Abstract Milk kefir is a fermented dairy product composed of a complex consortium of microorganisms, including lactic acid bacteria (LAB), yeasts, and acetic acid bacteria. Despite growing interest in its health-promoting properties, the synergistic interactions among the diverse microbial constituents of kefir remain insufficiently characterized. This study investigated the probiotic potential of milk kefir through a multi-omics approach. Two strains, Kluyveromyces marxianus SLAM 005Y and Lentilactobacillus kefiri SLAM 023B, were isolated from Korean household milk kefir. Both strains exhibited key probiotic properties, including antibacterial activity, adherence to intestinal epithelial cells, and resistance in simulated gastrointestinal conditions. Supplementation with SLAM 005Y, SLAM 023B, or their synergistic combination (SYN) significantly enhanced lifespan and immune function in Caenorhabditis elegans. Transcriptomic analysis further revealed upregulation of genes involved in longevity and antimicrobial defense, suggesting the activation of conserved stress-response pathways. In a dextran sulfate sodium (DSS)-induced murine colitis model, SYN administration led to marked improvement in clinical outcomes. SYN also restored the expression of inflammatory and anti-inflammatory cytokines and tight junction proteins, indicating improved intestinal barrier function and immune regulation. Importantly, multi-omics analysis revealed that SYN altered both gut microbial composition and fecal metabolite profiles, supporting a mechanistic link between microbiota modulation and host health. Taken together, milk kefir strains SLAM 005Y and SLAM 023B, particularly in combination, exhibit strong probiotic properties. Our findings highlight the therapeutic potential of milk kefir-originated probiotics as a functional food for improving intestinal health. Keywords: Kluyveromyces marxianus, Lentilactobacillus kefiri, Synergy effect, Gut microbiome, Inflammatory bowel disease, Multi-omics analysis Graphical abstract Image 1 [49]Open in a new tab Highlights * • Kefir strains showed probiotic traits including antibacterial activity, intestinal adherence, and gastrointestinal tolerance. * • Kefir derived strains activated stress-response pathways in Caenorhabditis elegans surrogate host model. * • Synbiotic kefir strains improved clinical outcomes in a dextran sulfate sodium (DSS)-induced murine colitis model. * • SYN with Kefir derived strains significantly altered both gut microbial composition and fecal metabolite profiles. 1. Introduction Inflammatory bowel disease (IBD), encompassing ulcerative colitis (UC) and Crohn's disease, comprises a group of chronic inflammatory disorders of the gastrointestinal tract ([50]Podolsky, 2002). Until present, the prevalence of IBD has surpassed 6.8 million cases, affecting individuals across diverse racial, geographic, and age groups ([51]Xia et al., 2023). The causes of IBD are still incompletely resolved, but many hypotheses exist, including genetic, environmental, and immunological factors ([52]Wirtz and Neurath, 2007). Approximately 95 % of the human microbiota resides in the gut, where microbial genes and metabolites play essential roles in maintaining host physiological homeostasis and have been implicated in disease pathogenesis ([53]Lee et al., 2021). Numerous studies have identified consistent alterations in the composition and function of the gut microbiota in individuals with IBD compared to healthy controls, highlighting a strong association between microbial dysbiosis and IBD development ([54]Metwaly et al., 2020; [55]Miele et al., 2009; [56]Rigottier-Gois, 2013; [57]Tamboli et al., 2004). Given that microbial imbalance is critical to the pathophysiology of IBD, strategies aimed at restoring homeostasis of gut microbiota are increasingly viewed as promising therapeutic approaches. Probiotics are defined as live microorganisms that, when administered in adequate amounts, confer health benefits on the host. Probiotics agents may contribute to the restoration of gut microbial balance, repair inflammation-induced epithelial barrier dysfunction, and normalize dysregulated intestinal motility ([58]Di Stefano et al., 2023; [59]Fischbach, 2018). Beyond their application in gastrointestinal disorders, probiotics have also demonstrated therapeutic potential in the context of systemic inflammatory diseases, including diabetes, obesity, and certain forms of cancer ([60]Bedada et al., 2020; [61]Bock et al., 2021; [62]Vallianou et al., 2020). Kefir, a traditional fermented milk product originating from the Caucasus and Tibetan regions, is produced through the fermentation of milk with kefir grains containing a complex microbial consortium ([63]Kazou et al., 2021). This probiotic-rich beverage typically comprises a diverse of bacteria and yeast ([64]Guangsen et al., 2021; [65]Leite et al., 2015; [66]Shavit, 2008; [67]TAŞ et al., 2012). Kefir has been reported to exhibit multiple health-promoting properties, including antioxidant, antibacterial, antifungal, anti-inflammatory, antidiabetic, and anti-atherosclerotic effects, thereby positioning it as a promising functional food ([68]Tzavaras et al., 2022). Recent experimental studies have demonstrated that kefir administration alleviates colonic inflammation in murine models of dextran sulfate sodium (DSS)-induced colitis ([69]da Silva et al., 2023). While these findings highlight the therapeutic potential of kefir in managing intestinal inflammation, the specific contributions and synergistic interactions among its constituent microbial species remain largely unexplored. In this study, we aimed to evaluate the probiotic potential of K. marxianus SLAM 005Y and L. kefiri SLAM 023B, isolated from traditional Korean milk kefir. We specifically investigated their individual and synergistic effects using both Caenorhabditis elegans (C. elegans) and a DSS-induced colitis mouse model. Through comprehensive in vivo and transcriptomic analyses, we sought to elucidate the mechanisms by which these strains modulate gut inflammation and host–microbe homeostasis. Our findings demonstrate that the combination of SLAM 005Y and SLAM 023B exerts synergistic effects in alleviating gut inflammation and maintaining microbial balance, highlighting their therapeutic potential as kefir-derived probiotics. 2. Materials and methods 2.1. Prepare of kefir grain and fermented kefir milk Three traditional kefir samples were collected from local households in Korea between September and December, including two from Seoul and one from Suwon. Kefir grains were carefully transported to the laboratory in pasteurized milk and immediately transferred into sterilized milk (Seoul Milk Co., Ltd., Seoul, Korea) for fermentation. To restore microbial activity, 2–3 cycles of subculturing were performed prior to the experiment. The grains were then cultivated in sterilized milk at 25 °C for 72 h, following previously described methods ([70]Kim et al., 2016, [71]2020). 2.2. Illumina sequencing for bacteria and yeast in kefir milk and kefir grain Following the fermentation process, kefir grains were separated via an autoclavable stainless steel sieve from kefir milk. Isolated kefir grain was washed with sterilized milk (Seoul Milk Co. Ltd.) to remove kefir milk remaining on its surface. Total DNA extraction from kefir milk (100 μl) and kefir grain (25 mg) were extracted using the DNeasy Blood & Tissue Kit (Qiagen, Hilden, Germany) according to the manufacturer's protocol. The V3-V4 region for bacteria and ITS region for yeast were amplified and sequenced using the MiSeq system platform (Macrogen Inc., Seoul, South Korea) based on the standard Illumina sequencing protocols. And then, fastq files obtained from Illumina sequencing data were analyzed using Mothur software (v. 1.44.3) according to MiSeq SOP protocol ([72]https://mothur.org/wiki/miseq_sop/) ([73]Kang et al., 2025) and MicrobiomeAnalyst. 2.3. Isolation and identification of lactic acid bacteria and yeast Bacteria and yeast are separately isolated from kefir milk and kefir grains. Kefir grains were placed in sterile stomacher bags containing saline solution and homogenized at high speed for 10 min using a stomacher. Subsequently, the suspension subjected to serial dilution and was plated onto Man, Rogosa, and Sharpe (MRS) agar (BD Difco, Sparks, MD, USA), Potato Dextrose Agar (PDA; BD Difco), and Yeast Extract Peptone Dextrose (YPD) agar (BD Difco). The plates were then incubated individually for 72 h at 37 °C and 48 h at 25 °C. To recover a broader spectrum of kefir-associated microorganisms, including coexisting lactic acid bacteria and yeasts, antifungal (amphotericin B) or antibacterial (chloramphenicol) agents were not supplemented in the media. This non-selective approach was employed to avoid artificial suppression of any microbial group and to better reflect the natural microbial composition of kefir. 2.4. Evaluating the fundamental characteristics of probiotics 2.4.1. Antimicrobial activity test 2.4.1.1. Pathogenic microbial strains and culture condition Staphylococcus aureus Newman and Listeria monocytogenes EGD-e were grown at 37 °C for 24 h in brain heart infusion (BHI) broth medium (BD Biosciences). Salmonella enterica serovar Typhimurium SL1344 was grown at 37 °C for 24 h in nutrient broth medium (BD Biosciences). Escherichia coli O157:H7 EDL933 was grown at 37 °C for 24 h in Luria– Bertani (LB) broth medium (BD Biosciences). Lactobacillus strain were grown at 37 °C for 48 h in MRS broth medium and Kluyveromyces strains were grown at 25 °C for 24 h. For the assay, pathogenic strains were prepared by inoculating overnight cultures into 0.4 % soft agar containing the respective broth medium at a final concentration of 5 % (v/v). 