Randomized Controlled Trial Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Mar 28, 2025; 31(12): 104081
Published online Mar 28, 2025. doi: 10.3748/wjg.v31.i12.104081
Randomized study of Lacticaseibacillus fermented milk in Indonesian elderly houses: Impact on gut microbiota and gut environment
I Nengah Sujaya, School of Public Health, Faculty of Medicine, Udayana University, Denpasar 80230, Bali, Indonesia
Mariyatun Mariyatun, Pratama Nur Hasan, Nancy Eka Putri Manurung, Putrika Citta Pramesi, Endang Sutriswati Rahayu, Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
Mariyatun Mariyatun, Pratama Nur Hasan, Nancy Eka Putri Manurung, Putrika Citta Pramesi, Tyas Utami, Endang Sutriswati Rahayu, Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
Putrika Citta Pramesi, Tyas Utami, Muhammad Nur Cahyanto, Endang Sutriswati Rahayu, Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
Mohammad Juffrie, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
Shuta Yamamoto, Takashi Asahara, Takuya Akiyama, Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi 186-8650, Tōkyō, Japan
Takuya Takahashi, Yakult Honsha European Research Center for Microbiology VOF, Ghent 9052, East Flanders, Belgium
ORCID number: I Nengah Sujaya (0000-0002-7826-2831); Mariyatun Mariyatun (0000-0002-7998-2531); Pratama Nur Hasan (0000-0001-7780-2421); Nancy Eka Putri Manurung (0000-0002-5182-7433); Putrika Citta Pramesi (0000-0003-0246-5189); Mohammad Juffrie (0000-0002-2862-3897); Tyas Utami (0000-0003-3600-6060); Muhammad Nur Cahyanto (0000-0002-1448-8762); Shuta Yamamoto (0000-0001-6236-3833); Takuya Takahashi (0000-0001-9998-2611); Takashi Asahara (0000-0001-5126-0636); Takuya Akiyama (0000-0002-3124-2040); Endang Sutriswati Rahayu (0000-0002-6101-3433).
Author contributions: Sujaya IN, Mariyatun M, Hasan PN, Manurung NEP, Pramesi PC, Yamamoto S, Takahashi T, and Asahara T contributed to the investigation of this manuscript; Sujaya IN, Cahyanto MN, and Rahayu ES were involved in the supervision of this manuscript; Sujaya IN and Rahayu ES contributed to the validation of this manuscript; Sujaya IN wrote the original draft; Mariyatun M, Hasan PN, and Akiyama T performed formal analysis; Mariyatun M and Akiyama T contributed to the data curation; Mariyatun M and Hasan PN participated in the resources; Mariyatun M, Juffrie M, Utami T, Yamamoto S, Takahashi T, Asahara T, Akiyama T, and Rahayu ES contributed to the methodology of this manuscript; Mariyatun M performed project administration and funding acquisition; Mariyatun M, Pramesi PC, and Akiyama T were involved in the writing - review and editing of this manuscript; Manurung NEP contributed to the data curated; Juffrie M, Utami T, Cahyanto MN, and Rahayu ES responsible in the conceptualization of this manuscript; Akiyama T contributed to the software.
Supported by the Yakult Honsha Co., Ltd., No. 1226/FTP-UGM/HK/2018.
Institutional review board statement: This study was approved by the Medical and Health Research Ethics Committee, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, with reference number KE/FK/0202/EC/2018 (approval date: March 16, 2018) and an amendment with reference number KE/FK/0202/EC/2018 (approval date: September 13, 2018).
Clinical trial registration statement: This study was registered at ClinicalTrials.gov. The registration number is NCT05308745.
Informed consent statement: All study participants or their legal guardians provided written informed consent before study enrollment.
Conflict-of-interest statement: Yamamoto S, Takahashi T, Asahara T, and Akiyama T are affiliated with Yakult Honsha Co., Ltd. The authors declare that the research was conducted in the absence of commercial or financial relationships that could be construed as potential conflicts of interest.
CONSORT 2010 statement: The authors have read the CONSORT 2010 Statement, and the manuscript was prepared and revised according to the CONSORT 2010 Statement.
Data sharing statement: The next-generation sequencing data from this study are publicly available through the DDBJ Sequence Read Archive under accession number DRA014200.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Endang Sutriswati Rahayu, PhD, Professor, Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Jl. Flora No. 1 Bulaksumur, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia. endangsrahayu@ugm.ac.id
Received: December 10, 2024
Revised: January 21, 2025
Accepted: February 24, 2025
Published online: March 28, 2025
Processing time: 107 Days and 4.2 Hours

Abstract
BACKGROUND

Health maintenance in elderly houses includes management of the gut microbiota and the environment. Lacticaseibacillus paracasei Shirota (LcS) is a probiotic strain that positively affects the human gut. However, the evidence of its effects on the Indonesian population remains limited.

AIM

To investigate the effect of LcS-fermented milk on the gut microbiota and environment of Indonesian elderly houses.

METHODS

This double-blind, randomized, placebo-controlled trial involved 112 participants from Indonesian elderly houses, spanning a 2-week baseline and 24-week treatment. Participants were randomly assigned to probiotic or placebo groups, consuming fermented milk with or without LcS (> 6.5 × 109 colony-forming units). Fecal samples were collected every three months. Gut microbiota analysis was performed using 16S rRNA gene sequencing and reverse transcription quantitative polymerase chain reaction, while gut environment was assessed by measuring fecal organic acids, amino acid metabolites, and stool frequency.

RESULTS

Analyses of 16S rRNA gene sequence data at the 3-month period revealed increased Bifidobacterium and Succinivibrio and decreased Rikenellaceae RC9 gut group in the probiotic group. These shifts were associated with significant differences in β-diversity metrics. The change in Bifidobacterium was confirmed by reverse transcription quantitative polymerase chain reaction, demonstrating higher abundance in the probiotic group than in the placebo group (8.5 ± 1.1 vs 8.0 ± 1.1, log10 bacterial cells/g; P = 0.044). At 6-month period, the differences in Succinivibrio and Rikenellaceae RC9 gut group persisted. The probiotic group showed higher butyrate levels than the placebo group at the 6-month period (5.04 ± 3.11 vs 3.95 ± 2.89, μmol/g; P = 0.048). The effect on amino acid metabolites and stool frequency was not significant.

CONCLUSION

Daily intake of LcS positively affects the gut microbiota and environment of people living in Indonesian elderly houses.

Key Words: Lacticaseibacillus paracasei Shirota; Fermented milk; Intestinal microbiota; Elderly; Probiotics

Core Tip: The daily consumption of fermented milk containing Lacticaseibacillus paracasei Shirota significantly altered the gut microbiota among individuals in Indonesian elderly houses. Notably, these changes included increases in Bifidobacterium and Succinivibrio and a decrease in the Rikenellaceae RC9 gut group. These modifications are associated with an improvement in the gut environment, as indicated by elevated butyrate levels.



INTRODUCTION

The increase in life expectancy is driving a substantial demographic shift toward an aging population worldwide. Indonesia, ranked the fourth most populous country, is not exempt from this phenomenon. The proportion of the elderly population (aged 60 years and older) is expected to increase to 11% by 2035[1]. The elderly population is growing at an annual rate of 4.7%, which exceeds the general population growth rate of 2.9%[2]. Simultaneously, the country’s socioeconomic and environmental development has resulted in changes to traditional community structures, prompting the elderly to relocate from rural to urban areas, where they often live independently. Given these demographic and lifestyle dynamics, the significance of elderly facilities (or houses) is increasing, playing a crucial role in preserving the health and well-being of the aging population.

The gut microbiome and environment are crucial factors affecting the health of elderly individuals. Unhealthy aging has been associated with a decline in gut microbial diversity, characterized by a reduction in beneficial bacteria (e.g., Bifidobacterium) and an increase in opportunistic and/or pro-inflammatory bacteria[3,4]. Such shifts affect microbial consortium metabolism, leading to decreased production of organic acids, including acetate and butyrate, excessive production of putrefactive metabolites, such as ammonia and phenols, and compromised immune function[3,4]. These alterations are driven by the complex interplay between the genetic, environmental, and lifestyle factors associated with aging, ultimately contributing to the loss of systemic homeostasis and functional decline.

Moreover, residents of elderly facilities face an increased risk of infection and colonization by multidrug-resistant organisms in the intestine[5]. Addressing these concerns involves regulating the microbiota surrounding the elderly, including that of individuals who regularly interact with them, and preserving the microbiota of the residents themselves. Haran et al[6] demonstrated that residents in close proximity share similar species and strains. This finding underscores the importance of preserving the gut microbiota and environment of elderly residents and individuals in proximity, such as facility staff, to mitigate the risk of horizontal transmission or infection by health-threatening strains within the facility.

