A spontaneous (bodily) alternation was defined as entries into all 3 artillery on sequent choices and was calculated as the ratio (%) of actual to possible alternations. A novel object recognition (NOR) chore was performed in the appliance consisting of a dark-open field box (45 × 45 × 50 cm) according to the method of Lee et al. (47). For the showtime trial, a mouse was placed in the box containing ii identical objects and the frequency of touching each object was recorded for 10 min. In the second trial conducted 24 h later the first trial, a mouse was placed in the box containing one of the erstwhile objects used in the first trial and a new object. Novel object recognition was calculated every bit the ratio of the number of times touching the new object to the sum of the touching frequencies. The Barnes maze was performed in the apparatus consisting of a round platform (89 cm in diameter) with 20 holes (five cm in diameter) situated evenly effectually the perimeter and an escape box located under only one of the holes below the platform according to the method of Patil et al. (48). The training/acquisition stage finished afterwards mouse entered the escape box or afterwards a maximum examination duration (5 min), following which mouse was allowed to stay in the box for 30 s and then moved to the cage. If mouse failed to find the escape box inside 5 min, it was led to the escape box. Mice were given two trials per twenty-four hours for five sequent days.

Assay of Myeloperoxidase Activeness

Myeloperoxidase activity in colon was assayed, every bit previously described (10). Colon tissues were homogenized with cold RIPA lysis buffer and centrifuged at 10,000 g for ten min (11). The supernatant was used as a crude enzyme solution. An aliquot of the supernatant was added in the reaction mixture containing 0.03% hydrogen peroxide and i.half-dozen mM tetramethylbenzidine and measured the absorbance at 650 nm time over 5 min. Activity was defined every bit the quantity degrading 1 μmol/mL of peroxide.

Enzyme-Linked Immunosorbent Analysis (ELISA) and Immunoblotting

For the cytokine assay, the supernatant of the hippocampus and colon homogenate and plasma was transferred to 96-well ELISA plates according to the method of Jang et al. (11). TNF-α concentrations were determined using commercial ELISA kits (Ebioscience, Atlanta, GA). For the immunoblotting analysis, the supernatants of tissue homogenates were resolved past sodium dodecyl sulfate polyacrylamide gel electrophoresis, transferred to nitrocellulose, and immunoblotted using diverse primary antibodies (Cell Signaling Applied science, Beverly, MA) (11). Ring densities were analyzed using the automatic imaging assay organisation, Quantity One (Bio-Rad, Hercules, CA).

Immunohistochemistry

Mice were trans-cardiacally perfused with 4% paraformaldehyde. Their brains and colons were mail service-fixed with iv% paraformaldehyde for 4 h, cytoprotected in xxx% sucrose solution, freezed, cut using a cryostat (Leica, Nussloch, Federal republic of germany), and immunostained according to the method of Jang et al. (xi). Briefly, the sections were washed with phosphate buffered saline, blocked with normal serum, incubated with antibodies for Iba1 (1:200, Abcam), LPS (1:200, Millipore), NF-κB (ane:100, Cell Signaling), CD11b (one:200, Abcam), CD11c (1:200, Abcam), and/or NeuN (ane:200, Millipore) overnight, and treated with the secondary antibodies for 2 h. Secondary antibodies conjugated with with Alexa Fluor 488 (ane:200, Invitrogen) or Alexa Fluor 594 (1:200, Invitrogen) were then treated to visualize. Nuclei were stained with four′,half dozen-diamidino-2-phenylindole, dilactate (DAPI, Sigma). Immunostained tissue slices were scanned with a confocal laser microscope.

Microbiota Sequencing

Genomic DNA was extracted from the fresh stools of five mice (not trans-cardiacally perfused with 4% paraformaldehyde for brain and colon tissue sections) using a commercial Dna isolation kit (QIAamp DNA stool mini kit), as previously reported (eleven, 47). Briefly, genomic DNA was extracted from the fresh stools of mice using a commercial DNA isolation kit (QIAamp DNA stool mini kit). Amplification of the genomic Deoxyribonucleic acid was performed using barcoded primers, which targeted the V4 region of the bacterial 16S rRNA factor, described in Supplementary Textile. Sequencing for each amplicon was performed using Illumina iSeq 100 (San Diego, CA). Predictive functional genes were analyzed using the phylogenetic investigation of communities past reconstruction of unobserved states (PICRUSt) (49). Linear discriminant analysis (LDA) analysis and cladograms were adult on family level data using LDA outcome size (LefSe) on Milky way platform (https://huttenhower.sph.harvard.edu/galaxy/) (50). Pyrosequencing reads have been deposited in the NCBI'south short read annal nether accession number PRJNA507690.