2.4.1.2. Antimicrobial activity screening Antimicrobial activity was evaluated using a three-layer agar diffusion method, modified from [74]Hossain et al. (2022). Briefly, 5 μL of overnight cultures of Kluyveromyces strains were spotted onto the bottom layer of YPD agar, and Lactobacillus strains were spotted onto MRS agar. These plates were incubated for 48 h at either 25 °C or 37 °C to allow probiotic growth. After incubation, a second layer consisting of 0.4 % soft agar seeded with the pathogenic strains was poured over the bottom layer and allowed to solidify. Subsequently, a third top layer of plain soft agar was added to stabilize diffusion and prevent desiccation. The plates were then incubated at 37 °C for an additional 15 h. Zones of inhibition were measured to determine antimicrobial activity as previously described method ([75]Pundir et al., 2010). 2.4.1.3. Acid and bile tolerance test The tolerance of Kluyveromyces strains and Lactobacillus strains to acid and bile salt was tested following a previous study with some modifications ([76]Cho et al., 2024; [77]Oh et al., 2018). Kluyveromyces strains were cultured for 24 h in YPD broth medium, and Lactobacillus strains were cultured for 48 h in MRS medium. To create an acidic environment simulating the stomach, the pH of YPD and MRS broth was adjusted to pH 2.5 using 6N HCl. After autoclaving, porcine gastric mucosa pepsin (Sigma-Aldrich, St. Louis, MO, USA) was added to achieve a concentration of 1000 units/mL and filtered through a 0.45-μm pore size syringe filter. In the acid tolerance test of Kluyveromyces strains and Lactobacillus strains, cultures were inoculated into acidic solution and incubated for 0 h and 3 h at 37 °C. The bile solution was prepared by suspending oxgall powder (Acumedia, Lansing, MI, USA) in YPD and MRS broth medium to a final concentration of 0.5 % (w/v). All cultures of Kluyveromyces strains and Lactobacillus strains were inoculated into 0.5 % bile solution and incubated for 24 h at 37 °C. The survival rate of each strain was compared to the initial viable counts on plates at 0 h and after 3 h and 24 h of incubation. Survival rate (%) = CFU of 3 h or 24 h/CFU of 0 h) X 100(%) 2.4.1.4. Adhesion ability using Caco-2 intestinal epithelial cell Human colon epithelial colorectal adenocarcinoma Caco-2 cells were obtained from the Korean Cell Line Bank (Korea). Cell maintenance was performed following a previous study ([78]Yoon et al., 2022). After activating the culture medium, cells were maintained in minimal essential medium (MEM) (Gibco BRL, USA), 10 % heat-inactivated fetal bovine serum (FBS) (WelGENE Inc., Korea), and penicillin-streptomycin (15140-122, Gibco). Caco-2 cells were inoculated in a 100 mm plate and incubated at 37 °C under 5 % CO[2] conditions. The medium was exchanged every two days, the cells were incubated until a monolayer formed. Caco-2 cells were maintained until the experiment. To remove the culture medium and non-attached cells before experiments, the cells were washed 3 times with PBS. Cell adhesion assays were performed using modified methods ([79]Heo et al., 2018). Yeast and bacterial strains were treated at 10^9 CFU/mL MEM medium without penicillin-streptomycin was incubated for 2 h at 37 °C under 5 % CO[2] conditions. After 2 h of incubation, the monolayers were washed 3 times with PBS to remove non-attached yeast and bacteria. Trypsin-EDTA was used to lyse the attached cells. After lysis, serial dilutions of the yeast and bacterial mixture were separately plated on YPD and MRS agar plates. Each plate was incubated for 48 h at 25 °C and 72 h at 37 °C. The Adhesion ability was evaluated by counting CFU/ml. 2.4.1.5. Functional experimental using C. elegans surrogate animal model Kuyveromyces marxianus SLAM 005Y and Saccharomyces boulardii SLAM 010SSB were grown at 25 °C for 24 h in YPD broth medium. Lentilactobacillus kefiri SLAM 023B and Lacticaseibacillus rhamnosus GG (LGG) were grown at 37 °C for 48 h in MRS broth. Escherichia coli OP50 (OP50) and E. coli O157:H7 were grown in LB broth medium. S. Typhimurium was grown at 37 °C for 24 h in nutrient broth medium. To prepare live microbial lawns for C. elegans feeding, each microbial pellet was collected by centrifugation at 6000 rpm for 10 min, washed twice with sterile M9 buffer. After washing, the bacterial pellet was suspended in M9 buffer and seeded on nematode growth medium (NGM) plates. 2.4.1.6. C. elegans lifespan, killing assay, and fluorescence analysis C. elegans were maintained on NGM agar at 15 °C using standard techniques until experiment ([80]Brenner, 1974). Various of C. elegans strains were used for experiment, including CF512(fer-15(b26)II;fem-1(hc17)IV) and AY102(acEx102 [vha-6p::pmk-1::GFP + rol-6(su1006)]), CF512, also known as temperature sensitive, was used for lifespan and killing assay and AY102 was employed for evaluating innate immunity, respectively. After Synchronization, L1 worms were grown on NGM plates, seeded with OP50 at a restrictive temperature (25 °C), to obtain sterile L4/young adult worms. To evaluate the microbial effect of kefir on C. elegans lifespan and immune response against pathogenic bacteria, we established methods based on a previous study ([81]Lee et al., 2024). For the lifespan assay, young adult L4 stage CF512 worms were transferred to 35 mm NGM agar plates and seeded with 100 μL of concentrated SLAM 005Y, SLAM 023B, SYN and OP50 using a platinum wire. For the killing assay, young adult L4 stage CF512 worms were transferred to 60 mm NGM agar plates seeded with 200 μL of concentrated SLAM 005Y, SLAM 023B, SYN, and OP50 using a platinum wire. After 48 h, worms were transferred to NGM plate seeded with pathogenic bacteria, S. Typhimurium, using a platinum wire. OP50 was used as normal control and positive control. The assessment of alive or dead C. elegans was conducted by gently touching them with a platinum wire, and the assay was conducted until all C. elegans died. The data from the C. elegans lifespan and killing assays were analyzed using the Kaplan-Meier method. The significance of differences between survival curves was assessed using the log-rank test with STATA6 (STATA, TX, USA). Other data were statistically analyzed and graphed using Prism 10.2.2 (Graphpad Software, USA). Statistical significance was set at p value < 0.05 (∗), <0.01 (∗∗), <0.001(∗∗∗), <0.0001 (∗∗∗∗). All data are expressed as means ± the standard error of the mean (SEM). In addition, C. elegans fluorescence analysis was conducted to examine immune response. To examine the induction of pmk-1::GFP (the key gene for the p38 type MAPK pathway) in the nematode intestine, AY102 transgenic worms were exposed to targeted strasin for 48 h. Then, individual worms (n = 10 per group) were transferred onto glass slides with 2 % agarose pads. C. elegans were anesthetized using 20 μL of 30 mM NaN[3] (Sigma-Aldrich), and the GFP expression rate was observed using fluorescence microscopy (IX53; Olympus, Tokyo, Japan). ImageJ (MacBiophotonics ImageJ version 1.42I + loci tool) was used to analyze the fluorescent intensity. 2.4.1.7. C. elegans total RNA extraction and transcriptomic analysis Transcriptomic analysis was performed based on a previous study ([82]Kang et al., 2022). Young adult L4 worms were exposed to plates seeded with 200 μL of concentrated OP50, SLAM 005Y, SLAM 023B, and SYN for 48 h. Total RNA was extracted using TRIZOL reagent (Invitrogen, Carlsbad, CA, USA) and a mini bead beater (Biospec, Bartlesville, OK), followed by purification with the RNeasy Mini Kit (Qiagen) according to the manufacturer's instructions. Quality and quantity checks were conducted by measuring absorbance (ratio = A260/280) on a SpectraMax ABS Plus. RNA sequencing and transcriptomic analysis were performed by Macrogen (Macrogen Inc., South Korea). The ClueGO plug-in in Cytoscape software (version 3.9.1) was used to analyze Biological Process, Cellular Component, and Molecular functions of differentially expressed genes. 2.4.1.8. Fermentation of Intestinal Microbiota Model (FIMM) The Fermenter for Intestine Microbiome Model (FIMM) is an in vitro batch fermentation system designed to simulate the human colonic environment under anaerobic conditions. This model was adapted from the Simulator of the Human Intestinal Microbial Ecosystem (SHIME) and further developed following previously reported study ([83]Choi et al., 2025; [84]Kang et al., 2022, [85]2024). In this study, fresh fecal samples from healthy human donors were pooled and homogenized under anaerobic conditions to prepare a 10 % (w/v) fecal slurry. The fermentation was carried out in modified Gifu Anaerobic Medium (mGAM; Himedia, Mumbai, India), with the pH adjusted to 7.0 to mimic the human colon. To evaluate the microbiota-modulating potential of kefir-derived probiotics, 1 % (v/v) of SLAM 005Y, SLAM 023B, or SYN was inoculated into the culture. The cultures were then incubated in anaerobic conditions for 48 h at 37 °C with shaking and subsequently stored at −80 °C until metagenome analysis. Metagenomic analysis was performed by described methods as above. 2.4.2. DSS-induced colitis mice model 2.4.2.1. Animal administration All experimental protocols and animal procedures were approved by the Institutional Animal Care and Use Committee of Seoul National University (certificate SNU-230718-5). C57BL/6, 5-week-old male mice (n = 25) were obtained from SamTako Bio (Korea) and housed 5 mice per cage. All mice were acclimatized without any intervention for a week under standard laboratory conditions (23 °C ± 1 °C with 55 % ± 5 % humidity, 12 h light/dark cycle), provided with normal chow diet and sterile water. After an acclimation period for one week, mice were placed in one of the following groups: (1) no-DSS control group (CONT; mice administered PBS only for 3-wk; no DSS induction of colitis). (2) DSS-alone (DSS), (3) SLAM 005Y with DSS (SLAM 005Y), (4) SLAM 023B with DSS (SLAM 023B), or (5) synbiotics (SYN). These mice were administered, respectively, PBS (200 μL of suspension per mouse), SLAM 005Y (1.0 × 10^9 CFU/day), SLAM 023B (1.0 × 10^9 CFU/day) or SYN (5.0 × 10^8 of SLAM 005Y + 5.0 × 10^8 of SLAM 023B CFU/day) every other day for 3-wk. Suspensions were given by oral gavage (Fuchigami Kikai, Kyoto, Japan). To induce experimental acute colitis, mice in the 4 treatment groups (DSS-C, SLAM 005Y, SLAM 023B, SYN) were provided drinking water containing low-molecular-weight DSS (2.5 % DSS with a molecular weight of 36,000–50,000 Da, MP Biomedicals, Solon, OH) for an additional week after the 2-wk study period. During the 3-wk period, daily clinical assessment of DSS-induced colitis was performed, including measuring BW and food intake and noting rectal bleeding, stool conditions, and blood in stool using the disease activity index (DAI) scoring system ([86]Chassaing et al., 2014). Stool bleeding score was determined using the Hema-Screen Lab Pack for fecal occult blood (EKF Diagnostics, TX, USA). Stool bleeding score was determined using the Hema-Screen Lab Pack for fecal occult blood (EKF Diagnostics, TX, USA). 