ARTICLE

Enhanced Longevity and Immunity in Caenorhabditis elegans through Ingestion of Lactiplantibacillus plantarum SKO-001: A Multi-Omics Study

Daniel Junpyo Lee1https://orcid.org/0000-0001-7224-3958, Ju Young Eor1https://orcid.org/0000-0002-3764-3339, Min-Jin Kwak1https://orcid.org/0000-0001-9832-3251, Junbeom Lee1https://orcid.org/0000-0001-6502-1556, An Na Kang1https://orcid.org/0000-0003-0208-6234, Daye Mun1https://orcid.org/0000-0002-3470-7632, Hyejin Choi1https://orcid.org/0000-0002-5977-2780, Min-Geun Kang1https://orcid.org/0000-0002-2204-6443, Youbin Choi1https://orcid.org/0000-0002-9444-3237, Hee Seo2https://orcid.org/0009-0007-4402-7642, Jae Yeong Ju2https://orcid.org/0009-0003-9232-7202, Minho Song3https://orcid.org/0000-0002-4515-5212, Jun-Mo Kim4https://orcid.org/0000-0002-6934-398X, Jungwoo Yang5https://orcid.org/0000-0003-3836-729X, Hyung Wook Kim6https://orcid.org/0009-0003-4839-3827, Sangnam Oh7https://orcid.org/0000-0002-2428-412X, Younghoon Kim1,*https://orcid.org/0000-0001-6769-0657
Author Information & Copyright
1Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Korea
2Food Science R&D Center, Kolmar BNH Co., Ltd., Seoul 06800, Korea
3Department of Animal Science and Biotechnology, Chungnam National University, Daejeon 34134, Korea
4Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Korea
5Department of Microbiology, College of Medicine, Dongguk University, Gyeongju 38066, Korea
6College of Life Sciences, Sejong University, Seoul 05006, Korea
7Department of Food and Nutrition, Jeonju University, Jeonju 55069, Korea
*Corresponding author : Sangnam Oh, Department of Food and Nutrition, Jeonju University, Jeonju 55069, Korea Tel: +82-63-220-3109 Fax: +82-63-220-2054 E-mail: osangnam@jj.ac.kr
*Corresponding author : Younghoon Kim, Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Korea Tel: +82-2-880-4808 Fax: +82-2-873-2271 E-mail: ykeys2584@snu.ac.kr

© Korean Society for Food Science of Animal Resources. This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Received: Aug 02, 2024 ; Revised: Sep 05, 2024 ; Accepted: Sep 12, 2024

Published Online: Sep 01, 2025

Abstract

Lactic acid bacteria are widely used as probiotics owing to their health-promoting properties. This study aimed to evaluate Lactiplantibacillus plantarum SKO-001 (SKO-001) as a probiotic candidate using Caenorhabditis elegans as a model organism. Our findings indicate that SKO-001 shows significantly stronger adhesive properties in C. elegans compared to Escherichia coli OP50, a standard dietary component used in laboratory settings for C. elegans, and the well-known probiotic Lacticaseibacillus rhamnosus GG (LGG). SKO-001 led to a significant increase in the longevity of C. elegans compared to those fed OP50. Additionally, pre-conditioning with SKO-001 significantly enhanced resistance to foodborne pathogenic bacteria. Transcriptomic analysis revealed that C. elegans fed with SKO-001 showed a significant increase in the expression of genes involved in the innate immune system, particularly those related to C-type lectins and lysozymes, compared to those fed with OP50. This suggests that feeding SKO-001 may boost immune responses against pathogens. Metabolomic analysis showed higher levels of lactic acid, L-valine, and L-isoleucine in C. elegans fed SKO-001 than in those fed OP50. Taken together, this research demonstrates the health-promoting potential of L. plantarum SKO-001 through multi-omics analysis, highlighting its capacity to extend lifespan and boost immune response in C. elegans.

Keywords: Lactiplantibacillus plantarum; Caenorhabditis elegans; longevity; immune response; multi-omics analysis

Introduction

Lactobacillus, a widely used probiotic, is recognized for its ability to extend lifespan, boost the immune system, and promote growth (Lee et al., 2022; Oh et al., 2023). Additionally, Lactobacillus species possess protective properties by preventing the invasion and colonization of pathogens and producing antipathogenic metabolites like lactic acid, hydrogen peroxide, bacteriocins, and phenylacetic acid (Chae et al., 2024; Cho et al., 2024; Eum et al., 2024; Han et al., 2024; Park et al., 2023b). They also provide immunological benefits by modulating the host’s immune function (Dimitrijevic et al., 2014; Jaafar et al., 2024). Within this group, Lactobacillus plantarum, recently denominated as Lactiplantibacillus plantarum stands out as a promising probiotic (Kim et al., 2023a; Ryu et al., 2023; Song et al., 2025; Yang et al., 2022). The safety of L. plantarum SKO-001, used in this study, has been confirmed in previous studies conducted on both mice and humans (Choi et al., 2023b; Shin et al., 2024).

Caenorhabditis elegans is an ideal surrogate animal model for studying microbe-host interactions (Kumar et al., 2020). C. elegans is extensively utilized as a model organism in research, owing to its numerous advantages, including a short lifespan, simple genetics, cost-effectiveness, ease of handling, and suitability for high-throughput screening (Choi et al., 2023a; Kang et al., 2024; Kim and Mylonakis, 2012; Lee et al., 2024b). Many research groups use C. elegans as a host model to evaluate the probiotic characteristics of various candidate strains (Kim and Mylonakis, 2012; Kim et al., 2021; Park et al., 2018). Bacteria commonly used as dietary resources for C. elegans can influence its phenotypes, including lifespan (Grompone et al., 2012; Kang et al., 2025) and immune response (Kim and Mylonakis, 2012; Park et al., 2020). Administering bacteria directly to C. elegans allows researchers to study microbe-host interactions while minimizing the influence of external nutrients. This makes C. elegans a suitable model organism for studying the characteristics of probiotics and their effects on hosts.

Whole-transcriptome analysis is commonly used to explore changes in multiple genetic pathways in C. elegans. C. elegans is well-suited for investigating genetic pathways and observing the effects of probiotics on aging and innate immunity (Kim et al., 2021; Lee et al., 2024b). Additionally, metabolomics has been used in C. elegans studies to identify metabolites linked to longevity and innate immunity (Lee et al., 2024a).