Quantitative Real Time–Polymerase Chain Reaction (qPCR)

Existent time PCRs for MUC1, MUC2, and GAPDH were performed on the Rotor-Gene Q® using Deoxyribonucleic acid polymerase and SYBR Light-green I (a reaction book, xx μL), as previously reported (11). Primers for qPCR are described in Table S3. The normalized expression of each target gene, equally for GAPDH, was calculated for all samples using Microsoft Excel.

qPCRs for Eastward. coli, L. mucosae, and 16S rRNA were performed on the Rotor-Cistron Q® using DNA polymerase and SYBR Greenish I (a reaction volume, 20 μL), as previously reported (11). Thermal cycling was performed at 95°C for xxx s by 42 cycles of denaturation at 95°C for 5 s and amplification 72°C for 30 s. Expression of genes were computed relatively to 16S rDNA, using Microsoft Excel. Primers for qPCR are described in Table S4. The normalized expression of each target cistron, equally for GAPDH, was calculated for all samples using Microsoft Excel.

LPS Assay

Blood and fecal endotoxin contents were adamant using the diazo-coupled limulus amoebocyte lysate (LAL) assays (Greatcoat Cod Inc., E. Falmouth, MA) according to the method of Kim et al. [(51), Supplementary Material].

Statistical Assay

Experimental results are indicated as mean ± standard deviation (SD), and were statistically analyzed using one-style analysis of variance followed by a Duncan multiple range test (p < 0.05). Pupil'due south t-tests were used to compare 2 groups. All p-values are indicated in Table S5.

Data Availability Statement

The data generated for this study are bachelor on asking to the respective author.

Ethics Statement

This study was performed according to the recommendation of the Kyung Hee University Creature Ethics Committee. The animal experimental protocol was reviewed and canonical past the institutional creature utilize committee [KHU IACUC No. KHUASP(SE)-18089] and human gut leaner collection protocol was reviewed and canonical by the institutional review board of Kyung Hee Academy Hospital (KMC IRB No KHUH 0922-08-A1). The patients/participants provided their written informed consent to participate in this written report.

Writer Contributions

J-KK and D-HK conceived and designed experiments and wrote the manuscript. J-KK, K-EL, S-AL, H-MJ, and D-HK analyzed information. J-KK, K-EL, and H-MJ contributed reagents, materials, and analysis tools.

Funding

This present study was supported past the Medical Research Program through the National Enquiry Foundation of Korea (NRF), funded past the Ministry building of Science and ICT (NRF- 2022R1A5A2014768).

Disharmonize of Interest

The authors declare that the research was conducted in the absence of whatsoever commercial or financial relationships that could be construed every bit a potential conflict of involvement.

Supplementary Material

The Supplementary Cloth for this article tin be constitute online at: https://www.frontiersin.org/manufactures/10.3389/fimmu.2020.00273/full#supplementary-material

References

1. Xu West, Anderson ME, Kotarba AE, Davis J, Robinson JK, Van Nostrand WE. Cerebral microvascular rather than parenchymal amyloid-β protein pathology promotes early cognitive impairment in transgenic mice. J Alzheimers Dis. (2014) 38:621–32. doi: 10.3233/JAD-130758

PubMed Abstract | CrossRef Full Text | Google Scholar

three. Neufeld KM, Kang Due north, Bienenstock J, Foster JA. Reduced anxiety-like behavior and central neurochemical change in germ-costless mice. Neurogastroenterol Motil. (2011) 23:255–64. doi: 10.1111/j.1365-2982.2010.01620.10

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Silverman MN, Sternberg EM. Glucocorticoid regulation of inflammation and its functional correlates: from HPA centrality to glucocorticoid receptor dysfunction. Ann N Y Acad Sci. (2012) 1261:55–63. doi: 10.1111/j.1749-6632.2012.06633.10

PubMed Abstruse | CrossRef Full Text | Google Scholar

five. Smith SM, Vale WW. The part of the hypothalamic-pituitary-adrenal axis in neuroendocrine responses to stress. Dialogues Clin Neurosci. (2006) eight:383–95.

