Next Article in Journal
Effects of Different Drying Methods on Drying Characteristics, Microstructure, Quality, and Energy Consumption of Apricot Slices
Previous Article in Journal
A Characterization of Biological Activities and Bioactive Phenolics from the Non-Volatile Fraction of the Edible and Medicinal Halophyte Sea Fennel (Crithmum maritimum L.)
Previous Article in Special Issue
An Update on Prebiotics and on Their Health Effects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Influence of Lactobacillus rhamnosus Supplementation on the Glycaemic Index, Lipid Profile, and Microbiome of Healthy Elderly Subjects: A Preliminary Randomized Clinical Trial

by
Chaiyavat Chaiyasut
1,
Bhagavathi Sundaram Sivamaruthi
1,2,
Subramanian Thangaleela
1,
Natarajan Sisubalan
1,2,
Muruganantham Bharathi
1,
Suchanat Khongtan
1,3,
Periyanaina Kesika
1,2,
Sasithorn Sirilun
1,3,
Thiwanya Choeisoongnern
4,
Sartjin Peerajan
5,
Pranom Fukngoen
1,2,
Phakkharawat Sittiprapaporn
4 and
Wandee Rungseevijitprapa
6,7,*
1
Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
2
Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
3
Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
4
Neuropsychological Research Laboratory, Neuroscience Research Center, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand
5
Health Innovation Institute, Chiang Mai 50200, Thailand
6
Department of Pharmaceutical Chemistry and Technology, Faculty of Pharmaceutical Sciences, Ubon Ratchathani University, Ubon Ratchathani 34190, Thailand
7
School of Cosmetic Science, Mae Fah Luang University, Muang District, Chiang Rai 57100, Thailand
*
Author to whom correspondence should be addressed.
Submission received: 20 March 2024 / Revised: 18 April 2024 / Accepted: 18 April 2024 / Published: 23 April 2024

Abstract

:
Aging is a time-dependent complex biological process of organisms with gradual deterioration of the anatomical and physiological functions. The role of gut microbiota is inevitable in the aging process. Probiotic interventions improve gut homeostasis and support healthy aging by enhancing beneficial species and microbial biodiversity in older adults. The present preliminary clinical trial delves into the impact of an 8-week Lactobacillus rhamnosus intervention (10 × 109 CFU per day) on the glycaemic index, lipid profile, and microbiome of elderly subjects. Body weight, body fat, fasting blood glucose, total cholesterol, triglyceride, high-density lipoprotein, and low-density lipoprotein (LDL) are assessed at baseline (Week 0) and after treatment (Week 8) in placebo and probiotic groups. Gaussian regression analysis highlights a significant improvement in LDL cholesterol in the probiotic group (p = 0.045). Microbiome analysis reveals numeric changes in taxonomic abundance at various levels. At the phylum level, Proteobacteria increases its relative frequency (RF) from 14.79 ± 5.58 at baseline to 23.46 ± 8.02 at 8 weeks, though statistically insignificant (p = 0.100). Compared to the placebo group, probiotic supplementations significantly increased the proteobacteria abundance. Genus-level analysis indicates changes in the abundance of several microbes, including Escherichia-Shigella, Akkermansia, and Bacteroides, but only Butyricimonas showed a statistically significant level of reduction in its abundance. Probiotic supplementations significantly altered the Escherichia-Shigella and Sutterella abundance compared to the placebo group. At the species level, Bacteroides vulgatus substantially increases after probiotic treatment (p = 0.021). Alpha and beta diversity assessments depict subtle shifts in microbial composition. The study has limitations, including a small sample size, short study duration, single-strain probiotic use, and lack of long-term follow-up. Despite these constraints, the study provides valuable preliminary insights into the multifaceted impact of L. rhamnosus on elderly subjects. Further detailed studies are required to define the beneficial effect of L. rhamnosus on the health status of elderly subjects.

1. Introduction

Aging is cellular senescence, a complex biological process with progressive decline in anatomical and physiological functions of multiple cells and tissues. The irreversible aging process is genetically marked and regulated by genetic and environmental factors [1]. The accumulated cellular senescence effects during aging produce damage and disturb somatic maintenance, which could cause certain cellular and molecular distractions leading to genetic instability, epigenetic alterations, mitochondrial dysfunction, proteostasis loss, stem cell exhaustion, cellular senescence, deregulation of nutrient sensing, and so on [2]. The intestine has been recognized as a crucial organ because age-related pathologies are associated with gut microbiota imbalances and gut-associated immune systems. The gut microbiota forms a biological ecosystem with trillions of bacteria and fungi and maintains the host’s health and energy homeostasis. The composition of gut microbiota is relatively stable throughout adult life, designs the health conditions of the host by balancing the pathogens, nutritional status, and energy expenditure, and substantially changes with aging and results in age-associated diseases [3]. The microbial imbalance causes cellular senescence, wherein the cells undergo irreversible growth arrest and accumulate senescent cells during aging due to the development of a senescent-associated secretory phenotype. This comprises the secretion of pro-inflammatory cytokines, growth factors, chemokines, proteases, and extracellular matrix components [4]. The accumulated senescent cells in the skin cause dermatological disorders mediated by the gut–skin axis influenced by the gut microbiota and their metabolites [4]. The mutual relationship between the gut microbiota and the human host helps to protect against the colonization of pathogenic bacteria, metabolic activities, synthesizing bioactive peptide compounds, vitamins, and hormones, and managing the immune system [5,6].
The gut microbiota is stable throughout adulthood and fluctuates with aging and disease conditions [3]. The gut microbial composition changes in the elderly population compared to adults due to various factors such as the aging process, nutritional habits, lifestyle, stress, reduced intestinal functions [3,7], changes in immune responses, lack of physical activity, infections, use of medications, and hospitalization [8,9,10]. Generally, the gut microbiota in adults mostly comprises Firmicutes and Bacteroides and smaller populations of Actinobacteria, Proteobacteria, and Verrucomicrobia [11]. Aged peoples’ gut microbiota is abundant with Bacteroides and Proteobacteria with a decrease in Firmicutes and Bifidobacteria [9,12,13]. Elderly subjects have reduced bacterial diversity and beneficial bacteria. Also, the elderly subjects have lower Bifidobacteria and Lactobacilli levels and increased Enterobacteriaceae and Clostridia levels than young subjects [14,15]. Older adults showed a reduced proportion of butyrate-producing Clostridium cluster XIVa, Roseburia and Ruminococcus [16]. In long-living people, their intestinal microbiota abundantly presented with Akkermansia, Bifidobacterium, and Christensenellaceae [17,18]. In elderly subjects, the gut microbial composition changes, and the microbial diversity is reduced because of various factors such as the accumulation of pro-inflammatory microbes and reduced beneficial microbes [19], aging, nutritional and lifestyle changes, decreased gut functions, and stress [3]. The age-dependent changes in the gut microbiota mean the gradual changes in the microbial diversity involving the decline in the abundance of core dominant microbial species like Bifidobacteria counts and the ratio of Firmicutes to Bacteroides and the increase in the abundance of sub-dominant microbial species like Proteobacteria [17,20].
During aging, the changes in lipid metabolism and their metabolite levels cause an increase in body adiposity. Excess adiposity can cause lipid toxicity, leading to cardiovascular diseases, arthritis, cancer, type 2 diabetes, and Alzheimer’s disease [21]. In humans, with an increase in age, the blood glyceride levels tend to increase, and the white blood lysophosphatidylcholine levels decrease. Human longevity is associated with specific sphingolipid and phospholipid blood profile changes. Furthermore, certain lipids and lipid-related compounds have been found to change depending on age. Lipid-related interventions in various model organisms are capable of modulating the lifespan. It shows that lipid metabolism is linked to the aging process and may enhance the health span. Blood lipids and lipid-related molecules can be biomarkers for human aging studies [22]. Lactic acid bacteria (LAB) and bifidobacteria are found commonly in the gut of humans, with potential physiological benefits, such as enhancing gut function and increasing the uptake of micronutrients, lowering cholesterol, and protecting the gastrointestinal tract from infection by modulating immune functions [23]. It is well-known that an individual’s gut microbiota is shaped right from the gestational stage. Increasing evidence states that probiotic interventions and dietary patterns significantly influence or modulate age-associated changes in gut microbiota and immune functions, promoting healthy aging outcomes [24]. Although several clinical studies evaluated every aspect of aging consequences, anti-aging, or healthy aging properties, the effect of each probiotic strain widely varies on gut composition, immune function, and metabolite synthesis. Certain other studies investigated the involvement of probiotics in gut health, cognitive functions, lipid metabolism, and other biomarkers. In this study, we evaluated the impact of Lactobacillus rhamnosus interventions on glycaemic indexes, lipid profiles, and differences in the microbial diversity of healthy Thai elderly subjects.