2.4.2.2. Morphological and biological analysis Spleen weight and colon length were measured after sacrifice. Spleen index was calculated by dividing the weight of the spleen (mg) by the weight of the mouse (g) at the time of dissection. Colon samples were washed with PBS and collected from the cecum to the rectum. After photographing the mouse colon tissue of each group with a ruler, the length of the colon was measured using ImageJ software version 1.8.0 (W. Rasband, USA). Colon samples from each mouse were washed with sterilized PBS. Samples were fixed in 10 % formalin for dehydration and then embedded in paraffin for staining with hematoxylin and eosin (H&E) analyzed by imaging with a digital slide scanning system (Konfoong bioinformation tech co., LTD). Histological score was based on previous studies ([87]Kyoung et al., 2025; [88]Ye et al., 2023). 2.4.2.3. RNA isolation and RT-qPCR Total RNA from colon tissues were extraction by Trizol reagent and a RNeasy Mini kit (QIAGEN) according to the manufacturer's instructions. Quantity and purity were confirmed by measuring absorbance (ratio = A260/280 and A260/230) on a SpectraMax ABS Plus spectrometer ([89]Kwak et al., 2024). All RNA samples were diluted to 1 μg for RT-qPCR using iScript cDNA synthesis Kit (Bio-rad, CA, USA). After then, real-time PCR was performed using SsoAdvanced Universal SYBR Green Supermix with CFX96™ System (Bio-rad, CA, USA). Expression of each gene was normalized to the housekeeping gene, Gapdh (2^−ΔΔCt) ([90]Kwak et al., 2022). Primer sequences used in experiments are listed in [91]Supplementary Table S1. RNA sequencing data have been deposited in the NCBI SRA under BioProject accession number PRJNA1288232. 2.4.2.4. Fecal metagenomic and metabolomic analysis Fecal samples were collected and stored at −80 °C until metabolomic analysis. Metagenomic analysis was based on previous method ([92]Lee et al., 2022). gDNA was homogenized and extracted using the Dneasy PowerSoil Pro Kit (Qiagen, Hilden, Germany). Metagenomic analysis were performed by described methods as above. All raw sequence reads from the 16S rRNA gene analysis have been submitted to the NCBI SRA under BioProject accession number PRJNA1288167. For metabolomic analysis, GC-MS analysis was conducted using a Thermo Trace 1310 GC (Waltham, MA, USA) coupled with a Thermo ISQ LT Single Quadrupole Mass Spectrometer (Waltham, MA, USA). A DB-5MS column (Agilent, Santa Clara, CA, USA). The sample was injected at 300 °C with a split ratio of 1:60 and a helium split flow of 7.5 mL/min. Mass spectra were acquired at a scan range of 35–650 m/z with a scan rate of 5 spectra per second. Ionization was performed in electron impact mode with the ion source temperature set at 270 °C. GC-MS spectra were deconvoluted and processed using the Automated Mass Spectral Deconvolution and Identification System (AMDIS) software ([93]Meyer et al., 2010) for peak detection and compound identification, and metabolites were identified by matching mass spectra and retention indices with the NIST Mass Spectral Search Tool (version 2.0, Gaithersburg, MD, USA) ([94]Lee et al., 2022). Further multivariate statistical analyses, including partial least squares-discriminant analysis (PLS-DA), were conducted using MetaboAnalyst 6.0 ([95]Pang et al., 2024) to evaluate group discrimination and determine key metabolites contributing to intergroup variation. 2.5. Statistics analysis All data were statistically analyzed and visualized using GraphPad Prism version 10.2.2 (GraphPad Software, San Diego, CA, USA). Multiple group comparisons were analyzed using one-way ANOVA or two-way ANOVA, as appropriate, followed by Tukey's post hoc test. Statistical significance was defined as p < 0.05 (∗), p < 0.01 (∗∗), p < 0.001 (∗∗∗), and p < 0.0001 (∗∗∗∗). All data are presented as means ± standard error of the mean (SEM). 3. Result 3.1. Metagenomic analysis for myco- and microbiota in kefir milk and kefir grain The relative abundance of fungal and bacterial communities in kefir milk and kefir grain samples from three individual samples is presented in [96]Fig. 1. Kefir-associated mycobiota and microbiota profiles were analyzed using next-generation sequencing (NGS). In the yeast community composition of kefir milk, KM1 was dominated by Kluyveromyces (55.65 %), followed by Issatchenkia (35.85 %) and Kazachstania (8.36 %). KM2 also exhibited high relative abundances of Kluyveromyces (42.89 %) and Issatchenkia (38.85 %), with lower levels of Kazachstania (2.53 %) and Pichia (0.79 %). Similarly, KM3 was composed predominantly of Kluyveromyces (42.88 %), followed by Issatchenkia (18.66 %), Kazachstania (0.37 %), and Pichia (1.19 %) ([97]Fig. 1A). In the corresponding kefir grains, distinct mycobiota patterns were observed. KG1 consisted almost exclusively of Kazachstania (99.28 %), with trace amounts of Kluyveromyces (0.39 %) and Issatchenkia (0.31 %). KG2 contained Kazachstania (81.95 %), Kluyveromyces (16.49 %), and Issatchenkia (0.66 %). Notably, KG3 exhibited a shift in dominance, with Kluyveromyces (70.78 %) as the most abundant genus, followed by Kazachstania (23.32 %), Pichia (0.44 %), and Issatchenkia (0.37 %) ([98]Fig. 1C). Regarding bacterial composition, KM1 was largely dominated by Lactobacillus (89.23 %), along with Lentilactobacillus (9.57 %), and minor contributions from Acetobacter (0.38 %) and Brevibacillus (0.63 %). In contrast, KM2 had a higher proportion of Acetobacter (56.32 %), with Lactobacillus (35.81 %) and Lentilactobacillus (7.76 %). KM3 consisted of Lactobacillus (56.39 %), Acetobacter (41.55 %), and Lentilactobacillus (2.04 %) ([99]Fig. 1B). The bacterial profiles of the kefir grains also observed consistency in Lactobacillus dominance. KG1 contained Lactobacillus (99.13 %), with minor contributions from Lentilactobacillus (0.59 %) and Acetobacter (0.26 %). KG2 followed a similar pattern, with Lactobacillus (98.42 %), Lentilactobacillus (1.14 %), and Acetobacter (0.42 %). KG3 was composed of Lactobacillus (97.22 %), Lentilactobacillus (2.31 %), and Acetobacter (0.33 %). Overall, these results indicate that Kluyveromyces and Lactobacillus were the predominant fungal and bacterial genera, respectively, in both kefir milk and grains across all Korean household kefir samples analyzed ([100]Fig. 1D). Our data revealed a core kefir microbiota composed mainly of Lactobacillus and Acetobacter species, and a core mycobiota dominated by Kluyveromyces, consistent with previous studies on traditional kefir fermentation ([101]Dertli and Çon, 2017; [102]Kim et al., 2020; [103]Plessas et al., 2017). We also observed inter-sample variability in the relative abundances of these taxa, particularly in the proportions of Lactobacillus and Kluyveromyces, which varied depending on the household source. Fig. 1. [104]Fig. 1 [105]Open in a new tab Metagenomic and culturomic analysis of kefir milk and kefir grain. The microbial composition of Korean household kefir was analyzed at the genus level following 72 h of fermentation. Relative abundances of fungal and bacterial taxa were determined using Illumina sequencing of the ITS region for yeasts and the 16S rRNA gene for bacteria. (A) Genus-level composition of yeasts in three kefir milk (KM) samples. (B) Genus-level composition of bacteria in the same KM samples. (C) Genus-level composition of yeasts in the corresponding kefir grain (KG) samples. (D) Genus-level composition of bacteria in the same KG samples. Subsequently, kefir samples were serially diluted from KM and KG and cultured for microbial isolation. A total of 43 yeast strains (E) and 44 bacterial strains (F) were isolated and taxonomically identified using ITS and 16S rRNA gene sequencing, respectively. 3.2. Isolation of potential probiotic yeast and bacteria strains Following the identification of Kluyveromyces and Lactobacillus as the predominant genera in both kefir milk (KM) and kefir grains (KG) of Korean household kefir samples using metagenomic analysis, serial dilutions of the KM and KG samples were plated on MRS agar, YPD agar, and PDA to isolate individual microbial colonies. Subsequent identification of isolates was performed using internal transcribed spacer (ITS) sequencing for yeasts and 16S rRNA gene sequencing for bacteria. A total of 43 yeast isolates and 44 bacterial isolates were obtained. Among the yeast isolates, six distinct strains of Kluyveromyces (SLAM 001Y, SLAM 002Y, SLAM 003Y, SLAM 004Y, SLAM 005Y, and SLAM 006Y) were identified ([106]Fig. 1E). For bacterial isolates, several functionally relevant lactic acid bacteria (LAB) species were identified. These included Leuconostoc mesenteroides SLAM 003B, Lactobacillus brevis SLAM 006B, Lacticaseibacillus paracasei SLAM 008B and SLAM 012B, Lactococcus lactis SLAM 010B, and Lentilactobacillus kefiri SLAM 023B ([107]Fig. 1F). These findings suggest that Kluyveromyces and various LAB strains represent dominant taxa in the kefir samples analyzed in this study, as confirmed by both metagenomic and culture-based approaches. 