Various studies with C. elegans have shown that the consumption of specific Lactobacillus strains can enhance both lifespan and immune health (Kim et al., 2021; Lee et al., 2024a; Park et al., 2018). This study aimed to assess the probiotic properties of the candidate strain L. plantarum SKO-001 in C. elegans using multi-omics analysis.

Materials and Methods

Bacterial strains and culture conditions

In all experiments, the C. elegans strain fer-15; fem-1, which cannot produce progeny at 25°C, was used. Escherichia coli OP50 (OP50) was cultured in Luria-Bertani broth (BD Biosciences, Sparks, MD, USA) at 37°C for 24 h. Lacticaseibacillus rhamnosus GG (LGG) and L. plantarum SKO-001 (Accession No. KCTC 14816BP) were grown in De Man-Rogosa-Sharpe broth (BD Biosciences, Sparks, MD, USA) at 37°C for 48 h. SKO-001 was isolated from Angelica gigas Nakai and obtained from Kolmar BNH (Seoul, Korea). Four foodborne pathogenic bacteria were cultured as follows: E. coli O157:H7 EDL933 in Luria-Bertani broth at 37°C for 24 h, Salmonella Typhimurium SL1344 in nutrient broth (BD Biosciences) at 37°C for 24 h, and Staphylococcus aureus Newman and Listeria monocytogenes EGD-e in Brain Heart Infusion broth (BD Biosciences) at 37°C for 24 h.

In vivo adhesive assay using Caenorhabditis elegans

To evaluate the colonization of SKO-001 in the C. elegans intestine, adhesion assays were conducted following established protocols (Lee et al., 2024a). C. elegans were cultured on NGM plates with OP50 until they contain eggs. Eggs were extracted using a sodium hypochlorite-sodium hydroxide solution, and synchronized L1 worms were grown on NGM plates with OP50 until the L4 stage at 25°C. Worms were then transferred to NGM plates seeded with OP50, SKO-001, or LGG [all at 8.0×109 colony-forming units (CFU/mL)]. After 48 h, 10 worms from each group were placed on Brain Heart Infusion agar with gentamycin (25 μg/mL) for 5 min. Worms were transferred to 1.5-mL Eppendorf tube containing M9 buffer with Triton X-100, then mechanically disrupted. Samples were spread on Luria-Bertani agar for OP50 and De Man-Rogosa-Sharpe agar for LGG and SKO-001, incubated at 37°C for 48 h. The experiment had 6 replicates per treatment, with 10 worms per replicate, totaling 60 worms per treatment group.

Caenorhabditis elegans life span and killing assay

To evaluate the impact of SKO-001 on C. elegans longevity and immune response to foodborne pathogens, slight modifications were made to previously established methods (Lee et al., 2024a; Park et al., 2018).

For lifespan assay, synchronized L1 stage worms were grown on NGM agar plates with OP50 until the L4 stage at 25°C. The worms were then individually transferred to 35-mm NGM agar plates seeded with OP50, SKO-001, or LGG (all at 8.0×109 CFU/mL). Each treatment group consisted of 90 worms, split across three plates (30 worms per plate), and kept at 25°C. Survival was recorded daily, and worms were moved to fresh plates every two days. Worms were assessed as alive or dead by gently touching them with a platinum wire. The experiment continued until all C. elegans in each group died.

For killing assay, L4 stage worms were transferred to 35-mm NGM agar plates seeded with OP50, SKO-001, or LGG (all at 8.0×109 CFU/mL) for a 48 h pre-conditioning period. Worms were then moved to NGM plates containing foodborne pathogens: E. coli O157:H7 EDL933, S. Typhimurium SL1344, S. aureus Newman, and L. monocytogenes EGD-e (all at 8.0×109 CFU/mL) and kept at 25°C. Each treatment group had 90 worms, divided across three plates (30 worms per plate). Worms were enumerated daily and moved to fresh plates every two days, with survival checked by gently touching them with a platinum wire. The experiment continued until all worms in each treatment group died.

Body size and locomotive activity

Locomotion and body dimensions were evaluated using Wormlab® software (MBF Bioscience, Williston, VT, USA), with slight modifications from the previous study (Shen et al., 2018). L4 stage C. elegans were exposed to OP50, SKO-001, or LGG (all at 8.0×109 CFU/mL) for 48 h, then moved to low-peptone NGM plates seeded with OP50. Filming began after a 10 min acclimation, with each video lasting 1 min for tracking analysis. Measurements included width, length, and peristaltic speed (μm/s). Ten worms per group were evaluated, and experiments were performed in triplicate.

The pharyngeal pumping rate, indicating food intake, was measured using a stereomicroscope by counting pharyngeal contractions over 30 s. At least 10 worms per group were measured, with all experiments conducted in triplicate.

RNA isolation and transcriptomic analysis

Transcriptomic analysis was conducted with slight modifications (Ryu et al., 2021). L4 stage C. elegans were placed on NGM plates containing either OP50 (8.0×109 CFU/mL) or SKO-001 (8.0×109 CFU/mL). After a 48 h, total RNA was extracted using TRIZOL (Invitrogen, Carlsbad, CA, USA) and purified with the RNeasy Mini Kit (QIAGEN, Valencia, CA, USA). RNA-seq was performed using a TruSeq RNA Sample Prep Kit v2 (Illumina, San Diego, CA, USA) and sequenced on an Illumina NovaSeq 6000 platform with paired-end reads (2×150 bp). Trimmomatic 0.38 was used for quality trimming (Bolger et al., 2014). Reads shorter than 36 bp were discarded. Hisat2 v2.1.0 was used to create the reference genome index, and uniquely mapped reads were quantified using Subread/featureCounts v1.5.1. Genes with |Log2-fold change|>1 and p-value<0.05 were considered significantly different. In this experiment, only genes with a significant difference (p-value<0.05) and a fold change greater than 2 were included in the transcriptomic analysis. Functions of differentially expressed genes were identified using Database for Annotation, Visualization, and Integrated Discovery (DAVID), with network analysis performed using Cytoscape.

Metabolites extraction and metabolomic analysis

Metabolomic analysis was performed to evaluate metabolite variations using the previous method (Lee et al., 2024b). L4 stage worms were provided with either OP50 (8.0×109 CFU/mL) or SKO-001 (8.0×109 CFU/mL) for 48 h. The worms were rinsed six times with sterile deionized water, homogenized, combined with ice-cold methanol, vortexed, and centrifuged at 10,000×g for 10 min at 4°C. The supernatants were then filtered through 0.2 μm syringe filters and vacuum dried.