2. Materials and Methods

The study protocols were approved by the Ethical Committee of Ubon Ratchathani University (Code: UBU-REC-44/2564). The participants’ consent was obtained before the study.

2.1. Study Group

Thai elderly adults aged ≥ 60 who were willing to participate and complete the study were included. Participants under any other medications or taking probiotics in the previous 2 weeks were omitted from the study. After this primary screening, participants (n = 50; male =12; female = 38) were included in the study. The participants were randomly allotted into placebo (n = 25) and probiotic (n = 25) groups. Participants were completely blind to the supplements. The samples (blood and fecal) were gathered from the study subjects at baseline (week 0) and after 8 weeks. Participants were asked to follow the allocated follow-up visits without absence. The changes in the glycemic, lipid parameters and microbial composition were studied. The study protocol is illustrated in Figure 1.

2.2. Probiotics Supplementation

Aluminum foil sachets containing 10 × 109 CFU of Lactobacillus rhamnosus received from Lactomason Co., Ltd. (Gyeongsangnam-do, Republic of Korea) were provided to the participants in the probiotic group. The placebo group participants were given a 10 g cornstarch sachet. All participants were instructed to take the supplement regularly by dissolving the contents of one sachet in a glass of water before breakfast. Instructions were given to store the sachet in a dry place at 2–8 °C. Participants were encouraged not to change their physical activities, nutrition, or lifestyle. The participants were advised to avoid the intake of any other probiotics, dietary supplements, or fermented food throughout the study.

2.3. Demographic Assessments

After assigning the participants to respective groups, the sociodemographic characteristics, including age, gender, smoking, alcohol drinking, and body weight, were recorded. Body and visceral fat were manually noted using an electronic scale (Picooc®, Model S1 Pro, Beijing, China) (Table 1). The blood parameters, including triglyceride, fasting blood glucose (FBS), total cholesterol (TC), high-density lipoprotein-cholesterol (HDL-C), and low-density lipoprotein-cholesterol (LDL-C), were determined from blood using the automated machine at AMS Clinical Service Center, Chiang Mai University, Chiang Mai, Thailand.

2.4. Next-Generation Sequencing (NGS)

The fecal genomic DNA was isolated, and DNA sequencing was conducted by the Omics Sciences and Bioinformatics Center, Faculty of Science, Chulalongkorn University, Thailand [25]. Due to the lack of quality DNA, we conducted NGS for only 12 samples each for the placebo and probiotic groups.

2.5. Statistical Analyses

The data are shown as the mean ± standard error of the mean (SEM) for continuous outcomes or as an absolute number and percentage for categorical outcomes. A paired t-test was performed for normally distributed data. The Wilcoxon signed-rank test analyzed the skewed data. The differentiation of outcomes between the placebo and probiotic groups was compared using a Mann–Whitney U test. The p-value < 0.05 was set as significant (two-tailed). The STATA version 15.1 for Windows (StataCorp, College Station, TX, USA) was utilized for statistical analysis.

3. Results

3.1. Changes in Biochemical Parameters

The basic parameters like body weight, body fat, visceral fat, and blood biochemical parameters like FBS, TC, TG, HDL, and LDL were measured at baseline and week 8 of the study. There were no significant differences in glycaemic or lipid parameters between the baseline and after treatment in the placebo group (Table 2). The marginal differences were observed in FBS, TG, HDL, and LDL after treatment in probiotic groups. These changes can be considered the initial state of functional changes due to the probiotic intervention. However, more duration is required to achieve significant effects in these parameters.
The statistical comparison of the considered parameters between the treatment and placebo groups was detailed (Table 3). The body weight (reduced; placebo (−0.06); probiotic (−0.68)), body fat (reduced; probiotic (−0.48)), visceral fat (reduced; probiotic (−1.45)), FBS (reduced; probiotic (−2.96)), and TC (reduced; probiotic (−14.36)), TG (reduced; probiotic (−7.88)), and LDL (reduced; probiotic (−5.44)) values were changed after the treatment in the placebo and probiotic groups, but not statistically significant (Table 3).
L. rhamnosus supplementation for 8 weeks substantially improved LDL (−69.85 to −0.79; p = 0.045) (Table 4). Besides LDL, no other parameters showed significant differences after 8 weeks of study.

3.2. Microbiome Analysis

The valid sequences were identified by matching the raw sequences with the corresponding barcode. The raw-sequence tags underwent analysis using QIIME 2™. Following chimera detection, the sequences were grouped into operational taxonomic units (OTUs) at a 97% sequence identity threshold. Quality assessment of pair-end reads was conducted by examining information in FASTQ files using DADA2. Reads failing to meet the default QIIME 2™ threshold values were filtered out, including a minimum quality score of 25, minimum/maximum length requirements of 200/1000, prohibition of ambiguous bases, and absence of mismatches in the primer sequence. The details of processed sequence reads are detailed in Table 5.
Changes in the phylum, genus, and species diversity between and within (baseline vs. after treatment) the placebo and probiotics group samples were compared. PW0 and PW8 indicate the baseline and after-treatment samples of the placebo group, respectively. TW0 and TW8 indicate the baseline and after-treatment samples of the probiotic group, respectively.

3.2.1. Alpha Diversity

The Shannon diversity index was calculated to evaluate substantial group variations and analyzed using the Kruskal–Wallis (pairwise) test. Berger–Parker analysis was carried out to measure dominance within the community and the proportion of individuals belonging to the most abundant species in the sample. There were no significant changes observed in Berger–Parker analysis (Figure 2A,B), dominance metrics (Figure 2C,D), or Shannon entropy analysis (Figure 2E,F) after treatment in placebo and probiotic groups compared to the respective baseline. The results indicate that, despite observing numerical changes, there was no alteration in microbial abundance or consistency following the treatments.

3.2.2. Beta Diversity

Principal coordinate analysis (PCoA) was employed to assess the relationships between samples, with visualization facilitated by QIIME 2™ View. PCoA plots were generated based on the first three principal coordinates, annotated according to their variance. In the placebo group, 25.79, 11.33, and 8.58% microbial variations were observed in axes 1, 2, and 3, respectively. Similarly, microbial variations of 18.29, 14.27, and 10.24% were observed in axes 1, 2, and 3, respectively, in the probiotic group. The scattered dots, representing baseline and after-treatment samples, indicated that microbial diversity changed after treatments (Figure 3).

3.2.3. Taxonomical Allocations and Determinations

A heat map was generated to illustrate the predicted bacterial taxonomy in the placebo and probiotic samples (Figure 4A,B).
The changes in microbial abundance after probiotic treatment are detailed in Table 6. There were numerical changes in the RF of the microbial abundance at the phylum level, but they were not statistically significant. Similarly, non-significant alteration of microbial abundance was observed at the genus and species level, except Butyricimonas (p = 0.017) and Bacteroides vulgatus (p = 0.021) (Table 6).
Table 7 details the changes in microbial abundance in the placebo group. The abundance of the phylum Actinobacteriota significantly (p = 0.023) increased after treatment. The abundance of Escherichia-Shigella (p = 0.050) was reduced, and the abundance of Collinsella (p = 0.045) and Sutterella (p = 0.047) increased significantly after treatment. No significant differences were reflected in species level in the placebo group (Table 7).
The comparison of placebo and probiotic groups after the treatment period indicated that proteobacteria (p = 0.015), Escherichia-Shigella (p = 0.024), Sutterella (p = 0.039), and Bacteroides vulgatus abundancy were altered significantly (Table 8).