3.3. Evaluating the fundamental characteristics of probiotics The antimicrobial activity was assessed by observing the formation of inhibition zones around the colonies of kefir-derived isolates ([108]Supplementary Table S2). Among the yeast isolates, K. marxianus strains SLAM 001Y through SLAM 005Y did not exhibit detectable antimicrobial activity against any of the tested pathogens under the experimental conditions. In contrast, several bacterial isolates demonstrated notable antibacterial effects. Most prominently, kefir-derived strains exhibited strong inhibitory activity against E. coli O157:H7, with strain Lentilactobacillus kefiri SLAM 023B and Lacticaseibacillus paracasei SLAM 008B also showing pronounced activity against S. Typhimurium. Probiotic functionality requires that candidate strains exhibit resistance to both gastric acidity and bile salts, which are critical for survival and adhesion within the human gastrointestinal tract ([109]Fig. 2A and B). To assess these attributes, we evaluated the acid and bile tolerance of selected kefir-derived strains under simulating gastrointestinal conditions. Initially, among the yeast isolates, K. marxianus SLAM 005Y demonstrated a high level of acid tolerance, with a survival rate of 87.04 %, comparable to that of the probiotic yeast reference strain Saccharomyces boulardii (SSB). Similarly, L. kefiri SLAM 023B showed a strong acid resistance profile, achieving a survival rate of 92.66 %, nearly identical to that of the well-established probiotic LGG strain. Moreover, in the bile tolerance assay, all K. marxianus strains revealed bile resistance comparable to SSB. Among the bacterial isolates, all strains except SLAM 012B maintained survival rates above 80 %. Notably, SLAM 010B and SLAM 023B demonstrated high bile tolerance, with survival rates of 93.65 % and 91.72 %, respectively, closely matching that of LGG. These findings highlight the robust acid and bile tolerance of kefir-derived microorganisms, particularly K. marxianus SLAM 005Y and L. kefiri SLAM 023B, suggesting their strong potential as viable probiotic candidates for gastrointestinal survival and functionality. Fig. 2. [110]Fig. 2 [111]Open in a new tab Evaluation of probiotic properties of yeast and bacterial strains isolated from Korean household kefir. The probiotic potential of kefir-derived microorganisms was assessed by measuring acid and bile tolerance, as well as adhesion capacity. (A) Survival rate after 3 h of exposure to acidic conditions (pH 2.5). (B) Survival rate after 24 h of exposure to 0.5 % (w/v) bile salts. (C) Adhesion capacity to Caco-2 cells, expressed as the percentage of adherent cells relative to baseline (0 h). Saccharomyces boulardii (SSB) and Lacticaseibacillus rhamnosus GG (LGG) were used as positive controls in all assays. Data are expressed as mean ± SEM from three independent experiments. Statistical significance was determined by one-way ANOVA followed by post hoc analysis; ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, and ∗∗∗∗p < 0.0001. Effective probiotic function requires not only gastrointestinal survivability but also the ability to adhere to the intestinal mucosal surface, which facilitates colonization, persistence, and exclusion of pathogenic microbes ([112]Pennacchia et al., 2006; [113]Xie et al., 2012). To evaluate the adhesion capacity of kefir-derived strains, we utilized the Caco-2 cell line as an in vitro model of the human intestinal epithelium. In this study, two established reference probiotics, L. rhamnosus GG (LGG), and Saccharomyces boulardii (SSB), commonly recognized for its superior mucosal adherence and functionality, served as positive controls ([114]Gu et al., 2022). As shown in [115]Fig. 2C, all tested kefir-derived microorganisms exhibited strong mucin-binding capacity, with adhesion rates exceeding 70 %. Of particular note, L. kefiri SLAM 023B displayed an adhesion rate above 80 %, surpassing that of several other tested strains, indicating a robust mucosal attachment potential. These results indicate that kefir isolates, especially L. kefiri SLAM 023B, exhibit strong adhesion to intestinal epithelial cells, supporting their potential for stable gut colonization. 3.4. Evaluation of functional properties using C. elegans To assess the functional effects of kefir-derived microorganisms on host physiology, we first evaluated their influence on the lifespan of C. elegans ([116]Supplementary Fig. S1). As shown in [117]Fig. 3A, several strains, including SLAM 003Y, SLAM 005Y, SLAM 006Y, and SLAM 003B, significantly extended the mean lifespan of C. elegans compared to the E. coil OP50 normal feeding group. These results suggest that specific kefir isolates possess enhanced longevity effects in the nematode model. To further examine their impact on host immune defense, we performed C. elegans pathogen killing assays ([118]Supplementary Fig. S2). After pre-exposure to kefir-derived strains for 48 h, worms were challenged with S. Typhimurium. Among the tested strains, SLAM 005Y showed a notable protective effect, reducing pathogen-induced mortality relative to the E. coli OP50 control group (p = 0.0242; [119]Fig. 3B). Similarly, strains SLAM 006B and SLAM 023B also conferred enhanced resistance to S. Typhimurium, suggesting an immunomodulatory role against Gram-negative bacterial infection. These functional assays that combined with previous results demonstrate acid resistance, bile tolerance, and strong epithelial adhesion, highlight K. marxianus SLAM 005Y and L. kefiri SLAM 023B as particularly promising probiotic candidates. Based on their superior performance in promoting lifespan extension and immune defense in C. elegans, subsequent experiments were focused on these two strains and their synergistic combination for further characterization. Fig. 3. [120]Fig. 3 [121]Open in a new tab Enhanced longevity and host immune response of Caenorhabditis elegans fed kefir-derived probiotic strains. (A) Lifespan analysis of C. elegans fed with kefir-derived Kluyveromyces marxianus SLAM 005Y and Lentilactobacillus kefiri SLAM 023B compared to the control diet of Escherichia coli OP50. (B) Survival of worms in the killing assay following infection with S. Typhimurium, assessing the protective effects of kefir-derived strains relative to OP50. (C) Fluorescence microscopy of AY102 transgenic worms carrying the pmk-1::GFP reporter, following 48 h exposure to kefir microorganisms. (D) Expression levels of immune-related genes following SYN treatment, with significantly upregulated genes defined as those showing a fold change >3 and p < 0.05 relative to OP50-fed controls. (E) Gene ontology enrichment analysis highlights the molecular response pathways activated by SYN supplementation. Lifespan data were analyzed using the Kaplan–Meier method with significance assessed by the log-rank test and immune and gene expression data were analyzed by one-way ANOVA. Statistical significance indicated as follows: p < 0.05 (∗), p < 0.01 (∗∗), p < 0.001 (∗∗∗), and p < 0.0001 (∗∗∗∗). Data represent means ± SEM from three independent experiments. Moreover, the PMK-1 signaling cascade, a central component of the p38 MAPK pathway, is recognized as a key regulator of innate immune responses in C. elegans ([122]Yang et al., 2019). Previous studies have demonstrated that certain probiotic strains enhance host immunity by up-regulating pmk-1 expression, thereby protecting the nematode from pathogenic infections. In the present study, kefir-derived microbial strains, particularly those in the SYN group—a synergistic combination of the two selected strains—elicited significantly higher expression levels of pmk-1 compared to the OP50-fed control group ([123]Fig. 3C and D). These findings suggest that kefir-derived microorganisms stimulate the host's innate immune system, which contributes to enhanced resistance against microbial pathogens. 3.5. Visualized and analyzed gene network using C. elegans RNA-sequencing To further investigate the molecular mechanisms underlying these probiotic effects, transcriptomic analysis was performed, and Cytoscape-based pathway enrichment analysis was conducted to elucidate the host transcriptional pathways modulated by kefir-derived microorganisms, in comparison with C. elegans fed the OP50 control. The RNA-sequencing results indicated that K. marxianus SLAM 005Y induced gene expression associated with metabolic defense responses to Gram-positive bacteria, as well as genes involved in stress regulation, cellular protective mechanisms, neuropeptide signaling, and cell growth. In contrast, L. kefiri SLAM 023B was primarily linked to reactive oxygen species (ROS)-associated pathways, including oxidative stress response, uronic acid metabolism, and protein localization within cells ([124]Supplementary Table S3). Notably, the SYN treatment group combined SLAM 005Y and SLAM 023B, exhibited enrichment in pathways related to regulation of response to stimulus, defense against microbial organisms, and cellular stress regulation ([125]Fig. 3E). The results enabled identification of genes associated with the observed lifespan extension and immune enhancement in Caenorhabditis elegans following individual or combined administration of K. marxianus SLAM 005Y and L. kefiri SLAM 023B isolated from kefir. These findings suggest that kefir-derived strains may confer beneficial effects on host physiology by modulating gene networks involved in immune responses and microbial defense or environmental stress adaptation. 