PubMed Abstract | Google Scholar

vii. Kelly JR, Borre Y, O' Brien C, Patterson E, El Aidy Southward, Deane J, et al. Transferring the blues: depression-associated gut microbiota induces neurobehavioural changes in the rat. J. Psychiatr. Res. (2016) 82:109–18. doi: 10.1016/j.jpsychires.2016.07.019

PubMed Abstract | CrossRef Total Text | Google Scholar

8. Singh RK, Chang HW, Yan D, Lee KM, Ucmak D, Wong Grand, et al. Influence of nutrition on the gut microbiome and implications for human health. J Transl Med. (2017) 15:73. doi: 10.1186/s12967-017-1175-y

PubMed Abstruse | CrossRef Total Text | Google Scholar

9. Topping DL, Clifton PM. Short-chain fatty acids and human colonic office: roles of resistant starch and nonstarch polysaccharides. Physiol Rev. (2001) 81:1031–64. doi: 10.1152/physrev.2001.81.3.1031

PubMed Abstruse | CrossRef Full Text | Google Scholar

ten. Jang SE, Lim SM, Jeong JJ, Jang HM, Lee HJ, Han MJ, et al. Gastrointestinal inflammation by gut microbiota disturbance induces retentiveness impairment in mice. Mucosal Immunol. (2018) 11:369–79. doi: 10.1038/mi.2017.49

PubMed Abstract | CrossRef Full Text | Google Scholar

eleven. Jang HM, Lee KE, Lee HJ, Kim DH. Immobilization stress-induced Escherichia coli causes anxiety by inducing NF-κB activation through gut microbiota disturbance. Sci Rep. (2018) viii:13897. doi: 10.1038/s41598-018-31764-0

PubMed Abstruse | CrossRef Total Text | Google Scholar

13. Buttini M, Limonta S, Boddeke HW. Peripheral assistants of lipopolysaccharide induces activation of microglial cells in rat brain. Neurochem Int. (1996) 29:25–35. doi: 10.1016/0197-0186(95)00141-vii

PubMed Abstruse | CrossRef Total Text | Google Scholar

14. Hasegawa-Ishii S, Inaba M, Umegaki H, Unno K, Wakabayashi One thousand, Shimada A. Endotoxemia-induced cytokine-mediated responses of hippocampal astrocytes transmitted by cells of the encephalon-immune interface. Sci Rep. (2016) half dozen:25457. doi: 10.1038/srep25457

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Goel R, Bhat SA, Hanif K, Nath C, Shukla R. Angiotensin 2 receptor blockers attenuate lipopolysaccharide-induced memory impairment past modulation of NF-κB-mediated BDNF/CREB expression and apoptosis in spontaneously hypertensive rats. Mol Neurobiol. (2018) 55:1725–39. doi: x.1007/s12035-017-0450-5

PubMed Abstract | CrossRef Total Text | Google Scholar

18. Onta T, Sashida M, Fujii North, Sugawara S, Rikiishi H, Kumagai K, et al. Induction of acute arthritis in mice by peptidoglycan derived from gram-positive leaner and its possible role in cytokine production. Microbiol Immunol. (1993) 37:573–82. doi: 10.1111/j.1348-0421.1993.tb01679.x

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Zhang G, Ghosh S. Molecular mechanisms of NF-kappaB activation induced by bacterial lipopolysaccharide through Toll-like receptors. J Endotoxin Res. (2000) 6:453–7. doi: x.1179/096805100101532414

PubMed Abstruse | CrossRef Full Text | Google Scholar

21. Lee HJ, Lim SM, Ko DB, Jeong JJ, Hwang YH, Kim DH. Soyasapogenol B and genistein attenuate lipopolysaccharide-induced retentiveness harm in mice by the modulation of NF-κB-mediated BDNF Expression. J Agric Nutrient Chem. (2017) 65:6877–85. doi: x.1021/acs.jafc.7b02569