4. Discussion

Age is a significant factor influencing the composition of the human intestinal microbiota, which changes throughout a person’s life [26,27,28,29]. These changes are influenced by factors such as physiological alterations in the gastrointestinal tract associated with aging, dietary habits specific to different countries, lifestyle choices, frailty conditions, antibiotic usage, and nutritional behaviors. Numerous studies have explored the impact of age-related physiological changes, lifestyle factors, and dietary habits on the gut microbiota [26,27,28,29,30]. Aging is accompanied by gut microbiome alterations that increase the various aging-associated diseases. It is difficult to conclude which factor initially contributed to this shift in the gut microbiome. However, lifestyle practices like increased medication, reduced mobility, and diet are found to manipulate the gut microbial composition. Understanding microbial manipulation in the elderly develops promising strategies to prevent age-associated diseases [31]. In the human body, gut microbiota acts as an important metabolic organ facilitating the metabolism of nutrients [32]. In addition to its association with metabolic diseases like obesity, liver diseases, intestinal diseases, neuropsychiatric diseases, diabetes, and cardiovascular diseases (CVD), the gut microbiota also acts as a reservoir for antibiotic-resistant genes [33]. The genotype of the host, diet, diseases, and age affect the composition and diversity of gut microbiota [34,35]. Therefore, employing microbial intervention to manipulate the gut microbiota represents an innovative strategy for impacting sleep and well-being [36]. Ingesting probiotics orally enables the restoration of functional activities within the gut microbiota [37].
Probiotic supplementation can protect gut integrity and restore its functions by initiating the growth of beneficial microbes, safeguarding the intestinal barrier, and positively modulating immune functions [1]. Costabile et al. investigated the effects of probiotic L. rhamnosus GG and pilus-deficient L. rhamnosus GG-PB12combined with soluble corn fiber on microbiota, immunity, metabolism, and blood lipids in healthy elder persons. Consumption of L. rhamnosus GG and prebiotics increased the natural killer cell activity compared to the baseline group. The fecal microbiota analysis showed that the synbiotic supplementation of L. rhamnosus GG with corn fiber and L. rhamnosus GG-PB12 with corn fiber significantly increased Parabacteroides. L. rhamnosus GG with corn fiber increased Ruminococcus, and Incertae Sedis and decreased the Oscillospira and Desulfovibrio. L. rhamnosus GG and corn fiber further reduced the total cholesterol and LDL. L. rhamnosus GG-PB12 with corn fiber treated volunteers showed a significant reduction in C-reactive protein compared to baseline. Thus, the dietary intervention with L. rhamnosus GG and corn fiber could positively enhance the immune response and microbial community [38]. The lipid status of an individual plays an important function in reducing the risk of CVD. Few observational studies suggested that hyperlipidemic subjects have a high risk of developing CVD. Additionally, it was found that reduced serum cholesterol, in turn, reduces the CVD risk. Clinical studies evaluated the beneficial effects of probiotic supplementation on serum lipid profiles [39,40,41]. Synbiotic interventions have been found to have more benefits in hypercholesterolemic patients than in normal people, and the reduction in total cholesterol and LDL levels is greater in the elderly than in younger individuals [38].
In this study, the difference between placebo and probiotic groups at the end of 8 weeks showed that the mean body weight reduction was almost the same. Reductions in visceral fat, FBS, TC, TG, and LDL were greater than placebo but not up to significant levels; simultaneously, there was a suggestive trend indicating improvement from baseline to week 8. However, after 8 weeks, this significance was reduced (p = 0.25) [36]. The difference in HDL level was increased 8 weeks after treatment (Table 3). Certain studies correlated that elevated serum TC, TG, and LDL and low HDL increase the risk for CVD [42,43,44]. Probiotics had no significant effects on the body, visceral fat, FBS, TC, TG, and HDL. The effect of probiotics on LDL depends on various factors, and a significant reduction in LDL was observed in the present study (Table 4). These results indicate that probiotic L. rhamnosus may improve the lipid profile when treated long or combined with other probiotics or prebiotic compounds.
In adults, Firmicutes are predominantly present in the gut, followed by Bacteroidetes [13]. Among the oldest adults, there is generally a decrease in Firmicutes and an increase in Bacteroidetes, which aligns with previous findings indicating a rise in the Firmicutes/Bacteroidetes ratio during adulthood followed by a decline in older age [45]. Vogt et al. [46] reported that Bacteroidetes showed an increase in abundance, whereas Firmicutes and the genus Bifidobacterium exhibited a decrease. Similarly, prior research has demonstrated that excessively high and excessively low Firmicutes/Bacteroidetes ratios can be linked to metabolic and gastrointestinal disorders [47]. These findings imply that a balanced distribution of these core phyla may signify good health and longevity, although specific environmental factors may partly influence this equilibrium. The abundance of Proteobacteria increased after L. rhamnosus supplementation; even though the changes were insignificant, it is noteworthy that L. rhamnosus supplementation could improve the microbiome positively in elderly subjects (Table 6). With the support of the evidence from the previous studies, proteobacteria, which is associated with increased gut inflammation and dysbiosis, was more abundant in older adults than in younger adults [48,49,50,51]. The Shannon–Wiener index considers fewer common species [37].
Following the treatment period, a comparison between the placebo and probiotic groups revealed significant alterations in the richness of Escherichia-Shigella (p = 0.024), Sutterella (p = 0.039), Agathobacter (p = 0.050), and Bacteroides vulgatus (p = 0.033) (Table 8). Laongkham et al. explained that the core gut microbiota of healthy Thai individuals comprises eleven species, including Firmicutes, Bacteroidetes, and Proteobacteria, shared by over 90% of subjects. Notably, Escherichia coli was found to be highly prevalent, especially among Thai elderly individuals. Age and PCA coordination were also correlated, particularly regarding the loading vector associated with the genus Escherichia/Shigella. However, no significant difference was observed in the abundance of this genus group between the adult and elderly groups [52]. After 8 weeks of probiotic intervention, non-significant changes were observed in the abundance of Akkermansia (p = 0.751), Prevotella (p = 0.609), and Bifidobacterium (p = 0.610) (Table 6). The alterations in Akkermansia, along with the changes in Bifidobacterium and the decline of Prevotella, have been proposed as biomarkers for PD [53].
In the case of the Agathobacter genus, López-García et al. mentioned that it did not meet statistical significance during the analysis. Still, there was a noticeable fluctuation in its occurrence throughout the clinical trial. In the Lactiplantibacillus pentosus-supplemented group, there was a rise in the average frequency of Agathobacter sequences from 4.67 to 4.98%. Conversely, there was a decrease in the placebo group from an initial frequency of 3.70% to a final frequency of 1.98% [54]. Similarly, in an investigation, nine healthy individuals were chosen to undergo a fasting regimen of approximately 17 h per day for 29 days. After the trial, no notable alterations in the measured values were reported. However, a noteworthy rise in the prevalence of both Akkermansia muciniphila and Bacteroides fragilis was reported [55].
Additionally, two clinical trials demonstrated a reduction in the abundance of B. vulgatus following the administration of probiotics [56,57]. Assessing intestinal microbiota composition poses a significant challenge due to its notable variability. This is primarily because modifications to commensal strains can occur within a short span of just a few days through changes in diet and lifestyle [58,59]. These findings underscore specific taxonomic changes within the probiotic group over the study period, emphasizing the importance of evaluating microbial dynamics at different taxonomic levels for a comprehensive understanding of the impact of probiotic intervention.

5. Limitations

The study has a few limitations that warrant consideration in interpreting its findings. The sample size of the study was relatively small, and the results may not be fully representative of the broader elderly population of Thailand. Individual variations among the study subjects may impact the generalizability of the observed effects. The study duration (8 weeks) was relatively short, with long-term effects. The sustainability of changes in glycaemic index, lipid profile, and microbiome may not be adequately captured within this timeframe.
Additionally, the exclusive use of a single strain, L. rhamnosus, as the probiotic intervention limits the understanding of potential synergistic effects that may arise from combining multiple strains. Furthermore, the study employed a single dosage of the probiotic, and exploring the impact of different dosage levels could provide insights into the dose–response relationship and optimal supplementation levels. The absence of follow-up research beyond the 8-week intervention period is another limitation, as longer-term assessments could reveal whether the observed effects persist or diminish over time.

6. Conclusions

The 8-week supplementation of L. rhamnosus improved the glycaemic index and lipid profile positively, but statistical significance was not observed. Gaussian regression analysis indicated that the probiotic supplementation significantly reduced the LDL level in the elderly subjects. Microbiome analysis revealed taxonomic shifts in both the placebo and probiotic groups. The results revealed that L. rhamnosus supplementation did not significantly affect the microbiome of the healthy elderly subjects. The study, however, is limited by its small sample size, short duration, and the use of a single probiotic strain. Future research with larger cohorts, extended study periods, and different probiotic formulations must confirm the findings. Addressing these limitations will contribute to a more comprehensive understanding of the potential benefits and mechanisms underlying probiotic interventions in the elderly population.

Author Contributions

Conceptualization, C.C., B.S.S., W.R., S.S., P.K. and P.S.; methodology, C.C., S.K., W.R. and T.C.; software, S.T., M.B., S.K. and B.S.S.; validation, C.C., M.B., P.K., W.R. and S.S.; formal analysis, T.C., S.T., M.B., P.K., P.F., W.R. and C.C.; investigation, S.K., S.S., S.T., W.R. and C.C.; resources, C.C.; data curation, S.T., M.B., T.C., P.F. and S.K.; writing—original draft preparation, B.S.S., S.T., C.C., P.K. and N.S.; writing—review and editing, B.S.S., S.T., C.C., P.K., P.S., W.R. and N.S.; supervision, W.R.; project administration, S.P. and C.C.; funding acquisition, C.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Fundamental Fund Research-2024, Chiang Mai University, Chiang Mai, Thailand.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in the study are included in the article, further inquiries can be directed to the corresponding author.

