Articles

  • KSBNS 2024

Article

Original Article

Exp Neurobiol 2023; 32(5): 328-342

Published online October 31, 2023

https://doi.org/10.5607/en23024

© The Korean Society for Brain and Neural Sciences

Extracellular Vesicles Released by Lactobacillus paracasei Mitigate Stress-induced Transcriptional Changes and Depression-like Behavior in Mice

Hyejin Kwon1, Eun-Hwa Lee1, Juli Choi1, Jin-Young Park1, Yoon-Keun Kim2* and Pyung-Lim Han1*

1Department of Brain and Cognitive Sciences, Scranton College, Ewha Womans University, Seoul 03760,
2MD Healthcare Inc., Seoul 03923, Korea

Correspondence to: *To whom correspondence should be addressed.
Yoon-Keun Kim, TEL: 82-70-7812-8065, FAX: 82-2-2655-0768
e-mail: ykkim@mdhc.kr
Pyung-Lim Han, TEL: 82-2-3277-4130, FAX: 82-2-3277-3419
e-mail: plhan@ewha.ac.kr

Received: July 19, 2023; Revised: September 4, 2023; Accepted: October 12, 2023

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.

Various probiotic strains have been reported to affect emotional behavior. However, the underlying mechanisms by which specific probiotic strains change brain function are not clearly understood. Here, we report that extracellular vesicles derived from Lactobacillus paracasei (Lpc-EV) have an ability to produce genome-wide changes against glucocorticoid (GC)-induced transcriptional responses in HT22 hippocampal neuronal cells. Genome-wide analysis using microarray assay followed by Rank-Rank Hypergeometric Overlap (RRHO) method leads to identify the top 20%-ranked 1,754 genes up- or down-regulated following GC treatment and their altered expressions are reversed by Lpc-EV in HT22 cells. Serial k-means clustering combined with Gene Ontology enrichment analyses indicate that the identified genes can be grouped into multiple functional clusters that contain functional modules of “responses to stress or steroid hormones”, “histone modification”, and “regulating MAPK signaling pathways”. While all the selected genes respond to GC and Lpc-EV at certain levels, the present study focuses on the clusters that contain Mkp-1, Fkbp5, and Mecp2, the genes characterized to respond to GC and Lpc-EV in opposite directions in HT22 cells. A translational study indicates that the expression levels of Mkp-1, Fkbp5, and Mecp2 are changed in the hippocampus of mice exposed to chronic stress in the same directions as those following GC treatment in HT22 cells, whereas Lpc-EV treatment restored stress-induced changes of those factors, and alleviated stress-induced depressive-like behavior. These results suggest that Lpc-EV cargo contains bioactive components that directly induce genome-wide transcriptional responses against GC-induced transcriptional and behavioral changes.


Keywords: Extracellular vesicles, Lactobacillus, Transcriptional responses, Stress-activated genes

Several lines of evidence suggest that gut microbiota play an important role in mental health [1-3]. Patients with depression have altered gut microflora. Conversely, supplementation with certain probiotics in patients with depression produces anti-depressant effects [2-8]. Mice exposed to chronic stress, which causes depressive-like behavior, have altered gut microbiota composition including a reduction of Lactobacillus [9, 10]. In contrast, administration of certain Lactobacillus species (Lactobacillus spp.), such as Lactobacillus plantarum, Lactobacillus casei, Lactobacillus reuteri, or Lactobacillus-derived factors, improve stress-induced depressive-like behaviors [8-16], suggesting that certain Lactobacillus bacteria have a potential to confer neurobehavioral effects by modification of stress-induced changes in the brain. Lactobacillus paracasei is a member of the genus Lactobacillus [17]. Recent reports show that supplementation of live or heat-killed Lactobacillus paracasei produced anti-depressive-like effects in mice chronically treated with corticosterone [18], in rats treated with maternal separation [19], and in mice exposed to chronic unpredictable stress [20]. Thus, different Lactobacillus species produce anti-depressive effects, although the mechanisms by which different Lactobacillus species modify brain function are not clearly understood.

Several mechanisms have been proposed for how probiotics affect brain function: a list of studies show that probiotics release bacterial metabolites, which stimulate residual gut immune cells to release neuroactive cytokines [8, 21-25]. Recent studies, including our own, have reported that bacteria-derived extracellular vesicles (EV) mediate the beneficial effects of probiotics [26-30]. Lactobacillus plantarum-derived EVs have an ability to restore glucocorticoid-induced down-regulation of Bdnf and Nt4/5, via up-regulation of Sirt1 in cultured hippocampal HT22 cells and produce anti-depressive effects in mice with stress-induced depression [10, 11]. However, the mechanisms by which different Lactobacillus strains modify brain function are not clearly understood.

In the present study, we investigated whether Lactobacillus paracasei-derived EV (Lpc-EV) produces genome-wide transcriptional responses in HT22 neuronal cells in vitro, and whether Lpc-EV treatment produces anti-depressive effects in a stress-induced animal model of depression.

Animals

Seven-week-old male C57BL6 mice were purchased from Daehan BioLink (Eumsung, Chungbuk, Republic of Korea). They were housed in pairs in standard clear plastic cages in a temperature (23~24°C)- and humidity (50~60%)-controlled environment under a 12-h light/dark cycle (lights on from 07:00 to 19:00), and were allowed ad libitum access to water and food.

Chronic stress was induced using restraints as described previously [31, 32]. Briefly, mice were individually restrained using a well-ventilated, 50-ml polypropylene conical tube for 2-h daily for 14 days. Control mice housed in pairs were maintained in their home cages without disturbance. Lpc-EV was administered to mice via the intraperitoneal route, each dose being 6 μg of Lpc-EV in 100 μl of injection volume, as described previously [11].

Mice were handled in accordance with the animal care guidelines of Ewha Womans University. The experimental procedures for treatment with restraint and EVs were approved by the Ewha Womans University Animal Care and Use Committee (IACUC 15-012).

Preparation of EVs from Lactobacillus paracasei

Bacterial culture and EV isolation were carried out as described previously [26]. In brief, Lactobacillus paracasei was cultured in MRS broth (MBCell, Seoul, Republic of Korea) for 18 h at 37°C with gentle shaking (150 rpm). The bacterial culture was centrifugated at 10,000×g for 20 min, and the supernatant was collected and passed through a 0.22-μm bottle-top filter (Corning, NY, USA) to remove remaining cells or cell debris. The filtrate was concentrated using a MasterFlex pump system (Cole-Parmer, IL, USA) and a 100-KDa Pellicon 2 Cassette filter membrane (Merck Millipore, MA, USA), and then passed through a 0.22-μm bottle-top filter again. EVs were pelleted from the resulting filtrate by ultracentrifugation at 150,000×g for 3 h at 4°C. Pellets were washed and resuspended in PBS (137 mM NaCl, 2.7 mM KCl, 10 mM Na2HPO4, and 1.8 mM KH2PO4). The relative amount of Lpc-EV was quantified on the basis of protein concentration, which was measured using a BCA protein assay kit (Thermo Fisher Scientific, MA, USA). Resuspended Lpc-EVs were stored at -80°C until use.