For the gas chromatography-mass spectrometry (GC-MS) analysis, each sample was treated with 30 μL of methoxyamine hydrochloride (Sigma-Aldrich, St. Louis, MO, USA) in pyridine (20 mg/mL) and incubated at 30°C for 90 min. Trimethylsilylation was then carried out by adding 50 μL of N,O-bis(trimethylsilyl)trifluoroacetamide (Sigma-Aldrich) and incubating at 60°C for 30 min, followed by 10 μL of fluoranthene adding (Sigma-Aldrich).

GC-MS analysis used a TRACE™ 1310 Gas Chromatograph (Thermo Fisher Scientific, Waltham, MA, USA) with an ISQ LT mass spectrometer (Thermo Fisher Scientific). Compounds were separated on a DB-5MS column (60 m×0.25 mm, 0.25-μm film thickness, Agilent, Santa Clara, CA, USA). The temperature program started at 50°C for 2 min, ramped to 180°C at 5°C/min (held for 8 min), then increased to 325°C at 2.5°C/min (held for 10 min). Samples were injected at 300°C with helium as the carrier gas at 1.5 mL/min and a split ratio of 1:60. Detection used electron ionization at 70 eV and an ion source temperature of 270°C. The mass spectrometer scanned from 30 to 450 m/z at 5 spectra/sec. Metabolites were identified using the NIST Mass Spectral Library (version 2.0, NIST, Gaithersburg, MD, USA) and data analyzed with MetaboAnalyst 5.0 (Pang et al., 2021). Only metabolites with a match score above 850 in the NIST library were included in the metabolomic analysis for this study.

Statistics

The Kaplan–Meier method was used to analyze the lifespan and killing assay data for C. elegans, and results were visualized with SigmaPlot 12.0 (Systat Software, Chicago, IL, USA). Statistical analysis of other datasets was performed using Prism 9 (GraphPad Software, San Diego, CA, USA). Significance levels were set at p-values of <0.05 (*), 0.01 (**), 0.001 (***), and 0.0001 (****). Graphs are presented as mean±SEM.

Data availability

The manuscript contains all the data needed to support the study’s conclusions and has been uploaded to the NCBI SRA database under Bioproject number PRJNA1132481.

Results

Evaluation of Lactiplantibacillus plantarum SKO-001 adhesion ability in Caenorhabditis elegans

We employed C. elegans to investigate the in vivo adhesion properties of Lactiplantibacillus plantarum SKO-001 (SKO-001). E. coli OP50 (OP50) served as a negative control, while LGG was used as a positive control. The adhesion assessment was performed following 48 h of exposure to OP50, SKO-001, or LGG. Our findings revealed that SKO-001 exhibited a significantly greater adhesion capability compared to both OP50 and LGG (p<0.0001 for each comparison with OP50 and LGG; Fig. 1). This suggests that SKO-001 shows remarkable adhesive properties in C. elegans in comparison to OP50 and LGG.

kosfa-45-5-1293-g1
Fig. 1. Adhesion ability of Lactiplantibacillus plantarum SKO-001 in Caenorhabditis elegans. Adhesion ability of OP50, SKO-001, or LGG in C. elegans strain fer-15; fem-1 after a 48 h exposure period. Data are expressed as means±SEM. Statistical analysis is conducted using a one-way analysis of variance, and statistical significance is considered when p-values are <0.05 (*), <0.01 (**), <0.001 (***), and <0.0001 (****). Statistical comparisons with SKO-001: p<0.0001 for both OP50 and LGG. OP50, Escherichia coli OP50; SKO-001, L. plantarum SKO-001; LGG, Lacticaseibacillus rhamnosus GG.
Download Original Figure
Analysis of the influence of Lactiplantibacillus plantarum SKO-001 on longevity and immune response in Caenorhabditis elegans

We explored the impact of SKO-001 on the lifespan of C. elegans by treating the worms with OP50, SKO-001, or LGG. The group receiving OP50 was labeled as the OP50 group, the group receiving SKO-001 was labeled as the SKO-001 group, and the group receiving LGG was labeled as the LGG group. C. elegans in the SKO-001 group had a significantly longer lifespan compared to those in the OP50 group (p=0.0000; Fig. 2A). Moreover, no significant difference in lifespan was observed between the SKO-001 and LGG groups (p=0.1506; Fig. 2A). These findings suggest that SKO-001 treatment significantly prolonged the lifespan of C. elegans.

kosfa-45-5-1293-g2
Fig. 2. Lifespan and killing assay of Caenorhabditis elegans fed with Lactiplantibacillus plantarum SKO-001. Lifespan of C. elegans strain fer-15; fem-1 fed OP50, SKO-001, and LGG. For the killing assay, C. elegans strain fer-15; fem-1 was pre-conditioned with OP50, SKO-001, or LGG for 48 h and then infected with foodborne pathogenic bacteria (two gram-negative and two gram-positive bacteria). (A) Lifespan assay of C. elegans, (B) killing assay using Escherichia coli O157:H7 EDL933 cells, (C) killing assay using Salmonella Typhimurium SL1344, (D) killing assay using Staphylococcus aureus Newman, (E) killing assay using Listeria monocytogenes EGD-e. Statistical analysis is conducted using Kaplan–Meier method, and differences are considered significant when the p-value is <0.05 (*) and <0.01 (**) compared to OP50. Survival statistics in the lifespan assay compared to SKO-001: p=0.0000 and p=0.1506 for OP50 and LGG, respectively. Survival statistics for the killing assay compared to SKO-001: E. coli O157:H7 EDL933, p=0.0003 and p=0.6531 for OP50 and LGG, respectively; S. Typhimurium SL1344, p=0.0000 and p=0.8388 for OP50 and LGG, respectively; S. aureus Newman, p=0.0000 and p=0.1506 for OP50 and LGG, respectively; L. monocytogenes EGD-e, p=0.0000 and p=0.3670 for OP50 and LGG, respectively. OP50, E. coli OP50; SKO-001, L. plantarum SKO-001; LGG, Lacticaseibacillus rhamnosus GG.
Download Original Figure