Acknowledgments

The authors gratefully acknowledge the Faculty of Pharmacy and Chiang Mai University, Chiang Mai, Thailand, for their support. T.C. wishes to acknowledge the support of the Postdoctoral Fellowship from Mae Fah Luang University, Thailand.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Landete, J.M.; Gaya, P.; Rodríguez, E.; Langa, S.; Peirotén, Á.; Medina, M.; Arqués, J.L. Probiotic Bacteria for Healthier Aging: Immunomodulation and Metabolism of Phytoestrogens. Biomed. Res. Int. 2017, 2017, 5939818. [Google Scholar] [CrossRef] [PubMed]
  2. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef]
  3. Lakshminarayanan, B.; Stanton, C.; O’Toole, P.W.; Ross, R.P. Compositional dynamics of the human intestinal microbiota with aging: Implications for health. J. Nutr. Health. Aging 2014, 18, 773–786. [Google Scholar] [CrossRef] [PubMed]
  4. Boyajian, J.L.; Ghebretatios, M.; Schaly, S.; Islam, P.; Prakash, S. Microbiome and Human Aging: Probiotic and Prebiotic Potentials in Longevity, Skin Health and Cellular Senescence. Nutrients 2021, 13, 4550. [Google Scholar] [CrossRef] [PubMed]
  5. Guarner, F.; Malagelada, J.R. Gut flora in health and disease. Lancet 2003, 361, 512–519. [Google Scholar] [CrossRef] [PubMed]
  6. Bäckhed, F.; Ley, R.E.; Sonnenburg, J.L.; Peterson, D.A.; Gordon, J.I. Host-bacterial mutualism in the human intestine. Science 2005, 307, 1915–1920. [Google Scholar] [CrossRef] [PubMed]
  7. Hickson, M. Malnutrition and ageing. Postgrad. Med. J. 2006, 82, 2–8. [Google Scholar] [CrossRef] [PubMed]
  8. Tiihonen, K.; Ouwehand, A.C.; Rautonen, N. Human intestinal microbiota and healthy ageing. Ageing. Res. Rev. 2010, 9, 107–116. [Google Scholar] [CrossRef]
  9. Claesson, M.J.; Cusack, S.; O’Sullivan, O.; Greene-Diniz, R.; de Weerd, H.; Flannery, E.; Marchesi, J.R.; Falush, D.; Dinan, T.; Fitzgerald, G.; et al. Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc. Natl. Acad. Sci. USA 2011, 108, 4586–4591. [Google Scholar] [CrossRef]
  10. Claesson, M.J.; Jeffery, I.B.; Conde, S.; Power, S.E.; O’Connor, E.M.; Cusack, S.; Harris, H.M.; Coakley, M.; Lakshminarayanan, B.; O’Sullivan, O.; et al. Gut microbiota composition correlates with diet and health in the elderly. Nature 2012, 488, 178–184. [Google Scholar] [CrossRef]
  11. Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.; Relman, D.A. Diversity of the human intestinal microbial flora. Science 2005, 308, 1635–1638. [Google Scholar] [CrossRef] [PubMed]
  12. Rondanelli, M.; Giacosa, A.; Faliva, M.A.; Perna, S.; Allieri, F.; Castellazzi, A.M. Review on microbiota and effectiveness of probiotics use in older. World J. Clin. Cases 2015, 3, 156–162. [Google Scholar] [CrossRef] [PubMed]
  13. Odamaki, T.; Kato, K.; Sugahara, H.; Hashikura, N.; Takahashi, S.; Xiao, J.Z.; Abe, F.; Osawa, R. Age-related changes in gut microbiota composition from newborn to centenarian: A cross-sectional study. BMC Microbiol. 2016, 16, 90. [Google Scholar] [CrossRef] [PubMed]
  14. Woodmansey, E.J. Intestinal bacteria and ageing. J. Appl. Microbiol. 2007, 102, 1178–1186. [Google Scholar] [CrossRef] [PubMed]
  15. Hopkins, M.J.; Macfarlane, G.T. Changes in predominant bacterial populations in human faeces with age and with Clostridium difficile infection. J. Med. Microbiol. 2002, 51, 448–454. [Google Scholar] [CrossRef] [PubMed]
  16. Mäkivuokko, H.; Tiihonen, K.; Tynkkynen, S.; Paulin, L.; Rautonen, N. The effect of age and non-steroidal anti-inflammatory drugs on human intestinal microbiota composition. Br. J. Nutr. 2010, 103, 227–234. [Google Scholar] [CrossRef] [PubMed]
  17. Biagi, E.; Franceschi, C.; Rampelli, S.; Severgnini, M.; Ostan, R.; Turroni, S.; Consolandi, C.; Quercia, S.; Scurti, M.; Monti, D.; et al. Gut Microbiota and Extreme Longevity. Curr. Biol. 2016, 26, 1480–1485. [Google Scholar] [CrossRef] [PubMed]
  18. Kong, F.; Hua, Y.; Zeng, B.; Ning, R.; Li, Y.; Zhao, J. Gut microbiota signatures of longevity. Curr. Biol. 2016, 26, R832–R833. [Google Scholar] [CrossRef] [PubMed]
  19. Du, Y.; Gao, Y.; Zeng, B.; Fan, X.; Yang, D.; Yang, M. Effects of anti-aging interventions on intestinal microbiota. Gut Microbes 2021, 13, 1994835. [Google Scholar] [CrossRef]
  20. Bischoff, S.C. Microbiota and aging. Curr. Opin. Clin. Nutr. Metab. Care 2016, 19, 26–30. [Google Scholar] [CrossRef]
  21. Chung, K.W. Advances in Understanding of the Role of Lipid Metabolism in Aging. Cells 2021, 10, 880. [Google Scholar] [CrossRef] [PubMed]
  22. Johnson, A.A.; Stolzing, A. The role of lipid metabolism in aging, lifespan regulation, and age-related disease. Aging Cell. 2019, 18, e13048. [Google Scholar] [CrossRef] [PubMed]
  23. Chiang, B.L.; Sheih, Y.H.; Wang, L.H.; Liao, C.K.; Gill, H.S. Enhancing immunity by dietary consumption of a probiotic lactic acid bacterium (Bifidobacterium lactis HN019): Optimization and definition of cellular immune responses. Eur. J. Clin. Nutr. 2000, 54, 849–855. [Google Scholar] [CrossRef] [PubMed]
  24. Sivamaruthi, B.S.; Kesika, P.; Chaiyasut, C. A review on anti-aging properties of probiotics. Int. J. Appl. Pharm. 2018, 10, 23–27. [Google Scholar] [CrossRef]
  25. Chaiyasut, C.; Sirilun, S.; Juntarachot, N.; Tongpong, P.; Ouparee, W.; Sivamaruthi, B.S.; Peerajan, S.; Waditee-Sirisattha, R.; Prombutara, P.; Klankeo, P.; et al. Effect of Dextranase and Dextranase-and-Nisin-Containing Mouthwashes on Oral Microbial Community of Healthy Adults-A Pilot Study. Appl. Sci. 2022, 12, 1650. [Google Scholar] [CrossRef]
  26. Mueller, S.; Saunier, K.; Hanisch, C.; Norin, E.; Alm, L.; Midtvedt, T. Differences in fecal microbiota in diferent European study populations in relation to age, gender, and country: A cross-sectional study. Appl. Environ. Microbiol. 2006, 72, 1027–1033. [Google Scholar] [CrossRef] [PubMed]
  27. Biagi, E.; Nylund, L.; Candela, M.; Ostan, R.; Bucci, L.; Pini, E.; Nikkïla, J.; Monti, D.; Satokari, R.; Franceschi, C.; et al. Through ageing, and beyond: Gut microbiota and infammatory status in seniors and centenarians. PLoS ONE 2010, 5, e10667. [Google Scholar] [CrossRef]
  28. Zhao, L.; Qiao, X.; Zhu, J.; Zhang, X.; Jiang, J.; Hao, Y.; Ren, F. Correlations of fecal bacterial communities with age and living region for the elderly living in Bama, Guangxi. China. J. Microbiol. 2011, 49, 186–192. [Google Scholar] [CrossRef] [PubMed]
  29. Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef]