Administration of probiotics is a promising method for sustaining the health of residents in elderly houses by modulating their gut microbiota and the environment. According to the International Scientific Association for Probiotics and Prebiotics, probiotics are live microorganisms that, when administrated in adequate amounts, confer a health benefit on the host[7]. Systematic reviews have shown the potential benefits of probiotics in improving gut health and mitigating age-related issues, such as constipation, common cold incidence/duration, and other infections, although some conflicting results exist owing to strain and study design variations[8,9]. The Lactobacillus casei strain Shirota, currently reclassified as Lacticaseibacillus paracasei Shirota (LcS), is a well-known probiotic that has a longstanding presence in the global market[10]. Clinical trials on the impact of LcS-fermented milk in nursing homes have shown positive outcomes, including increased levels of beneficial bacteria such as bifidobacteria and lactobacilli, and a reduction in potentially harmful bacteria such as Clostridioides difficile. These changes are associated with increased organic acid production, decreased intestinal pH (indicating gut environment improvement), and reduced risk of health complications in elderly residents, such as shorter duration of fever caused by norovirus infections and improved bowel movements[11,12]. Notably, Nagata et al[12] demonstrated that the positive impacts of LcS on the gut microbiota and environment extend not only to elderly residents but also to staff in the facility.

This indicates that LcS-fermented milk is beneficial for maintaining optimal gut conditions for individuals in elderly houses, thereby contributing to their overall health. However, the effects of LcS in the Indonesian population have not been adequately studied. Therefore, the present study aimed to investigate the effects of daily LcS-fermented milk intake on the gut microbiota and environment of individuals in Indonesian elderly houses, comprising residents and staff within a randomized controlled setting.

MATERIALS AND METHODS
Trial design

This was a multicenter, double-blind, placebo-controlled, parallel-group superiority study with balanced randomization (1:1) for the two arms.

Participants

Residents and staff of three elderly houses in Denpasar, Singaraja, and Tabanan, Bali, Indonesia, were studied. Bali is ranked third in Indonesia for its proportion of elderly individuals (13.5%) in 2022, followed by DI Yogyakarta (16.7%) and East Java (13.7%)[2]. Previous studies have reported that residents of elderly houses in Bali exhibit a less favorable gut microbiome than those in Yogyakarta, characterized by a higher abundance of potentially detrimental bacteria, such as Clostridium perfringens and Enterococcus[13]. The present study included individuals who provided informed consent and were confirmed to be physically and mentally healthy by doctors with no issues with regular meals. The exclusion criteria were excessive alcohol intake (> 28 glasses/week for men and > 21 glasses/week for women); narcotic/psychotropic drug use; recent antibiotic/laxative use (within 2 weeks prior to the study); recent consumption of probiotics, prebiotics, or fermented dairy foods (within 2 weeks); irritable bowel syndrome (diagnosed according to the Rome IV criteria); history of gastrointestinal surgery; pregnancy; and allergies to the study product. Additionally, individuals who could not avoid consuming fermented dairy products, probiotics, or prebiotics during the study period were excluded.

Interventions

Participants were randomized to receive either a fermented milk drink containing > 6.5 × 109 colony-forming units LcS/65 mL (Yakult®) or an acidified milk placebo matched in nutritional values and organoleptic characteristics. Following a 2-week baseline period, the participants consumed one bottle of their assigned product daily at breakfast for 24 weeks (6 months). The bacterial count used in this study reflected the composition of a commercially available product, which was designed to be consumed in one bottle per day. This dosing approach enables the evaluation of the intervention under conditions that closely simulate real-world use, offering practical insights into its applicability and effectiveness in the elderly community. During the study, the fermented milk samples were handled according to the manufacturer’s instructions to maintain the stability of the minimum viable cell count during the study. The products were stored in a refrigerator at 4-10 °C. Temperature was monitored using a thermohygrometer (TFA Dostmann, Germany). The consumption of the assigned product was monitored by collecting empty bottles.

Outcomes

The primary endpoint was the impact of the study product on the gut microbiome, which was assessed through fecal samples collected at the end of the 2-week baseline period and 3 and 6 months post-treatment (all with a ± 1-day window). The samples were subjected to 16S rRNA gene sequencing and reverse transcription quantitative polymerase chain reaction (RT-qPCR) to identify robust microbiome alterations. The secondary endpoint was the evaluation of gut environmental indicators through the measurement of organic and amino acid metabolite levels and fecal pH in the collected samples. In addition, stool frequency was assessed throughout the study.

Fecal sample collection and transportation

The protocol for fecal sample collection differed between the residents and staff. Elderly facility residents collected a portion of their stool onto trail paper (Eiken Chemical, Japan) and transferred it to a sterile 100 mL container using a disposable spoon. Subsequently, the filled container was delivered to the investigator and stationed at a pre-established clinical center within the facility. The investigator then aliquoted the sample into three sampling tubes labeled A, B, and C (cat. no. 80.734.001, Sarstedt, Germany). Tube A was prefilled with 2 mL of RNAlater (Thermo Fisher Scientific, Waltham, MA, United States) and zirconia beads. Immediately after addition of the stool, tube A was vigorously mixed using a vortex mixer. Staff residing outside the facilities received a collection kit packaged in an insulated box (cat. no. KC-3; Karux, Japan) containing two ice packs (cat. No. 160240; Yakult Shoji, Tokyo, Japan). They were instructed to collect their stool directly into three pre-labeled tubes (A, B, and C). Following sample collection, the staff delivered the samples to the investigator at their designated clinical center within 1 hour, maintaining cool conditions during transport. The samples collected in the evening were acceptable for delivery the following morning. Tube A was then delivered via refrigerated shipping to the rental P2 laboratory of Narita Animal Science Laboratory, Ltd. (Chiba, Japan) for subsequent nucleic acid extraction. After extraction, the samples were sent to the Yakult Central Institute (Tokyo, Japan) for microbiome analysis. Tubes B and C were delivered to the Prodia Clinical Laboratory/Prodia Occupational Health Institute, Jakarta, Indonesia, in a frozen state for organic acid and amino acid metabolite analyses.

Gut microbiota analysis

16S rRNA gene sequencing and taxonomic assignment were performed as previously described[14]. Fecal DNA was extracted by bead-beating in phenol[15]. Subsequently, variable regions 1 and 2 (V1 and V2) of the 16S rRNA gene were amplified using the primers 27Fmod2-MiSeq and 338R-MiSeq (Supplementary Table 1). Amplicons were sequenced on a MiSeq System using a paired-end 2 × 250 bp cycle run and the MiSeq Reagent Kit v2 (Illumina, CA, United States). The Quantitative Insights into Microbial Ecology 2 (QIIME2) software package (version 2018.8)[16] was used to analyze the amplicon sequence reads. DADA2 was employed to filter low-quality sequences, denoise, concatenate 16S rRNA reads, and remove potential chimeric sequences[17]. The resulting features were clustered into phylotypes (clustered features) using q2-vsearch for open reference clustering against the Living Tree Database (LPTs132_SSU)[18]. The taxonomic assignment for each phylotype was achieved using the QIIME feature classifier against the SILVA database (Release 138)[19] with a minimum bootstrap threshold of 50%. Alpha diversity (the number of OTUs observed, Chao1, Shannon index, and Faith’s phylogenetic diversity) and β-diversity (Bray-Curtis, Jaccard, Weighted UniFrac, Unweighted UniFrac, and Aitchison distance) were estimated for 6500 randomly selected sequences.

16S and 23S rRNA-targeted RT-qPCR was performed using the Yakult Intestinal Flora-Scan system (YIF-SCAN®)[20]. Fecal RNA was isolated as previously described[11,12]. A standard curve was generated using the threshold cycle (Ct), the cycle number at which the threshold fluorescence was reached, and the corresponding cell count, which was determined microscopically using 4,6-diamidino-2-phenylindole (Vector Laboratories, Burlingame, CA, United States) staining for the dilution series of standard strains described elsewhere[11,12]. To determine the bacteria present in the samples, three serial dilutions of the extracted RNA samples were used for RT-qPCR. Ct values in the linear range of the assay were applied to the standard curve generated in the same experiment to obtain the corresponding bacterial cell count for each nucleic acid sample, which was then converted to the number of bacteria per sample. The specificity of the RT-qPCR assay using group-, genus-, or species-specific primers was determined as previously described[11,12]. The primers used in this study and their annealing temperatures are listed in Supplementary Table 2.

Organic acid and pH analysis

Organic acids, including acetate, formate, propionate, isobutyrate, butyrate, isovalerate, valerate, isocaproate, caproate, heptanoate, lactate, and succinate, were analyzed following established protocols with minor modifications[21-24]. Briefly, a 200 mg fecal sample was suspended in 1 mL of ultrapure water, homogenized with a vortex, and sonicated for 20 minutes. The homogenate was centrifuged at 10000 × g for 5 minutes, and the resulting clear brownish supernatant was transferred to separate tubes for analysis. For the organic acids except succinate and lactate, 300 μL of ultrapure water containing the internal standard (2,2-dimethyl butyric acid; Merck, Germany) was added to 100 μL of fecal homogenate supernatant. Then, 100 μL of the mixture was combined with 600 μL of a diluent containing isopropanol (Merck) and 1.5 M hydrochloric acid (Merck) and transferred to gas chromatography (GC) vials. For succinate and lactate, 75 μL of 3 M hydrochloric acid in methanol was added to 25 μL of fecal homogenate supernatant. Subsequently, 400 μL of methanol (Merck) was added to the 100 μL derivatized mixture and transferred to GC vials. External standards were prepared to create calibration curves. Samples were injected into a 7890 Gas Chromatography System coupled with a 5977 mass-selective detector equipped with an electron impact source (Agilent Technologies, CA, United States). Organic acids were separated using a Nukol column (film thickness: 30 m × 0.25 mm × 0.25 μm; Supelco, PA, United States). The GC oven temperature was increased from 60 °C to 150 °C at a rate of 10 °C/minute. The mass-selective detector was operated in scan mode, acquiring data within a mass range of 40-150 m/z. The fecal pH was measured by inserting the probe of a pH meter (Eutech Instruments, Singapore) into the fecal sample.