HT22 cell culture and drug treatment

HT22 cells were cultured as described previously [11, 33]. HT22 cells were grown in DMEM (LM-001-05; Wel Gene Inc., Gyeongsan-si, Republic of Korea) supplemented with 10% heat-inactivated fetal bovine serum (FB02-500; Serum Source, NC, USA) and antibiotics (penicillin and streptomycin) (LS-202-02; Wel Gene Inc.) at 37°C in a humidified incubator gassed with 95% air and 5% CO2. Cells grown to 70~80% confluence were treated with corticosterone (400 ng/ml) or Lpc-EV (10 μg/ml) in DMEM containing 1% fetal bovine serum. After 24 h, cells were washed with phosphate-buffered saline (PBS) and harvested.

Microarray analysis

Microarray analysis was carried out as described previously [32, 34, 35]. Briefly, control HT22 cells (CON), HT22 cells treated with GC (GC), and HT22 cells treated with GC and Lpc-EV (GC+Lpc-EV) groups were prepared. Total RNA from each group was isolated using RNeasy Mini Kit columns (Qiagen, Hilden, Germany). The quality and quantity of the purified RNA were measured using the Nanodrop 1000 V3.7.1 Spectrophotometer; NanoDrop Technologies, Wilmington, DE, USA) and, additionally, Agilent Bioanalyzer 2100 Expert (Agilent Technologies, Palo Alto, CA, USA). Purified total RNA had a quality between 1.8 and 2.0 for the 28s/18s rRNA ratio and between 1.8 and 1.9 for the A260/280 ratio.

Purified total RNA (400 ng) was converted to first- and second-strand cRNA, which was then converted to biotin-labeled cRNA samples. Biotin-labeled cRNAs (7.5 µg) were fragmented in an array fragmentation buffer by heating to 94°C for 35 min. Each of the fragmented, biotin-labeled cRNA samples (6.0 µg) was hybridized to an Agilent SurePrint G3 Mouse GE 8×60K Microarray, containing 29,116 mouse gene transcript counts. After washing, the array signals were amplified with Amersham Fluorolink streptavidin-Cy3 (GE Healthcare Bio-Sciences, Little Chalfont, UK) and scanned using GeneChip® HT Scanner and AGCC software (Affymetrix GeneChip® Command Console, Version 3.2.2). Microarray data were obtained from two independent sets of samples and were analyzed.

Microarray signals were converted into log2 scale values and normalized with Robust Multi-array Average (RMA) method implemented in Affymetrix® Power Tools (APT). False discovery rate (FDR) was obtained by adjusting p-value using Benjamini-Hochberg algorithm. The log2 expression values of microarray data were used to determine the differences in the expression levels of genes in control (CON) vs. GC, and GC vs. GC+Lpc-EV. Macrogen Inc. (Seoul, South Korea) carried out the microarray process of hybridization, raw data collection, including signaling reading of the internal quality control probes and extraction of the scanned raw data.

Gene Ontology enrichment analysis

Converted microarray signal values were analyzed using the Rank-Rank Hypergeometric Overlap (RRHO) method as described previously [35-37]. Microarray signal values of CON vs. GC and GC vs. GC+Lpc-EV were ranked by signed log10-transformed p-values. A RRHO geographic map of microarray signal values was constructed by placing the gene expression profile of the CON vs. GC group on the x-axis and that of the GC+Lpc-EV group on the y-axis using the RRHO website (http://systems.crump.ucla.edu/rankrank/). The ranked value was colored red if the mean expression level was higher than that of the comparison group, and blue if it was lower. A total of 29,116 transcript signals were analyzed and plotted on a RRHO geographic map. The genes that were up- or down-regulated by GC or Lpc-EV were selected by a top 20% cutoff. For the selected genes, serial hierarchical clustering was used to construct a dendrogram to visualize GC- and Lpc-EV-dependent gene expression profiles with a heatmap using TIGR Multiexperiment Viewer (MeV), version 4.9 (http://www.tm4.org/mev.html).

Serial k-means clustering and following by Gene Ontology (GO) enrichment analyses were carried out as described previously [32, 35]. Identified genes were grouped into multiple functional clusters using k-means clustering with increasing k values, followed by Gene Ontology (GO) enrichment analysis. K-means clustering with k=7 was used to form clusters of genes with representative GO terms, such as “Response to steroid hormones”, “Histone modification”, and/or the known key stress signature genes Mkp-1, Fkbp5, and Mecp2. The GO term hierarchy for the significant probe list was assigned based on the Mouse Genome-Database (http://www.informatics.jax.org), and GO was assessed via STRING (http://string-db.org).

Quantitative real-time PCR

Real-time PCR was used to verify gene expression patterns for selected genes in vitro and in vivo. Quantitative real-time PCR was carried out as described previously [10, 11]. Briefly, hippocampal tissues were homogenized using pellet pestles (Z359971, Sigma-Aldrich) in TRIzol reagent (15596-018, Invitrogen). HT22 cells grown in 6-well plates were harvested and homogenized in TRIzol reagent, and the total RNA was isolated. After treating with DNase I to avoid genomic DNA contamination, 2 μg of total RNA was used for conversion to cDNA using a reverse transcriptase system (Promega, Madison, WI, USA). Quantitative real-time PCR was carried out with 4 μl of 1/8 diluted cDNA, 10 μl of 2X iQTM SYBR Green Supermix (Bio-Rad Laboratories, Foster City, CA, USA), and 1 μl each of 5 pmol/μl forward and reverse primers in 20 μl of volume using the CFX 96 Real-Time PCR System Detector (Bio-Rad Laboratories). Transcript levels were normalized relative to Gapdh levels. Expression levels were analyzed by calculating the delta delta Ct values, which are the difference between the delta Ct values of the treated group and the control group. The delta delta Ct values were then expressed as 2 raised to the power of delta delta Ct using the CFX Manager Software (Bio-Rad Laboratories).

The following primers were used:

tBdnf (total form), forward 5’-TGGCTGACACTTTTGAGCAC-3’ and reverse 5’-GTTTGCGGCATCCAGGTAAT-3’; Bdnf1, forward 5’-CAGTGACAGGCGTTGAGAAA-3’ and reverse 5’-AACGCCCTCATTCTGAGAGA-3’; Bdnf2, forward 5’-GCAAGTGTTTATCACCAGGATCTAGC-3’ and reverse 5’-CGCCTTCATGCAACCGAAGTATG-3’; Bdnf3, forward 5’-GCTGCCTTGATGTTTACTTTGAC-3’ and reverse 5’-GCAACCGAAGTATGAAATAACATAG-3’; Bdnf4, forward 5’-GCTTTGTGTGGACCCTGAGTT C-3’ and reverse 5’-GCCGCCTTCATGCAACCG-3’; Nt3, forward 5’-TACTACGGCAACAGAGACG-3’ and reverse 5’-GTTGCCCACATAATCCTCC-3’; Nt4/5, forward 5’-AGCGTTGCCTAGGAATACAGC-3’ and reverse 5’-GGTCATGTTGGATGGGAGGTATC-3’; Ngf, forward 5’-AGCATTCCCTTGACACAG-3’ and reverse 5’-GGTCTACAGTGATGTTGC-3’; Nt3, forward 5’-TACTACGGCAACAGAGACG-3’ and reverse 5’-GTTGCCCACATAATCCTCC-3’; TrkB, forward 5’-AAGGACTTTCATCGGGAAGCTG-3’ and reverse 5’-TCGCCCTCCACACAGACAC-3’; Mkp-1, forward 5’-GCGCTCCACTCAAGTCTTCT-3’ and reverse 5’-AGAGGGGTACTACAGGAGCT-3’; Fkbp5, forward 5’-AATCAAACGGAAAGGCGAGG-3’ and reverse 5’- -3’; Hsp90a, forward 5’- -3’ and reverse 5’-CCAATCGGAATGTCGTGGTC-3’; p53, forward 5’-TCACTGCATGGACGATCTGT-3’ and reverse 5’-GACAAAAGATGACAGGGGCC-3’; Mecp2, forward 5’-ACAGCGGCGCTCCATTATC-3’ and reverse 5’-CCCAGTTACCGTGAAGTCAAAA-3’; Cbp, forward 5’-GGTTGCCTATGCTAAGAAAGT-3’ and reverse 5’-GATGCCTTGCTTATGTAAACG-3’; p300, forward 5’-GACTGAGCAGCGATAATG-3’ and reverse 5’-CAAGGTGTCTCTAGTGTATG-3’; Hdac1, forward 5’-CAGTGTGGCTCAGATTCCCT-3’ and reverse 5’-GGGCAGCTCATTAGGGATCT-3’; Hdac2, forward 5’- GGGACAGGCTTGGTTGTTTC-3’ and reverse 5’-GAGCATCAGCAATGGCAAGT-3’; Hdac3, forward 5’-AGAGAGGTCCCGAGGAGAAC-3’ and reverse 5’-ACTCTTGGGGACACAGCATC-3’; Sirt1, forward 5’-GATCCTTCAGTGTCATGGTTC-3’ and reverse 5’-ATGGCAAGTGGCTCATCA-3’; Sirt7, forward 5’-CTGGAGATTCCTGTCTACAACCG-3’ and reverse 5’- AGTGACTTCCTACTGTGGCTGC-3’; Kdm4a, forward 5’-GTCTGGCCTCTTCACTCAGT-3’ and reverse 5’- TACCATTCACGTCTGCTCCA-3’; Kdm4b, forward 5’-CGGGGCTTTTCACCACAGTAC-3’ and reverse 5’- GTACAGGGAGCCACTGATGT-3’; SUV39H1, forward 5’-TGGTTAAGTGGCGTGGGTAT-3’ and reverse 5’- TTGTTCCCAACGCTGAAGTG-3’; Setdb1, forward 5’-GGTGGTTGAAGAGCTGGGTA-3’ and reverse 5’-TCACTTCCCTGGATGCATCA-3’; Gapdh, forward 5’-AGAAGGTGGTGAAGCAGGCATC-3’ and reverse 5’-CGAAGGTGGAAGA GTGGGAGTTG-3’.

Behavioral tests

All behavioral tests were monitored with a video tracking system (SMART; Panlab, Barcelona, Spain) and/or a webcam recording system (HD Webcam C210, Logitech, Newark, CA, USA).

Sociability test

The sociability test was carried out as described previously [32, 33]. Briefly, a U-shaped two-choice field was prepared by partitioning an open field (40×40 cm2) with a wall (20 cm in width and 20 cm in height) at the center point. A subject mouse was allowed to freely explore the two-choice field with an empty circular grid cage (12 cm in diameter and 33 cm in height) on each side for 5 min and was then returned to the home cage. After 10 min, a social target was loaded into a circular grid cage on one side, and the subject mouse was allowed to explore both fields for 10 min. The trajectory of the mouse’s movements and time spent in each field were recorded using a video tracking system. The field with the circular grid cage containing a social target and the field containing an empty grid cage were defined as the target field and non-target field, respectively.

Tail suspension test

The tail suspension test was carried out as described previously [32, 33]. Mice were suspended individually by fixing their tails with adhesive tape to the ceiling of an enclosed shelf 50 cm above a bottom floor. While recording using a webcam recording system, cumulative immobility time was measured for 6 min. Immobility was defined as the time the animal spent suspended with all limbs motionless.

Forced swim test

The forced swim test was performed as described previously [32, 33]. Mice were placed in a Plexiglas cylinder (15 cm in diameter×27 cm height) containing water at a temperature of 24°C. Mice were placed in the cylinder for 6 min, and the cumulative immobility time was measured for the last 5 min. Immobility was defined as the time the animal spent floating with all limbs motionless.

Statistical analysis

Two-sample comparison was carried out using the student’s t-test. Multiple comparisons were performed by one-way ANOVA followed by the Newman-Keuls post hoc test. All data are represented as the mean±SEM, and statistical significance was accepted at the 5% level.

Lpc-EV counteracted glucocorticoid-induced genomic responses in HT22 cells

We investigated whether Lpc-EV has the ability to modify glucocorticoid (GC)-induced changes in the expression of neurotrophic factors. GC (corticosterone) treatment in HT22 neuronal cells decreased the expression of Bdnf, Nt3, Nt4/5, Ngf, and TrkB. In contrast, Lpc-EV treatment counteracted GC-induced decreased expression of Bdnf, Nt3, Ngf, and TrkB (Fig. 1A).

Next, we explored the underlying mechanisms by which Lpc-EV restored GC-induced changes in the expression of neurotrophic factors. To the end, we applied two different approaches. First, we tested whether any known factors regulating the expression of neurotrophic factors are involved in Lpc-EV effects. Several classes of stress-related cellular or epigenetic factors, such as Mkp1, Fkbp5, Hsp90a, p53, MeCP2, Hdac2, and Sirt1 have been examined for their role in regulating the expression of neurotrophic factors [11, 33, 38-40]. GC treatment in HT22 cells decreased or tended to decrease the expression of p53, Mecp2, and Sirt1, while increasing the expression of Mkp1, Fkbp5, and Hsp90a. In contrast, Lpc-EV treatment blocked GC-induced reduced expression of p53 and Mecp2, and GC-induced increased expression of Mkp1 and Fkbp5 but not significantly Hsp90a, Hdac2, and Sirt1 (Fig. 1B, C).

Recently, it has been reported that MeCP2 is a critical player in regulating the expression of Mkp1 and Fkbp5, and neurotrophoic factors [39-41]. Therefore, it was investigated whether MeCP2 is a key regulator of the effects of Lpc-EV on the expression of neurotrophic factors. siRNA-mediated knockdown of Mecp2 blocked Lpc-EV-induced recovery of GC-induced changes of Bdnf, Nt4/5, TrkB, p53, Mkp1, and Fkbp5 but not Nt3 and Ngf (Fig. 1D~G). These results suggest that Lpc-EV has the ability to counteract GC-induced genomic changes of those genes through Mecp2 upregulation.

Second, we used a genome-wide approach to identify genes whose expression was changed by GC and their altered expression was reversed by Lpc-EV. We obtained the microarray signal values of the genes whose expression was changed in HT22 cells treated with GC (GC) compared to control HT22 cells (CON) (GC vs. CON), and the genes whose expression was changed in HT22 cells treated with GC and Lpc-EV (GC+Lpc-EV) compared to HT22 cells treated with GC (GC+Lpc-EV vs. GC). Then, we constructed the geographic map of gene expression profiles using the obtained the signal values (Fig. 2A, B) Using the Rank-Rank Hypergeometric Overlap (RRHO) method [36, 37]. Of the total 29,116 transcripts in the microarray, 8,158 transcripts (28.0%) were upregulated by GC, and their altered expression was reversed by Lpc-EV treatment and these transcripts were located in Quadrant A of the RRHO map. Another 8,592 transcripts (29.5%) were down-regulated by GC and their altered expression was reversed by Lpc-EV treatment, and these transcripts were located in Quadrant D of the RRHO map (Fig. 2A~C). Of those transcripts, the top 20% of transcripts by rank based on expression differences in each quadrant were selected. Then, 1,632 in Quadrant A and 1,728 in Quadrant D were selected further by annotating the 20%-ranked transcripts with Mus musculus genes. A cutoff of 0.4, which is a medium confidence interaction score in the STRING database, was used to finally select 998 transcripts in Quadrant A and 756 transcripts in Quadrant D that were most likely to be actively responding transcripts to GC and Lpc-EV. These transcripts were then placed in further analyses.

Serial K-means clustering and Gene Ontology (GO) enrichment analysis indicated that the selected genes (998 genes in Quadrant A and 756 genes in Quadrant D) could be grouped into multiple functional clusters that enriched genes involved in stress responses and epigenetic modifications. In principle, all the selected genes actively responded to both GC and Lpc-EV. In the present study, we focused on the following gene enrichment pathways. The 998 genes in Quadrant A could be grouped into 7 clusters (Fig. 2D; Supplemental Table S1 and S2), one of which (Cluster 5) included a number of the genes covered by the GO terms “regulation of neuron death”, “regulation of stress-activated MAPK cascade”, “response to steroid hormone”, “regulation of oxidative stress-induced cell deaths” and “Histone H3 acetylation” (Fig. 2D, F, G; Supplemental Table S2). Cluster 5 contained the stress signature genes Mkp1 (Dusp1) and Fkbp5 (Supplemental Table S2), whose expression was increased by GC and their increased expression was reversed by Lpc-EV treatment (Fig. 1).

The 756 genes in Quadrant D could be grouped into 8 clusters (See also Supplemental Table S1 and S3), two of which (Clusters 3 and 4) included a list of the genes covered by the GO terms, “histone modification” and “histone acetylation” (Cluster 3); and “regulation of neuron death”, “stress-activated protein kinase signaling cascade”, “regulation of transcription from RNA polymerase II promoter in response to stress” and “regulation of long-term neuronal synaptic plasticity” (Cluster 4) (Fig. 2E, H; Supplemental Table S3). Cluster 4 contained Ngf and Mecp2 (Supplemental Table S3), whose expression was decreased by GC and their decreased expression was reversed by Lpc-EV in HT22 cells (Fig. 1).

GC- and Lpc-EV-dependent changes in the expression of the selected genes were analyzed using a dendrogram. A dendrogram of the expression patterns of the 135 genes in Cluster 5 (Quadrant A) highlighted the genes for stress-activated Mapk cascades and neuronal cell death and whose expression was upregulated by GC and their altered expression was reversed by Lpc-EV (Fig. 3A). Similar analyses of the 101 genes in Cluster 3 and the 99 genes in Cluster 4 (Quadrant D) were used to build the dendrograms of genes for histone modification factors and stress-activated protein kinase signaling cascade and whose expression exhibited GC-dependent downregulation and Lpc-EV-induced reversion of the altered expression (Fig. 3B, C).

We focused on and investigated the changes in the expression of the genes with the functional modules representing histone modification factors in Clusters 3 and 4 in Quadrant D (Fig. 3B, C; Supplemental Table S1 and S3). Real-time PCR analysis indicated that GC treatment decreased the expression of p300, Sirt1, Sirt7, and Suv39h1 and increased the expression of Hdac2. In contrast, Lpc-EV treatment blocked GC-induced down-regulation of p300, Sirt1, and Suv39h1 but not Sirt7, also inhibiting GC-induced up-regulation of Hdac2 (Fig. 4A, B). siRNA-mediated knockdown of Mecp2 blocked Lpc-EV-induced restoration of GC-induced changes of p300, Hdac2, and Sirt1, whereas its effect on Suv39h1 was insignificant (Fig. 4C, D). These results suggest that Lpc-EV counteracts GC-induced changes in the expression of histone modification factors through Mecp2 upregulation.

Lpc-EV treatment reversed stress-induced changes in the expression of Mkp-1, Fkbp5, and Mecp2 in the hippocampus

Next, we investigated whether Lpc-EV treatment produces genomic responses against stress-induced changes in the brain of mice. Mice exposed to chronic restraint stress (CRST) for 2-h daily over 14 days exhibit persistent depressive-like behaviors [10, 33, 34]. CRST-treated mice had increased expression of Mkp-1 and Fkbp5, and decreased expression of Mecp2 in the hippocampus (Fig. 5A~C). In contrast, post-stress treatment with Lpc-EV in CRST-treated mice reversed stress-induced increased expression of Mkp-1 and Fkbp5, also reversing decreased Mecp2 expression (Fig. 5A~C).

The epigenetic factors, Mecp2, Sirt1, Suv39h1, and Hdac2 act as upstream regulators of Mkp-1, Bdnf, Nt3, Nt4/5, and TrkB in the brains of CRST mice [11, 33, 41]. We tested whether Lpc-EV treatment had an impact on stress-induced changes of epigenetic factors. CRST mice had decreased expression of Cbp, p300, Sirt1, Sirt7, and Suv39h1, and increased expression of Hdac2. In contrast, post-stress treatment with Lpc-EV in CRST-treated mice partially rescued stress-induced changes in the expression of Hdac2, Sirt1 and Suv39h1 (Fig. 5D, E). Lpc-EV also rescued stress-induced changes in the expression of Bdnf, Nt3, Nt4/5, Ngf, and TrkB in the hippocampus (Fig. 5F).

Collectively, these results suggest that Lpc-EV cargo contents have the ability to counteract stress-induced changes in the expression of Mkp-1, Fkbp5, Mecp2, Sirt1 and Suv39h1, also counteracting stress-induced changes in the expression of Bdnf, Nt3, Nt4/5, Ngf, and TrkB in the hippocampus.

Lpc-EV treatment relieved stress-induced depressive-like behavior

Mice exposed to CRST showed reduced time to explore the cage with a target mouse over an empty cage in the social interaction test (SIT) (Fig. 6A~C), thus exhibiting reduced social interaction in the SIT, and showed increased immobility time in the tail suspension test (TST) and the forced swim test (FST) (Fig. 6D, E), which are consistent with previous reports [10, 33, 34]. In contrast, post-stress treatment with Lpc-EV for 14 days in CRST mice reversed stress-induced reduced social interaction (Fig. 6A~C), and stress-induced increased immobility in the TST and FST (Fig. 6D, E). These results suggest that Lpc-EV produces anti-depressive effects at the behavioral level.

Probiotics are believed to be beneficial for psychiatric disorders, yet the underlying mechanisms by which specific probiotic strains produce neuroactive effects are not clearly understood. In the present study, we demonstrate that Lpc-EV produced genome-wide changes against GC-induced transcriptional responses in HT22 cells. Microarray analysis followed by the RRHO method led us to identify 1,754 genes that were up- or down-regulated by GC, and their altered expression was reversed by Lpc-EV treatment in HT22 cells (Fig. 1~4; Supplemental Table S1~S3). Lpc-EV-induced actively responding genes include those which contain functional modules representing “responses to stress or steroid hormones”, “histone modification”, and “regulating MAPK signaling pathways”. Our results raise the following important issues on Lpc-EV effects (Fig. 2; Supplemental Table S3).

First, Lpc-EV induced genome-wide transcriptional changes by directly acting on HT22 neuronal cells without intermediate mediators. Considering that bacterial EVs contain proteins/peptides, carbohydrates, lipids, nucleic acids and bacterial metabolites [42-44], the bioactive components in Lpc-EV should contain some of such components, although which cargo components in Lpc-EV produce those massive transcriptional responses yet remains to be characterized in the future. More discussion will be presented in the following sections.

Second, Lpc-EV cargo contents have the ability to reverse GC-induced changes in the expression of epigenetic factors. Lpc-EV-induced up-regulation against GC-induced changes in neurotrophic factors, Mecp2, and Sirt1 (Fig. 1) are similar to those of Lactobacillus plantarum-derived EVs [10, 11]. Lpc-EV also up-regulated other epigenetic factors, including Hdac1, Hdac2, and Suv39h1 (Fig 4; Supplemental Table S3). Considering that epigenetic factors can serve as the basic regulators of chromosome structures and the transcription of numerous genes, Lpc-EV-induced changes in epigenetic regulation of genes likely contribute genome-wide effects.

Third, Lpc-EV counteracted GC-induced transcriptional responses. Genome-wide microarray analysis followed by the RRHO method led to the identification of 1,754 GC-responsive genes (998 genes in Quadrant A and 756 genes in Quadrant D) and their altered expression was reversed by Lpc-EV treatment (Fig. 1~4). Regarding the massive and diverse genome-wide effects of Lpc-EV, it is possible that bioactive components in Lpc-EV likely drive multiple arrays of signaling events to restore a homeostatic ability in GC-challenged HT22 cells.

The findings that Lpc-EV produced similar effects on the expression of Mkp-1, Fkbp5, Mecp2, Sirt1, and Suv39h1 epigenetic and nuclear factors, and of Bdnf, Nt3, Nt4/5, Ngf, and TrkB in HT22 cells (Fig. 1~4) and in the hippocampus of CRST-treated mice (Fig. 5) raise the possibility that Lpc-EVs contain neuroactive components that can directly modify brain cells via transcriptional responses. Bacterial EVs carry various molecular cargo materials, such as bacterial nucleic acids, enzymes, peptides, short-chain fatty acids, and biometabolites [42, 43]. Because bacterial EVs consist of lipid bilayer-encapsulated vesicles with a diameter of 20~250 nm, they can easily cross the mucosal barrier of the intestine and cell membrane [42]. Indeed, orally delivered Cy7-labeled Staphylococcus aureus EVs [43] or Cy7-labeled Helicobacter pylori EVs [45] in mice were distributed in the brain and many other tissues. Treatment with Lactobacillus casei-derived EVs in the human colorectal cell line Caco-2 decreases TLR9 gene expression and increases the anti-inflammatory cytokines IL-4 and IL-10 [46]. Treatment with Lactobacillus plantarum-derived EVs increases BDNF expression in HT22 cells [11]. These results suggest that bacterial EVs contain cargo materials that may induce signaling events and transcriptional responses by directly acting on eukaryotic host cells including brain cells. Recently, it has been reported that EVs derived from Lactobacillus acidophilus or Escherichia coli exhibit antitumor activity, which is dependent on interferon-gamma (IFN-γ) production. Trypsin-sensitive EV surface proteins appear to be the active component of EVs that induce IFN-γ production [47]. Propionibacterium freudenreichii EVs have anti-inflammatory properties, and the surface proteins in these EVs are responsible for their probiotic effects [48]. However, it is not known at present whether EVs from different Lactobacillus species contain distinct cargo materials that induce similar or distinct transcriptional responses or whether EVs from different Lactobacillus species have similar or common cargo materials that induce the same pathways leading to the observed transcriptional responses.

In the present study, we delivered EVs through the intraperitoneal (IP) route, which is physiologically linked to the circulatory system, and thereby attempted to abate a possible complication involving intestine-related mechanisms. IP-injected Lactobacillus plantarum-derived EVs increased BDNF expression in the hippocampus of mice, similarly as that induced in cultured neuronal cells [10, 11]. IP-injected molecules in rodents rapidly and effectively enter the circulatory system, and, therefore, IP administration can be chosen as a justifiable route for proof-of-concept studies [49]. IP-injected small molecules (MW<5,000) and fluids are predominantly absorbed from the visceral peritoneum by diffusion through the mesenteric capillaries, drain into the portal vein, and enter the systemic circulatory system. Whereas IP-injected large molecules (MW>5,000), proteins, blood and immune cells are taken up mainly through the lymphatic system [49]. Although it needs to be investigated which is a preferred path for IP-injected probiotic-EVs to enter the circulatory system, we speculate they might go through any of the two routes as EVs are nano-size lipid vesicles. IP-injected fluorescent lipophilic dye-labeled EVs are accumulated in many organs in the body, including the brain, liver, and kidney, although accumulation levels vary among different regions [50]. Therefore, the results of the present study, together with those from our recent studies [10, 11, 44, 45], support the notion that bacterial EVs can serve as important mediators for communication from gut microbiota to the brain.

This study was supported by a grant (2022M3E5E8017561) for PL Han, a grant (2022R1C1C2011198) for EH Lee, and a grant (2022R1I1A1A01070983) for JY Park from the Ministry of Science, ICT and Future Planning, Republic of Korea and partly by MD Healthcare Inc.