  30. Hayashi, H.; Sakamoto, M.; Kitahara, M.; Benno, Y. Molecular analysis of fecal microbiota in elderly individuals using 16S rDNA library and T-RFLP. Microbiol. Immunol. 2003, 47, 557–570. [Google Scholar] [CrossRef]
  31. Hutchinson, A.N.; 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. [Google Scholar] [CrossRef] [PubMed]
  32. Liu, Y.H.; Kang, L. Progress in researches on gut microbiota and metabolism. Chin. J. Microecol. 2013, 3, 362–364+367. [Google Scholar]
  33. Sommer, M.O.; Dantas, G. Antibiotics and the resistant microbiome. Curr. Opin. Microbiol. 2011, 14, 556–563. [Google Scholar] [CrossRef] [PubMed]
  34. Gajer, P.; Brotman, R.M.; Bai, G.; Sakamoto, J.; Schütte, U.M.; Zhong, X.; Koenig, S.S.; Fu, L.; Ma, Z.S.; Zhou, X.; et al. Temporal dynamics of the human vaginal microbiota. Sci. Transl. Med. 2012, 4, 132ra52. [Google Scholar] [CrossRef] [PubMed]
  35. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [PubMed]
  36. Patterson, E.; Tan, H.T.T.; Groeger, D.; Andrews, M.; Buckley, M.; Murphy, E.F.; Groeger, J.A. Bifidobacterium longum 1714 improves sleep quality and aspects of well-being in healthy adults: A randomized, double-blind, placebo-controlled clinical trial. Sci. Rep. 2024, 14, 3725. [Google Scholar] [CrossRef]
  37. Vicariotto, F.; Malfa, P.; Viciani, E.; Dell’Atti, F.; Squarzanti, D.F.; Marcante, A.; Castagnetti, A.; Ponchia, R.; Governini, L.; De Leo, V. Efficacy of Lactiplantibacillus plantarum PBS067, Bifidobacterium animalis subsp. lactis BL050, and Lacticaseibacillus rhamnosus LRH020 in the Amelioration of Vaginal Microbiota in Post-Menopausal Women: A Prospective Observational Clinical Trial. Nutrients 2024, 16, 402. [Google Scholar] [CrossRef] [PubMed]
  38. Costabile, A.; Bergillos-Meca, T.; Rasinkangas, P.; Korpela, K.; de Vos, W.M.; Gibson, G.R. Effects of Soluble Corn Fiber Alone or in Synbiotic Combination with Lactobacillus rhamnosus GG and the Pilus-Deficient Derivative GG-PB12 on Fecal Microbiota, Metabolism, and Markers of Immune Function: A Randomized, Double-Blind, Placebo-Controlled, Crossover Study in Healthy Elderly (Saimes Study). Front. Immunol. 2017, 8, 1443. [Google Scholar] [PubMed]
  39. Sabatine, M.S.; Wiviott, S.D.; Im, K.; Murphy, S.A.; Giugliano, R.P. Efficacy and Safety of Further Lowering of Low-Density Lipoprotein Cholesterol in Patients Starting With Very Low Levels: A Meta-analysis. JAMA Cardiol. 2018, 3, 823–828. [Google Scholar] [CrossRef]
  40. Chen, Z.Y.; Jiao, R.; Ma, K.Y. Cholesterol-lowering nutraceuticals and functional foods. J. Agric. Food Chem. 2008, 56, 8761–8773. [Google Scholar] [CrossRef]
  41. Shimizu, M.; Hashiquchi, M.; Shiga, T.; Tamura, H.O.; Mochizuki, M. Meta analysis: Effects of probiotic supplementation on lipid profiles in normal to mildly hypercholesterolemic individuals. PLoS ONE 2015, 10, e0139795. [Google Scholar] [CrossRef] [PubMed]
  42. Gould, A.L.; Davies, G.M.; Alemao, E.; Yin, D.D.; Cook, J.R. Cholesterol reduction yields clinical benefits: Meta-analysis including recent trials. Clin. Ther. 2007, 29, 778–794. [Google Scholar] [CrossRef] [PubMed]
  43. Law, M.R.; Wald, N.J.; Rudnicka, A.R. Quantifying effect of statins on low density lipoprotein cholesterol, ischaemic heart disease, and stroke: Systematic review and meta-analysis. BMJ 2003, 326, 1423–1429. [Google Scholar] [CrossRef] [PubMed]
  44. De Caterina, R.; Scarano, M.; Marfisi, R.; Lucisano, G.; Palma, F.; Tatasciore, A.; Marchioli, R. Cholesterol-lowering interventions and stroke: Insights from a meta-analysis of randomized controlled trials. J. Am. Coll. Cardiol. 2010, 55, 198–211. [Google Scholar] [CrossRef] [PubMed]
  45. Mariat, D.; Firmesse, O.; Levenez, F.; Guimarăes, V.; Sokol, H.; Doré, J.; Corthier, G.; Furet, J.P. The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol. 2009, 9, 123. [Google Scholar] [CrossRef]
  46. Vogt, N.M.; Kerby, R.L.; Dill-McFarland, K.A.; Harding, S.J.; Merluzzi, A.P.; Johnson, S.C.; Carlsson, C.M.; Asthana, S.; Zetterberg, H.; Blennow, K.; et al. Gut microbiome alterations in Alzheimer’s disease. Sci. Rep. 2017, 7, 13537. [Google Scholar] [CrossRef] [PubMed]
  47. Walters, W.A.; Xu, Z.; Knight, R. Meta-analyses of human gut microbes associated with obesity and IBD. FEBS Lett. 2014, 588, 4223–4233. [Google Scholar] [CrossRef] [PubMed]
  48. Rizzatti, G.; Lopetuso, L.; Gibiino, G.; Binda, C.; Gasbarrini, A. Proteobacteria: A common factor in human diseases. BioMed Res. Int. 2017, 2017, 9351507. [Google Scholar] [CrossRef] [PubMed]
  49. Kim, B.S.; Choi, C.W.; Shin, H.; Jin, S.P.; Bae, J.S.; Han, M.; Seo, E.Y.; Chun, J.; Chung, J.H. Comparison of the gut microbiota of centenarians in longevity villages of South Korea with those of other age groups. J. Microbiol. Biotechnol. 2019, 29, 429–440. [Google Scholar] [CrossRef]
  50. Wu, L.; Zeng, T.; Zinellu, A.; Rubino, S.; Kelvin, D.J.; Carru, C. A Cross-Sectional Study of Compositional and Functional Profiles of Gut Microbiota in Sardinian Centenarians. mSystems 2019, 4, e00325-19. [Google Scholar] [CrossRef]
  51. Yu, X.; Wu, X.; Qiu, L.; Wang, D.; Gan, M.; Chen, X.; Wei, H.; Xu, F. Analysis of the intestinal microbial community structure of healthy and long-living elderly residents in Gaotian Village of Liuyang City. Appl. Microbiol. Biotechnol. 2015, 99, 9085–9095. [Google Scholar] [CrossRef] [PubMed]
  52. Laongkham, O.; Nakphaichit, M.; Nakayama, J.; Keawsompong, S.; Nitisinprasert, S. Age-related changes in the gut microbiota and the core gut microbiome of healthy Thai humans. 3 Biotech 2020, 10, 276. [Google Scholar] [CrossRef] [PubMed]
  53. Vidal-Martinez, G.; Chin, B.; Camarillo, C.; Herrera, G.V.; Yang, B.; Sarosiek, I.; Perez, R.G. A Pilot Microbiota Study in Parkinson’s Disease Patients versus Control Subjects, and Effects of FTY720 and FTY720-Mitoxy Therapies in Parkinsonian and Multiple System Atrophy Mouse Models. J. Parkinsons Dis. 2020, 10, 185–192. [Google Scholar] [CrossRef] [PubMed]
  54. López-García, E.; Benítez-Cabello, A.; Arenas-de Larriva, A.P.; Gutierrez-Mariscal, F.M.; Pérez-Martínez, P.; Yubero-Serrano, E.M.; Garrido-Fernández, A.; Arroyo-López, F.N. Oral intake of Lactiplantibacillus pentosus LPG1 Produces a Beneficial Regulation of Gut Microbiota in Healthy Persons: A Randomized, Placebo-Controlled, Single-Blind Trial. Nutrients 2023, 15, 1931. [Google Scholar] [CrossRef] [PubMed]
  55. Ozkul, C.; Yalinay, M.; Karakan, T. Islamic fasting leads to an increased abundance of Akkermansia muciniphila and Bacteroides fragilis group: A preliminary study on intermittent fasting. Turk. J. Gastroenterol. 2019, 30, 1030–1035. [Google Scholar] [CrossRef] [PubMed]