Amino acid metabolite analysis

Indole, p-cresol, phenol, and ammonia levels were quantified by distillation, followed by GC with flame ionization detection (Agilent Technologies, CA, United States) for indole, p-cresol, and phenol, and spectrophotometry for ammonia. Briefly, fecal samples (0.5 g) were homogenized with 0.5 g of phosphate buffer, and Milli-Q water (ultrapure/type 1 water) up to 5 g was added. The homogenate was transferred to a distillation apparatus and distilled at 110 °C for 180 minutes. In measuring indole, p-cresol, and phenol, the distillate was collected in a 4 mL test tube containing 500 μL of methanol. Subsequently, the collected distillate was diluted to 5 mL with Milli-Q water, and 1 mL of the diluted solution was transferred to a 2 mL vial for GC with flame ionization detection. For ammonia measurement, the distillate was collected in a 4 mL test tube containing 500 μL of 12.04 N hydrochloric acid. Using Milli-Q water, the collected distillate was diluted to 5 mL, followed by 20-fold dilution. Subsequently, 5 mL of the 20-fold diluted sample was mixed with 0.2 mL of phenol solution (mixture of liquefied phenol (≥ 89%) and 95% v/v ethyl alcohol, in 1:8.009 ratio, respectively), 0.2 mL of 0.5% w/v sodium nitroprusside solution, and 0.5 mL of oxidizing solution (alkaline citrate solution and sodium hypochlorite mixture, in 4:1 ratio, respectively) in a 12 mL test tube. The test tube was covered and incubated for 1 hour at 22-27 °C (or room temperature). Absorbance was measured at 640 nm using a spectrophotometer (Agilent, China).

Stool frequency

The participants reported their daily stool frequency and compliance with the study protocol throughout the baseline and intervention periods using a self-completed diary.

Sample size

The sample size was determined to replicate the findings of a previous study by Nagata et al[12], which included all elderly residents and staff in an elderly care facility. We hypothesized that administering probiotics to the entire community might improve the gut microbiome, such as, partially by reducing the transmission of pathogenic bacteria. Accordingly, we planned to recruit all eligible residents and staff from participating facilities, resulting in a total of 112 participants. The rationale for this sample size was further supported by a previous intervention study, which reported mean lactobacilli counts of 6.20 ± 0.82 Log10 bacterial cells/g in the treatment group and 5.73 ± 0.66 Log10 bacterial cells/g in the control group following four weeks of commercial probiotic consumption. Using a two-sided test with a 5% significance level and 85% power, the required sample size was calculated to be 45 participants per group. After accounting for a 20% dropout rate, the adjusted sample size was 56 participants per group (112 participants in total). This calculation ensured a balanced sample size sufficient to yield reliable and generalizable results.

Randomization

A statistician from Prodia, the CRO (Jakarta, Indonesia), who was independent of participant recruitment, generated a random allocation sequence using block randomization with a block size of four, stratified by participant type (resident/staff) and study site (Denpasar/Singaraja/Tabanan). PT Yakult Indonesia Persada (Jakarta, Indonesia) concealed the allocation sequence and dispensed blinded investigational products.

Blinding

To maintain participant and investigator blinding, the treatment allocation was concealed using a randomization code. The investigational products were labeled with unique codes (“SBMC” and “SBMD”), one corresponding to the placebo and the other to LcS fermented milk. However, the labels did not disclose which code represented which intervention, ensuring that the participants and site personnel could not identify their assigned treatment based on appearance or taste. Furthermore, fecal tubes were labeled with unique identifiers corresponding to the randomization code to ensure blinded fecal sample analysis.

Statistical analysis

This study employed an intention-to-treat approach, incorporating all participants regardless of loss to follow-up, noncompliance, or withdrawal. Differential abundance analysis on genus-level microbiome data was conducted at each time point using ALDEx2, ANCOM, and ANCOM-BC, with adjusting for participant type (resident/staff) and study site (Denpasar/Singaraja/Tabanan) effects[25-29]. The top and bottom 20 bacteria were identified using the effect size (ALDEx2), centered log ratio mean difference (ANCOM), and log-fold change (ANCOM-BC), and their significance testing information was reported. This methodological approach adheres to the recommendation of employing multiple methods to interpret differential abundance data in microbial communities[30]. Genera were considered differentially abundant if they showed significance in at least one analysis with consistent ordering in all analyses. Significance testing for global changes in community composition (β-diversity) was performed using permutational multivariate analysis of variance based on Bray-Curtis, Jaccard, Weighted UniFrac, Unweighted UniFrac, and Aitchison distance metrics. The analysis was adjusted for participant type and study site using the Adonis function in the vegan package[31]. Between-group differences for continuous outcomes were evaluated using the Student’s t-test after log or square root transformation, if necessary, for data normalization. Alternatively, the nonparametric Mann-Whitney U test was used for non-normalized data. Categorical variables were compared using the Fisher’s exact test. Mixed linear (or logistic) models were constructed based on the framework of Twisk et al[32], adjusting for baseline values, participant type, and study site with the appropriate link function applied. A subgroup analysis was conducted to assess the differences in treatment responses between residents and staff by evaluating the interactions between participant type and treatment. P < 0.05 was considered statistically significant. All analyses were performed using R software (version 4.1.1; The R Foundation for Statistical Computing, Vienna, Austria).

RESULTS
Participant flow and recruitment

Figure 1 shows a participant flow diagram. Eligible participants were recruited from three elderly houses in Bali, Indonesia in September 2018. Overall, 112 participants were randomized using a stratified design according to participant type (resident/staff) and facility location (Denpasar/Singaraja/Tabanan). This resulted in 56 participants (33 residents and 23 staff) allocated to the placebo group and 56 participants (34 residents and 22 staff) allocated to the probiotic group. Following a 2-week baseline period in October 2018, participants received daily investigational products for 6 months (October 2018 to March 2019). Two participants in the placebo group withdrew before receiving their assigned drinks because of death and missing baseline fecal samples. During the intervention period, four participants in the placebo group and seven in the probiotic group were lost to follow-up. Reasons for withdrawal included unavoidable circumstances such as death unrelated to the study products, hospitalization, relocation from the facility, and noncompliance with the protocol (missed stool sample collections or refusal to consume the assigned drinks). An intention-to-treat analysis was performed on participants who received the allocated interventions, including 54 participants in the placebo group and 56 in the probiotic group.

Figure 1
Figure 1 Flow diagram representing the study.
Baseline data

Table 1 shows the demographic and baseline characteristics of the participants. The baseline data were well balanced between the groups, indicating effective randomization.

Table 1 Baseline characteristics of the participants, n (%).

Placebo (n = 54)
Probiotic (n = 56)
Participant type, resident/staff31 (57.4)/23 (42.6)34 (60.7)/22 (39.3)
Age, median (IQR), year65.0 (47.0-61.7)65.5 (50.8-61.7)
Height, mean ± SD, cm153.8 ± 10.6155.0 ± 9.8
Weight, median (IQR), kg53.9 (41.6-64.6)52.4 (41.8-66.6)
BMI, median (IQR), kg/m222.6 (19.0-25.7)22.3 (18.6-26.1)
Sex, male/female15 (27.8)/39 (72.2)18 (32.1)/38 (67.9)
Nutrient intake, mean ± SD, day
Energy, mean ± SD, kcal893.30 ± 154.37901.39 ± 154.51
Water, mean ± SD, g696.86 ± 134.93735.78 ± 154.77
Protein, mean ± SD, g42.28 ± 10.1141.60 ± 10.31
Fat, mean ± SD, g38.50 ± 9.0339.40 ± 10.44
Carbohydrate, mean ± SD, g95.23 ± 20.5596.00 ± 15.83
Dietary fiber, mean ± SD, g5.25 ± 1.935.55 ± 1.86
Alcohol, mean ± SD, g0.00 ± 0.000.00 ± 0.01
Polyunsaturated fatty acid, mean ± SD, g6.87 ± 2.007.37 ± 2.56
Cholesterol, mean ± SD, mg190.50 ± 85.76202.28 ± 92.92
Vitamin A, mean ± SD, μg563.72 ± 344.67536.09 ± 273.61
Carotene, mean ± SD, mg0.08 ± 0.110.09 ± 0.12
Vitamin E, mean ± SD, mg2.75 ± 0.832.81 ± 0.86
Vitamin B1, mean ± SD, mg0.45 ± 0.150.44 ± 0.14
Vitamin B2, mean ± SD, mg0.52 ± 0.160.54 ± 0.15
Vitamin B6, mean ± SD, mg0.72 ± 0.200.74 ± 0.22
Folic acid, mean ± SD, μg109.34 ± 38.93110.76 ± 35.88
Vitamin C, mean ± SD, mg30.02 ± 24.7630.48 ± 25.23
Sodium, mean ± SD, mg392.78 ± 375.76312.71 ± 287.12
Potassium, mean ± SD, mg1030.62 ± 998.85919.93 ± 267.01
Calcium, mean ± SD, mg211.76 ± 96.06221.41 ± 90.79
Magnesium, mean ± SD, mg167.40 ± 107.23159.73 ± 36.73
Phosphorus, mean ± SD, mg483.79 ± 132.22469.54 ± 106.26
Iron, mean ± SD, mg6.02 ± 2.995.74 ± 1.50
Zinc, mean ± SD, mg4.22 ± 0.954.35 ± 0.99
Effect of probiotic drink on the gut microbiota

The intervention showed no significant effect on α-diversity metrics throughout the study (Supplementary Table 3). Conversely, several β-diversity measures, including Aitchison distance and Jaccard dissimilarity index, revealed significant alterations in the overall microbial community following 3 months of probiotic drinking (P = 0.045 and 0.035, respectively) (Supplementary Table 4). However, these effects were no longer detected at 6-month period.

Differential abundance analysis using three methods (i.e., ALDEx2, ANCOM, and ANCOM-BC) revealed consistent shifts in the microbial composition at the genus level (Figure 2). At 3-month period, all methods identified a significant increase in Bifidobacterium in the probiotic group compared to that in the placebo group, with the largest effect size (Figure 2A). Additionally, the methods consistently indicated that Succinivibrio was the second-most induced genus and that the Rikenellaceae RC9 gut group was the most suppressed, both reaching significance in ANCOM-BC. At 6-month period, these changes in Succinivibrio and Rikenellaceae RC9 gut group persisted (Figure 2B).

Figure 2
Figure 2 The top and bottom 20 differentially abundant bacterial genera at three months and six months, compared to the placebo group, identified by three methods (ALDEx2, ANCOM, and ANCOM-BC) with their respective metrics. A: Three months; B: Six months. The number next to the ANCOM bar represents the W value.

The transient induction of Bifidobacterium at 3 months in the probiotic group compared to the placebo group was further corroborated by RT-qPCR using the YIF-SCAN® system (8.5 ± 1.1 vs 8.0 ± 1.1, log10 bacterial cells/g, P = 0.044) (Table 2)[20]. Additionally, the analysis revealed an exponential increase in the abundance of Lacticaseibacillus in the probiotic group at three and six months, reaching 106 bacterial cells/g. This is possibly due to the presence of “live” LcS derived from the probiotic intervention, with intact RNA molecules detectable by RT-qPCR. Notably, studies in several countries have shown that LcS in Yakult® can remain viable at > 106 bacterial cells/g in human stool[33-36]. Another study showed that 12 weeks of LcS consumption in the elderly increased the abundance of the genus Lacticaseibacillus in the probiotic group compared to that at baseline and in the placebo group[37]. The study findings indicate that LcS from the probiotic drink significantly impacts the gut microbiota after three months, as evidenced by increases in Bifidobacterium and Succinivibrio and a decrease in the Rikenellaceae RC9 gut group, leading to significant changes in several β-diversity metrics. Although the increase in Bifidobacterium appeared transient, alterations in the other two genera persisted after six months.

Table 2 Changes in microbiota measured by reverse transcription quantitative polymerase chain reaction.
Placebo
Probiotic
Baseline

3 months

6 months

Baseline

3 months

6 months

Total bacteria9.3 ± 1.598%9.9 ± 0.898%9.8 ± 1.498%9.8 ± 0.9100%9.9 ± 1.098%9.8 ± 1.0100%
Bacillota
Clostridium coccoides group8.8 ± 1.389%9.2 ± 0.996%9.3 ± 0.994%9.0 ± 1.1100%9.4 ± 0.794%9.1 ± 1.098%
Clostridium leptum subgroup8.6 ± 1.496%9.2 ± 0.994%9.4 ± 0.794%9.1 ± 1.0100%9.1 ± 1.098%9.2 ± 1.0100%
Clostridium perfringens6.3 ± 1.591%6.0 ± 1.388%5.5 ± 1.292%6.4 ± 1.498%5.9 ± 1.292%5.5 ± 1.194%
Clostridioides difficile6.0 ± 1.76%6.9 ± 0.96%4.9 ± 1.96%3.9 ± 0.45%< 2.30%< 2.30%
Total lactobacilli16.4 ± 1.793%7.0 ± 1.396%7.1 ± 1.694%6.7 ± 1.698%7.1 ± 1.398%7.2 ± 1.298%
Lactobacillus25.4 ± 1.576%5.6 ± 1.490%5.8 ± 1.588%5.7 ± 1.688%5.8 ± 1.581%5.5 ± 1.592%
Levilactobacillus brevis23.3 ± 0.624%3.5 ± 0.637%3.5 ± 1.238%3.4 ± 0.838%4.0 ± 1.023%3.3 ± 0.429%
Lacticaseibacillus24.6 ± 1.419%5.2 ± 1.431%4.2 ± 1.036%4.9 ± 1.332%6.1 ± 1.2a94%b6.3 ± 0.8b88%b
Limosilactobacillus fermentum25.6 ± 1.056%5.3 ± 0.771%4.9 ± 0.956%5.4 ± 0.771%5.5 ± 0.971%4.9 ± 0.665%
Fructilactobacillus fructivorans23.12%3.1 ± 0.46%2.72%< 2.30%3.0 ± 0.26%2.8 ± 0.46%
Lactiplantibacillus24.1 ± 1.185%4.4 ± 1.292%4.6 ± 1.094%4.2 ± 1.091%4.4 ± 1.283%4.6 ± 1.198%
Limosilactobacillus except L. fermentum24.7 ± 1.380%5.1 ± 1.290%4.7 ± 1.394%5.2 ± 1.382%5.3 ± 1.287%4.9 ± 1.190%
Liquorilactobacillus and Ligilactobacillus26.8 ± 2.135%7.3 ± 1.648%7.7 ± 1.548%7.1 ± 2.036%6.9 ± 2.058%7.3 ± 1.753%
Latilactobacillus24.7 ± 0.44%4.6 ± 0.415%4.0 ± 0.816%4.3 ± 0.89%4.0 ± 0.912%3.9 ± 0.816%
Enterococcus6.8 ± 1.578%7.5 ± 0.885%7.5 ± 1.086%6.9 ± 1.179%7.1 ± 1.1c81%7.2 ± 1.290%
Streptococcus5.1 ± 1.183%5.0 ± 0.896%5.3 ± 0.892%5.1 ± 0.996%5.0 ± 0.894%5.3 ± 0.998%
Staphylococcus7.4 ± 1.970%8.0 ± 1.077%8.2 ± 0.972%8.1 ± 1.080%8.1 ± 1.271%8.0 ± 1.284%
Bacteroidota
Bacteroides fragilis group7.6 ± 0.980%8.0 ± 0.992%8.2 ± 0.894%7.7 ± 1.093%8.0 ± 0.994%8.2 ± 1.098%
Prevotella8.3 ± 1.780%8.9 ± 1.388%8.9 ± 1.474%8.8 ± 1.280%9.1 ± 1.583%9.1 ± 1.178%
Actinomycetota
Atopobium cluster8.7 ± 1.294%8.9 ± 0.896%8.8 ± 0.996%9.1 ± 0.8100%8.9 ± 0.898%8.8 ± 0.8100%
Bifidobacterium8.1 ± 1.181%8.0 ± 1.194%8.2 ± 1.190%8.3 ± 1.198%8.5 ± 1.1a94%8.1 ± 0.998%
Pseudomonadota
Enterobacteriaceae7.7 ± 1.489%8.2 ± 1.096%8.2 ± 0.994%7.7 ± 1.2100%8.1 ± 1.096%8.0 ± 1.098%
Pseudomonas4.3 ± 1.030%3.9 ± 0.950%4.1 ± 0.776%3.8 ± 0.727%3.9 ± 0.942%4.0 ± 0.578%
Effect of probiotic drink on the gut environment

Stool butyrate concentration significantly increased after six months in the probiotic group compared to that in the placebo group (5.04 ± 3.11 vs 3.95 ± 2.89 μmol/g, P = 0.048) (Table 3). The probiotic drinks did not significantly affect the levels of other organic acid species, pH, and amino acid metabolites. Nonetheless, mixed linear effects modeling adjusted for baseline values, participant type, and study site revealed potential trends (Supplementary Table 5). The model demonstrated a transient increase in acetate and total organic acid levels three months after the administration of the probiotic drink (P = 0.061 and P = 0.069, respectively) and a lower stool pH (P = 0.053). Furthermore, ammonia levels tended to decrease and indole levels tended to increase at six months (P = 0.068 and P = 0.071, respectively). However, further investigations are required to confirm these trends.

Table 3 Changes in stool organic acids and amino acid metabolites.
Placebo
Probiotic
Baseline
3 months
6 months
Baseline
3 months
6 months
Organic acids, μmol/g
Acetate, mean ± SD13.75 ± 4.8215.88 ± 8.0114.63 ± 5.5414.24 ± 5.4917.97 ± 8.2115.81 ± 4.85
Formate, mean ± SD1.91 ± 0.401.71 ± 0.302.44 ± 0.241.90 ± 0.381.72 ± 0.172.43 ± 0.21
Propionate, mean ± SD4.44 ± 2.435.56 ± 3.515.43 ± 3.814.68 ± 2.666.46 ± 4.095.82 ± 3.27
Isobutyrate, mean ± SD0.41 ± 0.230.50 ± 0.390.48 ± 0.280.48 ± 0.320.51 ± 0.420.52 ± 0.27
Butyrate, mean ± SD3.56 ± 2.234.32 ± 3.113.95 ± 2.894.29 ± 3.035.21 ± 3.05b5.04 ± 3.11a
Isovalerate, mean ± SD0.36 ± 0.240.45 ± 0.350.42 ± 0.270.44 ± 0.340.48 ± 0.390.43 ± 0.24
Valerate, mean ± SD0.65 ± 0.410.63 ± 0.470.57 ± 0.350.73 ± 0.440.68 ± 0.390.67 ± 0.37
Isocaproate, median (IQR)0.01 (0.01-0.03)0.02 (0.01-0.03)0.01 (0.00-0.01)0.01 (0.01-0.02)0.02 (0.01-0.04)0.01 (0.01-0.01)
Caproate, median (IQR)0.07 (0.04-0.14)0.06 (0.01-0.23)0.05 (0.03-0.12)0.05 (0.03-0.16)0.06 (0.02-0.11)0.05 (0.03-0.17)
Heptanoate, median (IQR)0.01 (0.01-0.03)0.01 (0.00-0.10)0.01 (0.00-0.01)0.02 (0.01-0.03)0.00 (0.00-0.06)0.01 (0.00-0.02)
Lactate, median (IQR)0.03 (0.02-0.05)0.03 (0.02-0.04)0.03 (0.02-0.04)0.03 (0.01-0.04)0.02 (0.02-0.03)0.02 (0.02-0.04)
Succinate, median (IQR)0.01 (0.01-0.02)0.02 (0.01-0.04)0.01 (0.01-0.03)0.01 (0.01-0.03)0.03 (0.01-0.06)0.02 (0.01-0.03)
Total organic acids, mean ± SD25.31 ± 9.0829.38 ± 13.8328.11 ± 11.8127.00 ± 9.5433.33 ± 13.8630.95 ± 10.11
pH
pH, median (IQR)6.12 (5.86-6.30)5.99 (5.80-6.35)6.06 (5.81-6.39)6.08 (5.75-6.41)5.89 (5.60-6.38)5.93 (5.75-6.18)b
Amino acid metabolites, μg/g
Ammonia, median (IQR)383.7 (245.4-479.5)467.6 (361.5-568.1)2015.4 (651.5-3263.7)358.9 (260.8-506.0)470.2 (374.6-585.3)1401.8 (335.8-3116.6)
Indole, median (IQR)15.92 (9.19-30.40)13.46 (8.14-22.35)3.82 (2.78-10.28)11.97 (7.29-23.70)14.70 (11.50-23.87)5.94 (2.95-12.01)
Phenol, median (IQR)5.24 (4.35-8.67)5.74 (1.91-14.24)3.71 (1.96-5.46)7.02 (2.87-15.46)3.65 (1.66-10.27)2.31 (1.40-4.07)
p-cresol, median (IQR)16.36 (7.42-26.30)13.03 (8.54-29.98)6.94 (4.33-11.47)12.45 (9.46-26.09)12.44 (7.94-25.91)8.09 (5.64-12.06)
Effect of probiotic drink on stool frequency

Probiotic consumption did not significantly influence stool frequency during the intervention period, despite exhibiting higher frequencies in the probiotic group than in the placebo group for most months. Notably, all participants reported near-optimal bowel movement frequency before the start of the intervention (Table 4).

Table 4 Changes in stool frequency, mean ± SD.
MonthsStool frequency (times/week)
Stool frequency (days/week)
Placebo
Probiotic
Placebo
Probiotic
Baseline6.24 ± 2.616.20 ± 2.145.59 ± 1.625.69 ± 1.63
16.05 ± 1.726.07 ± 1.435.85 ± 1.495.89 ± 1.33
26.19 ± 1.626.27 ± 1.375.98 ± 1.386.08 ± 1.22
36.25 ± 1.476.21 ± 1.326.02 ± 1.296.08 ± 1.26
46.18 ± 1.496.50 ± 1.146.03 ± 1.406.27 ± 1.00
56.32 ± 1.406.56 ± 1.056.10 ± 1.276.32 ± 0.87
66.50 ± 1.256.78 ± 1.026.29 ± 1.156.51 ± 0.77
Subgroup analysis

Subgroup analyses were conducted to evaluate potential variations in probiotic efficacy according to the participant type (residents vs staff). Therefore, the interaction effects between treatment group and participant type were assessed for all outcome measures, followed by least squares (LS) mean difference calculations within each participant type. Significant interactions were observed in the RT-qPCR outcomes (Table 5). Bifidobacterium levels exhibited significant interaction effects at three (P = 0.031) and six months (P = 0.044). Notably, the 95% confidence interval of the LS mean difference at 3 months was positive for residents and overlapped with zero for staff, indicating a greater effect of the probiotic drink on increasing Bifidobacterium levels in residents than in staff. Additionally, a significant interaction was identified for Enterococcus levels at six months (P = 0.003). The mean differences in LS at three and six months indicated a notable reduction in Enterococcus abundance in the staff population following probiotic consumption.

Table 5 Subgroup analyses on reverse transcription quantitative polymerase chain reaction outcomes.
Participant types3 months
6 months
LS mean difference (95%CI)
P value1
LS mean difference (95%CI)
P value1
Total bacteriaResident0.12 (-0.33 to 0.57)0.488-0.05 (-0.68 to 0.59)0.752
Staff-0.12 (-0.65 to 0.41)0.11 (-0.62 to 0.83)
Bacillota
Clostridium coccoides groupResident0.30 (-0.14 to 0.74)0.2790.08 (-0.42 to 0.58)0.068
Staff-0.07 (-0.60 to 0.45)-0.62 (-1.20 to -0.05)
Clostridium leptum subgroupResident0.13 (-0.35 to 0.61)0.1980.06 (-0.42 to 0.54)0.159
Staff-0.36 (-0.93 to 0.22)-0.47 (-1.03 to 0.09)
Clostridium perfringensResident0.07 (-0.62 to 0.75)0.4510.22 (-0.41 to 0.86)0.491
Staff-0.34 (-1.14 to 0.46)-0.12 (-0.87 to 0.64)
Total lactobacilliResident0.37 (-0.31 to 1.06)0.2730.51 (-0.22 to 1.25)0.067
Staff-0.21 (-1.01 to 0.59)-0.55 (-1.43 to 0.32)
EnterococcusResident-0.13 (-0.70 to 0.43)0.1440.32 (-0.26 to 0.90)0.003b
Staff-0.77 (-1.42 to -0.12)-1.04 (-1.70 to -0.37)
StreptococcusResident-0.06 (-0.47 to 0.36)0.9640.08 (-0.37 to 0.53)0.698
Staff-0.07 (-0.56 to 0.42)-0.06 (-0.57 to 0.46)
StaphylococcusResident0.17 (-0.52 to 0.86)0.966-0.01 (-0.65 to 0.62)0.340
Staff0.19 (-0.57 to 0.94)-0.48 (-1.21 to 0.26)
Bacteroidota
Bacteroides fragilis groupResident0.04 (-0.42 to 0.50)0.6390.10 (-0.35 to 0.55)0.167
Staff-0.13 (-0.68 to 0.43)-0.38 (-0.90 to 0.14)
PrevotellaResident0.44 (-0.29 to 1.16)0.2060.15 (-0.65 to 0.96)0.911
Staff-0.28 (-1.13 to 0.57)0.22 (-0.61 to 1.04)
Actinomycetota
Atopobium clusterResident0.00 (-0.41 to 0.40)0.904-0.05 (-0.49 to 0.38)0.969
Staff-0.04 (-0.53 to 0.44)-0.07 (-0.57 to 0.44)
BifidobacteriumResident0.84 (0.26 to 1.42)0.031a0.25 (-0.26 to 0.75)0.044a
Staff-0.14 (-0.81 to 0.53)-0.54 (-1.11 to 0.04)
Pseudomonadota
EnterobacteriaceaeResident-0.03 (-0.54 to 0.49)0.3900.10 (-0.4 to 0.61)0.102
Staff-0.37 (-0.97 to 0.23)-0.54 (-1.12 to 0.05)
PseudomonasResident-0.19 (-0.74 to 0.36)0.721-0.21 (-0.56 to 0.15)0.865
Staff-0.39 (-1.35 to 0.58)-0.16 (-0.62 to 0.31)
Adverse events

Table 6 summarizes the Unified Medical Language System/Medical Dictionary for Regulatory Activities-coded adverse events, focusing on gastrointestinal disorders. No significant differences were found in overall adverse events between the groups (Table 6). Bloating had the highest odds ratio compared to the placebo group (odds ratio = 5.00; P = 0.496), whereas respiratory, thoracic, and mediastinal disorders had the lowest odds ratio (odds ratio = 0.23; P = 0.202).

Table 6 Number and percentage of participants with adverse events, n (%).

Placebo
Probiotic
Odds ratio
P value
Any adverse event22 (40.7)21 (38.9)0.870.845
Gastrointestinal disorders8 (14.8)6 (11.1)0.690.577
Diarrhea2 (3.7)2 (3.7)0.961.000
Stomatitis1 (1.9)0 (0)0.320.491
Stomachache2 (3.7)1 (1.9)0.470.615
Toothache1 (1.9)1 (1.9)0.961.000
Pain of epigastrium1 (1.9)2 (3.7)1.960.514
Bloated0 (0)2 (3.7)5.000.496
Gastric pain0 (0)1 (1.9)2.951.000
Dyspepsia syndrome1 (1.9)0 (0)0.320.491
Lower abdominal pain1 (1.9)0 (0)0.320.491
Nausea0 (0)1 (1.9)2.951.000
Nervous system disorders5 (9.3)8 (14.8)1.630.557
Musculoskeletal and connective tissue disorders5 (9.3)6 (11.1)1.181.000
General disorders and administration site conditions4 (7.4)5 (9.3)1.230.523
Respiratory, thoracic, and mediastinal disorders4 (7.4)1 (1.9)0.230.202
Infections and infestations2 (3.7)2 (3.7)0.961.000
Skin and subcutaneous tissue disorders2 (3.7)1 (1.9)0.470.615
Eye disorders1 (1.9)1 (1.9)0.961.000
Metabolism and nutrition disorders0 (0)1 (1.9)2.951.000
Vascular disorders0 (0)1 (1.9)2.951.000
Psychiatric disorders1 (1.9)0 (0)0.320.491
DISCUSSION

This study investigated the impact of daily consumption of fermented milk containing > 6.5 × 109 colony-forming units LcS on the gut microbiota and metabolite profile of individuals in elderly houses, including residents and staff, over a 6-month period. After three months, significant gut microbiota changes were observed, characterized by an increase in Bifidobacterium and Succinivibrio and a decrease in the Rikenellaceae RC9 gut group. These alterations coincided with a shift in β-diversity metrics, indicating a change in overall microbial community. Although limited changes were detected in the metabolite profile at this time point, increased total organic acids and acetate and decreased fecal pH were observed. This was followed by a remarkable induction of butyrate production at six months.

The metabolite profile alterations were consistent with changes in the microbial community. Succinivibrio has been reported to exhibit a strong correlation with butyrate production[38,39]. It is a plant polysaccharide-fermenting bacterium that produces succinate and acetate, typically enriched in the Prevotella-type gut microbial community, which is an enterotype prevalent in the Indonesian population[13,40,41]. The observed increase in this genus in our relatively older study group (median age: 65 years) demonstrated that LcS consumption may have shifted the putrefactive-prone gut microbial metabolism toward a more saccharolytic one, potentially leading to higher organic acid production and lower pH. Additionally, Bifidobacterium, which produces acetate from mono- or polysaccharides, may have contributed to the increase in organic acids and lower fecal pH at three months and subsequent butyrate induction at six months. Intriguingly, the acetate produced by Bifidobacterium can be converted to butyrate by other colonic bacteria through cross-feeding interactions[42-45]. Thus, the early modifications in bacterial communities and metabolism at three months may have gradually created an environment favoring cross-feeding interactions that ultimately led to increased butyrate production at six months. Future metatranscriptomic analyses could further elucidate these hypotheses by providing deeper insights into microbial gene expression patterns.

These observed shifts in the gut microbiome have been associated with positive effects on gut and systemic health. Bifidobacterium have been linked to improved outcomes due to their immunomodulatory properties and acetate production[46-49]. Acetate fuels the colonocytes, maintains a robust gut barrier, and prevents pathogen infiltration[50]. Similarly, butyrate is a critical energy source for intestinal cells, reduces inflammation, supports immune function, preserves gut barrier integrity, and may lower the risk of colorectal cancer[50]. Furthermore, a recent study utilizing Mendelian randomization identified Rikenellaceae RC9 gut group as a detrimental bacterium that contributes to ischemic stroke[51]. This bacterium has also been implicated in Parkinson’s disease through its role in modulating the gut branched-chain and aromatic amino acid ratios, potentially influencing immune responses[52]. This collective evidence indicates that LcS has beneficial effects on the gut microbiota and environment, leading to improved human health.

Our findings corroborate those of previous studies. Notably, a randomized, double-blind, placebo-controlled trial in Japan observed similar increases in Bifidobacterium and organic acids following six months of LcS-fermented milk consumption by elderly facility residents and staff[12]. Moreover, this study linked Bifidobacterium changes to elevated stool acetate levels and decreased stool pH, mirroring our results. However, the current study noted these alterations only at three months, whereas the previous study sustained them for six months. These discrepancies in responsiveness may stem from differences in administered bacterial counts or participant characteristics. Specifically, the optimal stool frequency observed in our participants indicated a potentially more favorable baseline gut environment than that of participants in the previous study. This favorable environment may explain the diminished overall responsiveness and selective Bifidobacterium increase observed in the older resident subgroup.

A significant finding in this study was the decrease in Enterococcus in the staff subgroup after LcS consumption. Enterococci pose a growing concern in long-term care facilities owing to their propensity to cause antibiotic-resistant hospital infections[53,54]. These bacteria acquire and disseminate resistance by the horizontal transfer of mobile genetic elements, mediating vancomycin resistance transfer from enterococci to methicillin-resistant Staphylococcus aureus[54]. Notably, antibiotic-resistant Enterococcus can spread directly through patient contact or indirectly through transient carriage on the hands of healthcare personnel, contaminated surfaces, and equipment. The observed reduction in Enterococcus raises the possibility of using LcS as a preventive measure to mitigate the horizontal transmission of antibiotic-resistant Enterococcus strains in elderly care facilities. Further studies that directly investigate the impact on antibiotic resistance genes are warranted to confirm this potential benefit.

This study has several limitations. First, bacterial counts from RT-qPCR analysis were unexpectedly lower than those reported in a previous preliminary study[13]. For example, the prevalence of the Clostridium coccoides group decreased from 100% in a preliminary study conducted in Yogyakarta and Bali, to 89% in the current study. Similarly, the mean total bacterial count decreased from 1010 cells/g to 109 cells/g. This discrepancy is likely due to RNA degradation at the various stages of fecal sample handling, including collection, storage, transport, and extraction. Because this degradation occurred randomly across samples, it may not have significantly impacted the statistical comparisons between the study groups, although the absolute bacterial counts could be biased. Notably, DNA degradation was less of a concern, as indicated by the consistent bacterial counts obtained by qPCR (data not shown). The agreement between 16S rRNA gene sequencing and RT-qPCR results further supports the robustness of our findings. Additionally, the ammonia measurements in some samples may have been biased because of the bumping reaction during the heating step. Another limitation of this study was that it could not establish a cause-and-effect relationship between the altered gut microbiota, observed gut environment changes, and the health outcomes of the participants. However, a previous study conducted in an elderly facility in Japan found that LcS administration reduced fever duration during a mass norovirus gastroenteritis outbreak, possibly due to an improved gut environment and immune-enhancing effects[11]. This specific effect could not be evaluated in our study because of the small number of participants experiencing similar health issues; however, the observed lower odds ratio for respiratory, thoracic, and mediastinal disorders shows a possible link between LcS-mediated improvements in the gut environment and immune modulation. Future trials in Indonesian populations focusing on clinically relevant outcomes may further elucidate this potential benefit.

CONCLUSION

Daily consumption of LcS-fermented milk significantly increased Bifidobacterium and Succinivibrio levels and decreased the Rikenellaceae RC9 gut group in the guts of individuals in Indonesian elderly houses. These alterations were accompanied by favorable modifications in stool metabolite markers, particularly increased butyrate levels, indicating an improved gut environment. These findings reveal that regular consumption of LcS-fermented milk could benefit the gut microbiota and overall gut health of the elderly population. Further trials are warranted to assess their effects on clinically relevant health outcomes.

ACKNOWLEDGEMENTS

We would like to thank Dina Aulia Nurfiana and Fathyah Hanum P for their assistance in analyzing food record data, Devin Varian Wiryohanjoyo and Bedri Sekar Nurmadhani for their helpful assistance during participant screening, and Monika Dwi Adkhayati and Suharman for their helpful assistance during the sample collection period of this study.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: Indonesia

Peer-review report’s classification

Scientific Quality: Grade B, Grade B, Grade C, Grade C, Grade C

Novelty: Grade B, Grade B, Grade B, Grade C, Grade C

Creativity or Innovation: Grade B, Grade B, Grade B, Grade C, Grade C

Scientific Significance: Grade B, Grade B, Grade B, Grade C, Grade C

P-Reviewer: Cao QG; Lin DT; Xie YF S-Editor: Wang JJ L-Editor: A P-Editor: Zheng XM

References
1.  Basrowi RW, Rahayu EM, Khoe LC, Wasito E, Sundjaya T. The Road to Healthy Ageing: What Has Indonesia Achieved So Far? Nutrients. 2021;13:3441.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
2.  Arifin EN, Ananta A.   The Past Three Population Censuses: A Deepening Ageing Population in Indonesia. In: Guilmoto C, Jones G. Contemporary Demographic Transformations in China, India and Indonesia. Cham: Springer, 2016.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Ghosh TS, Shanahan F, O'Toole PW. The gut microbiome as a modulator of healthy ageing. Nat Rev Gastroenterol Hepatol. 2022;19:565-584.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in RCA: 304]  [Article Influence: 101.3]  [Reference Citation Analysis (0)]
4.  Ragonnaud E, Biragyn A. Gut microbiota as the key controllers of "healthy" aging of elderly people. Immun Ageing. 2021;18:2.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in RCA: 198]  [Article Influence: 49.5]  [Reference Citation Analysis (0)]
5.  Dumyati G, Stone ND, Nace DA, Crnich CJ, Jump RL. Challenges and Strategies for Prevention of Multidrug-Resistant Organism Transmission in Nursing Homes. Curr Infect Dis Rep. 2017;19:18.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in RCA: 66]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
6.  Haran JP, Bucci V, Dutta P, Ward D, McCormick B. The nursing home elder microbiome stability and associations with age, frailty, nutrition and physical location. J Med Microbiol. 2018;67:40-51.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in RCA: 68]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
7.  Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, Morelli L, Canani RB, Flint HJ, Salminen S, Calder PC, Sanders ME. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol. 2014;11:506-514.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 4055]  [Cited by in RCA: 5265]  [Article Influence: 478.6]  [Reference Citation Analysis (2)]
8.  Hutchinson AN, Bergh C, Kruger K, Sűsserová M, Allen J, Améen S, Tingö L. The Effect of Probiotics on Health Outcomes in the Elderly: A Systematic Review of Randomized, Placebo-Controlled Studies. Microorganisms. 2021;9:1344.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
9.  Rondanelli M, Giacosa A, Faliva MA, Perna S, Allieri F, Castellazzi AM. Review on microbiota and effectiveness of probiotics use in older. World J Clin Cases. 2015;3:156-162.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in CrossRef: 80]  [Cited by in RCA: 84]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
10.  Zheng J, Wittouck S, Salvetti E, Franz CMAP, Harris HMB, Mattarelli P, O'Toole PW, Pot B, Vandamme P, Walter J, Watanabe K, Wuyts S, Felis GE, Gänzle MG, Lebeer S. A taxonomic note on the genus Lactobacillus: Description of 23 novel genera, emended description of the genus Lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae. Int J Syst Evol Microbiol. 2020;70:2782-2858.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1986]  [Cited by in RCA: 1683]  [Article Influence: 336.6]  [Reference Citation Analysis (0)]
11.  Nagata S, Asahara T, Ohta T, Yamada T, Kondo S, Bian L, Wang C, Yamashiro Y, Nomoto K. Effect of the continuous intake of probiotic-fermented milk containing Lactobacillus casei strain Shirota on fever in a mass outbreak of norovirus gastroenteritis and the faecal microflora in a health service facility for the aged. Br J Nutr. 2011;106:549-556.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in RCA: 87]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
12.  Nagata S, Asahara T, Wang C, Suyama Y, Chonan O, Takano K, Daibou M, Takahashi T, Nomoto K, Yamashiro Y. The Effectiveness of Lactobacillus Beverages in Controlling Infections among the Residents of an Aged Care Facility: A Randomized Placebo-Controlled Double-Blind Trial. Ann Nutr Metab. 2016;68:51-59.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in RCA: 42]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
13.  Rahayu ES, Utami T, Mariyatun M, Hasan PN, Kamil RZ, Setyawan RH, Pamungkaningtyas FH, Harahap IA, Wiryohanjoyo DV, Pramesi PC, Cahyanto MN, Sujaya IN, Juffrie M. Gut microbiota profile in healthy Indonesians. World J Gastroenterol. 2019;25:1478-1491.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in CrossRef: 19]  [Cited by in RCA: 25]  [Article Influence: 4.2]  [Reference Citation Analysis (3)]
14.  Tsukuda N, Yahagi K, Hara T, Watanabe Y, Matsumoto H, Mori H, Higashi K, Tsuji H, Matsumoto S, Kurokawa K, Matsuki T. Key bacterial taxa and metabolic pathways affecting gut short-chain fatty acid profiles in early life. ISME J. 2021;15:2574-2590.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in RCA: 156]  [Article Influence: 39.0]  [Reference Citation Analysis (0)]
15.  Matsuki T, Yahagi K, Mori H, Matsumoto H, Hara T, Tajima S, Ogawa E, Kodama H, Yamamoto K, Yamada T, Matsumoto S, Kurokawa K. A key genetic factor for fucosyllactose utilization affects infant gut microbiota development. Nat Commun. 2016;7:11939.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 215]  [Cited by in RCA: 252]  [Article Influence: 28.0]  [Reference Citation Analysis (0)]
16.  Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet CC, Al-Ghalith GA, Alexander H, Alm EJ, Arumugam M, Asnicar F, Bai Y, Bisanz JE, Bittinger K, Brejnrod A, Brislawn CJ, Brown CT, Callahan BJ, Caraballo-Rodríguez AM, Chase J, Cope EK, Da Silva R, Diener C, Dorrestein PC, Douglas GM, Durall DM, Duvallet C, Edwardson CF, Ernst M, Estaki M, Fouquier J, Gauglitz JM, Gibbons SM, Gibson DL, Gonzalez A, Gorlick K, Guo J, Hillmann B, Holmes S, Holste H, Huttenhower C, Huttley GA, Janssen S, Jarmusch AK, Jiang L, Kaehler BD, Kang KB, Keefe CR, Keim P, Kelley ST, Knights D, Koester I, Kosciolek T, Kreps J, Langille MGI, Lee J, Ley R, Liu YX, Loftfield E, Lozupone C, Maher M, Marotz C, Martin BD, McDonald D, McIver LJ, Melnik AV, Metcalf JL, Morgan SC, Morton JT, Naimey AT, Navas-Molina JA, Nothias LF, Orchanian SB, Pearson T, Peoples SL, Petras D, Preuss ML, Pruesse E, Rasmussen LB, Rivers A, Robeson MS 2nd, Rosenthal P, Segata N, Shaffer M, Shiffer A, Sinha R, Song SJ, Spear JR, Swafford AD, Thompson LR, Torres PJ, Trinh P, Tripathi A, Turnbaugh PJ, Ul-Hasan S, van der Hooft JJJ, Vargas F, Vázquez-Baeza Y, Vogtmann E, von Hippel M, Walters W, Wan Y, Wang M, Warren J, Weber KC, Williamson CHD, Willis AD, Xu ZZ, Zaneveld JR, Zhang Y, Zhu Q, Knight R, Caporaso JG. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat Biotechnol. 2019;37:852-857.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 12449]  [Cited by in RCA: 10698]  [Article Influence: 1783.0]  [Reference Citation Analysis (0)]
17.  Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJ, Holmes SP. DADA2: High-resolution sample inference from Illumina amplicon data. Nat Methods. 2016;13:581-583.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 18515]  [Cited by in RCA: 16445]  [Article Influence: 1827.2]  [Reference Citation Analysis (0)]
18.  Yilmaz P, Parfrey LW, Yarza P, Gerken J, Pruesse E, Quast C, Schweer T, Peplies J, Ludwig W, Glöckner FO. The SILVA and "All-species Living Tree Project (LTP)" taxonomic frameworks. Nucleic Acids Res. 2014;42:D643-D648.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 1629]  [Cited by in RCA: 2030]  [Article Influence: 169.2]  [Reference Citation Analysis (0)]
19.  Quast C, Pruesse E, Yilmaz P, Gerken J, Schweer T, Yarza P, Peplies J, Glöckner FO. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 2013;41:D590-D596.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 15104]  [Cited by in RCA: 17244]  [Article Influence: 1437.0]  [Reference Citation Analysis (0)]
20.  Tsuji H, Matsuda K, Nomoto K. Counting the Countless: Bacterial Quantification by Targeting rRNA Molecules to Explore the Human Gut Microbiota in Health and Disease. Front Microbiol. 2018;9:1417.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in RCA: 30]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
21.  Chen HM, Yu YN, Wang JL, Lin YW, Kong X, Yang CQ, Yang L, Liu ZJ, Yuan YZ, Liu F, Wu JX, Zhong L, Fang DC, Zou W, Fang JY. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma. Am J Clin Nutr. 2013;97:1044-1052.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in RCA: 218]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
22.  Ohigashi S, Sudo K, Kobayashi D, Takahashi O, Takahashi T, Asahara T, Nomoto K, Onodera H. Changes of the intestinal microbiota, short chain fatty acids, and fecal pH in patients with colorectal cancer. Dig Dis Sci. 2013;58:1717-1726.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in RCA: 120]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
23.  Suh JW, Lee SH, Chung BC. GC-MS determination of organic acids with solvent extraction after cation-exchange chromatography. Clin Chem. 1997;43:2256-2261.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in RCA: 27]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
24.  Tangerman A, Nagengast FM. A gas chromatographic analysis of fecal short-chain fatty acids, using the direct injection method. Anal Biochem. 1996;236:1-8.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in RCA: 70]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
25.  Fernandes AD, Macklaim JM, Linn TG, Reid G, Gloor GB. ANOVA-like differential expression (ALDEx) analysis for mixed population RNA-Seq. PLoS One. 2013;8:e67019.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 334]  [Cited by in RCA: 486]  [Article Influence: 40.5]  [Reference Citation Analysis (0)]
26.  Fernandes AD, Reid JN, Macklaim JM, McMurrough TA, Edgell DR, Gloor GB. Unifying the analysis of high-throughput sequencing datasets: characterizing RNA-seq, 16S rRNA gene sequencing and selective growth experiments by compositional data analysis. Microbiome. 2014;2:15.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 595]  [Cited by in RCA: 727]  [Article Influence: 66.1]  [Reference Citation Analysis (0)]
27.  Gloor GB, Macklaim JM, Fernandes AD. Displaying Variation in Large Datasets: Plotting a Visual Summary of Effect Sizes. J Comput Graph Stat. 2016;25:971-979.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in RCA: 17]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
28.  Mandal S, Van Treuren W, White RA, Eggesbø M, Knight R, Peddada SD. Analysis of composition of microbiomes: a novel method for studying microbial composition. Microb Ecol Health Dis. 2015;26:27663.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 589]  [Cited by in RCA: 999]  [Article Influence: 99.9]  [Reference Citation Analysis (0)]
29.  Lin H, Peddada SD. Analysis of compositions of microbiomes with bias correction. Nat Commun. 2020;11:3514.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 540]  [Cited by in RCA: 925]  [Article Influence: 185.0]  [Reference Citation Analysis (0)]
30.  Nearing JT, Douglas GM, Hayes MG, MacDonald J, Desai DK, Allward N, Jones CMA, Wright RJ, Dhanani AS, Comeau AM, Langille MGI. Microbiome differential abundance methods produce different results across 38 datasets. Nat Commun. 2022;13:342.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in RCA: 350]  [Article Influence: 116.7]  [Reference Citation Analysis (0)]
31.  Dixon P. VEGAN, a package of R functions for community ecology. J Veg Sci. 2003;14:927-930.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 2962]  [Cited by in RCA: 3209]  [Article Influence: 200.6]  [Reference Citation Analysis (0)]
32.  Twisk J, Bosman L, Hoekstra T, Rijnhart J, Welten M, Heymans M. Different ways to estimate treatment effects in randomised controlled trials. Contemp Clin Trials Commun. 2018;10:80-85.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in RCA: 306]  [Article Influence: 43.7]  [Reference Citation Analysis (0)]
33.  Cook CM, Makino H, Kato K, Blonquist T, Derrig L, Shibata H. The probiotic Lacticaseibacillus paracasei strain Shirota (LcS) in a fermented milk beverage survives the gastrointestinal tract of generally healthy U.S. Adults. Int J Food Sci Nutr. 2023;74:645-653.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
34.  Cox AJ, Makino H, Cripps AW, West NP. Recovery of Lactobacillus casei strain Shirota (LcS) from faeces with 14 days of fermented milk supplementation in healthy Australian adults. Asia Pac J Clin Nutr. 2019;28:734-739.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
35.  Mai TT, Hop DV, Anh TT, Lam NT. Recovery of Lactobacillus casei strain Shirota (LcS) from the intestine of healthy Vietnamese adults after intake of fermented milk. Asia Pac J Clin Nutr. 2017;26:72-77.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
36.  Wang R, Chen S, Jin J, Ren F, Li Y, Qiao Z, Wang Y, Zhao L. Survival of Lactobacillus casei strain Shirota in the intestines of healthy Chinese adults. Microbiol Immunol. 2015;59:268-276.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in RCA: 27]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
37.  Yang X, He X, Xu S, Zhang Y, Mo C, Lai Y, Song Y, Yan Z, Ai P, Qian Y, Xiao Q. Effect of Lacticaseibacillus paracasei strain Shirota supplementation on clinical responses and gut microbiome in Parkinson's disease. Food Funct. 2023;14:6828-6839.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 31]  [Reference Citation Analysis (0)]
38.  Xue MY, Sun HZ, Wu XH, Guan LL, Liu JX. Assessment of rumen bacteria in dairy cows with varied milk protein yield. J Dairy Sci. 2019;102:5031-5041.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in RCA: 63]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
39.  Iqbal MW, Zhang Q, Yang Y, Zou C, Li L, Liang X, Wei S, Lin B. Ruminal fermentation and microbial community differently influenced by four typical subtropical forages in vitro. Anim Nutr. 2018;4:100-108.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in RCA: 19]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
40.  De Filippo C, Di Paola M, Ramazzotti M, Albanese D, Pieraccini G, Banci E, Miglietta F, Cavalieri D, Lionetti P. Diet, Environments, and Gut Microbiota. A Preliminary Investigation in Children Living in Rural and Urban Burkina Faso and Italy. Front Microbiol. 2017;8:1979.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in RCA: 181]  [Article Influence: 22.6]  [Reference Citation Analysis (0)]
41.  Nakayama J, Yamamoto A, Palermo-Conde LA, Higashi K, Sonomoto K, Tan J, Lee YK. Impact of Westernized Diet on Gut Microbiota in Children on Leyte Island. Front Microbiol. 2017;8:197.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in RCA: 90]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
42.  Rivière A, Gagnon M, Weckx S, Roy D, De Vuyst L. Mutual Cross-Feeding Interactions between Bifidobacterium longum subsp. longum NCC2705 and Eubacterium rectale ATCC 33656 Explain the Bifidogenic and Butyrogenic Effects of Arabinoxylan Oligosaccharides. Appl Environ Microbiol. 2015;81:7767-7781.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in RCA: 141]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
43.  Rivière A, Selak M, Lantin D, Leroy F, De Vuyst L. Bifidobacteria and Butyrate-Producing Colon Bacteria: Importance and Strategies for Their Stimulation in the Human Gut. Front Microbiol. 2016;7:979.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 979]  [Cited by in RCA: 1054]  [Article Influence: 117.1]  [Reference Citation Analysis (0)]
44.  De Vuyst L, Leroy F. Cross-feeding between bifidobacteria and butyrate-producing colon bacteria explains bifdobacterial competitiveness, butyrate production, and gas production. Int J Food Microbiol. 2011;149:73-80.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 210]  [Cited by in RCA: 235]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
45.  De Vuyst L, Van Kerrebroeck S, Harth H, Huys G, Daniel HM, Weckx S. Microbial ecology of sourdough fermentations: diverse or uniform? Food Microbiol. 2014;37:11-29.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 253]  [Cited by in RCA: 269]  [Article Influence: 22.4]  [Reference Citation Analysis (0)]
46.  van den Munckhof ICL, Kurilshikov A, Ter Horst R, Riksen NP, Joosten LAB, Zhernakova A, Fu J, Keating ST, Netea MG, de Graaf J, Rutten JHW. Role of gut microbiota in chronic low-grade inflammation as potential driver for atherosclerotic cardiovascular disease: a systematic review of human studies. Obes Rev. 2018;19:1719-1734.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in RCA: 176]  [Article Influence: 25.1]  [Reference Citation Analysis (0)]
47.  Stražar M, Temba GS, Vlamakis H, Kullaya VI, Lyamuya F, Mmbaga BT, Joosten LAB, van der Ven AJAM, Netea MG, de Mast Q, Xavier RJ. Gut microbiome-mediated metabolism effects on immunity in rural and urban African populations. Nat Commun. 2021;12:4845.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in RCA: 29]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
48.  Arenas-Padilla M, Duarte-Gutiérrez JL, Mata-Haro V. Bifidobacterium animalis ssp. lactis Bb12 induces IL-10 through cell membrane-associated components via TLR2 in swine. J Appl Microbiol. 2018;125:1881-1889.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in RCA: 11]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
49.  Imaoka A, Shima T, Kato K, Mizuno S, Uehara T, Matsumoto S, Setoyama H, Hara T, Umesaki Y. Anti-inflammatory activity of probiotic Bifidobacterium: enhancement of IL-10 production in peripheral blood mononuclear cells from ulcerative colitis patients and inhibition of IL-8 secretion in HT-29 cells. World J Gastroenterol. 2008;14:2511-2516.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in CrossRef: 127]  [Cited by in RCA: 121]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
50.  Silva YP, Bernardi A, Frozza RL. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front Endocrinol (Lausanne). 2020;11:25.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 780]  [Cited by in RCA: 1440]  [Article Influence: 288.0]  [Reference Citation Analysis (0)]
51.  Dai XC, Yu Y, Zhou SY, Yu S, Xiang MX, Ma H. Assessment of the causal relationship between gut microbiota and cardiovascular diseases: a bidirectional Mendelian randomization analysis. BioData Min. 2024;17:6.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
52.  Yan Z, Yang F, Cao J, Ding W, Yan S, Shi W, Wen S, Yao L. Alterations of gut microbiota and metabolome with Parkinson's disease. Microb Pathog. 2021;160:105187.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in RCA: 46]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
53.  Bonomo RA. Multiple antibiotic-resistant bacteria in long-term-care facilities: An emerging problem in the practice of infectious diseases. Clin Infect Dis. 2000;31:1414-1422.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in RCA: 141]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
54.  Palmer KL, Kos VN, Gilmore MS. Horizontal gene transfer and the genomics of enterococcal antibiotic resistance. Curr Opin Microbiol. 2010;13:632-639.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 188]  [Cited by in RCA: 194]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]