3.6. Dynamics of microbial composition using FIMM assay To evaluate the modulatory effects of kefir-derived microorganisms on the intestinal microbial community, we conducted a Fermentation of Intestinal Microbiota Model (FIMM) experiment using a colon-mimicking culture system ([126]Fig. 4). Following 24 h of co-culture with fecal samples and selected kefir microbial strains, beta diversity analysis via UniFrac revealed distinct clustering patterns among treatment groups, suggesting microbial compositional shifts ([127]Fig. 4A–B). Alpha diversity analysis using the Shannon index demonstrated a significant increase in microbial diversity in the kefir-treated groups compared to the untreated control (CONT) (p < 0.0001) ([128]Fig. 4C). However, no significant differences were observed in species richness based on the Chao index (p = 0.1577) ([129]Fig. 4D). At the phylum level, administration of kefir strains resulted in a reduction of Proteobacteria, with L. kefiri SLAM 023B exhibiting the most pronounced decrease (∼4 %) ([130]Fig. 4E). At the family level, SLAM 023B notably increased the abundance of Bifidobacteriaceae, surpassing that of the positive control LGG, and also elevated Lactobacillaceae levels relative to the CONT group ([131]Fig. 4F). Genus-level analysis revealed a significant increase in Lactobacillus abundance and a marked reduction in the relative abundance of pathogenic Escherichia_Shigella in kefir-treated samples, particularly in the SLAM 023B group. Furthermore, the combination of SLAM 005Y and SLAM 023B exhibited the highest enrichment of Akkermansia, a genus associated with mucosal integrity and anti-inflammatory effects ([132]Fig. 4G). These results indicate that the administration of kefir-derived probiotics, either alone or in combination, promotes the growth of beneficial taxa such as Bifidobacteriaceae and Akkermansia, while suppressing pathogenic bacteria such as Escherichi_Shigella. These findings suggest that these strains may beneficially reshape the intestinal microbiota, promoting a more balanced and health-associated microbial composition. While the FIMM model does not capture host physiological responses such as epithelial interaction, immune modulation, or systemic metabolism, it provides a controlled and reproducible environment for assessing direct microbiota–microbe interactions. This makes it a suitable platform for preliminary screening of probiotic effects on microbial composition and metabolic output in a human-relevant colonic context. Fig. 4. [133]Fig. 4 [134]Open in a new tab Kefir-derived microbial supplementation modulates gut microbiota composition in an in vitro digesting system. To evaluate the influence of kefir derived microorganisms on gut microbial communities, fermentor for intestine microbiota model (FIMM) was established by co-culturing healthy human fecal samples with kefir-derived strains. Microbial composition was assessed through next-generation sequencing of the 16S rRNA gene. (A–B) Principal Coordinates Analysis (PCoA) plots based on weighted and unweighted UniFrac distances reveal group-level clustering patterns. Statistical significance was determined using PERMANOVA, with F-values, R^2, and p-values indicated. (C–D) Alpha diversity indices (Shannon and Chao1) were calculated to assess microbial richness and evenness. Data are presented as mean ± SEM. (E–G) Relative abundance of microbial taxa at the phylum, family, and genus levels illustrates compositional changes across treatment groups. 3.7. Effect of kefir-derived probiotic strains in DSS-induced colitis mouse Body weight was monitored as a general indicator of health status in mice ([135]Fig. 5A). After exposure to the DSS solution, the percent changes in body weight relative to baseline (pre-DSS exposure) were as follows: 103.42 % for the CONT group, 91.30 % for the DSS group, 96.19 % for the SLAM 005Y group, 97.34 % for the SLAM 023B group, and 95.45 % for the SYN group. Although the CONT group maintained the highest relative body weight compared to the DSS group, the difference did not reach statistical significance (p = 0.2051; 95 % CI: −8.98 to 33.23; [136]Fig. 5B). These results suggest that kefir-derived strains, particularly SLAM 005Y and SLAM 023B, may attenuate DSS-induced weight loss, potentially reflecting a protective effect against colitis-associated physiological decline. In addition, Disease Activity Index (DAI) was used to quantitatively assess the progression and severity of colitis, incorporating weight loss, stool consistency, and the presence of rectal bleeding as evaluation parameters. Importantly, SLAM 005Y, SLAM 023B, and SYN treatment groups exhibited significantly reduced DAI scores compared to the DSS-only group (p < 0.0001, p = 0.0061, and p < 0.0001, respectively), while the CONT group showed no pathological signs and maintained the lowest score ([137]Fig. 5C). These results indicate that the kefir-derived strains SLAM 005Y and SLAM 023B, either individually or in combination, significantly attenuate DSS-induced weight loss and disease activity, suggesting their potential to alleviate colitis-associated clinical symptoms. Fig. 5. [138]Fig. 5 [139]Open in a new tab Effects of kefir-derived microorganisms on clinical and physiological gut health in a DSS-induced colitis mouse model. (A) Experimental schematic of DSS-induced acute colitis and kefir microbial intervention. Mice were orally administered 200 μL of treatment daily: the CONT group received autoclaved tap water with PBS; the DSS group received 2.5 % DSS in drinking water with PBS; SLAM 005Y group received Kluyveromyces marxianus SLAM 005Y with DSS; SLAM 023B group received Lactobacillus kefiri SLAM 023B with DSS; and SYN group received a combination of SLAM 005Y and SLAM 023B with DSS. (B) Disease Activity Index (DAI) scores were assessed based on weight loss, stool consistency, and rectal bleeding. (C) Spleen index was calculated to evaluate systemic inflammation.(D) Colon length was measured as a surrogate marker for colonic inflammation and tissue damage. Statistical significance was determined using one-way or two-way ANOVA. Asterisks indicate significance relative to the DSS group: p < 0.05 (∗), p < 0.01 (∗∗), p < 0.001 (∗∗∗), and p < 0.0001 (∗∗∗∗). The spleen index, a well-established marker for evaluating systemic inflammatory responses, was used to assess the physiological impact of DSS-induced colitis. As expected, DSS exposure resulted in a notable increase in the spleen index relative to CONT group. While the SLAM 005Y, SLAM 023B, and SYN combined groups also exhibited elevated spleen indices compared to CONT, treatment with kefir-derived microorganisms led to a significant reduction in the spleen index relative to the DSS group, particularly in the SLAM 023B (p = 0.0443) and SYN (p = 0.0064) groups ([140]Fig. 5D). These results suggest that kefir microbial treatments can attenuate systemic inflammation induced by DSS. Furthermore, colon length was measured as a direct indicator of colonic inflammation and mucosal damage, with shortening reflecting the severity of tissue injury. As expected, DSS exposure led to a marked reduction in colon length across all treated groups. However, probiotics treated groups showed a protective effect, as evidenced by longer colon lengths compared to the DSS group. This effect was most pronounced in the SYN group, which exhibited a statistically significant improvement (p = 0.0013) ([141]Fig. 5E). Taken together, these findings suggest that the co-administration of kefir-derived strains mitigates DSS-induced colonic tissue damage and systemic inflammation, supporting their potential role in alleviating colitis-related inflammatory responses. 3.8. Morphological and biological impacts of kefir-derived probiotics Hematoxylin and eosin (H&E) staining was employed to evaluate histopathological changes in colonic tissue following DSS-induced colitis. This widely utilized method enables detailed visualization of tissue architecture and inflammatory cell infiltration, allowing for assessment of mucosal integrity and inflammatory severity. As shown in [142]Fig. 6A, DSS treatment resulted in pronounced epithelial disruption, goblet cell depletion, and increased inflammatory infiltrates. In contrast, the kefir-treated groups including SLAM 005Y, SLAM 023B, and SYN, exhibited markedly improved tissue morphology, with reduced inflammatory cell infiltration and preservation of epithelial structure, similar to the non-treated CONT group. Quantitative histological scoring confirmed these observations ([143]Fig. 6B), with the DSS group displaying significantly elevated histopathological scores compared to all kefir strains-treated groups (p < 0.0001; 95 % CI: 2.042 to 3.958). These findings indicate that administration of SLAM 005Y and SLAM 023B, either individually or in combination, reduced histopathological signs of DSS-induced colonic damage, as evidenced by improved colon architecture and lower histological scores. Taken together, these results suggest that kefir-derived probiotics may exert beneficial effects in alleviating colonic tissue injury associated with DSS-induced colitis. Fig. 6. [144]Fig. 6 [145]Open in a new tab Histological and intestinal barrier fuunction analysis of colonic tissue following kefir-derived microbial supplementation in a DSS-induced colitis mouse model. (A) Representative images of hematoxylin and eosin (H&E)-stained colon sections at 40 × and 200 × magnification showing mucosal architecture and inflammatory features. (B) Histopathological scoring was performed based on inflammatory cell infiltration, mucosal disruption, and glandular integrity. Quantitative real-time PCR (RT-qPCR) was used to evaluate gene expression of pro-inflammatory cytokines: (C) TNF-α, (D) IL-6, and (E) IL-1β; the anti-inflammatory cytokine (F) IL-10; tight junction proteins: (G) claudin-1, (H) occludin, and (I) ZO-1; and (J) mucin-2 (MUC-2). Gene expression levels were normalized to GAPDH as housekeeping gene and calculated using the 2^-ΔΔCt method. Statistical analyses were performed using one-way or two-way ANOVA, with comparisons made to the DSS group. Significance levels are indicated as follows: p < 0.05 (∗), p < 0.01 (∗∗), p < 0.001 (∗∗∗), and p < 0.0001 (∗∗∗∗). 