Fig. 1. Lpc-EV treatment blocked GC-induced changes in the expression of neurotrophic factors, TrkB, Mkp1, Fkbp5, Hsp90a1, p53, and Mecp2 in HT22 cells. (A~C) Transcript levels of total Bdnf (tBdnf), Bdnf1, Bdnf3, Bdnf4, Nt3, Nt4/5, Ngf, and TrkB (A); Mkp1, Fkbp5, Hsp90a1, and p53 (B); Mecp2, Hdac2, and Sirt1 (C) in HT22 cells treated for 24 h with glucocorticoid (GC, corticosterone; 400 ng/ml) or GC plus Lpc-EV (10 μg/ml). (D~G) siRNA-mediated knockdown of Mecp2 (D). Transcript levels of tBdnf, Bdnf1, Bdnf3, Bdnf4, Nt3, Nt4/5, Ngf, and TrkB (E); Mkp1, Fkbp5, Hsp90a, and p53 (F); and Mecp2 (G) in HT22 cells treated for 24 h with GC (400 ng/ml), GC and Lpc-EV plus control-siRNA (siCON), and GC and Lpc-EV plus Mecp2-siRNA. Data are presented as the mean±SEM (n=8). *p<.05; **p<.01 (one-way ANOVA followed by Newman-Keuls post hoc test, and two-way ANOVA followed by Bonferroni post hoc test).
Fig. 2. Microarray analysis of gene expression profiles changed by GC and the reversion of their gene expression by Lpc-EV in HT22 cells. (A) A geometric map consisting of hypothetical gene expression signatures of CON vs. GC on the x-axis and GC vs. GC+Lpc-EV on the Y-axis. The x-axis and the y-axis represent the ratios of the logarithms (log10) of the gene expression levels of GC to Con and GC+Lpc-EV to GC, respectively. The features and locations of up- or down-regulated expression of genes are explained in four quadrants. (B) A geometric map constructed with gene expression signatures of CON vs. GC on the x-axis and GC vs. GC+Lpc-EV on the y-axis. The genes that were up-regulated by GC and subsequently down-regulated by Lpc-EV are located in Quadrant A. Conversely, the genes that were down-regulated by GC and subsequently up-regulated by Lpc-EV are located in Quadrant D. Dotted lines: 20% cut-off. Blue-to Red color gradient: the number of genes counted; blue, rare or few; red, many numbers indicated. (C) A summary and experimental flow of the sequential analysis of gene expression profiles of CON vs. GC on the x-axis and GC vs. GC+Lpc-EV on the y-axis. The numbers of the genes located in Quadrants A and D of the RRHO map, the number of the top 20%-ranked genes defined by RRHO in Quadrants A and D, the number of the top 20%-ranked genes that had mouse gene annotations in the STRING database, and the number of the top 20%-ranked genes predicted to form functional interaction with defined gene members with a medium confidence strength of 0.400 are indicated. (D, E) The identified 998 genes of Quadrant A were grouped by K-means clustering into seven clusters (D), of which Cluster 5 contained the stress signature genes Mkp1 and Fkbp5. Cluster 1 (162 genes), red; cluster 2 (158 genes), brown; cluster 3 (156 genes), olive; cluster 4 (149 genes), green; cluster 5 (135 genes), blue; cluster 6 (131 genes), light blue; cluster 7 (101 genes), medium blue. See also Supplemental Table S1 and S2. The 756 genes of Quadrant D were grouped by K-means clustering into eight clusters (E), of which Cluster 4 carried Mecp2. Cluster 1 (111 genes), red; cluster 2 (107 genes), brown; cluster 3 (101 genes), olive; cluster 4 (99 genes), green; cluster 5 (94 genes), lime green; cluster 6 (90 genes), cyan; cluster 7 (78 genes), blue; cluster 8 (76 genes), purple. The number 5 in (D) and the numbers 3 and 4 in (E) indicates the cluster names. See also Supplemental Table S1 and S3. (F~H) A summary of key features of Cluster 5 of Quadrant A (F) and Clusters 3 and 4 of Quadrant D (G, H).
Fig. 3. Dendrograms exhibiting GC-induced up- or downregulated genes and the reversion of their altered expression by Lpc-EV in HT22 cells. (A) A dendrogram showing the expression patterns of the 135 genes of Cluster 5 in Quadrant A. The heatmap highlighted the up-regulation of the genes by GC and the reversal of their altered expression by Lpc-EV in duplicate sample groups. Red and blue represent up-regulation and down-regulation, respectively, in the heatmap. Expression levels, log2 values. The stress signature genes Mkp1 (Dusp1) and Fkbp5 were marked. See also Supplemental Table S1 and S2. (B, C) Dendrograms showing the expression patterns of the 101 genes in Cluster 3 (B) and the 99 genes in Cluster 4 (C) in Quadrant D. The heatmap exhibiting GC-dependent downregulation and Lpc-EV-induced reversion of their altered expression in duplicate sample groups. Red and blue represent up-regulation and down-regulation, respectively, as above. Expression levels, log2 values. Ngf, Vgefa, and Mecp2 were marked. See also Supplemental Table S1 and S3.
Fig. 4. Lpc-EV treatment counteracted GC-induced altered expression of epigenetic factors in HT22 cells. (A, B) Expression levels of Cbp, p300, Hdac1, Hdac2, Hdac3, Sirt1, and Sirt7 (A), and Kdm4a, Kdm4b, Suv39h1, and Setdb1 (B) in HT22 cells treated for 24 h with GC (400 ng/ml) or GC plus Lpc-EV (10 μg/ml). HATs, Histone Acetyltransferases; HDACs, Histone Deacetylases; HMTs, Histone methyltransferases; HDMTs, histone demethylases. (C, D) Expression levels of Cbp, p300, Hdac1, Hdac2, Hdac3, Sirt1, and Sirt7 (C), and Kdm4a, Kdm4b, Suv39h1, and Setdb1 (D) in HT22 cells treated for 24 h with GC (400 ng/ml), GC plus Lpc-EV (10 μg/ml) and control-siRNA, or GC plus Lpc-EV and Mecp2-siRNA. Data are presented as the mean±SEM (n=6~8). *p<.05; **p<.01 (one-way ANOVA followed by Newman-Keuls post hoc test, and two-way ANOVA followed by Bonferroni post hoc test).
Fig. 5. Post-stress treatment with Lpc-EV reversed stress-induced changes in the expression of MeCP2, Sirt1 and neurotrophic factors in the hippocampus of CRST mice. (A) Experimental design. CRST mice were intraperitoneally (i.p.) treated with Lpc-EV for 14 days at a dose of 2 μg/100 μl/mouse. n=6~12 animals. (B, C) Expression levels of Mkp1, Fkbp5, and Hsp90a1 (B), and Mecp2 (C) in the hippocampus of CRST mice treated with Lpc-EV. Veh, vehicle (n=6~8 qPCR repeats). (D, E) Expression levels of Cbp, p300, Hdac1, Hdac2, Hdac3, Sirt1, and Sirt7 (D), and Kdm4a, Kdm4b, Suv39h1, and Setdb1 (E) in the hippocampus of CRST mice treated with Lpc-EV. Veh, vehicle (n=6~8 qPCR repeats). (F) Expression levels of total Bdnf (tBdnf), Bdnf1, Bdnf2, Bdnf3, Nt3, Nt4/5, Ngf, and TrkB in the hippocampus of mice treated with Lpc-EV. Veh, vehicle (n=6~8 qPCR repeats). Data are presented as the mean±SEM. *p<0.05; **p<0.01 (one-way ANOVA followed by Newman-Keuls post hoc test).
Fig. 6. Post-stress treatment with Lpc-EV improved stress-induced depressive-like behavior in CRST mice. (A) Experimental design. CRST mice were treated with imipramine (IMI) or Lpc-EV for 14 days. Behavior tests were performed on post-stress days 15~16 (p15~p16). Control mice injected with saline (CON+veh), mice treated with CRST (CRST+veh), CRST treated with IMI (CRST+IMI), and CRST treated with Lpc-EV (CRST+Lpc-EV). Lpc-EV, 2 μg/100 ul/mouse/day (i.p.). n=10~12 animals. (B, C) Social interaction time of the indicated groups spent in the target vs. non-target fields in the U-shaped two-choice sociability test. A diagram of the U-shaped two-choice field (right panel). (D, E) Immobility time in the TST (D) and FST (E) for the indicated groups. Data are presented as the mean±SEM. *p<0.05; **p<0.01 (one-way ANOVA followed by Newman-Keuls post hoc test).
  1. Sharon G, Sampson TR, Geschwind DH, Mazmanian SK (2016) The central nervous system and the gut microbiome. Cell 167:915-932
    Pubmed KoreaMed CrossRef
  2. van Zyl WF, Deane SM, Dicks LMT (2020) Molecular insights into probiotic mechanisms of action employed against intestinal pathogenic bacteria. Gut Microbes 12:1831339
    Pubmed KoreaMed CrossRef
  3. Nikolova VL, Smith MRB, Hall LJ, Cleare AJ, Stone JM, Young AH (2021) Perturbations in gut microbiota composition in psychiatric disorders: a review and meta-analysis. JAMA Psychiatry 78:1343-1354
    Pubmed KoreaMed CrossRef
  4. Karl JP, Hatch AM, Arcidiacono SM, Pearce SC, Pantoja-Feliciano IG, Doherty LA, Soares JW (2018) Effects of psychological, environmental and physical stressors on the gut microbiota. Front Microbiol 9:2013
    Pubmed KoreaMed CrossRef
  5. Rea K, Dinan TG, Cryan JF (2020) Gut microbiota: a perspective for psychiatrists. Neuropsychobiology 79:50-62
    Pubmed CrossRef
  6. Wallace CJK, Milev R (2017) The effects of probiotics on depressive symptoms in humans: a systematic review. Ann Gen Psychiatry 16:14
    Pubmed KoreaMed CrossRef
  7. Ansari F, Pourjafar H, Tabrizi A, Homayouni A (2020) The effects of probiotics and prebiotics on mental disorders: a review on depression, anxiety, alzheimer, and autism spectrum disorders. Curr Pharm Biotechnol 21:555-565
    Pubmed CrossRef
  8. Yong SJ, Tong T, Chew J, Lim WL (2020) Antidepressive mechanisms of probiotics and their therapeutic potential. Front Neurosci 13:1361
    Pubmed KoreaMed CrossRef
  9. Luo Y, Zeng B, Zeng L, Du X, Li B, Huo R, Liu L, Wang H, Dong M, Pan J, Zheng P, Zhou C, Wei H, Xie P (2018) Gut microbiota regulates mouse behaviors through glucocorticoid receptor pathway genes in the hippocampus. Transl Psychiatry 8:187
    Pubmed KoreaMed CrossRef
  10. Choi J, Kwon H, Kim YK, Han PL (2022) Extracellular vesicles from gram-positive and gram-negative probiotics remediate stress-induced depressive behavior in mice. Mol Neurobiol 59:2715-2728
    Pubmed CrossRef
  11. Choi J, Kim YK, Han PL (2019) Extracellular vesicles derived from Lactobacillus plantarum increase BDNF expression in cultured hippocampal neurons and produce antidepressant-like effects in mice. Exp Neurobiol 28:158-171
    Pubmed KoreaMed CrossRef
  12. Chevalier G, Siopi E, Guenin-Macé L, Pascal M, Laval T, Rifflet A, Boneca IG, Demangel C, Colsch B, Pruvost A, Chu-Van E, Messager A, Leulier F, Lepousez G, Eberl G, Lledo PM (2020) Effect of gut microbiota on depressive-like behaviors in mice is mediated by the endocannabinoid system. Nat Commun 11:6363
    Pubmed KoreaMed CrossRef
  13. Barros-Santos T, Silva KSO, Libarino-Santos M, Elisangela Gouveia Cata-Preta, Reis HS, Tamura EK, de Oliveira-Lima AJ, Berro LF, Uetanabaro APT, Marinho EAV (2020) Effects of chronic treatment with new strains of Lactobacillus plantarum on cognitive, anxiety- and depressive-like behaviors in male mice. PLoS One 15:e0234037
    Pubmed KoreaMed CrossRef
  14. Dhaliwal J, Singh DP, Singh S, Pinnaka AK, Boparai RK, Bishnoi M, Kondepudi KK, Chopra K (2018) Lactobacillus plantarum MTCC 9510 supplementation protects from chronic unpredictable and sleep deprivation-induced behaviour, biochemical and selected gut microbial aberrations in mice. J Appl Microbiol 125:257-269
    Pubmed CrossRef
  15. Xie R, Jiang P, Lin L, Jiang J, Yu B, Rao J, Liu H, Wei W, Qiao Y (2020) Oral treatment with Lactobacillus reuteri attenuates depressive-like behaviors and serotonin metabolism alterations induced by chronic social defeat stress. J Psychiatr Res 122:70-78
    Pubmed CrossRef
  16. Ma X, Shin YJ, Park HS, Jeong JW, Kim JY, Shim JJ, Lee JL, Kim DH (2023) Lactobacillus casei and its supplement alleviate stress-induced depression and anxiety in mice by the regulation of BDNF expression and NF-κB activation. Nutrients 15:2488
    Pubmed KoreaMed CrossRef
  17. Dempsey E, Corr SC (2022) Lactobacillus spp. for gastrointestinal health: current and future perspectives. Front Immunol 13:840245
    Pubmed KoreaMed CrossRef
  18. Wei CL, Wang S, Yen JT, Cheng YF, Liao CL, Hsu CC, Wu CC, Tsai YC (2019) Antidepressant-like activities of live and heat-killed Lactobacillus paracasei PS23 in chronic corticosterone-treated mice and possible mechanisms. Brain Res 1711:202-213
    Pubmed CrossRef
  19. Liao JF, Hsu CC, Chou GT, Hsu JS, Liong MT, Tsai YC (2019) Lactobacillus paracasei PS23 reduced early-life stress abnormalities in maternal separation mouse model. Benef Microbes 10:425-436
    Pubmed CrossRef
  20. Xu M, Tian P, Zhu H, Zou R, Zhao J, Zhang H, Wang G, Chen W (2022) Lactobacillus paracasei CCFM1229 and Lactobacillus rhamnosus CCFM1228 alleviated depression- and anxiety-related symptoms of chronic stress-induced depression in mice by regulating xanthine oxidase activity in the brain. Nutrients 14:1294
    Pubmed KoreaMed CrossRef
  21. Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, Pettersson S (2012) Host-gut microbiota metabolic interactions. Science 336:1262-1267
    Pubmed CrossRef
  22. Schirmer M, Smeekens SP, Vlamakis H, Jaeger M, Oosting M, Franzosa EA, Ter Horst R, Jansen T, Jacobs L, Bonder MJ, Kurilshikov A, Fu J, Joosten LAB, Zhernakova A, Huttenhower C, Wijmenga C, Netea MG, Xavier RJ (2016) Linking the human gut microbiome to inflammatory cytokine production capacity. Cell 167:1125-1136.e8
    Pubmed KoreaMed CrossRef
  23. Plaza-Diaz J, Ruiz-Ojeda FJ, Gil-Campos M, Gil A (2019) Mechanisms of action of probiotics. Adv Nutr 10(Suppl 1):S49-S66
    Pubmed KoreaMed CrossRef
  24. Rutsch A, Kantsjö JB, Ronchi F (2020) The gut-brain axis: how microbiota and host inflammasome influence brain physiology and pathology. Front Immunol 11:604179
    Pubmed KoreaMed CrossRef
  25. Silva YP, Bernardi A, Frozza RL (2020) The role of short-chain fatty acids from gut microbiota in gut-brain communication. Front Endocrinol (Lausanne) 11:25
    Pubmed KoreaMed CrossRef
  26. Choi JH, Moon CM, Shin TS, Kim EK, McDowell A, Jo MK, Joo YH, Kim SE, Jung HK, Shim KN, Jung SA, Kim YK (2020) Lactobacillus paracasei-derived extracellular vesicles attenuate the intestinal inflammatory response by augmenting the endoplasmic reticulum stress pathway. Exp Mol Med 52:423-437
    Pubmed KoreaMed CrossRef
  27. Haas-Neill S, Forsythe P (2020) A budding relationship: bacterial extracellular vesicles in the microbiota-gut-brain axis. Int J Mol Sci 21:8899
    Pubmed KoreaMed CrossRef
  28. Díaz-Garrido N, Badia J, Baldomà L (2021) Microbiota-derived extracellular vesicles in interkingdom communication in the gut. J Extracell Vesicles 10:e12161
    Pubmed KoreaMed CrossRef
  29. Ñahui Palomino RA, Vanpouille C, Costantini PE, Margolis L (2021) Microbiota-host communications: bacterial extracellular vesicles as a common language. PLoS Pathog 17:e1009508
    Pubmed KoreaMed CrossRef
  30. Cuesta CM, Guerri C, Ureña J, Pascual M (2021) Role of microbiota-derived extracellular vesicles in gut-brain communication. Int J Mol Sci 22:4235
    Pubmed KoreaMed CrossRef
  31. Choi J, Kwon HJ, Seoh JY, Han PL (2021) Hyperoxygenation ameliorates stress-induced neuronal and behavioral deficits. Exp Neurobiol 30:415-429
    Pubmed KoreaMed CrossRef
  32. Lee EH, Park JY, Kwon HJ, Han PL (2021) Repeated exposure with short-term behavioral stress resolves pre-existing stress-induced depressive-like behavior in mice. Nat Commun 12:6682
    Pubmed KoreaMed CrossRef
  33. Lee JE, Kwon HJ, Choi J, Seo JS, Han PL (2020) Aging increases vulnerability to stress-induced depression via upregulation of NADPH oxidase in mice. Commun Biol 3:292
    Pubmed KoreaMed CrossRef
  34. Kim TK, Lee JE, Kim JE, Park JY, Choi J, Kim H, Lee EH, Han PL (2016) G9a-mediated regulation of OXT and AVP expression in the basolateral amygdala mediates stress-induced lasting behavioral depression and its reversal by exercise. Mol Neurobiol 53:2843-2856
    Pubmed CrossRef
  35. Park JY, Lee EH, Kwon HJ, Baek IS, Seo JS, Kim KS, Han PL (2022) Behavioral engagement with playable objects resolves stress-induced adaptive changes by reshaping the reward system. Biol Psychiatry 91:676-689
    Pubmed CrossRef
  36. Plaisier SB, Taschereau R, Wong JA, Graeber TG (2010) Rank-rank hypergeometric overlap: identification of statistically significant overlap between gene-expression signatures. Nucleic Acids Res 38:e169
    Pubmed KoreaMed CrossRef
  37. Cahill KM, Huo Z, Tseng GC, Logan RW, Seney ML (2018) Improved identification of concordant and discordant gene expression signatures using an updated rank-rank hypergeometric overlap approach. Sci Rep 8:9588
    Pubmed KoreaMed CrossRef
  38. Crochemore C, Michaelidis TM, Fischer D, Loeffler JP, Almeida OF (2002) Enhancement of p53 activity and inhibition of neural cell proliferation by glucocorticoid receptor activation. FASEB J 16:761-770
    Pubmed CrossRef
  39. Zannas AS, Wiechmann T, Gassen NC, Binder EB (2016) Gene-stress-epigenetic regulation of FKBP5: clinical and translational implications. Neuropsychopharmacology 41:261-274
    Pubmed KoreaMed CrossRef
  40. Lee JE, Kwon HJ, Choi J, Han PL (2019) Stress-induced epigenetic changes in hippocampal Mkp-1 promote persistent depressive behaviors. Mol Neurobiol 56:8537-8556
    Pubmed CrossRef
  41. Choi J, Kwon HJ, Lee JE, Lee Y, Seoh JY, Han PL (2019) Hyperoxygenation revitalizes Alzheimer's disease pathology through the upregulation of neurotrophic factors. Aging Cell 18:e12888
    Pubmed KoreaMed CrossRef
  42. Yang J, Kim EK, McDowell A, Kim YK (2018) Microbe-derived extracellular vesicles as a smart drug delivery system. Transl Clin Pharmacol 26:103-110
    Pubmed KoreaMed CrossRef
  43. Gill S, Catchpole R, Forterre P (2019) Extracellular membrane vesicles in the three domains of life and beyond. FEMS Microbiol Rev 43:273-303
    Pubmed KoreaMed CrossRef
  44. Choi SJ, Kim MH, Jeon J, Kim OY, Choi Y, Seo J, Hong SW, Lee WH, Jeon SG, Gho YS, Jee YK, Kim YK (2015) Active immunization with extracellular vesicles derived from Staphylococcus aureus effectively protects against staphylococcal lung infections, mainly via Th1 cell-mediated immunity. PLoS One 10:e0136021
    Pubmed KoreaMed CrossRef
  45. Choi HI, Choi JP, Seo J, Kim BJ, Rho M, Han JK, Kim JG (2017) Helicobacter pylori-derived extracellular vesicles increased in the gastric juices of gastric adenocarcinoma patients and induced inflammation mainly via specific targeting of gastric epithelial cells. Exp Mol Med 49:e330
    Pubmed KoreaMed CrossRef
  46. Vargoorani ME, Modarressi MH, Vaziri F, Motevaseli E, Siadat SD (2020) Stimulatory effects of Lactobacillus casei derived extracellular vesicles on toll-like receptor 9 gene expression and cytokine profile in human intestinal epithelial cells. J Diabetes Metab Disord 19:223-231
    Pubmed KoreaMed CrossRef
  47. Kim OY, Park HT, Dinh NTH, Choi SJ, Lee J, Kim JH, Lee SW, Gho YS (2017) Bacterial outer membrane vesicles suppress tumor by interferon-γ-mediated antitumor response. Nat Commun 8:626
    Pubmed KoreaMed CrossRef
  48. Le Maréchal C, Peton V, Plé C, Vroland C, Jardin J, Briard-Bion V, Durant G, Chuat V, Loux V, Foligné B, Deutsch SM, Falentin H, Jan G (2015) Surface proteins of Propionibacterium freudenreichii are involved in its anti-inflammatory properties. J Proteomics 113:447-461
    Pubmed CrossRef
  49. Al Shoyaib A, Archie SR, Karamyan VT (2019) Intraperitoneal route of drug administration: should it be used in experimental animal studies?. Pharm Res 37:12
    Pubmed KoreaMed CrossRef
  50. Garofalo M, Villa A, Rizzi N, Kuryk L, Mazzaferro V, Ciana P (2018) Systemic administration and targeted delivery of immunogenic oncolytic adenovirus encapsulated in extracellular vesicles for cancer therapies. Viruses 10:558
    Pubmed KoreaMed CrossRef