PubMed Abstract | CrossRef Total Text | Google Scholar

22. Lee HJ, Jeong JJ, Han MJ, Kim DH. Lactobacillus plantarum C29 alleviates TNBS-induced memory damage in mice. J Microbiol Biotechnol. (2018) 28:175–ix. doi: 10.4014/jmb.1709.09042

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Wiley NC, Dinan TG, Ross RP, Stanton C, Clarke Thousand, Cryan JF. The microbiota-gut-brain axis equally a central regulator of neural office and the stress response: Implications for human and animal health. J Anim Sci. (2017) 95:3225–46. doi: 10.2527/jas2016.1256

PubMed Abstract | CrossRef Total Text | Google Scholar

27. Jeong MY, Jang HM, Kim DH. High-fat diet causes psychiatric disorders in mice by increasing Proteobacteria population. Neurosci Lett. (2019) 698:51–7. doi: ten.1016/j.neulet.2019.01.006

PubMed Abstruse | CrossRef Total Text | Google Scholar

28. Brebner K, Hayley S, Zacharko R, Merali Z, Anisman H. Synergistic effects of interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha: central monoamine, corticosterone, and behavioral variations. Neuropsychopharmacology. (2000) 22:566–80. doi: x.1016/S0893-133X(99)00166-9

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Frühauf-Perez PK, Temp FR, Pillat MM, Signor C, Wendel AL, Ulrich H, et al. Spermine protects from LPS-induced memory deficit via BDNF and TrkB activation. Neurobiol Larn Mem. (2018) 149:135–43. doi: x.1016/j.nlm.2018.02.012

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Aoki C, Kojima N, Sabaliauskas North, Shah Fifty, Ahmed Thursday, Oakford J, et al. Drebrin a knockout eliminates the rapid form of homeostatic synaptic plasticity at excitatory synapses of intact adult cerebral cortex. J Comp Neurol. (2009) 517:105–21. doi: 10.1002/cne.22137

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Janz R, Südhof TC, Hammer RE, Unni V, Siegelbaum SA, Bolshakov VY. Essential roles in synaptic plasticity for synaptogyrin I and synaptophysin I. Neuron. (1999) 24:687–700. doi: 10.1016/S0896-6273(00)81122-eight

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Li JD, Dohrman AF, Gallup M, Miyata S, Gum JR, Kim YS, et al. Transcriptional activation of mucin by Pseudomonas aeruginosa lipopolysaccharide in the pathogenesis of cystic fibrosis lung disease. Proc Natl Acad Sci U.S.A. (1997) 94:967–72. doi: ten.1073/pnas.94.three.967

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Periasamy S, Lin CH, Nagarajan B, Sankaranarayanan NV, Desai UR, Liu MY. Mucoadhesive office of tamarind xyloglucan on inflammation attenuates ulcerative colitis. J Funct Foods. (2018) 47:one–10. doi: ten.1016/j.jff.2018.05.035

PubMed Abstruse | CrossRef Full Text | Google Scholar

36. Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J, et al. The intestinal microbiota bear upon central levels of brain-derived neurotropic gene and beliefs in mice. Gastroenterology. (2011) 141:599–609. doi: 10.1053/j.gastro.2011.04.052

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Davis DJ, Doerr HM, Grzelak AK, Busi SB, Jasarevic E, Ericsson Air-conditioning, et al. Lactobacillus plantarum attenuates anxiety-related beliefs and protects confronting stress-induced dysbiosis in adult zebrafish. Sci Rep. (2016) half dozen:33726. doi: 10.1038/srep33726

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Desbonnet L, Garrett L, Clarke G, Kiely B, Cryan JF, Dinan TG. Effects of the probiotic Bifidobacterium infantis in the maternal separation model of depression. Neuroscience. (2010) 170:1179–88. doi: ten.1016/j.neuroscience.2010.08.005