  56. Nicolucci, A.C.; Hume, M.P.; Martínez, I.; Mayengbam, S.; Walter, J.; Reimer, R.A. Prebiotics reduce body fat and alter intestinal microbiota in children who are overweight or with obesity. Gastroenterology 2017, 153, 711–722. [Google Scholar] [CrossRef] [PubMed]
  57. Dewulf, E.M.; Cani, P.D.; Claus, S.P.; Fuentes, S.; Puylaert, P.G.; Neyrinck, A.M.; Bindels, L.B.; de Vos, W.M.; Gibson, G.R.; Thissen, J.P.; et al. Insight into the prebiotic concept: Lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women. Gut 2013, 62, 1112–1121. [Google Scholar] [CrossRef]
  58. Ciobârcă, D.; Cătoi, A.F.; Copăescu, C.; Miere, D.; Crișan, G. Bariatric Surgery in Obesity: Effects on Gut Microbiota and Micronutrient Status. Nutrients 2020, 12, 235. [Google Scholar] [CrossRef]
  59. Alhusain, F. Microbiome: Role and Functionality in Human Nutrition Cycle. Saudi Med. J. 2021, 42, 146–150. [Google Scholar] [CrossRef]
Figure 1. The illustration shows the study protocol (created using Biorender.com; accessed on 4 March 2024).
Figure 1. The illustration shows the study protocol (created using Biorender.com; accessed on 4 March 2024).
Foods 13 01293 g001
Figure 2. Alpha diversity estimation between the baseline and post-treatment of placebo (PW0 and PW8) and probiotic groups (TW0 and TW8). The results of Berger–Parker (A,B) analysis, dominance metrics (C,D), and Shannon entropy (E,F).
Figure 2. Alpha diversity estimation between the baseline and post-treatment of placebo (PW0 and PW8) and probiotic groups (TW0 and TW8). The results of Berger–Parker (A,B) analysis, dominance metrics (C,D), and Shannon entropy (E,F).
Foods 13 01293 g002
Figure 3. Beta diversity of placebo (A) and probiotic (B) group. The circle dots indicate the outlier samples. The red and blue dots indicate the baseline and treatment samples of the placebo group. The purple and orange dots indicate the baseline and treatment samples of the probiotic group.
Figure 3. Beta diversity of placebo (A) and probiotic (B) group. The circle dots indicate the outlier samples. The red and blue dots indicate the baseline and treatment samples of the placebo group. The purple and orange dots indicate the baseline and treatment samples of the probiotic group.
Foods 13 01293 g003
Figure 4. The heat-map represents the taxonomy estimated for the placebo (A) and probiotic (B) samples after week 0 and week 8 of treatment. The diversity was represented with log10 frequency.
Figure 4. The heat-map represents the taxonomy estimated for the placebo (A) and probiotic (B) samples after week 0 and week 8 of treatment. The diversity was represented with log10 frequency.
Foods 13 01293 g004
Table 1. Sociodemographic data of the study participants.
Table 1. Sociodemographic data of the study participants.
No.VariablesGroupp-Value
Placebo (n = 25)Probiotic (n = 25)
1Age64.96 ± 0.8663.00 ± 1.090.165 a
2Male, n (%)3 (12.00)9 (36.00)0.095 b
Female, n (%)22 (88.00)16 (64.00)
3Smoking 0.490 b
No, n (%)25 (100.00)23 (92.00)
Yes, n (%)0 (0.00)2 (8.00)
4Alcohol drinking 0.235 b
No, n (%)25 (100.00)22 (88.00)
Yes, n (%)0 (0.00)3 (12.00)
5Body weight (kg)63.80 ± 2.2959.40 ± 2.170.170 a
6Body fat (%)32.77 ± 1.4127.76 ± 1.450.036 *a
7Visceral fat (%)15.18 ± 0.5713.55 ± 1.490.060 c
8FBS (mg/dL)106.12 ± 8.77104.00 ± 5.970.843 a
9TC (mg/dL)211.68 ± 9.41209.12 ± 8.550.841 a
10TG (mg/dL)134.48 ± 11.20157.36 ± 12.490.179 a
11HDL (mg/dL)53.04 ± 1.9552.72 ± 3.090.931 a
12LDL (mg/dL)131.08 ± 8.35120.01 ± 6.220.293 a
FBS: fasting blood sugar; TC: total cholesterol; TG: triglycerides; HDL: high-density lipoprotein; LDL: low-density lipoprotein. Data are mean ± SE. * indicates the significant difference in p-value at a 95% confidence interval. a p-value from the independent t-test, b p-value from Fischer’s exact test, c p-value from the Mann–Whitney U test.
Table 2. Biochemical parameters at baseline (week 0) and treatment (week 8) in the placebo and probiotic groups.
Table 2. Biochemical parameters at baseline (week 0) and treatment (week 8) in the placebo and probiotic groups.
ParametersPlacebo (n = 25)p-ValueProbiotic (n = 25)p-Value
Baseline
(P-Week 0)
Treatment
(P-Week 8)
Baseline
(T-Week 0)
Treatment
(T-Week 8)
Body weight (kg)63.80 ± 2.2963.74 ± 2.300.884 a59.40 ± 2.1758.73 ± 2.110.058 a
Body fat (%)32.77 ± 1.4134.95 ± 1.530.127 a27.76 ± 1.4527.28 ± 1.670.657 a
Visceral fat (%)15.18 ± 0.5715.27 ± 0.660.402 b13.55 ± 1.4912.09 ± 1.130.098 b
FBS (mg/dL)106.12 ± 8.77109.92 ± 6.950.504 a104.00 ± 5.97106.96 ± 5.840.375 a
TC (mg/dL)211.68 ± 9.41201.96 ± 11.980.178 a209.12 ± 8.55194.76 ± 7.630.055 a
TG (mg/dL)134.48 ± 11.20149.80 ± 13.620.184 a157.36 ± 12.49149.48 ± 12.620.463 a
HDL (mg/dL)53.04 ± 1.9553.52 ± 3.550.858 a52.72 ± 3.0956.28 ± 3.460.137 a
LDL (mg/dL)131.08 ± 8.35128.42 ± 8.460.728 a120.01 ± 6.22114.57 ± 8.000.439 a
P-week 0: Placebo-week 0; P-week 8: Placebo-week 8; T-week 0: Treatment-week 0; T-week 8: Treatment-week 8; Baseline-Week 0; Treatment-Week 8; TC: total cholesterol; TG: triglycerides; HDL: high-density lipoprotein; LDL: low-density lipoprotein. Data are mean ± SE. a p-value from Paired t-test, b p-value from the Wilcoxon signed-rank test.
Table 3. The differentiation of biochemical parameters between the placebo and probiotic groups at the end of the study.
Table 3. The differentiation of biochemical parameters between the placebo and probiotic groups at the end of the study.
VariablesDifferencep-Value *
Placebo (n = 25)Probiotic (n = 25)
Body weight (kg)−0.06−0.680.252
Body fat (%)2.17−0.480.163
Visceral fat (%)0.09−1.450.104
FBS (mg/dL)3.80−2.960.734
TC (mg/dL)−9.72−14.360.869
TG (mg/dL)15.32−7.880.479
HDL (mg/dL)0.483.560.214
LDL (mg/dL)−2.66−5.440.823
FBS: Fasting blood sugar; TC: total cholesterol; TG: triglycerides; HDL: high-density lipoprotein; LDL: low-density lipoprotein. Data are mean. * p-value from the Mann–Whitney U test.
Table 4. Gaussian regression analysis of the probiotic treatment group after 8 weeks of study.
Table 4. Gaussian regression analysis of the probiotic treatment group after 8 weeks of study.
ParametersCoefficient95% Confidence Intervalp-Value
Body weight (kg)−0.65−2.24 to 0.940.409
Body fat (%)−2.48−6.48 to 1.520.214
Visceral fat (%)−0.39−2.14 to 1.360.650
FBS (mg/dL)−5.19−22.15 to 11.780.536
Total cholesterol (mg/dL)−4.99−28.91 to 18.930.675
Triglyceride (mg/dL)−25.07−74.60 to 24.470.308
HDL (mg/dL)9.76−2.43 to 21.940.112