3.9. Transcriptional regulation of inflammatory cytokine and tight junction genes DSS is a well-established agent for inducing acute inflammation in the large intestine and serves as a reliable model for studying colitis. To elucidate the effects of kefir-derived microorganisms on inflammation and intestinal barrier function, we assessed the expression levels of pro- and anti-inflammatory cytokines, as well as tight junction-related genes, in colonic tissues using RT-qPCR. As expected, pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β), were markedly elevated in the DSS group compared to the CONT group, reflecting acute intestinal inflammation. Notably, mice treated with SLAM 005Y and SLAM 023B showed a modest attenuation of these inflammatory markers, while the SYN group exhibited a more substantial reduction in TNF-α, IL-6, and IL-1β expression relative to the DSS group ([146]Fig. 6C–E). In contrast, expression of the anti-inflammatory cytokine IL-10, which modulates immune responses and plays a pivotal role in suppressing inflammation, was significantly downregulated in the DSS group compared to the CONT group. However, IL-10 levels were restored in all kefir-treated groups, with the SYN group showing a statistically significant increase (p = 0.0277) compared to DSS alone ([147]Fig. 6F). These results suggest that kefir-derived strains, particularly the SYN combination, downregulate pro-inflammatory cytokine genes and upregulate anti-inflammatory genes, thereby contributing to the attenuation of inflammation in DSS-induced colonic tissues. To further investigate the protective effects of kefir-derived probiotics on intestinal barrier function, we analyzed the expression levels of key tight junction (TJ) proteins that are critical for maintaining colonic epithelial integrity. TJ proteins enhance intercellular adhesion and play a vital role in preventing the translocation of harmful luminal substances. Compared to the DSS group, expression of TJ-related genes was elevated in all kefir-treated groups. Notably, claudin-1 and ZO-1 expression was significantly upregulated in the SYN group (p < 0.0001), suggesting enhanced epithelial barrier function ([148]Fig. 6G–I). In addition, MUC-2, the principal component of intestinal mucus that contributes to mucosal protection, was significantly increased in both the SLAM 005Y and SLAM 023B groups compared to DSS controls (p < 0.0001 and p = 0.0141, respectively) ([149]Fig. 6J). These findings indicate that kefir-derived strains promote the expression of tight junction-related genes, as well as mucin-producing gene, thereby contributing to the restoratif epithelial barrier integrity and enhancement of mucosal defense in DSS-induced colitis. 3.10. Multi-omics analysis with metagenomic and metabolomic approaches To evaluate the impact of kefir-derived microorganisms on gut microbiota composition in DSS-induced colitis, we performed metagenomic analyses using fecal samples collected on day 7 of DSS administration ([150]Fig. 7). Beta diversity, as visualized by PCoA based on both weighted and unweighted UniFrac distances, revealed distinct clustering patterns across treatment groups, indicating divergent microbial communities ([151]Fig. 7A–B). Alpha diversity was assessed using both the Shannon and Chao1 indices. The SYN group displayed the highest Shannon diversity, while the SLAM 023B group showed the lowest ([152]Fig. 7C); interestingly, the Chao1 index presented the inverse trend (p = 0.015 and p = 0.017, respectively) ([153]Fig. 7D). At the phylum level, DSS exposure was associated with a non-significant reduction in Firmicutes across all DSS-treated groups (p = 0.6581 for DSS, p = 0.9703 for SLAM 005Y, p = 0.8520 for SLAM 023B, and p = 0.9026 for SYN) ([154]Fig. 7E). At the family level, the abundance of Muribaculaceae (formerly S24-7) significantly declined across all DSS-treated groups, with the greatest reduction observed in the SYN group (p = 0.0004) ([155]Fig. 7F). Bifidobacteriaceae levels increased in the SLAM 005Y and SLAM 023B groups compared to the DSS group (p = 0.9957 and p = 0.9967, respectively), while Lachnospiraceae and Ruminococcaceae were significantly enriched in the SLAM 005Y group (p < 0.0001 and p = 0.0044, respectively). At the genus level, Aerococcus and Corynebacterium were elevated in the DSS group relative to CONT (p = 0.9368 and p = 0.6846), while Bacteroides decreased in all DSS-treated groups (p = 0.1005 for DSS, p = 0.9537 for SLAM 005Y, p = 0.1439 for SLAM 023B, and p = 0.1797 for SYN). Notably, Oscillospira, Ruminococcus, and Bifidobacterium were elevated in the SLAM 005Y group (p = 0.6548, p = 0.9965, and p = 0.7527, respectively), whereas Corynebacterium was significantly reduced in both the SLAM 023B and SYN groups (p = 0.0063 and p = 0.0145, respectively) ([156]Fig. 7G). Fig. 7. [157]Fig. 7 [158]Open in a new tab Multi-omics anaylsis with of fecal microbiota and fecal metabolites in DSS-induced colitis mice treated with kefir-derived microbial strains. (A–B) Principal Coordinate Analysis (PCoA) plots based on weighted and unweighted UniFrac distances revealed distinct clustering patterns, indicating significant differences in beta diversity between treatment groups. (C–D) Alpha diversity was assessed using Shannon and Chao1 indices to evaluate microbial richness and evenness; data are presented as mean ± SEM. (E–G) Taxonomic composition of gut microbiota was analyzed at the phylum (E), family (F), and genus (G) levels based on 16S rRNA gene sequencing. (H–J) Partial least squares discriminant analysis (PLS-DA) of fecal metabolomic profiles was performed on GC-MS–derived fecal metabolomic data using MetaboAnalyst 6.0 and revealed clear separation among treatment groups: (H) overall comparisons across all groups, (I) comparisons between DSS-treated groups, and (J) DSS vs. SYN groups, highlighting metabolic shifts following kefir microbial intervention. (K) Bar plot showing log[2] fold changes of significantly altered metabolites in the SYN group relative to the DSS group. To further explore metabolic shifts in gut environments, fecal metabolomic profiling was conducted using GC-MS. PLS-DA analysis demonstrated clear separation between groups, with distinct clustering of the CONT and DSS groups, while SLAM 005Y, SLAM 023B, and SYN formed a separate cluster ([159]Fig. 7H). When restricted to DSS-treated samples, SLAM 005Y and SLAM 023B clustered closely ([160]Fig. 7I), whereas SYN formed a distinct cluster from the DSS group ([161]Fig. 7J). Comparison of the SYN group with DSS controls revealed upregulation of 19 metabolites, including L-valine, L-isoleucine, 5-aminovaleric acid, aspartic acid, L-threonine, succinic acid, batyl alcohol, L-5-oxoproline, glycerol, coprostanol, and lactic acid ([162]Fig. 7K). These results suggest that kefir-derived microorganisms help restore the gut microbial balance disrupted by DSS-induced colitis, as evidenced by increased microbial diversity and enrichment of beneficial taxa such as Bifidobacterium, which are known to support intestinal health. Furthermore, metabolomic profiling revealed an increased abundance of metabolites, particularly branched-chain amino acids, in the SYN group, suggesting that kefir-derived microbes not only reshape the gut microbial community but also contribute to the restoration of metabolic homeostasis. These results collectively highlight the role of kefir-derived strains in regulating both microbial composition and metabolic profiles, supporting their potential as therapeutic candidates for gut dysbiosis and intestinal inflammation. 4. Discussion This study aimed to evaluate the probiotic potential of kefir-derived microorganisms, specifically Kluyveromyces marxianus SLAM 005Y and Lactobacillus kefiri SLAM 023B and to explore the synergistic effects of their combination (SYN). kefir is a fermented dairy product traditionally produced through the symbiotic activity of various lactic acid bacteria and yeasts ([163]Garrote et al., 2001). Among its dominant microbial residents are Lactobacillus kefiranofaciens, L. kefiri, K. marxianus, and Saccharomyces cerevisiae, which collectively contribute to its probiotic functionality and unique sensory properties ([164]Kim et al., 2020; [165]Youn et al., 2022). K. marxianus, which is known to dominate approximately 95 % of the yeast population in kefir, has been reported to possess probiotic potential due to its high survivability in the intestinal environment and strong adhesion capability to intestinal epithelial cells ([166]Maccaferri et al., 2012; [167]Shiby and Mishra, 2013; [168]Youn et al., 2022). K. marxianus has been reported to modulate the intestinal microbiota composition following oral administration in BALB/c mice ([169]Youn et al., 2023). Moreover, in a murine model of high-fat diet–induced obesity, K. marxianus administration significantly reshaped gut microbial communities and mitigated lipid metabolic dysregulation, suggesting its potential role in restoring gut–metabolic axis homeostasis ([170]Tang et al., 2025). L. kefiri, one of the most prominent lactobacilli isolated from kefir, has been shown to protect intestinal epithelial cells against Salmonella infection through its surface-layer proteins ([171]Carasi et al., 2014; [172]Golowczyc et al., 2007). In vivo studies further demonstrated that oral administration of L. kefiri stimulated gut mucosal immunity and modulated the composition of the intestinal microbiota in mice ([173]Carasi et al., 2015). Furthermore, the exopolysaccharide produced by L. kefiri has been reported to exert anticancer effects by inducing apoptosis in HT-29 human colorectal cancer cells ([174]Rajoka et al., 2019). Clinically, administration of L. kefiri was also associated with improvements in diarrhea symptoms among cancer patients ([175]Ghidini et al., 2021). The present investigation examined also investigated the synergistic effects between kefir-derived yeasts and lactic acid bacteria. In the kefir microbial consortium, the metabolic interplay between yeasts and lactic acid bacteria exemplifies mutualistic cooperation, wherein yeast-derived amino acids and peptides foster an environment conducive to LAB growth and activity ([176]Ponomarova et al., 2017; [177]Simova et al., 2006). Previous studies have reported that co-culturing kefir-derived L. kefiranofaciens and K. marxianus enhances extracellular polysaccharide (EPS) production, thereby improving microbial survivability in simulated digestion models ([178]González-Orozco et al., 2023). Similarly, co-cultivation of L. helveticus with K. marxianus has been shown to exhibit greater antioxidant activity compared to individual treatments ([179]Wang et al., 2023). In line with previous observations, the present study revealed that both K. marxianus SLAM 005Y and L. kefiri SLAM 023B, as well as their synergistic combination, significantly enhanced host immune responses in the C. elegans model and alleviated DSS-induced colitis by restoring gut microbial and metabolic homeostasis. In this study, the surrogate model organism C. elegans was employed. Due to its short lifespan, genetic tractability, and ease of cultivation, C. elegans serves as a valuable system for studying host–microbe interactions, aging, and innate immunity ([180]Amrit et al., 2014; [181]Anderson et al., 2003; [182]Choi et al., 2023; [183]Yoo et al., 2022). When compared to the normal feed E. coli OP50, exposure to SLAM 005Y, SLAM 006Y, and SLAM 003B significantly prolonged the lifespan of C. elegans, indicating that these kefir-derived strains may promote longevity and enhance host resilience. In addition to lifespan extension, the ability of kefir strains to modulate early immune responses was also considered. C. elegans mounts rapid innate immune defenses to combat microbial infections, and intestinal colonization by pathogens can negatively impact host survival ([184]Martineau et al., 2021; [185]Scott et al., 2019). Prior research has demonstrated that the probiotic yeast Saccharomyces boulardii can colonize the C. elegans gut and exert antibacterial activity against enteric pathogens ([186]Chelliah et al., 2021). However, studies specifically investigating the probiotic efficacy of other yeast strains in C. elegans remain limited, underscoring the need for further functional validation ([187]Kunyeit et al., 2021). Similarly, various Lactobacillus species have been shown to exert protective effects in vivo by enhancing host immune responses and suppressing pathogenic colonization ([188]Lee et al., 2011; [189]Zhou et al., 2014). Consistent with these findings, our results suggest that both SLAM 005Y and SLAM 023B confer health-promoting effects in C. elegans, supporting their candidacy as novel probiotic strains. These data further highlight the utility of C. elegans as a preliminary in vivo platform for screening probiotic functionality prior to mammalian validation. Building upon our findings in C. elegans, which demonstrated the anti-aging and immunomodulatory properties of kefir-derived microorganisms, we next sought to evaluate their protective effects in a mouse animal model of intestinal inflammation. Specifically, we examined whether K. marxianus SLAM 005Y, L. kefiri SLAM 023B, and their combination (SYN) could ameliorate colonic damage in a DSS-induced colitis mouse model. This chemically induced model is widely accepted as a preclinical platform for investigating inflammatory bowel disease (IBD), as it recapitulates many clinical and histopathological features of human ulcerative colitis, including weight loss, diarrhea, rectal bleeding, mucosal barrier disruption, and colonic inflammation ([190]Goyal et al., 2014; [191]Sann et al., 2013). While kefir microorganism treatment attenuated DSS-induced weight loss, no statistically significant differences were observed compared to the DSS group. This could be due to the relatively short duration of DSS exposure and the treatment period with kefir strains, which may not have been sufficient to observe significant physiological recovery. A longer pre-treatment or extended recovery phase may enhance the manifestation of kefir's protective effects against colitis-induced symptoms. Colon length, a commonly used marker of inflammation and mucosal injury, was significantly preserved in the SYN group compared to DSS-only controls, indicating a protective effect against colitis-associated tissue damage ([192]Chen et al., 2017; [193]Duary et al., 2012). Histological analysis further supported these findings. Consistent with previous reports of DSS-induced IBD, the colons of untreated DSS mice exhibited epithelial distortion, crypt loss, and extensive infiltration of inflammatory cells ([194]Xu et al., 2020). In contrast, kefir-treated groups showed substantially less histopathological damage and inflammatory cell infiltration. Pro-inflammatory cytokines including TNF-α, IL-1β, and IL-6, were significantly elevated in the DSS group, in line with prior studies ([195]Lacruz-Guzman et al., 2013). Treatment with SLAM 005Y, SLAM 023B, and SYN led to marked reductions in these cytokines. Moreover, expression of the anti-inflammatory cytokine IL-10, which was suppressed by DSS exposure, was restored in all kefir-treated groups, with the SYN group showing the greatest increase ([196]Barbara et al., 2000). These findings suggest that kefir-derived strains modulate colonic inflammation by downregulating proinflammatory cytokines, while concurrently enhancing the expression of anti-inflammatory mediators. This modulation may contribute to the restoration of immune homeostasis and epithelial inflammation in the DSS-induced colitis model. Collectively, these results support the therapeutic potential of SLAM 005Y, SLAM 023B, and their combination in attenuating intestinal inflammation and enhancing epithelial repair in inflammatory bowel disease. Although the precise etiology of IBD remains incompletely understood, disruption of the intestinal epithelial barrier is recognized as a hallmark feature of disease pathology ([197]Pope et al., 2014). TJ proteins play a crucial role in maintaining intestinal barrier integrity and regulating paracellular permeability ([198]Lee, 2015). In DSS-induced colitis models, expression of these proteins is typically downregulated, leading to increased intestinal permeability and inflammation ([199]Liu et al., 2023). In the present study, kefir microbial treatment significantly restored TJ protein expression levels suppressed by DSS exposure. Notably, Claudin-1 and ZO-1 were most strongly upregulated in the SYN-treated group compared to the DSS group, consistent with previous findings highlighting their loss in colitis. However, it should be acknowledged that the expression patterns and roles of specific TJ proteins may vary depending on the type and severity of intestinal disease ([200]Sawada, 2013). In addition to tight junction proteins, the mucus layer provides a critical line of defense against pathogenic infiltration. Among mucin glycoproteins, MUC2, a secretory mucin produced by goblet cells, is the principal component of this layer and is essential for mucosal protection in both humans and mice ([201]Van der Sluis et al., 2006). MUC2 expression is typically reduced in DSS-induced colitis, contributing to barrier dysfunction and increased susceptibility to infection ([202]Bankole et al., 2021). Our findings demonstrated that kefir microbial administration significantly enhanced colonic MUC2 gene expression, as determined by qPCR analysis. This elevation in MUC2 levels suggests enhanced goblet cell activity and mucin production, which are critical for forming a protective mucus barrier. This increase was especially pronounced in the SLAM 005Y group and was associated with increased goblet cell numbers, consistent with previous studies showing similar outcomes following probiotic administration ([203]Morampudi et al., 2016). Collectively, these results suggest that SLAM 005Y, SLAM 023B, and their synergistic combination promote epithelial barrier restoration through upregulation of both tight junction and mucin genes. These effects likely contribute to improved intestinal structural integrity and a stabilized mucosal environment in the context of colitis. Such findings support the therapeutic potential of kefir-derived probiotics in managing IBD by enhancing mucosal defense mechanisms. Transcriptomic analysis in C. elegans revealed that kefir-derived strains modulate the expression of genes associated with bacterial defense, external stimulus response, and immune activation including PMK-1. More importantly, oral administration of these strains in mice resulted in upregulated expression of MUC-2 and Il-10, indicating enhanced mucosal protection and anti-inflammatory responses. These findings suggest that the observed probiotic properties—such as mucosal adhesion and antimicrobial activity—may contribute to the therapeutic effects. However, the precise molecular pathways mediating these effects remain to be elucidated and warrant further mechanistic investigations. The gut microbiome is increasingly recognized as a key factor in the pathogenesis and progression of IBD, playing a dual role in either promoting or attenuating intestinal inflammation ([204]Manichanh et al., 2012). In murine models, including DSS-induced colitis, microbial dysbiosis and a marked reduction in gut microbial diversity are commonly observed ([205]Munyaka et al., 2016). Similar patterns of decreased microbial richness and compositional shifts have been documented in both human IBD patients and animal models ([206]Samanta et al., 2012). In the present study, the administration of kefir-derived microorganisms, particularly SLAM 005Y and SYN, resulted in increased alpha diversity compared to the DSS group, suggesting a partial restoration of microbial diversity disrupted by colitis. Furthermore, beta diversity analysis revealed distinct clustering between the SYN and DSS groups, indicating significant shifts in microbial community composition following kefir microbial treatment. The dominant bacterial phyla in the mammalian gut microbiota include Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria ([207]Jo et al., 2024; [208]Tlaskalova-Hogenova et al., 2011; [209]Yu et al., 2022). A consistent hallmark of IBD-associated dysbiosis is the expansion of Proteobacteria, which is thought to reflect a destabilized gut ecosystem and heightened inflammatory tone ([210]Ni et al., 2017). In line with previous findings, the DSS group in this study exhibited an increased relative abundance of Proteobacteria. However, kefir microbial treatment led to a notable reduction in Proteobacteria abundance. In contrast, the abundance of S24-7, a commensal bacterial group typically enriched in healthy mice, increased significantly in the SLAM 023B and SYN groups. The observed decrease in Proteobacteria following kefir treatment further supports its anti-inflammatory potential, consistent with earlier studies linking Proteobacterial overgrowth to intestinal inflammation and colitis ([211]Mukhopadhya et al., 2012; [212]Rizzatti et al., 2017; [213]Shin et al., 2015). Moreover, genera such as Bifidobacterium and Parabacteroides, known for their probiotic activity and considered next-generation probiotic candidates ([214]Gomes and Malcata, 1999; [215]Menberu et al., 2022; [216]Sun et al., 2023), were increased in the SLAM 023B and SYN groups. These changes suggest that kefir microorganisms contribute to the restoration of gut microbial balance following DSS-induced dysbiosis, as evidenced by increased microbial diversity and the enrichment of beneficial taxa. Taken together, these findings support the potential of SLAM 005Y, SLAM 023B, and their combination to modulate gut microbial communities in colitis, thereby promoting microbial and metabolic homeostasis. Further mechanistic investigations are required to delineate the specific host–microbe interactions through which kefir-induced alterations in the gut microbiota mediate immunomodulatory effects and facilitate epithelial regeneration in the context of intestinal inflammation. In conjunction with our metagenomic findings, metabolomic profiling was conducted to further explore host responses to kefir microbial supplementation. Comparative analysis of fecal metabolites between the SYN and DSS groups revealed several distinct changes, particularly in amino acid profiles. Amino acids are central regulators of numerous metabolic pathways, and our results showed that BCAAs including L-valine and L-isoleucine, as well as aspartic acid and L-threonine, were significantly upregulated in the kefir-treated group. Prior studies have demonstrated that BCAAs play key roles in modulating inflammation, cellular stress responses, energy production, muscle metabolism, and immune function ([217]Xie et al., 2021; [218]Yan et al., 2023). Previous studies have demonstrated that dietary supplementation with BCAAs enhances intestinal development and epithelial cell proliferation by increasing the intestinal absorption of amino acids and glucose ([219]Zhou et al., 2018). In mice, BCAAs restriction has been shown to impair immune function and increase susceptibility to pathogenic infections ([220]Jose and Good, 1973). Amino acids such as L-valine and L-isoleucine are known to serve as substrates for immune cells and influence protein synthesis, thereby contributing to immune competence ([221]Calder, 2006). Moreover, BCAAs have been reported to regulate immune responses through activation of the mTOR signaling pathway, further underscoring their role in maintaining mucosal immunity ([222]Kimball and Jefferson, 2006). Notably, aspartic acid has been implicated in the development of visceral pain associated with intestinal inflammation, suggesting a possible link between elevated levels and inflammatory signaling in the gut ([223]Zhou and Nicholas Verne, 2008). Among the differentially regulated metabolites, arachidonic acid that a lipid mediator known to support mucosal repair, was also elevated following kefir microbial intervention ([224]Xu et al., 2021). These findings indicate that kefir microorganisms may contribute to the restoration of metabolic homeostasis, possibly through the modulation of key metabolites involved in intestinal regeneration and energy metabolism. The upregulation of BCAAs and other bioactive metabolites suggests that kefir supplementation could counteract the metabolic dysregulation associated with DSS-induced colitis. While these metabolomic alterations provide important insights into the potential mechanisms underlying the protective effects of kefir microbes, several limitations must be acknowledged. The precise microbial–host interactions responsible for these metabolic shifts remain to be clarified, and the causal links between specific metabolites and disease outcomes require further investigation. Future studies should focus on elucidating the microbial enzymatic pathways involved in metabolite production and their downstream effects on host physiology and immune regulation. A deeper mechanistic understanding—particularly of the microbial metabolic pathways and their downstream effects on epithelial repair and immune modulation—will be critical for developing targeted probiotic strategies to treat IBD using kefir-derived strains. 5. Conclusion Our study highlights the promising potential of K. marxianus SLAM 005Y, L. kefiri SLAM 023B, and their synergistic combination as multifunctional probiotic candidates isolated from milk kefir capable of enhancing host immune responses, extending lifespan, and ameliorating intestinal inflammation ([225]Fig. 8). Through an integrative approach combining gene expression profiling, microbiota analysis, and metabolomic evaluation, we provide mechanistic insight into how these kefir-derived microorganisms exert their beneficial effects. This study provides a foundational framework for future research investigating the specific mechanistic pathways through which kefir-derived microorganisms interact with the gut microbiota to regulate host immune responses and promote gastrointestinal health. Importantly, while the current study demonstrates efficacy in C. elegans and murine models of colitis, translational validation through human clinical trials is essential to determine their therapeutic relevance in the prevention and management of IBD. Further exploration of the interactions between kefir microbes and the host gut microbiome will be critical for understanding their broader impact on intestinal homeostasis and systemic health. Our work contributes to the growing body of evidence supporting the use of probiotic yeasts and bacteria as functional agents with therapeutic potential. By demonstrating consistent benefits across multiple biological models, this study supports the advancement of kefir-based probiotics as novel interventions for gut health and inflammation-related disorders. Fig. 8. [226]Fig. 8 [227]Open in a new tab Schematic representation of the protective effects of the synergistic combination of K. marxianus SLAM 005Y and L. kefiri SLAM 023B in a DSS-induced colitis mouse model. The left panel illustrates the pathological features associated with DSS-induced colitis, including dysbiosis, increased pro-inflammatory cytokines, disrupted intestinal barrier integrity, and reduced mucin and tight junction (TJ) protein expression. In contrast, the right panel depicts the therapeutic effects of SYN administration. SYN modulates the gut microbiota composition, leading to elevated production of beneficial metabolites such as branched-chain amino acids, amino acids, and fatty acids. These changes enhance mucin secretion and upregulate the expression of TJ proteins (claudin-1, occludin, ZO-1), thereby reinforcing epithelial barrier integrity and limiting microbial translocation. Furthermore, SYN suppresses pro-inflammatory cytokines (TNF-α, IL-6, IL-1β) while enhancing anti-inflammatory cytokine IL-10 expression, collectively contributing to reduced intestinal inflammation. This schematic highlights the role of milk kefir-derived microbial supplementation in restoring gut homeostasis and mitigating DSS-induced colitis via microbiome, metabolite, and immune modulation. CRediT authorship contribution statement Se Hyun Lim: Writing – original draft, Methodology, Investigation, Formal analysis, Conceptualization. Visualization, Software, Writing – review & editing. Daye Mun: Writing – original draft, Methodology, Investigation, Formal analysis, Conceptualization. Visualization, Software, Writing – review & editing. Sangdon Ryu: Writing – original draft, Methodology, Investigation, Formal analysis, Conceptualization. Min-Geun Kang: Writing – review & editing, Visualization, Software, Methodology. Anna Kang: Writing – review & editing, Visualization, Software, Methodology. Daniel Junpyo Lee: Writing – review & editing, Visualization, Software, Methodology, Data curation. Eunsol Seo: Writing – review & editing, Visualization, Software, Methodology. Seon-hui Son: Writing – review & editing, Visualization, Software, Methodology. Soyeon Kim: Writing – review & editing, Visualization, Software, Methodology. Ki Beom Jang: Writing – review & editing, Visualization, Software, Methodology. Bum-Keun Kim: Visualization, Software, Methodology. Dong-Jun Park: Visualization, Software, Methodology. Sangnam Oh: Writing – review & editing, Supervision, Project administration, Investigation, Conceptualization. Younghoon Kim: Writing – review & editing, Supervision, Project administration, Investigation, Funding acquisition, Conceptualization. Ethics approval and consent to participate Not applicable. Consent for publication Not applicable. Data availability Additional data is available from the authors upon request. Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. Acknowledgments