PubMed Abstruse | CrossRef Total Text | Google Scholar

39. Jang HM, Jang SE, Han MJ, Kim DH. Anxiolytic-like event of Bifidobacterium adolescentis IM38 in mice with or without immobilisation stress. Benef Microbes. (2018) 9:123–32. doi: 10.3920/BM2016.0226

CrossRef Total Text | Google Scholar

forty. Kobayashi Y, Sugahara H, Shimada K, Mitsuyama E, Kuhara T, Yasuoka A, et al. Therapeutic potential of Bifidobacterium breve strain A1 for preventing cognitive impairment in Alzheimer's disease. Sci Rep. (2017) seven:13510. doi: 10.1038/s41598-017-13368-2

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Hwang YH, Park S, Paik JW, Chae SW, Kim DH, Jeong DG, et al. Efficacy and safety of Lactobacillus plantarum C29-fermented soybean (DW2009) in individuals with mild cerebral impairment: a 12-week, multi-center, randomized, double-blind, placebo-controlled clinical trial. Nutrients. (2019) 11:E305. doi: 10.3390/nu11020305

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Rudzki L, Ostrowska 50, Pawlak D, Małus A, Pawlak Yard, Waszkiewicz N, et al. Probiotic Lactobacillus plantarum 299v decreases kynurenine concentration and improves cognitive functions in patients with major low: a double-blind, randomized, placebo controlled report. Psychoneuroendocrinology. (2019) 100:213–22. doi: 10.1016/j.psyneuen.2018.10.010

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM, Dinan TG, et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U.South.A. (2011) 108:16050–v. doi: 10.1073/pnas.1102999108

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Jang HM, Han SK, Kim JK, Oh SJ, Jang HB, Kim DH. Lactobacillus sakei alleviates high-fat-diet-induced obesity and anxiety in mice by inducing AMPK activation and SIRT1 expression and inhibiting gut microbiota-mediated NF-κB activation. Mol Nutr Nutrient Res. (2019) 63:e1800978. doi: x.1002/mnfr.201800978

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Jang SE, Han MJ, Kim SY, Kim DH. Lactobacillus plantarum CLP-0611 ameliorates colitis in mice by polarizing M1 to M2-similar macrophages. Int Immunopharmacol. (2014) 21:186–92. doi: ten.1016/j.intimp.2014.04.021

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Dunn AJ, Swiergiel AH. Effects of interleukin-1 and endotoxin in the forced swim and tail suspension tests in mice. Pharmacol Biochem Behav. (2005) 81:688–93. doi: 10.1016/j.pbb.2005.04.019

PubMed Abstruse | CrossRef Full Text | Google Scholar

47. Lee HJ, Hwang YH, Kim DH. Lactobacillus plantarum C29-fermented soybean (DW2009) alleviates retention impairment in 5XFAD transgenic mice by regulating microglia activation and but microbiota composition. Mol Nutr Food Res. (2018) 62:e1800359. doi: 10.1002/mnfr.201800359

PubMed Abstract | CrossRef Total Text | Google Scholar

48. Patil SS, Sunyer B, Hoger H, Lubec G. Evaluation of spatial memory of C57BL/6J and CD1 mice in the Barnes maze, the multiple T-maze and in the Morris water maze. Behav Brain Res. (2009) 198:58–68. doi: x.1016/j.bbr.2008.10.029

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Langille MG, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, et al. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotechnol. (2013) 31:814–21. doi: x.1038/nbt.2676

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, et al. Metagenomic biomarker discovery and explanation. Genome Biol. (2011) 12:R60. doi: 10.1186/gb-2011-12-6-r60

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Kim KA, Gu W, Lee IA, Joh EH, Kim DH. Loftier fat diet-induced gut microbiota exacerbates inflammation and obesity in mice via the TLR4 signaling pathway. PLoS ONE. (2012) vii:e47713. doi: x.1371/journal.pone.0047713

PubMed Abstract | CrossRef Full Text | Google Scholar

Source: https://www.frontiersin.org/articles/10.3389/fimmu.2020.00273/full

0 Response to "How To Increase E Coli In Gut"

Post a Comment

Iklan Atas Artikel

Iklan Tengah Artikel 1

Iklan Tengah Artikel 2

Iklan Bawah Artikel