LDL (mg/dL)−35.32−69.85 to −0.790.045 *
FBS: Fasting blood sugar; TC: total cholesterol; TG: triglycerides; HDL: high-density lipoprotein; LDL: low-density lipoprotein. Data are mean. * indicates the significant difference in p-value at a 95% confidence interval.
Table 5. The 16s rRNA amplicon sequences of the placebo (Baseline: PW0, After-treatment: PW8) and probiotics (Baseline: TW0, After-treatment: TW8) groups.
Table 5. The 16s rRNA amplicon sequences of the placebo (Baseline: PW0, After-treatment: PW8) and probiotics (Baseline: TW0, After-treatment: TW8) groups.
Sample-IDInputFilteredDenoisedMergedNon-Chimeric
Placebo group
PW0-1-02228,670156,586155,663152,50884,553
PW0-1-1376,71650,51949,90648,82141,576
PW0-1-15121,42388,25487,61285,70952,312
PW0-1-2465,60749,26248,99448,20228,559
PW0-1-3572,98150,93650,57649,17136,557
PW0-1-36121,09682,24981,56279,97668,897
PW0-1-3948,33234,30534,01233,22525,337
PW0-1-4283,99962,26261,82060,45140,175
PW0-1-4362,70243,38942,81841,42330,313
PW0-1-6364,51754,97554,78953,89435,429
PW0-1-6969,46357,12056,96556,29742,940
PW0-1-7882,03367,79367,53966,81648,736
PW8-2-6334,39714,82514,70814,53111,610
PW8-2-6941,79019,35819,14718,87713,859
PW8-2-7854,62425,33825,05324,42118,908
PW8-3-0299,99768,32167,63766,19646,325
PW8-3-1365,99046,65246,32745,63236,879
PW8-3-1550,67232,26231,96131,48924,307
PW8-3-24128,32793,30092,84990,84870,533
PW8-3-3550,17236,30235,95534,84025,906
PW8-3-3688,83964,98864,50763,08551,453
PW8-3-3984,64260,54560,06559,02344,259
PW8-3-42264,836208,317207,379203,133145,732
PW8-3-43100,42574,39673,84672,27952,322
Probiotic group
TW0-1-22138,660101,667101,21899,82974,148
TW0-1-23108,78578,70277,92276,38251,645
TW0-1-26148,589111,060110,479108,55571,637
TW0-1-30145,969108,395107,827106,32479,072
TW0-1-33124,551100,19699,96299,23762,441
TW0-1-467,80048,00247,44446,49336,199
TW0-1-10192,77977,62677,26776,43565,627
TW0-1-28123,20798,27597,84995,99957,353
TW0-1-4493,55576,10875,72774,92757,845
TW0-1-6479,34263,33063,01062,27345,977
TW0-1-8780,50265,05364,68963,70846,194
TW0-1-9385,01767,24667,04366,11254,043
TW8-2-10129,90415,10914,92514,67813,074
TW8-2-2837,61918,60418,24917,97714,104
TW8-2-4439,04719,01118,85418,65015,916
TW8-2-6435,48417,17017,01716,84913,546
TW8-2-8729,23213,55313,40513,2989193
TW8-2-9351,54825,06024,82624,40419,697
TW8-3-2280,47557,92357,47556,47649,362
TW8-3-2388,81566,88066,59265,66150,791
TW8-3-26240,804181,521180,569177,129145,464
TW8-3-3087,28664,38164,02763,25450,862
TW8-3-33137,03794,96194,18192,39981,152
TW8-3-4128,89593,09392,46090,94976,221
Table 6. The statistical differences in the phylum, genus, and species between the baseline and after-treatment samples in the probiotic group.
Table 6. The statistical differences in the phylum, genus, and species between the baseline and after-treatment samples in the probiotic group.
TaxonomyBaseline (TW0)After Treatment (TW8)p-Value
Phyla
Proteobacteria14.79 ± 5.5823.46 ± 8.020.100
Verrucomicrobiota5.54 ± 3.712.39 ± 1.840.475
Bacteroidota27.77 ± 7.9024.61 ± 6.310.938
Actinobacteriota9.27 ± 4.006.66 ± 1.840.875
Firmicutes41.09 ± 6.8141.48 ± 6.570.754
Fusobacteriota1.32 ± 0.801.27 ± 0.800.969
Desulfobacterota0.19 ± 0.070.11 ± 0.050.388
Patescibacteria0.04 ± 0.020.01 ± 0.0040.254
Genera
Escherichia-Shigella13.71 ± 5.9021.90 ± 8.630.117
Akkermansia5.63 ± 3.772.49 ± 1.900.751
Bacteroides16.81 ± 6.7618.21 ± 4.900.272
Bifidobacterium5.80 ± 3.864.30 ± 1.520.610
Phascolarctobacterium4.66 ± 1.872.66 ± 1.150.724
Prevotella9.58 ± 3.404.96 ± 2.780.609
Faecalibacterium2.85 ± 0.745.75 ± 1.960.182
Blautia5.52 ± 2.783.77 ± 2.170.239
Collinsella1.70 ± 0.612.22 ± 0.770.388
Weissella3.89 ± 3.700.07 ± 0.030.308
Subdoligranulum0.79 ± 0.321.32 ± 0.480.638
Agathobacter0.41 ± 0.131.13 ± 0.410.100
Romboutsia0.52 ± 0.210.39 ± 0.141.000
Roseburia0.72 ± 0.220.99 ± 0.550.784
Alistipes0.45 ± 0.170.42 ± 0.360.325
Paraprevotella2.05 ± 1.172.79 ± 2.400.305
Streptococcus1.43 ± 0.901.38 ± 0.870.433
Fusobacterium1.14 ± 0.850.38 ± 0.160.969
Slackia0.84 ± 0.371.76 ± 1.120.906
UCG 0020.69 ± 0.171.14 ± 0.280.410
Dorea1.53 ± 1.241.09 ± 0.530.255
Lactobacillus0.08 ± 0.020.39 ± 0.160.784
Monoglobus0.05 ± 0.031.31 ± 1.180.365
Parasutterella0.57 ± 0.430.55 ± 0.420.184
Enterococcus0.22 ± 0.080.64 ± 0.330.723
Butyricicoccus0.91 ± 0.740.08 ± 0.040.388
Olsenella0.20 ± 0.110.72 ± 0.320.305
CAG 3520.81 ± 0.390.30 ± 0.150.076
Holdemanella0.33 ± 0.170.35 ± 0.110.145
Fusicatenibacter0.45 ± 0.170.42 ± 0.360.583
Parabacteroides0.66 ± 0.220.61 ± 0.210.969
Coprococcus0.28 ± 0.140.69 ± 0.270.383
Lachnoclostridium0.50 ± 0.150.30 ± 0.120.153
Barnesiella0.38 ± 0.150.07 ± 0.040.178
Odoribacter0.32 ± 0.100.17 ± 0.100.267
Atopobium0.52 ± 0.510.06 ± 0.050.344
Megamonas0.85 ± 0.350.39 ± 0.270.268
Clostridia UCG 0140.09 ± 0.060.49 ± 0.370.767
Klebsiella1.32 ± 0.630.48 ± 0.210.289
Ruminococcus0.38 ± 0.240.24 ± 0.120.969
Erysipelotrichaceae UCG 0030.31 ± 0.130.08 ± 0.040.234
Enterobacteriaceae0.35 ± 0.180.19 ± 0.120.456
Anaerostipes0.08 ± 0.030.38 ± 0.160.132
Enterobacter0.21 ± 0.070.31 ± 0.200.505
Veillonella0.37 ± 0.151.25 ± 1.140.326
Flavonifractor0.10 ± 0.040.13 ± 0.101.000
UBA18190.09 ± 0.030.13 ± 0.070.875
Lachnospiraceae UCG 0100.13 ± 0.090.04 ± 0.020.692
Hungatella0.15 ± 0.120.15 ± 0.090.966
Butyricimonas0.18 ± 0.050.06 ± 0.040.017 *
Bilophila0.11 ± 0.060.11 ± 0.060.784
Lachnospiraceae0.23 ± 0.060.26 ± 0.130.724
Sutterella0.10 ± 0.040.15 ± 0.111.000
Species
Bacteroides stercoris10.31 ± 7.831.96 ± 1.010.422
Bacteroides vulgatus12.34 ± 3.9428.81 ± 8.310.021 *
Bacteroides fragilis3.26 ± 1.6113.96 ± 7.300.222
Bacteroides uniformis10.70 ± 3.765.32 ± 2.280.365
Lachnospiraceae NK4A136 group4.77 ± 2.026.29 ± 2.670.634
Eubacterium hallii group12.40 ± 5.9812.80 ± 5.460.610
Ruminococcus torques group7.95 ± 2.236.66 ± 2.750.326
Slackia isoflavoniconvertens7.76 ± 4.493.97 ± 1.770.579
Bacteroides massiliensis6.64 ± 3.711.14 ± 0.620.148
Eubacterium eligens group2.30 ± 0.961.12 ± 0.470.222
Incertae sedis0.66 ± 0.220.56 ± 0.290.555
Eubacterium ramulus0.48 ± 0.210.95 ± 0.420.969
Parabacteroides distasonis1.49 ± 0.610.88 ± 0.490.422
The Wilcoxon signed-rank test was used to determine the statistical significance. * Statistically significant if p ≤ 0.05.
Table 7. The statistical differences in the phylum, genus, and species between the baseline and after-treatment samples in the placebo group.
Table 7. The statistical differences in the phylum, genus, and species between the baseline and after-treatment samples in the placebo group.