We next performed killing assays to assess whether SKO-001 improved the ability of C. elegans to defend against various foodborne pathogenic bacteria. After a 48 h pre-conditioning period with OP50, SKO-001, or LGG, the worms were placed on NGM plates seeded with foodborne pathogens. E. coli O157:H7 EDL933 and S. Typhimurium SL1344, both gram-negative bacteria, and S. aureus Newman and L. monocytogenes EGD-e, both gram-positive bacteria, were used. C. elegans that had been pre-conditioned with SKO-001 showed a significantly reduced susceptibility to the gram-negative bacteria compared to those pre-conditioned with OP50 (p=0.0003 for E. coli O157:H7 EDL933 and p=0.0000 for S. Typhimurium SL1344; Figs. 2B and C). However, no significant differences in susceptibility were observed between C. elegans pre-conditioned with SKO-001 and those pre-conditioned with LGG (p=0.6531 for E. coli O157:H7 EDL933 and p=0.8388 for S. Typhimurium SL1344; Figs. 2B and C). In experiments involving gram-positive bacteria, C. elegans pre-conditioned with SKO-001 showed significantly better survival rates compared to those pre-conditioned with OP50 (p=0.0000 for S. aureus Newman and p=0.0000 for L. monocytogenes EGD-e; Figs. 2D and E). No notable difference in survival was observed between C. elegans pre-conditioned with SKO-001 and those pre-conditioned with LGG (p=0.1506 for S. aureus Newman and p=0.3670 for L. monocytogenes EGD-e; Figs. 2D and E). Overall, these findings suggest that pre-conditioning with SKO-001 improves the resistance of C. elegans to infections caused by both gram-negative and gram-positive pathogenic bacteria.

Evaluation of the impact of Lactiplantibacillus plantarum SKO-001 on Caenorhabditis elegans phenotype

To evaluate the impact of SKO-001 on C. elegans’ phenotype, we assessed body size and locomotive activity. Worms fed SKO-001 exhibited significantly larger body dimensions, including both length and width, compared to those fed OP50 (p<0.0001 for both length and width) and LGG (p<0.0001 for both length and width; Figs. 3A and B). However, no significant difference in peristaltic speed, a measure of worm activity, was observed between the SKO-001 and OP50 groups (p=0.7777; Fig. 3C). Similarly, the peristaltic speed was comparable between the SKO-001 and LGG groups (p=0.9783; Fig. 3C). In the pumping rate assay, which reflects food intake, worms in the SKO-001 group showed a significant increase compared to those in the OP50 and LGG groups (p<0.0001 for OP50 and p=0.0015 for LGG; Fig. 3D). Overall, these findings suggest that SKO-001 enhances both body size and pumping rate in C. elegans.

kosfa-45-5-1293-g3
Fig. 3. Body size and locomotive activity of Caenorhabditis elegans fed with Lactiplantibacillus plantarum SKO-001. Body size and locomotive activity of C. elegans strains fer-15; fem-1 after a 48-h exposure period with OP50, SKO-001, or LGG (A) length, (B) width, (C) peristaltic speed, and (D) pumping rate. Data are expressed as means±SEM. Statistical analysis is conducted using a one-way analysis of variance, and statistical significance is considered when p-values are <0.05 (*), <0.01 (**), <0.001 (***), and <0.0001 (****). Statistics compared to SKO-001: length, p<0.0001 and p<0.0001 for OP50 and LGG, respectively; width, p<0.0001 and p<0.0001 for OP50 and LGG, respectively; peristaltic speed, p=0.7777 and p=0.9783 for OP50 and LGG, respectively; pumping rate, p<0.0001 and p=0.0015 for OP50 and LGG, respectively. OP50, Escherichia coli OP50; SKO-001, L. plantarum SKO-001; LGG, Lacticaseibacillus rhamnosus GG.
Download Original Figure
Transcriptomic analysis of Caenorhabditis elegans after exposure to Lactiplantibacillus plantarum SKO-001

A transcriptomic analysis was conducted to investigate the gene expression alterations in C. elegans induced by SKO-001 feeding in comparison to OP50. Genes showing more than a 2-fold increase in expression with SKO-001 were identified and examined using DAVID to determine associated upregulated pathways. The top 10 pathways related to these significantly upregulated genes are detailed in Table 1. Consistent with the findings from the killing assays, pathways related to the innate immune response and defense mechanisms against both gram-positive and gram-negative bacteria were notably upregulated in SKO-001-fed C. elegans. Specifically, genes linked to C-type lectins (clec-41, clec-66, clec-86, clec-186, and clec-187) and lysozymes (lys-1, lys-2, lys-3, lys-7, and lys-8) were significantly upregulated in response to SKO-001 (Table 2). To identify the Kyoto Encyclopedia of Genes and Genomes pathways upregulated by feeding SKO-001, Cytoscape was performed with genes that exhibited more than a 2-fold increase in C. elegans fed SKO-001 compared to those fed OP50. The results identified several pathways that were significantly upregulated with SKO-001, including those involved in drug metabolism, tryptophan metabolism, lysosomes, glycine, serine, and threonine metabolism, sphingolipid metabolism, glycerophospholipid metabolism, longevity-regulating pathways, and arginine and proline metabolism (Fig. 4). These results collectively indicate that SKO-001 enhances immune response and promotes longevity.

Table 1. Transcriptomic analysis of Caenorhabditis elegans fed with Lactiplantibacillus plantarum SKO-001
Term1) Gene count (%) p-value
Innate immune response 75 (8.4) 0.000
Defense response to gram-positive bacteria 20 (2.2) 0.000
Anatomical structure development 23 (2.6) 0.000
Peptidoglycan catabolic process 6 (0.7) 0.000
Cell wall macromolecule catabolic process 6 (0.7) 0.000
Defense response to gram-negative bacteria 12 (1.3) 0.000
Glutathione metabolic process 10 (1.1) 0.000
Proteolysis 26 (2.9) 0.002
Lipid metabolic process 19 (2.1) 0.003
Lipid transport 7 (0.8) 0.003

1) The top 10 pathways associated with genes that are significantly upregulated by >2.0 folds in C. elegans after 48 h of exposure to L. plantarum SKO-001 compared to Escherichia coli OP50.

Download Excel Table
Table 2. Transcriptomic analysis of Caenorhabditis elegans fed with Lactiplantibacillus plantarum SKO-001
Group and gene1) Gene number Fold change p-value Description
C-type lectin-related
clec-41 CELE_B0365.6 5.315288 0.000 C-type lectin
clec-66 CELE_F35C5.9 2.534151 0.000 C-type lectin
clec-86 CELE_C54D1.2 6.759153 0.000 C-type lectin
clec-186 CELE_ZK896.7 5.362350 0.000 C-type lectin
clec-187 CELE_ZK896.6 4.920694 0.000 C-type lectin
Lysozyme-related
lys-1 CELE_Y22F5A.4 2.592985 0.000 Lysozyme
lys-2 CELE_Y22F5A.5 6.513251 0.000 Lysozyme
lys-3 CELE_Y22F5A.6 2.661261 0.000 Lysozyme
lys-7 CELE_C02A12.4 3.604976 0.000 Lysozyme
lys-8 CELE_C17G10.5 2.087275 0.000 Lysozyme

1) The list of genes associated with the innate immune response pathway that are significantly upregulated by >2.0 folds in C. elegans after 48 h of exposure to L. plantarum SKO-001 compared to Escherichia coli OP50.

Download Excel Table
kosfa-45-5-1293-g4
Fig. 4. Transcriptomic analysis of Caenorhabditis elegans fed with Lactiplantibacillus plantarum SKO-001. The identification of Kyoto Encyclopedia of Genes and Genomes pathways related to genes is significantly upregulated by >2.0 folds in C. elegans after 48 h of exposure to L. plantarum SKO-001 as compared to Escherichia coli OP50. Cytoscape is used for the analysis.
Download Original Figure
Metabolomic analysis of Caenorhabditis elegans after exposure to Lactiplantibacillus plantarum SKO-001

Metabolomic analysis was conducted to evaluate the effect of SKO-001 on the metabolite composition of C. elegans. The partial least squares-discriminant analysis revealed distinct clustering of metabolite profiles between C. elegans fed SKO-001 and those fed OP50 (Fig. 5A). A heatmap of the top 12 most significantly altered metabolites revealed increased levels of carbamic acid, lactic acid, L-valine, and L-isoleucine in C. elegans receiving SKO-001, compared to the OP50 group (Fig. 5B). Quantitative analysis highlighted that metabolites such as lactic acid, succinic acid, L-aspartic acid, and 3-oxaoct-4-en-2-imine were elevated by more than 2-fold in SKO-001 fed C. elegans (Fig. 5C). Additionally, a volcano plot showed that seven metabolites lactic acid, carbamic acid, L-isoleucine, L-valine, 3-oxaoct-4-en-2-imine, nonanoic acid, and L-aspartic acid were significantly upregulated in the SKO-001 group compared to the OP50 group (Fig. 5D). Overall, these findings indicate that SKO-001 alters the metabolite profile of C. elegans, with a notable increase in several key metabolites, including lactic acid.

kosfa-45-5-1293-g5
Fig. 5. Metabolomic analysis of Caenorhabditis elegans fed with Lactiplantibacillus plantarum SKO-001. Comparison of the metabolite composition of C. elegans after 48 h of exposure to L. plantarum SKO-001 and Escherichia coli OP50. (A) PLS-DA, (B) volcano plot, (C) the top 12 enriched heat maps, (D) quantitative graph depicting metabolites that changes by >2.0 folds.
Download Original Figure

Discussion

Lactic acid bacteria, especially those belonging to the Lactobacillus genus, are well-known for their beneficial effects on health and are frequently used as probiotics. Lactobacillus plantarum has been reported to positively influence longevity and immune responses in various studies (Kim et al., 2022; Kumar et al., 2022; Yu et al., 2022). Therefore, we investigated the potential of L. plantarum SKO-001 (SKO-001) as a probiotic candidate using C. elegans.

Previous studies have demonstrated that the ability of probiotic bacteria to adhere to the host’s gastrointestinal tract is a key criterion in their selection. This adhesive ability facilitates colonization and enhances immunomodulatory effects by stimulating the gut barrier and metabolic function (Kim et al., 2023b; Kim et al., 2024; Song et al., 2023). Consequently, probiotics can survive, proliferate, and deliver numerous health benefits to their host (Kinara et al., 2024; Oh et al., 2022; Park et al., 2023a; Park et al., 2024). In our study, SKO-001 demonstrated significantly higher adhesive ability than OP50 and LGG. Lifespan measurements are extensively used to study aging processes. C. elegans, with its short lifespan, is a suitable in vivo model for measuring the ability of candidate probiotic bacteria (Tissenbaum, 2015). In the lifespan assay, SKO-001 significantly increased the lifespan of C. elegans compared to OP50, showing no significant difference from LGG. This result supports earlier findings that Lactobacillus species with probiotic properties can increase the lifespan of C. elegans (Heo et al., 2018; Lee et al., 2024a). Similarly, the killing assay revealed that pre-conditioning with SKO-001 significantly improved the immune response of C. elegans against both gram-negative and gram-positive pathogenic bacteria. This observation aligns with previous research, which underscores the strong antimicrobial properties of L. plantarum against pathogens and its capacity to boost the immune response in C. elegans (Li et al., 2017; Mun et al., 2019).

The quality of food affects worm phenotypes (Shtonda and Avery, 2006). Additionally, different bacteria can impact the growth of C. elegans to varying degrees (Avery and You, 2018). Therefore, we measured worm size and locomotor activity to assess the quality of SKO-001 as a food source and to determine whether it could alter the phenotype of the worms. Worms fed SKO-001 exhibited a significant increase in both length and width compared to those fed OP50 and LGG. Furthermore, SKO-001 also improved the pumping rate more effectively than OP50 and LGG. These results indicate that SKO-001 not only caused notable phenotypic changes in the worms but also enhanced their growth performance.

Our study indicates that pre-conditioning C. elegans with SKO-001 enhances its immune defense against foodborne pathogens. We hypothesized that pre-conditioning upregulates specific immune-related genes. Transcriptomic analysis revealed that genes with more than a 2-fold increase in C. elegans fed SKO-001, compared to those fed OP50, were predominantly associated with innate immunity.

Notably, genes related to C-type lectins (clec-41, clec-66, clec-86, clec-186, and clec-187) and lysozymes (lys-1, lys-2, lys-3, lys-7, and lys-8) showed significant upregulation following SKO-001 treatment. In C. elegans, clec genes encode a variety of proteins with C-type lectin-like domains, which play a role in pathogen defense (Schulenburg et al., 2008). Previous studies have shown that clec-41 plays a vital role in the resistance to the gram-positive pathogen Bacillus thuringiensis MYBt18247 (Pees et al., 2021). Similarly, clec-86 has been demonstrated to be essential for defense against the gram-positive pathogen Microbacterium nematophilum (O’Rourke et al., 2006). These results suggested that clec-41 and clec-86 play a crucial role in resistance against pathogenic bacteria. Consistent with prior studies, the expression levels of both clec-41 and clec-86 were notably higher in C. elegans fed SKO-001 compared to those fed OP50. Lysozymes function as antimicrobial agents within the C. elegans gut, breaking down bacterial cells (Ciancio, 2016). Genes related to lysozymes, including lys-1, lys-3, and lys-7, are essential for the defense mechanisms in C. elegans (Schulenburg et al., 2008). We found that these genes were significantly upregulated following SKO-001 feeding. This suggests that the enhanced expression of both C-type lectin and lysozyme-related genes induced by SKO-001 contributes to a more robust immune response against pathogenic bacteria.

In the metabolomic analysis, C. elegans fed SKO-001 showed a notable increase in lactic acid compared to those fed OP50. Lactic acid, often produced by lactic acid bacteria, is associated with enhanced defense and resistance in C. elegans (Fernández et al., 2003). The higher levels of lactic acid observed with SKO-001 treatment likely contributed to an improved immune response against pathogens. Additionally, the branched-chain amino acids L-isoleucine and L-valine, which were elevated in SKO-001 fed C. elegans, are crucial for various physiological processes. Previous studies have demonstrated that supplementing C. elegans with L-valine and L-isoleucine can significantly prolong their lifespan (Wang and Zhang, 2018; Wang et al., 2018). Collectively, the increased levels of metabolites observed with SKO-001 feeding may have contributed to enhanced longevity and improved immune response in C. elegans.

Conclusion

In conclusion, we investigated the probiotic potential of L. plantarum SKO-001 (SKO-001) using C. elegans as a model organism. SKO-001 showed superior adhesion capabilities compared to OP50 and LGG, suggesting its effectiveness in gastrointestinal colonization. Additionally, SKO-001 significantly prolonged the lifespan of C. elegans, improved its resistance to foodborne pathogens, and supported its growth. Transcriptomic analysis revealed notable upregulation of genes related to the innate immune system, particularly those involved in C-type lectins and lysozymes. Metabolomic analysis showed increased levels of lactic acid, L-valine, and L-isoleucine in C. elegans treated with SKO-001. Overall, our findings suggest that L. plantarum SKO-001 is a promising probiotic with potential benefits for improving longevity and boosting immune function.

Conflicts of Interest

The authors declare no potential conflicts of interest.

Acknowledgements

This study was supported by a National Research Foundation of Korea Grant, funded by the Korean government (MEST) (NRF-2021R1A2C3011051), and by the Korea Institute of Planning and Evaluation for Technology in Food, Agriculture, Forestry and Fisheries (IPET-321037-5) and by the Korea Evaluation Institute of Industrial Technology (KEIT, 20012411).

Author Contributions

Conceptualization: Lee DJ, Eor JY, Kwak MJ, Lee J, Kang AN, Mun D, Choi H, Kang MG, Choi Y, Seo H, Ju JY, Song M, Kim JM, Yang J, Kim HW, Oh S, Kim Y. Investigation: Lee DJ, Eor JY, Kwak MJ, Lee J, Kang AN, Mun D, Choi H, Kang MG, Choi Y, Seo H, Ju JY. Writing - original draft: Lee DJ, Eor JY, Kwak MJ, Lee J, Kang AN, Mun D, Choi H, Kang MG, Choi Y, Seo H, Ju JY, Song M, Kim JM, Yang J, Kim HW, Oh S, Kim Y. Writing - review & editing: Lee DJ, Eor JY, Kwak MJ, Lee J, Kang AN, Mun D, Choi H, Kang MG, Choi Y, Seo H, Ju JY, Song M, Kim JM, Yang J, Kim HW, Oh S, Kim Y.

Ethics Approval

This article does not require IRB/IACUC approval because there are no human and animal participants.

References

1.

Avery L, You YJ. 2018. C. elegans feeding. Available from: https://www.ncbi.nlm.nih.gov/books/NBK116080/Accessed at Jan 30, 2024.

2.

Bolger AM, Lohse M, Usadel B. 2014; Trimmomatic: A flexible trimmer for illumina sequence data. Bioinformatics. 30:2114-2120

3.

Chae JP, Vasquez R, Song JH, Pajarillo E, Hwang IC, Kang DK. 2024 Surface displayed porcine epidemic diarrhea virus membrane epitopes on Lactiplantibacillus plantarum stimulates antibody production in mice. J Anim Sci Technol (in press).

4.

Cho E, Yoo Y, Yoon Y. 2024 Antimicrobial activity of Pediococcus pentosaceus strains against diarrheal pathogens isolated from pigs and effect on paracellular permeability of HT-29 cells. J Anim Sci Technol (in press).

5.

Choi H, Mun D, Ryu S, Kwak M, Kim BK, Park DJ, Oh S, Kim Y. 2023a; Molecular characterization and functionality of rumen-derived extracellular vesicles using a Caenorhabditis elegans animal model. J Anim Sci Technol. 65:652-663

6.

Choi MJ, Yu H, Kim JI, Seo H, Kim JG, Kim SK, Lee HS, Cheon HG. 2023b; Anti-obesity effects of Lactiplantibacillus plantarum sko-001 in high-fat diet-induced obese mice. Eur J Nutr. 62:1611-1622

7.

Ciancio A. 2016 Defense and immune systems. In Invertebrate bacteriology: Function, evolution and biological ties. Ciancio A. edSpringer. Dordrecht, The Netherlands:

8.

Dimitrijevic R, Ivanovic N, Mathiesen G, Petrusic V, Zivkovic I, Djordjevic B, Dimitrijevic L. 2014; Effects of Lactobacillus rhamnosus LA68 on the immune system of C57Bl/6 mice upon oral administration. J Dairy Res. 81:202-207

9.

Eum BG, Elnar A, Jang Y, Kim GB. 2024 Complete genome sequence of Ligilactobacillus agilis LDTM47, bacteriocin-producing lactic acid bacteria isolated from broiler gastrointestinal tract. J Anim Sci Technol (in press).

10.

Fernández MF, Boris S, Barbés C. 2003; Probiotic properties of human lactobacilli strains to be used in the gastrointestinal tract. J Appl Microbiol. 94:449-455

11.

Grompone G, Martorell P, Llopis S, González N, Genovés S, Mulet AP, Fernández-Calero T, Tiscornia I, Bollati-Fogolín M, Chambaud I. 2012; Anti-inflammatory Lactobacillus rhamnosus CNCM I-3690 strain protects against oxidative stress and increases lifespan in Caenorhabditis elegans. PLOS ONE. 7e52493

12.

Han S, Elnar AG, Lim C, Kim GB. 2024; Complete genome sequence of bacteriocin-producing Ligilactobacillus salivarius B4311 isolated from fecal samples of broiler chicken with anti-listeria activity. J Anim Sci Technol. 66:232-236

13.

Heo J, Shin D, Chang SY, Bogere P, Park MR, Ryu S, Lee WJ, Yun B, Lee HK, Kim Y, Oh S. 2018; Comparative genome analysis and evaluation of probiotic characteristics of Lactobacillus plantarum strain JDFM LP11. Korean J Food Sci Anim Resour. 38:878-888

14.

Jaafar MH, Xu P, Mageswaran UM, Balasubramaniam SD, Solayappan M, Woon JJ, Teh CSJ, Todorov SD, Park YH, Liu G, Liong MT. 2024; Constipation anti-aging effects by dairy-based lactic acid bacteria. J Anim Sci Technol. 66:178-203

15.

Kang A, Kwak MJ, Lee DJ, Lee JJ, Kim MK, Song M, Lee M, Yang J, Oh S, Kim Y. 2024; Dietary supplementation with probiotics promotes weight loss by reshaping the gut microbiome and energy metabolism in obese dogs. Microbiol Spectr. 12e0255223

16.

Kang AN, Lee J, Eor JY, Kwak MJ, Kim YA, Oh S, Kim Y. 2025; A comprehensive assessment of immunomodulatory potentials of Korean antler velvet extract in mouse and neurodegenerative Caenorhabditis elegans models. J Anim Sci Technol. 67:421-438

17.

Kim B, Kim K, Xu X, Lee H, Pathiraja D, Park DJ, Choi IG, Oh S. 2023a; Complete genome and two plasmids sequences of Lactiplantibacillus plantarum l55 for probiotic potentials. J Anim Sci Technol. 65:1341-1344

18.

Kim B, Meng Z, Xu X, Baek S, Pathiraja D, Choi IG, Oh S. 2023b; Complete genome sequence of Limosilactobacillus fermentum JNU532 as a probiotic candidate for the functional food and feed supplements. J Anim Sci Technol. 65:271-274

19.

Kim B, Xu X, Lee H, Pathiraja D, Jae-Young K, Choi YH, Choi IG, Kim SH. 2024; Complete genome sequence of candidate probiotic Limosilactobacillus fermentum KUFM407. J Anim Sci Technol. 66:859-862

20.

Kim H, Shin M, Ryu S, Yun B, Oh S, Park DJ, Kim Y. 2021; Evaluation of probiotic characteristics of newly isolated lactic acid bacteria from dry-aged Hanwoo beef. Food Sci Anim Resour. 41:468-480

21.

Kim JY, Kim JY, Kim H, Moon EC, Heo K, Shim JJ, Lee JL. 2022; Immunostimulatory effects of dairy probiotic strains Bifidobacterium animalis ssp. lactis HY8002 and Lactobacillus plantarum HY7717. J Anim Sci Technol. 64:1117-1131

22.

Kim Y, Mylonakis E. 2012; Caenorhabditis elegans immune conditioning with the probiotic bacterium Lactobacillus acidophilus strain NCFM enhances Gram-positive immune responses. Infect Immun. 80:2500-2508

23.

Kinara E, Moturi J, Mun J, Hosseindoust A, Tajudeen H, Ha S, Park SR, Lee S, Kim JS. 2024 Dietary supplementation of Lactobacillus salivarius in suckling and weanling piglets modulates intestinal microbiota, morphology and improves growth performance. J Anim Sci Technol (in press).

24.

Kumar A, Baruah A, Tomioka M, Iino Y, Kalita MC, Khan M. 2020; Caenorhabditis elegans: A model to understand host–microbe interactions. Cell Mol Life Sci. 77:1229-1249

25.

Kumar A, Joishy T, Das S, Kalita MC, Mukherjee AK, Khan MR. 2022; A potential probiotic Lactobacillus plantarum JBC5 improves longevity and healthy aging by modulating antioxidative, innate immunity and serotonin-signaling pathways in Caenorhabditis elegans. Antioxidants. 11:268

26.

Lee D, Goh TW, Kang MG, Choi HJ, Yeo SY, Yang J, Huh CS, Kim YY, Kim Y. 2022; Perspectives and advances in probiotics and the gut microbiome in companion animals. J Anim Sci Technol. 64:197-217

27.

Lee DJ, Eor JY, Kwak MJ, Lee J, Kang AN, Mun D, Choi H, Song M, Kim JN, Kim JM, Yang J, Kim HW, Oh S, Kim Y. 2024a; Metabolic regulation of longevity and immune response in Caenorhabditis elegans by ingestion of Lacticaseibacillus rhamnosus IDCC 3201 using multi-omics analysis. J Microbiol Biotechnol. 34:1109-1118

28.

Lee DJ, Kang AN, Lee J, Kwak MJ, Mun D, Lee D, Oh S, Kim Y. 2024b; Molecular characterization of Fusarium venenatum-based microbial protein in animal models of obesity using multi-omics analysis. Commun Biol. 7:133

29.

Li M, Lee K, Hsu M, Nau G, Mylonakis E, Ramratnam B. 2017; Lactobacillus-derived extracellular vesicles enhance host immune responses against vancomycin-resistant enterococci. BMC Microbiol. 17:66

30.

Mun SY, Kim SK, Woo ER, Chang HC. 2019; Purification and characterization of an antimicrobial compound produced by Lactobacillus plantarum em showing both antifungal and antibacterial activities. LWT-Food Sci Technol. 114:108403

31.

Oh HJ, Lee JP, Lee JH, Kim YJ, An JW, Chang SY, Go YB, Song DC, Cho HA, Jeon MG, Yoon YH, Cho JH. 2022; Effects of Pediococcus pentosaceus strains isolated from three different types of kimchi in ICR mice infected with Escherichia coli or Salmonella typhimurium. Korean J Agric Sci. 49:1-10

32.

Oh YJ, Lee J, Lim SK, Kwon MS, Lee S, Choi SP, Yu D, Oh Y, Park J, Choi HJ. 2023; Complete genome sequence of probiotic Lactobacillus johnsonii 7409N31 isolated from a healthy Hanwoo calf. J Anim Sci Technol. 65:890-893

33.

O’Rourke D, Baban D, Demidova M, Mott R, Hodgkin J. 2006; Genomic clusters, putative pathogen recognition molecules, and antimicrobial genes are induced by infection of Celegans with M. nematophilum. Genome Res. 16:1005-1016

34.

Pang Z, Chong J, Zhou G, de Lima Morais DA, Chang L, Barrette M, Gauthier C, Jacques PÉ, Li S, Xia J. 2021; MetaboAnalyst 5.0: Narrowing the gap between raw spectra and functional insights. Nucleic Acids Res. 49:W388-W396

35.

Park MR, Ryu S, Maburutse BE, Oh NS, Kim SH, Oh S, Jeong SY, Jeong DY, Oh S, Kim Y. 2018; Probiotic Lactobacillus fermentum strain JDFM216 stimulates the longevity and immune response of Caenorhabditis elegans through a nuclear hormone receptor. Sci Rep. 8:7441

36.

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

37.

Park S, Kim JA, Jang HJ, Kim DH, Kim Y. 2023a; Complete genome sequence of functional probiotic candidate Lactobacillus amylovorus CACC736. J Anim Sci Technol. 65:473-477

38.

Park S, Park MA, Jang HJ, Kim DH, Kim Y. 2024 Complete genome sequence of potential probiotic Ligilactobacillus ruminis CACC881 isolated from swine. J Anim Sci Technol (in press).

39.

Park S, Son S, Park MA, Kim DH, Kim Y. 2023b; Complete genome sequence of Latilactobacillus curvatus CACC879 and its functional probiotic properties. J Anim Sci Technol. 66:630-634

40.

Pees B, Yang W, Kloock A, Petersen C, Peters L, Fan L, Friedrichsen M, Butze S, Zárate-Potes A, Schulenburg H, Dierking K. 2021; Effector and regulator: Diverse functions of Celegans C-type lectin-like domain proteins. PLOS Pathog. 17e1009454

41.

Ryu S, Doo H, Kim ES, Keum GB, Kwak J, Pandey S, Choi Y, Kang J, Kim S, Kim HB, Lee JH. 2023 Complete genome sequence of Lactiplantibacillus plantarum strain GA_C_14 with potential characteristics applicable in the swine industry. J Anim Sci Technol (in press).

42.

Ryu S, Shin M, Yun B, Lee W, Choi H, Kang M, Oh S, Kim Y. 2021; Bacterial quality, prevalence of pathogens, and molecular characterization of biofilm-producing Staphylococcus aureus from Korean dairy farm environments. Animals. 11:1306

43.

Schulenburg H, Hoeppner MP, Weiner III J, Bornberg-Bauer E. 2008; Specificity of the innate immune system and diversity of C-type lectin domain (CTLD) proteins in the nematode Caenorhabditis elegans. Immunobiology. 213:237-250

44.

Shen P, Kershaw JC, Yue Y, Wang O, Kim KH, Mcclements DJ, Park Y. 2018; Effects of conjugated linoleic acid (CLA) on fat accumulation, activity, and proteomics analysis in Caenorhabditis elegans. Food Chem. 249:193-201

45.

Shin SM, Park JS, Kim SB, Cho YH, Seo H, Lee HS. 2024; A 12-week, single-centre, randomised, double-blind, placebo-controlled, parallel-design clinical trial for the evaluation of the efficacy and safety of Lactiplantibacillus plantarum SKO-001 in reducing body fat. Nutrients. 16:1137

46.

Shtonda BB, Avery L. 2006; Dietary choice behavior in Caenorhabditis elegans. J Exp Biol. 209:89-102

47.

Song D, Lee J, Kim K, Oh H, An J, Chang S, Cho H, Park S, Jeon K, Yoon Y, Yoo Y, Cho Y, Cho J. 2023; Effects of dietary supplementation of Pediococcus pentosaceus strains from kimchi in weaned piglet challenged with Escherichia coli and Salmonella enterica. J Anim Sci Technol. 65:611-626

48.

Song D, Lee J, Yoo Y, Oh H, Chang S, An J, Park S, Jeon K, Cho Y, Yoon Y, Cho J. 2025; Effects of probiotics on growth performance, intestinal morphology, intestinal microbiota weaning pig challenged with Escherichia coli and Salmonella enterica. J Anim Sci Technol. 67:106-136

49.

Tissenbaum HA. 2015; Using Celegans for aging research. Invertebr Reprod Dev. 59:59-63

50.

Wang H, Wang J, Zhang Z. 2018; Leucine exerts lifespan extension and improvement in three types of stress resistance (thermotolerance, anti-oxidation and anti-UV irradiation) in Celegans. J Food Nutr Res. 6:665-673

51.

Wang HY, Zhang ZZ. 2018; Evidences that intake of L-valine may affect the lifespan-specific local gene network pattern in caenorhabditis elegans. Proceedings of the 2017 2nd International Conference on Biological Sciences and Technology (BST 2017). Zhuhai, China. pp p. 161-167

52.

Yang S, Deng C, Li Y, Li W, Wu Q, Sun Z, Cao Z, Lin Q. 2022; Complete genome sequence of Lactiplantibacillus plantarum ST, a potential probiotic strain with antibacterial properties. J Anim Sci Technol. 64:183-186

53.

Yu DY, Oh SH, Kim IS, Kim GI, Kim JA, Moon YS, Jang JC, Lee SS, Jung JH, Park J, Cho KK. 2022; Intestinal microbial composition changes induced by Lactobacillus plantarum GBL 16, 17 fermented feed and intestinal immune homeostasis regulation in pigs. J Anim Sci Technol. 64:1184-1198