TaxonomyBaseline (PW0)After Treatment (PW8)p-Value
Phyla
Proteobacteria19.71 ± 6.457.66 ± 1.860.071
Bacteroidota30.95 ± 8.5124.05 ± 4.910.754
Firmicutes44.75 ± 6.7460.11 ± 2.450.136
Actinobacteriota1.68 ± 0.464.35 ± 1.570.023 *
Fusobacteriota1.69 ± 1.170.80 ± 0.500.938
Verrucomicrobiota1.02 ± 0.392.85 ± 2.240.609
Desulfobacterota0.21 ± 0.090.18 ± 0.060.969
Genera
Escherichia-Shigella16.30 ± 6.585.85 ± 2.140.050 *
Bacteroides14.37 ± 4.1414.92 ± 3.881.000
Faecalibacterium3.22 ± 0.833.46 ± 1.240.530
Blautia6.07 ± 3.245.11 ± 0.880.182
Prevotella14.65 ± 6.526.97 ± 2.540.609
Agathobacter1.90 ± 0.830.57 ± 0.240.224
Collinsella0.98 ± 0.293.62 ± 1.890.045 *
Subdoligranulum2.49 ± 1.041.20 ± 0.530.158
Streptococcus2.13 ± 1.535.47 ± 3.560.695
Roseburia1.33 ± 0.282.41 ± 0.830.195
Phascolarctobacterium2.61 ± 0.553.78 ± 1.090.433
Romboutsia0.46 ± 0.161.22 ± 0.510.081
Klebsiella1.54 ± 0.590.63 ± 0.220.170
Fusobacterium1.75 ± 1.210.92 ± 0.570.938
Ruminococcus0.92 ± 0.331.11 ± 0.460.906
Dorea0.64 ± 0.171.20 ± 0.530.254
Akkermansia1.12 ± 0.433.92 ± 3.040.609
Lachnoclostridium0.77 ± 0.141.28 ± 0.490.814
UCG-0021.50 ± 0.550.95 ± 0.440.289
Fusicatenibacter0.68 ± 0.280.35 ± 0.250.209
Enterobacter1.29 ± 0.600.09 ± 0.030.090
Veillonella0.21 ± 0.084.91 ± 2.590.100
Alistipes1.23 ± 0.540.96 ± 0.380.480
CAG-3520.39 ± 0.120.52 ± 0.220.969
Enterobacteriaceae1.88 ± 1.100.35 ± 0.170.209
Coprococcus0.70 ± 0.230.77 ± 0.220.410
Sutterella0.49 ± 0.241.64 ± 0.990.047 *
Anaerostipes0.41 ± 0.180.64 ± 0.220.556
Parabacteroides0.90 ± 0.291.04 ± 0.350.875
Holdemanella0.68 ± 0.370.53 ± 0.320.906
Butyricicoccus0.52 ± 0.141.03 ± 0.250.100
Enterococcus0.15 ± 0.092.62 ± 1.830.057
Lachnospira0.41 ± 0.200.22 ± 0.100.428
Lachnospiraceae0.62 ± 0.241.04 ± 0.310.255
Paraprevotella0.58 ± 0.390.42 ± 0.250.969
Muribaculaceae0.38 ± 0.180.95 ± 0.540.937
Haemophilus0.10 ± 0.050.26 ± 0.150.178
Bifidobacterium0.13 ± 0.050.50 ± 0.220.170
UBA18190.06 ± 0.020.10 ± 0.050.969
Lachnospiraceae UCG0100.16 ± 0.040.14 ± 0.050.692
Odoribacter0.15 ± 0.060.17 ± 0.100.937
Oscillibacter0.05 ± 0.010.13 ± 0.080.937
Bilophila0.13 ± 0.100.07 ± 0.030.783
Butyricimonas0.26 ± 0.120.15 ± 0.060.844
Species
Bacteroides uniformis7.72 ± 2.215.48 ± 3.860.158
Bacteroides vulgatus7.99 ± 2.238.17 ± 2.280.875
Bacteroides plebeius5.44 ± 3.844.16 ± 2.490.937
Lachnospiraceae NK4A136 group2.39 ± 1.723.78 ± 1.520.665
Bacteroides coprophilus3.10 ± 2.962.03 ± 1.321.000
Eubacterium hallii group11.47 ± 6.499.85 ± 5.060.754
Ruminococcus gnavus group0.84 ± 0.387.56 ± 4.520.812
Ruminococcus bicirculans1.81 ± 0.821.70 ± 0.800.906
Lactobacillus salivarius7.42 ± 7.294.51 ± 4.360.902
Prevotella stercorea3.32 ± 1.812.87 ± 1.500.475
Prevotella copri1.55 ± 1.3610.04 ± 4.890.475
Ruminococcus torques group2.35 ± 0.483.51 ± 1.050.194
Bacteroides stercoris4.60 ± 1.841.11 ± 0.590.194
Parabacteroides merdae1.30 ± 0.430.90 ± 0.300.255
Eubacterium eligens group1.28 ± 0.361.36 ± 0.630.433
Parabacteroides distasonis2.05 ± 1.141.41 ± 0.810.692
Ruminococcus gauvreauii group0.71 ± 0.361.75 ± 1.450.410
Alistipes shahii0.87 ± 0.450.46 ± 0.230.194
The Wilcoxon signed-rank test was used to determine the statistical significance. * Statistically significant if p ≤ 0.05.
Table 8. The differentiation of microbiome between the placebo and probiotic groups at the end of the study.
Table 8. The differentiation of microbiome between the placebo and probiotic groups at the end of the study.
TaxonomyPlacebo vs. Treatmentp-Value
Phylum
Proteobacteria−12.068.670.015 *
Bacteroidota−6.90−3.150.773
Firmicutes15.360.390.166
Actinobacteriota2.67−2.610.564
Fusobacteriota−0.89−0.050.817
Verrucomicrobiota1.84−3.150.384
Desulfobacterota−0.03−0.070.686
Genus
Escherichia-Shigella−10.458.200.024 *
Bacteroides0.551.400.326
Faecalibacterium0.242.900.273
Blautia−0.96−1.760.106
Prevotella−7.68−4.610.908
Agathobacter−1.330.720.050
Collinsella2.630.520.525
Subdoligranulum−1.290.530.133
Streptococcus3.340.750.225
Roseburia1.080.270.237
Phascolarctobacterium1.18−1.990.419
Romboutsia0.76−0.120.138
Klebsiella−0.91−0.830.977
Fusobacterium−0.83−0.050.817
Ruminococcus0.19−0.140.862
Dorea0.550.450.817
Akkermansia2.80−3.140.487
Lachnoclostridium0.50−0.200.453
UCG-002−0.550.920.260
Fusicatenibacter−0.330.020.204
Enterobacter−1.190.100.355
Veillonella4.700.880.106
Alistipes−0.28−0.251.000
CAG-3520.130.520.148
Enterobacteriaceae−1.53−0.160.419
Coprococcus0.070.400.977
Sutterella1.150.050.039 *
Anaerostipes0.230.300.470
Parabacteroides0.13−0.040.908
Holdemanella−0.16−0.500.386
Butyricicoccus0.510.430.326
Enterococcus2.47−0.020.182
Lachnospiraceae0.410.030.312
Paraprevotella−0.16−0.030.541
Bifidobacterium0.37−1.500.795
UBA18190.040.040.729
Lachnospiraceae UCG-010−0.02−0.090.931
Odoribacter0.03−0.150.309
Bilophila−0.060.000.862
Butyricimonas−0.11−0.120.271
Species
Bacteroides uniformis−2.24−5.380.908
Bacteroides vulgatus0.1816.470.033 *
Lachnospiraceae NK4A136 group1.391.521.000
Bacteroides stercoris−3.49−8.350.727
Parabacteroides distasonis−0.64−0.610.447
* Statistically significant if p ≤ 0.05.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chaiyasut, C.; Sivamaruthi, B.S.; Thangaleela, S.; Sisubalan, N.; Bharathi, M.; Khongtan, S.; Kesika, P.; Sirilun, S.; Choeisoongnern, T.; Peerajan, S.; et al. Influence of Lactobacillus rhamnosus Supplementation on the Glycaemic Index, Lipid Profile, and Microbiome of Healthy Elderly Subjects: A Preliminary Randomized Clinical Trial. Foods 2024, 13, 1293. https://0-doi-org.brum.beds.ac.uk/10.3390/foods13091293

AMA Style

Chaiyasut C, Sivamaruthi BS, Thangaleela S, Sisubalan N, Bharathi M, Khongtan S, Kesika P, Sirilun S, Choeisoongnern T, Peerajan S, et al. Influence of Lactobacillus rhamnosus Supplementation on the Glycaemic Index, Lipid Profile, and Microbiome of Healthy Elderly Subjects: A Preliminary Randomized Clinical Trial. Foods. 2024; 13(9):1293. https://0-doi-org.brum.beds.ac.uk/10.3390/foods13091293

Chicago/Turabian Style

Chaiyasut, Chaiyavat, Bhagavathi Sundaram Sivamaruthi, Subramanian Thangaleela, Natarajan Sisubalan, Muruganantham Bharathi, Suchanat Khongtan, Periyanaina Kesika, Sasithorn Sirilun, Thiwanya Choeisoongnern, Sartjin Peerajan, and et al. 2024. "Influence of Lactobacillus rhamnosus Supplementation on the Glycaemic Index, Lipid Profile, and Microbiome of Healthy Elderly Subjects: A Preliminary Randomized Clinical Trial" Foods 13, no. 9: 1293. https://0-doi-org.brum.beds.ac.uk/10.3390/foods13091293

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop