Next Article in Journal
MLLT11 Regulates Endometrial Stroma Cell Adhesion, Proliferation and Survival in Ectopic Lesions of Women with Advanced Endometriosis
Previous Article in Journal
Dissecting CYP1A2 Activation by Arylalkanoic Acid Prodrugs toward the Development of Anti-Inflammatory Agents
Previous Article in Special Issue
Unique Gut Microbiome Signatures among Adult Patients with Moderate to Severe Atopic Dermatitis in Southern Chinese
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Direct and Indirect Evidence of Effects of Bacteroides spp. on Obesity and Inflammation

1
Department of Rehabilitation Medicine of Korean Medicine, Ilsan Hospital of Dongguk University, Goyang 10326, Republic of Korea
2
Department of Rehabilitation Medicine of Korean Medicine, Bundang Hospital of Dongguk University, Seongnam 13601, Republic of Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(1), 438; https://doi.org/10.3390/ijms25010438
Submission received: 13 November 2023 / Revised: 22 December 2023 / Accepted: 23 December 2023 / Published: 28 December 2023

Abstract

:
Metabolic disorders present a significant public health challenge globally. The intricate relationship between the gut microbiome, particularly Bacteroides spp. (BAC), and obesity, including their specific metabolic functions, remains partly unresolved. This review consolidates current research on BAC’s role in obesity and lipid metabolism, with three objectives: (1) To summarize the gut microbiota’s impact on obesity; (2) To assess BAC’s efficacy in obesity intervention; (3) To explore BAC’s mechanisms in obesity and lipid metabolism management. This review critically examines the role of BAC in obesity, integrating findings from clinical and preclinical studies. We highlight the changes in BAC diversity and concentration following successful obesity treatment and discuss the notable differences in BAC characteristics among individuals with varying obesity levels. Furthermore, we review recent preclinical studies demonstrating the potential of BAC in ameliorating obesity and related inflammatory conditions, providing detailed insights into the methodologies of these in vivo experiments. Additionally, certain BAC-derived metabolites have been shown to be involved in the regulation of host lipid metabolism-related pathways. The enhanced TNF production by dendritic cells following BAC administration, in response to LPS, also positions BAC as a potential adjunctive therapy in obesity management.

1. Introduction

The human gastrointestinal tract is a dense microbial habitat hosting approximately 4 × 1013 bacteria, predominantly Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, and Verrucomicrobia [1,2]. Firmicutes and Bacteroidetes are dominant constituents of the gut microbiota [3]. Bacteroides (BAC), a prominent genus within the Bacteroidetes phylum, is characterized by anaerobic, bile-resistant, non-spore-forming, Gram-negative rods [4]. Significant taxonomic revisions in recent decades have expanded the genus to encompass over 20 distinct species [5,6]. Prominent members of BAC in the colon include B. vulgatus, B. distasonis, and B. thetaiotaomicron (at levels of 1010 per g dry weight of feces); species such as B. fragilis, B. ovatus, B. eggerthii, B. uniforms, and B. “B5-21” are less abundant but still present in substantial concentrations (109 per g dry weight of feces). BAC are integral to the ecological balance of the colon [7,8]. Their robust adaptability within the host and potential benefits have garnered significant attention from researchers, positioning them as valuable models or communities for studies in the realm of gut microbiology.
Recent studies have highlighted the potential of prebiotic and probiotic supplementation for weight management, with negligible adverse effects [9]. For instance, supplementation with Lactobacilli (phylum Firmicutes) has exhibited favorable effects on blood glucose levels, lipid profiles, weight, and body fat in overweight adults and those with obesity, as corroborated by several clinical studies [10,11]. Bifidobacterium spp. has also effectively reduced fat and body weight in preclinical and clinical studies of obesity and type 2 diabetes [12,13]. Furthermore, BAC have been shown to activate fat oxidation in adipose tissue via the bile acid-TGR5-PPARα axis, thus increasing energy consumption [14]. A previous review also emphasized the ability of BAC to mitigate various diseases, such as Helicobacter hepaticus infection, oxazolone-induced experimental colitis, Vibrio parahaemolyticus infection, antibiotic-associated diarrhea, high-fat diet-induced cardiovascular disease, and cancer [15]. BAC-derived compounds and metabolites interact with various immune cells and contribute to immune response activation. Notably, Polysaccharide A (PSA), derived from the human commensal B. fragilis NCTC9343, is a symbiotic factor that stimulates immune development in mammalian hosts, stimulates dendritic cell maturation and T-cell proliferation, in addition to possessing abscess-inducing properties [16,17].
BAC engage in both competitive and symbiotic interactions with adjacent bacterial species, thereby markedly influencing gut microbiota dynamics. BAC employ two distinct strategies for polysaccharide breakdown: they either prevent other bacteria from accessing the products of polysaccharide degradation or make these products available to other species. For example, B. thetaiotaomicron exhibits limited degradation of the yeast cell wall polysaccharide α-mannan outside the cell, transporting the relatively large α-mannan polysaccharides into the periplasm for further breakdown into manno-oligosaccharides and mannose [18]. This strategy discourages the utilization of partially degraded α-mannan by bacterial species lacking the requisite transporter for this unique polysaccharide, thereby favoring the proliferation of species equipped to utilize it [19]. However, previous research records show that BAC is identical to some commercial conventional probiotics [20,21,22]. The health-promoting properties also depend on the strain. Although the PSA produced by non-enterotoxigenic B. fragilis attenuates infections and aids colonization, it also serves as a symbiont for bacterial growth [23,24]. Despite the diversity of research directions, there is a common thread among numerous previous studies that have focused on BAC, with most findings supporting their beneficial effects against obesity, metabolic disorders, and inflammation.
Recent investigations have elucidated that BAC exhibited anti-inflammatory properties within the body [25]. Notably, the interplay between obesity and inflammation is well-documented, with both conditions often presenting concurrently [26]. Despite these findings, the direct impact of BAC in mitigating obesity in humans has not been thoroughly explored, underscoring the need for comprehensive research in this area.
In our analysis, we primarily referenced both direct and indirect experiments in previous studies that explored the effects of BAC on weight loss, encompassing animal models and human clinical trials. Given the absence of clinical trials that directly elucidate the mechanisms whereby BAC supplementation modulates obesity in humans, we aimed to extrapolate the potential mechanisms by summarizing the outcomes of preclinical and clinical studies associated with increased concentration of BAC in the gut. Our review included an exploration of the efficacy and potential mechanisms of BAC related to its anti-obesity and lipid-reducing properties, encompassing the impact of BAC on the structure and diversity of the host gut microbiota as well as the potential involvement of BAC-derived metabolites in the regulation of host metabolic signaling pathways. This analysis is pivotal for advancing targeted obesity interventions.

2. Materials and Methods

2.1. Data Source and Search Strategy

We conducted a comprehensive literature search in November 2022 using the PubMed, Google Scholar, Cochrane, Scopus, and Web of Science databases within a restricted time period between 1 January 2000 and 30 November 2022. We employed specific search strings combining terms and Boolean operators, such as “Bacteroides AND obesity AND trial”. These terms were adapted for each literature database and combined with database-specific filters for human clinical trials and preclinical trials in animals.

2.2. Inclusion and Exclusion Criteria

We searched for studies demonstrating that BAC effectively treated obesity, metabolic diseases, and inflammation. Because of the lack of studies that focused on the efficacy of BAC for obesity, the searched articles included both clinical trials and preclinical studies using animal models. However, the screening and selection were conducted separately, according to the below criteria. There were no language restrictions, and only the literature with abstracts was reviewed. In addition, studies with incorrect BAC descriptions or those without statistical analyses were excluded.
The inclusion criteria for clinical trials with indirect evidence were as follows: (1) human studies wherein participants were clearly defined as being overweight or obese at baseline; (2) participants who were overweight or obese and had a significant reduction in body weight, body fat, and other relevant body metrics after treatment; and (3) the richness, diversity, abundance, and diversity of BAC changed as the body metrics gradually normalized. The exclusion criteria were as follows: (1) unavailability of the full text; (2) no weight loss with treatment; (3) animal models; and (4) failure to provide results related to BAC. For the article type, it was not limited to randomized controlled trials. Observational studies, which compared the obese characteristics to healthy subjects’ characteristics, were also included. In the case of intervention, any reasonable treatments for obesity were included, and there was no limitation for the comparator. The before–after studies for a single group were also included; however, reviews, case series, or case reports were excluded.
The inclusion criteria for preclinical trials with direct certification were as follows: (1) animal model; (2) involvement of a control group; and (3) treatment or intervention with BAC supplementation. The following exclusion criteria were used: (1) not an original paper; (2) lack of comparison; and (3) not using animal models such as human studies; no direct results demonstrated that BAC exhibited anti-obesity effects. The key data for inclusion are summarized in Figure 1.

2.3. Data Extraction and Data Analysis

In the initial phase of our research, we amalgamated the search results sourced from various databases to compile a comprehensive dataset. Subsequently, this dataset underwent a rigorous process of de-duplication, ensuring the removal of any redundant entries. The task of evaluating and determining the relevance of the collated data was undertaken collaboratively by a trio of researchers. In cases where differing viewpoints emerged, these discrepancies were diligently addressed through in-depth discussions, leading to a consensus. This methodical approach not only ensured the accuracy and reliability of our data selection but also underscored the collaborative integrity of our research methodology.
From each selected publication, data extraction was performed separately based on the characteristics of the studies.
For the clinical trials used for indirect proof, the following data were extracted: information about sex; age; physical data related to obesity; and the changes caused by BAC were obtained for human participants. For the preclinical studies, information on study design, interventions, comparisons, strains and doses, intervention periods, and primary outcomes were extracted.
The primary outcome indicator for indirect evidence in clinical trials was a change in the abundance of BAC, whereas the primary outcome indicators for direct evidence in preclinical studies were body weight, liver weight, body fat, blood lipids, liver injury, steatosis, hepatic lipid metabolism, and insulin levels.
For studies utilizing BAC in combination therapies, a sub-analysis was conducted to isolate the effects attributable to BAC alone based on available data. Studies that indicated improved health markers without weight loss were subjected to a separate analytical process to determine the potential role of BAC in metabolic health.
Given the established anti-inflammatory properties of BAC in human subjects, this study aimed to explore the broader implications of BAC on obesity management. The data analysis is, therefore, organized into four distinct sections: The first part comprises studies that indirectly demonstrated changes in BAC after treatment with probiotics, herbs, dietary supplements, and synthetic probiotic supplements, either alone or in combination with other treatments, resulting in weight loss in overweight human participants and patients with obesity using medical subject terms (Bacteroides spp.). The second section explores the differences in the abundance of BAC in the gut microbiota of individuals with obesity and healthy individuals using medical subject headings (Bacteroides spp., gut microbiota, clinical trials, overweight, obesity, and 16S rRNA gene sequencing). Because there are no clinical trials of BAC directly treating obesity, the third part comprises information regarding preclinical studies demonstrating that BAC can treat obesity using medical subject headings (Bacteroides spp., mice, preclinical studies, overweight, obesity, weight loss). The fourth section comprises preclinical wherein BAC were found to directly affect inflammation (Bacteroides spp., mice, preclinical studies, inflammation).

2.4. Quality Assessment

We adapted the Downs and Black checklist to suit the nuances of the reviewed studies, focusing on parameters critical for assessing the reliability and applicability of obesity and microbe-related research [27]. The checklist contained 27 items across the following five domains: reporting; external validity; internal validity (bias); internal validity (confounding); and efficacy. After assessment, the studies were categorized according to their scores as excellent (25–27), good (20–24), fair (15–19), or poor (<14). If no patients were lost to follow-up, the ninth analyzed indicator was deleted, and an extra point was added to the scoring results. The quality of the included studies was presented using Review Manager version 5.3. (Copenhagen, the Nordic Cochrane Centre, the Cochrane Collaboration, 2014).

3. Results

3.1. Study Description

The primary objective of our study was to investigate the impact of BAC on human obesity. Our research commenced with a systematic search for clinical trials specifically addressing the direct treatment of obesity using BAC strains. Regrettably, no relevant clinical trials were identified. However, we identified pertinent preclinical trials and collected clinical trials for indirect evidence. We curated clinical trials where BAC strains indirectly influenced obesity treatment. Of the 31 relevant studies, 19 reported that interventions leading to weight loss in human participants resulted in increased BAC abundance. These findings imply a correlation between BAC and weight management, although the directionality of this relationship requires further investigation. Interestingly, two studies documented weight loss concurrent with a decrease in BAC abundance, suggesting that other unidentified factors might influence the interaction between the gut microbiota and weight regulation or that different BAC strains might have varying effects. The ten comparative microbiota analyses across individuals with different demographic backgrounds and body compositions further improved our understanding of the role of BAC. Although they did not provide direct evidence of weight loss, they contributed significant insights into the natural variance of BAC in the human gut and its correlation with obesity, underscoring the need for personalized approaches to microbiota-targeted treatments. The key data for inclusion are summarized in Table 1, Table 2 and Table 3.
The direct evidence analysis identified 142 potentially relevant articles, of which 12 met the inclusion criteria. Six of these studies, wherein BAC were directly administered via gavage, reported a consistent decrease in body weight, suggesting the potential anti-obesity effect of these bacterial strains. The more details are presented in Table 4.
The data summarized in Table 5 from relevant studies indicate that BAC, in combination with various pharmacological agents, contribute to reducing the inflammatory response in experimental animal models. This section always reflects the mechanisms by which obesity, inflammation, and BAC may be related. In this section, only clinical weight loss trials after pharmacological interventions evaluate the specific drug of the intervention, the number of patients involved in the problem, the type of intervention, and the duration. The second part of the review considers only articles reporting fat reduction upon direct gavage of BAC strains were considered, and the types of interventions considered included specific strains, types of experimental animals, types of interventions, and duration of interventions.

3.2. Indirect Evidence of the Anti-Obesity Effect of BAC from Clinical Studies

Among the 18 included clinical trials, the shortest duration of intervention was 29 days, and the longest was 16 weeks. There were 13 cases wherein weight, waist circumference (WC), or body mass index (BMI) achieved significant reduction after different interventions, such as probiotics, herbs, and low-carbohydrate diets [28,29,30,31,32,33,34,35,36,37,38,39,40]; additionally, the richness and diversity of BAC strains increased to varying degrees with a significant reduction in weight [28,30,31,32,34,36,37,38,39,40,41,42,43,44]. In two studies, although no significant changes in obesity-related parameters were induced, both studies showed a decreasing trend; the intervention in both cases increased the abundance of BAC spp. [29,33].
A clinical trial conducted on Italian patients with obesity, wherein most of the included patients had metabolic dysfunction, showed a statistically significant reduction in body weight and adiposity after the administration of a healthy diet based on caloric intake, macronutrient composition, and fiber intake. There was a statistically significant increase in the abundance of BAC spp., especially B. uniformis (p = 0.005) [35]. In another study, nine healthy volunteers were selected to fast for approximately 17 h per day for 29 days; at the end of the trial, there were no significant changes in obesity-related values, but a significant increase in the abundance of both A. muciniphila (p = 0.0039) and B. fragilis (p = 0.0078) was observed [45]. Notably, an increase in the abundance of BAC spp. was reported in all these studies (Table 1).
However, a few clinical trials have yielded conflicting results in that obesity improved while BAC spp. abundance decreased. Furthermore, two clinical trials in children and women with obesity showed a decrease in B. vulgatus abundance after the administration of probiotics [46,47] (Table 2).
Table 1. Indirect evidence of the anti-obesity effect of BAC.
Table 1. Indirect evidence of the anti-obesity effect of BAC.
Research PurposeNumbers
of Patients
Randomized/
Analyzed
Intervention
Type (Regimen)
Treatment
Period
Main Outcome MeasureMain
Conclusion
Reference
Exploration of the anti-obesity efficacy of steamed Rehmannia glutinosa root.Middle-aged female participants with obesity (40–65 years)
12/12
Nor group (n = 25)
LCD group (n = 26)
8 weeksBAC↑
Waist circumference
Roseburia
Dorea
BAC levels are negatively correlated with waist circumference.[28]
Assessment of metabolic markers related to obesity post-Schisandra chinensis fruit administrationParticipants with obesity
40/28
SCF group (n = 13)
Placebo group (n = 15)
12 weeksBAC↑
Akkermansia
Roseburia
Prevotella
Bifidobacterium
Ruminococcus
Relationship with BAC:
Fat mass
BAC levels are negatively correlated with fat mass and aspartate aminotransferase and/or alanine aminotransferase levels.[29]
Examination of the correlation between gut microbial modulation and plasma lipopolysaccharide-binding protein (LBP) levels.Participants who were overweight and obese
49/49
Pomegranate extract (PE)
Placebo
3 weeksBAC↑
Plasma LBP
High-sensitivity C-reactive protein
Faecalibacterium
Butyricicoccus
Odoribacter
Butyricimonas
BAC levels are positively correlated with the levels of other beneficial bacteria and health indicators.[30]
Determination of the effects of metformin on anthropometry and gut microbiota in non-diabetic women with obesity under a low-calorie regimen.Women with obesity
46/36
Metformin + LCD group (n = 20)
Placebo + LCD group (n = 16)
8 weeksBAC↑
BMI
Insulin concentration
Escherichia/Shigella abundance
Roseburia
Blautia
Butyrivibrio
BAC levels are negatively correlated with BMI and insulin concentrations.[31]
Evaluation of the role of daily avocado intake within a hypocaloric diet on weight management, inflammation biomarkers, and gut microbiota diversity.Healthy men and women who were overweight/obese
63/51
Avocado daily intake (n = 24)
hypocaloric diet (n = 27)
12 weeksBAC↑
BW
BMI
Total body fat
Visceral adipose tissue
Serum glucose levels
Serum hepatic growth factor levels
IL-1β
C-reactive protein
Clostridium↑
Methanosphaera
Candidatus Soleaferrea
BAC levels are negatively correlated with BW.[32]
Assessment of the efficacy of probiotic and synbiotic supplementation on intestinal microbiota modulation in adults with prediabetes.Patients with prediabetes
120/85
Probiotic group (n = 27)
Synbiotic group (n = 29)
Placebo group (n = 28)
6 weeksProbiotics:
B. fragilis/E. coli
Firmicutes/Bacteroidetes
BAC levels are positively correlated with the levels of other beneficial bacteria.[33]
Assessment of a low-fat vegan diet’s influence on gut microbiota and its correlation with weight and insulin resistance in an overweight cohort.Participants
Age: 25~75 years old
BMI: 28~40 kg/m2
168/115
Vegan group (n = 84)
Control group (n = 84)
16 weeksVegan group:
B. fragilis
BW
Fat mass
Visceral fat
PREDIM
Faecalibacterium prausnitzii
BAC levels are negatively correlated with BW.[34]
Assessment of the influence of a calorie-restricted Mediterranean diet on the gut microbial composition in individuals who are overweight and obese.Patients with obesity
23/23
NW
OB
3 weeksB. cellulosilyticus
B. uniformis
BW
Waist circumference
Body mass index
Fat mass
Daily caloric intake
Prevotella stercorea
BAC levels are negatively correlated with BW.[35]
Investigation of comparative gut microbiota profiles in individuals with dyslipidemia who were overweight and obese: Responses to orlistat and ezetimibe interventions.174 Volunteers (96 volunteers with dyslipidemia who were overweight and obese)
174/116
Control group (n = 31)
Patient group (n = 27)
Orlistat group (n = 32)
Ezetimibe group (n = 26)
12 weeksBAC↑
Waist circumference
TG
FBG
Actinomyces
BAC levels are negatively correlated with waist circumference.[36]
Evaluation of the prebiotic impact of omega-3 supplementation on the gut microbiome.Patients with obesity
69/69
20 g of inulin fiber
500 mg of omega-3 supplements daily
6 weeksBAC↑
Butyric acid
Iso-butyric acid
Iso-valeric acid
Total fatty acids
Total omega-3/total fatty acids
Coprococcus spp.↑
BAC levels are positively correlated with increased levels of SCFAs.[37]
Investigation of predictive alterations in gut microbiota post-short-term low-carbohydrate dietary intervention in patients with obesity.Participants who were overweight or obese
51/51
ND group (n = 25)
LCD group (n = 26)
12 weeksBAC↑
BW
Ruminococcaceae Oscillospira
Odoribacteraceae Butyricimonas
Porphyromonadaceae Parabacteroides
BAC levels are negatively correlated with BW.[38]
Investigation of the combined effects of a Medical Food Therapy plant-based diet and intermittent energy restriction on glycemic parameters.T2D patients without obesity
39/39
20 CMNT group (received a low-calorie human CMNT diet for five consecutive days, 10 days of habitual eating per cycle for 90 days)
19 Control group (continued normal diet)
8 weeksBAC↑
Hemoglobin A1C
BMI
FBG
Postprandial
blood glucose levels
Medication usage
Parabacteroides
Roseburia
BAC levels are negatively correlated with BMI.[39]
Comparative analysis of the effects of traditional vs. commercial kochujang on obesity metrics in adults who are overweight.Participants
62/48
High-dose Kochujang (n = 19)
Low-dose Kochujang (n = 18)
Commercial Kochujang (CK; n = 17)
6 weeksHigh-dose Kochujang and Low-dose Kochujang groups
BAC↑
TC
Low-density lipoprotein cholesterol
High-density lipoprotein cholesterol
Triglyceride levels
Waist circumference
Lactobacillus spp.↑
Bifidobacterium spp.↑
Lactococcus lactis
Enterococcus faecium
BAC levels are positively correlated with positive health knots, such as reduced waist circumference.[40]
Evaluation of the effect of Lactobacillus salivarius Ls-33 on the fecal microbiota composition in adolescents with obesity.Adolescents with obesity
51/50
1 × 1010 CFU L. salivarius Ls-33ATCCSD5208 (n = 27)
Placebo: maltodextrin (n = 23)
12 weeksB. fragilis
SCFAs↑
E. rectale
BAC levels were positively correlated with the increase in SCFA levels.[41]
Detailed analysis of propionate-induced alterations in glucose homeostasis, gut microbiota, plasma metabolome, and immune responses.Non-diabetic adults who were overweight and obese
14/12
Cellulose
Inulin
IPE
42 days ×3Inulin and IPE:
B. uniformis
B. caccae
B. xylanisolvens
Insulin resistance
IgG
IL-8
BAC levels are positively correlated with insulin sensitivity and inversely correlated with inflammatory indicators.[42]
Microbiota shifts in individuals with obesity post very low-energy dietary intervention.Finnish participants with obesity
16/16
Participants with obesity (BMI > 30 kg/m2; six men and ten women)12 weeksBAC↑BAC levels positively correlate with health indicators.[43]
Examination of the interplay between Korean red ginseng’s effects on metabolic syndrome and gut microbiota alterations.Patients with metabolic syndrome
60/50
KRG group (n = 25)
placebo group (n = 25)
8 weeksBAC↑
BMI
Prevotella
Ruminococcaceae
Sutterella
Odobacter
BAC levels are negatively correlated with the BMI and insulin concentrations.[44]
Assessment of the impact of intermittent fasting on the composition of gut microbiome.Health volunteers
9/9
Female (n = 7)
Male (n = 2)
B. fragilis
A. muciniphila
Serum fasting glucose levels
TC
BAC levels are positively correlated with other beneficial bacteria levels.[45]
The results presented in the table demonstrate statistical significance. IPE: Inulin-propionate ester; TC: Total cholesterol; BMI: Body Mass Index; T2D: Type 2 diabetes; PREDIM: Predicted clamp-derived insulin sensitivity index from a standard meal test; IL-1β: Interleukin 1 beta; FBG: Fasting blood glucose; LBP: Lipopolysaccharide-binding protein; IL-8: Interleukin-8; IgG: Immunoglobulin G; SCFAs: Short-chain fatty acids; BW: Body weight. An upward arrow (↑) signifies an increase in the abundance of the specified gut microbe, while a downward arrow (↓) indicates a decrease in the abundance of the specified gut microbe.
Table 2. Indirect evidence that BAC do not have an anti-obesity effect.
Table 2. Indirect evidence that BAC do not have an anti-obesity effect.
Research PurposeNumbers of Patients
Randomized/Analyzed
Intervention
Type (Regimen)
Treatment
Period
Main Outcome MeasureMain
Conclusion
Reference
To investigate the effects of prebiotic supplementation on body composition, inflammatory markers, bile acids in stool samples, and gut microbiota composition in overweight or children with obesity.Children who were overweight or obese
42/42
Prebiotic group (n = 22)
Placebo (n = 20)
16 weeksB. vulgatus
BW
BMI
Percent body fat
Percent trunk fat
Bifidobacterium
IL-6
BAC levels are negatively correlated with the beneficial effects of prebiotic supplementation.[46]
To investigate the impact of dietary inulin-type fructans (ITF prebiotics) on host metabolism through modulation of the gut microbiota in women with obesity.Females with obesity
30/30
Inulin/oligofructose 50/50 mix; (n = 15)
placebo (maltodextrin; n = 15)
12 weeksB. intestinalis
B. vulgatus
Bifidobacterium
Faecalibacterium
Rausnitzii
Propionibacterium
Serum lipopolysaccharide levels
BAC levels are inversely correlated with metabolic improvement due to ITF probiotics.[47]
BMI: Body mass index; IL-6: Interleukin 6. An upward arrow (↑) signifies an increase in the abundance of the specified gut microbe, while a downward arrow (↓) indicates a decrease in the abundance of the specified gut microbe.

3.3. Comparison of Gut Microbiota between Individuals with Obesity and Healthy Individuals Demonstrates an Association between BAC and Obesity

To further demonstrate the relationship between BAC and obesity, we included nine clinical trials that analyzed the differences in gut microbiota between populations with and without obesity. Seven of these were studies on adults; two were on children and adolescents; seven studies, while not directly stating their significance, reported lower BAC abundance and diversity in obese populations than in non-obese populations, and three studies directly indicated significance.
The results of a survey of the gut microbiota of 46 pairs of twins and their mothers were revealed. The obese gut microbiota was significantly different (p = 0.003) and less diverse than the lean core gut microbiome [48]. In a study by Kasai et al., (2015), a comparison of gut microbiota composition between individuals with and without obesity in a Japanese population showed that B. faecichinchillae and B. thetaiotaomicron were virtually undetectable in the feces of individuals with obesity and that the proportion of these BAC was significantly higher in individuals without obesity [49]. Additional details are provided in Table 3.
Table 3. Comparison of gut microbiota between individuals with obesity and healthy individuals demonstrating the indirect metabolism-modulation function of BAC.
Table 3. Comparison of gut microbiota between individuals with obesity and healthy individuals demonstrating the indirect metabolism-modulation function of BAC.
Research PurposeMain Outcome MeasureResults (Relationship with BAC)Main ConclusionReference
Objective: To characterize the gut microbiota and short-chain fatty acids (SCFAs) in participants with obesity from Xinjiang, China.
Summary: This study highlights regional variations in gut microbial composition related to obesity.
16S rRNA sequencing and microbial diversity and community analyses
68 individuals with obesity
31 controls
Individuals with obesity↑
B. fragilis
Gemmiger
Dialister
Megamonas
Anaerostipe
Blautia
Gemmiger
Bifidobacterium
Prevotella
The gut microbiota profile of obese patients was characterized by enrichment of Lactobacillus and the reduction in the diversity and the depletion of Actinobacteria, BAC, Bifidobacterium, and B. fragilis.[36]
Objective: To identify core gut microbiota in obese and lean twins and their association with obesity.
Summary: This study investigated twins discordant in obesity to reveal key microbial players associated with obesity.
16S rRNA gene sequencing
31 monozygotic twin pairs
23 dizygotic twin pairs and their mothers (n = 46)
Participants with obesity:
BAC↓
Actinobacteria
Carbohydrate, lipid, and amino acid metabolism↓
The deviations from a core microbiome at function level are associated with different physiological states.[48]
FObjective: To determine the relationship between gut microbiota composition and obesity in a Japanese population.
Summary: This study explores unique microbial patterns associated with obesity in the Japanese demographic.
T-RFLP analysis and 16S rRNA sequencing
23 participants without obesity
BMI < 20 kg/m2
33 participants with obesity
BMI ≥ 25 kg/m2
Participants without obesity:
B. faecichinchillae
B. thetaiotaomicron
Blautia wexlerae
Clostridium bolteae
Flavonifractor plautii
Gut microbial properties differ between obese and non-obese subjects, suggesting that gut microbiota composition is related to obesity.[49]
Objective: To elucidate the gut microbial composition in normal adolescents and those with obesity.
Summary: This research uncovered microbial signatures specific to obesity during adolescence.
16S rRNA gene sequencing
67 adolescents with obesity (BMI ≥ 30 kg/m2 or ≥ 99th BMI percentile)
67 normal adolescents (BMI < 25 kg/m2 or <85th BMI percentile)
Adolescents with obesity
BAC↓
Prevotella↓
TG↑
TC↑
hs-CRP↑
A significant association between the composition of several bacterial taxa and childhood obesity was revealed.[50]
Objective: To understand the mechanisms of obesity development by comparing the gut microbiota of children with obesity with that of healthy controls.
Summary: This research highlights distinct gut microbial features associated with obesity.
16S rRNA gene sequencing, enterotypes
Children with obesity aged 3 to 18 years (n = 87)
healthy children aged 3 to 18 years (n = 56)
Children with obesity:
BAC↓
Firmicutes/Bacteroidetes
Enterococcus
Blautia
Sutterella
Klebsiella
Parabacteroides
Anaerotruncus
Coprobacillus
BAC are associated with weight loss, and are recommended as a dietary prebiotic and probiotic supplement as an adjunct to obesity treatment.[51]
Objective: To identify microbial and metabolic factors associated with obesity through MWAS and serum metabolomics profiling.
Summary: This comprehensive approach uncovers microbial and metabolic contributors to obesity in young Chinese individuals.
shotgun sequencing
72 individuals with obesity (body mass index (BMI), 36.78 ± 4.46 kg/m2; age, 23.6 ± 3.7 years)
79 controls (BMI, 20.2 ± 1.3 kg/m2; age, 23.2 ± 1.8 years)
Controls
B. thetaiotaomicron
B. uniformis
B. xylanisolvens
B. ovatus
Akkermansia muciniphila
Fecalibacterium prausnitzii
The abundance and diversity of BAC were significantly lower in obese individuals than in normal individuals.[52]
Objective: To study gut microbiota alterations specific to childhood obesity.
Summary: This research reveals microbial markers and shifts associated with obesity during childhood.
16S rRNA gene sequencing
Children with obesity aged 6 to 16 years (n = 42)
Children of normal weight (n = 36)
Children with obesity:
B. vulgatus
B. stercoris
Firmicutes/Bacteroidetes↑ (p < 0.0001)
Reduced numbers of BAC in the gut may be linked to childhood obesity.[53]
Objective: To identify unique gut microbiota characteristics in patients who were overweight/obese and compare them with those in normal weight controls in Sardinia.
Summary: This research uncovers microbial signatures specific to obesity in this geographical region.
16S rRNA gene sequencing
Patients who were overweight/obese (n = 46)
normal weight participants (n = 46)
Patients who were overweight/obese:
BAC↓
Flavobacteriaceae
Porphyromonadaceae
Sphingobacteriaceae
Flavobacterium
Rikenella spp.↓
Pedobacter spp.↓
Parabacteroides spp.↓
Body fatness and waist circumference were negatively correlated with BAC.[54]
Objective: To explore interactions between the gut microbiome, metabolites, and brain network metrics.
Summary: This investigation sheds light on how the gut microbiota and its metabolic products influence brain function, potentially linking gut–brain interactions to obesity.
16S rRNA gene sequencing
287 participants with obesity and without obesity (male n = 99, female n = 198)
Participants with obesity:
BAC↓
Microbial diversity↓
Prevotella/BAC↑
Fecal tryptophan↓
Observed significant taxonomic changes at the genus level associated with obesity.
BAC↓ (p < 0.001)
[55]
The results presented in the table demonstrate statistical significance. BMI: Body mass index; hs-CRP: High-sensitivity C-reactive protein; SCFAs: Short-chain fatty acids; TC: Total cholesterol; TG: Total triglyceride; T-RFLP: Terminal restriction fragment length polymorphism. An upward arrow (↑) signifies an increase in the abundance of the specified gut microbe, while a downward arrow (↓) indicates a decrease in the abundance of the specified gut microbe.

3.4. In Vivo Direct Evidence of the Anti-Obesity Effects of BAC

Animal experiments published in 2021 had a high frequency of publications; five were conducted on C57BL/6J mice, and only one on rats; >90% of the studies used male animals. The duration of the intervention ranged from 7 to 18 weeks. Consistent evidence from various animal studies indicates that certain BAC strains can significantly reduce key obesity-related metrics, including body weight, fat accumulation, and adverse serum profiles.
The six preclinical studies involved B. uniformis CECT 7771, B. uniformis CBA7346, B. vulgatus SNUG 40005, B. acidifaciens JCM10556, B. dorei DSM17855, and B. vulgatus ATCC8482. Cano et al., and Agusti et al., compared the ability of different BAC strains to produce cytokines. Notably, administration of B. uniformis CECT 7771 was found to significantly reduce body weight gain in HFD-fed mice (p < 0.05) and caloric intake during the binge phase (p < 0.001) [56,57]. In another study, administration of B. vulgatus SNUG 40,005 for 18 weeks significantly reduced the body weights of obese mice (p = 0.004) [58]. In the study by Yang et al., (2017), oral administration of B. acidifaciens resulted in a significant reduction in body weight (p < 0.001) and adiposity (p < 0.05) in Atg7 ΔCD11c mice [14]. Administration of B. uniformis CBA7346 significantly reduced body and liver weights (p < 0.05) [59]. Yoshida et al., (2021) found that combined gavage of B. dorei DSM17855 and B. vulgatus ATCC8482 significantly suppressed obesity and weight gain [60]. Further details are listed in Table 4.
Table 4. In vivo evidence of anti-obesity effect of BAC.
Table 4. In vivo evidence of anti-obesity effect of BAC.
Bacteroides StrainSourceSubjectIntervention
Type (Regimen)
Treatment
Period
Main
Outcome
Measure
Main
Conclusion
Reference
B. acidifaciens JCM10556Human fecesC57BL/6, CD11c-Cre, Villine-Cre, Atg7f/f and LysM-Cre miceHFD + PBS
HFD + 5 × 109 cfu B. acidifaciens JCM10556
10 weeksBW and fat mass
Insulin
Bile acid-TGR5-PPARα axis
Dipeptidyl peptidase-4
Glucagon-like peptide-1
B. acidifaciens JCM10556 helps prevent metabolic disorders such as diabetes and obesity.[14]
B. uniformis CECT7771Infant stoolMale C57BL/6J miceNor
HFD
Nor + 5.0 × 108 cfu B. uniformis CECT 7771
HFD + 5.0 × 108 cfu B. uniformis CECT 7771
7 weeksBW
Adipose tissue weight
Hepatic steatosis
Macrophage functionality
Dendritic cell functionality
B. uniformis CECT7771 improves metabolic and immune dysfunction associated with intestinal ecological dysregulation.[56]
B. uniformis CECT7771Infant stoolAdult male Wistar Kyoto rats (170–200 g)SD
IF
IF + 1 × 108 cfu B. uniformis CECT 7771
18 daysCaloric intake
Anxiety-like behavior
Dopamine
B. uniformis CECT 7771 helps control compulsive eating by affecting brain reward systems.[57]
B. uniformis CBA7346Human fecesMale C57BL/6J miceNor + PBS
Nor + 1 × 106 cfu B. uniformis
HFD + PBS
HFD + 1 × 106 cfu B. uniformis
12 weeksBody and liver weights
Blood lipids
Liver injury and steatosis
Hepatic lipid metabolism
B. uniformis CBA7346 mitigates HFD-induced NAFLD, thereby modulating LPS release, lipid-related proteins, and insulin sensitivity.[59]
B. dorei DSM17855 and B. vulgatus ATCC8482Human fecesMale C57BL/6J miceNor
HFD
HFD + 2.5 × 109 cfu B. dorei + B. vulgatus mix
12 weeksBW
Blood glucose
BAT weight
UCP1
Macrophage number
B. dorei DSM17855 and B. vulgatus ATCC8482 enhance branched-chain amino acid catabolism in brown adipose tissue[60]
The results presented in the table demonstrate statistical significance. UCP1: Uncoupling protein 1; BAT: Brown adipose tissue; BW: Body weight.

3.5. Studies on Direct Anti-Inflammatory Therapy with BAC

Recent studies have shown that white adipose tissue in patients with obesity develops low-grade chronic inflammation, which induces insulin resistance [61]. Due to the close relationship between obesity and inflammation, we included six preclinical studies in which BAC were directly gavaged. All experiments were conducted with mice; the minimum duration of the experiment was five days, and the maximum was 21 days. Only three experiments clearly labeled the gavaged BAC concentration, which ranged from 1 × 109 CFU/mL to 7 × 109 CFU/mL. In these studies, BAC significantly upregulated the expression of anti-inflammatory cytokines and downregulated the expression of pro-inflammatory cytokines. These studies suggest that BAC have therapeutic potential for preventing intestinal inflammatory diseases in animal models (Table 5).
Table 5. Study of direct anti-inflammation treatments with BAC.
Table 5. Study of direct anti-inflammation treatments with BAC.
Bacteroides StrainSourceSubjectIntervention
Type (Regimen)
Treatment
Period
Main
Outcome
Measure
Main
Conclusion
Reference
B. fragilis NCTC9343Human fecesMale C57BL/6J miceDSS
DSS + 5 × 109~7 × 109 CFU B. fragilis NCTC9343
8 daysIL-1β
CCR5
Number of colonic tumors
TLR2 signaling
This study highlights B. fragilis’ protective role against weight loss, colonic histopathological changes, and inflammation in colitis-associated colon cancer.[62]
B. thetaiotaomicron DSM 2079Human fecesFemale and male C57BL/6J miceControl
DSS
DSS + 3 × 1010 CFU B. thetaiotaomicron DSM 2079
8–14 daysinflammation
BW
Colon length
This study demonstrates that B. ovatus monotherapy is a consistent and effective approach for colitis management, surpassing traditional FMT.[63]
B. fragilis (HCK-B3, ATCC25285) and B. ovatus (ELH-B2, JCM5824)Human fecesFemale C57BL/6J mice1 × 109 CFU B. fragilis HCK
1 × 109 CFU B. fragilis 25285
1 × 109 CFU B. ovatus ELH
1 × 109 CFU B. ovatus JCM5824
5 daysTNF
IL-10
FITC-Dextran
NF-κB
This research underscores the promise of BAC as a therapeutic agent for intestinal inflammatory diseases.[64]
B. ovatus ATCC 8483Human fecesMale C57BL/6J miceB. ovatus
B. thetaiotaomicron
B. vulgatus
B. ovatus + B. thetaiotaomicron + B. vulgatus mix
9 daysIncreased proliferation of epithelial cells
Hyperplastic crypts
inflammation
This study demonstrates that B. ovatus monotherapy is a consistent and effective approach for colitis management, surpassing traditional FMT.[65]
B. fragilis NCTC9343Human fecesWT (129S6/SvEvTac) and Rag2−/− (129S6/SvEvTac-Rag2tm1Fwa) miceWT + B. fragilis
WT + PSA
Rag + PSA
WT + PBS
Rag + PBS
21 daysT-cell populations
IL-10
IFNγ
This research underscores the protective role of B. fragilis NCTC9343 against nervous system inflammatory diseases.[66]
B. vulgatus FJS7K1Human fecesMale C57BL/6J miceControl
LPS
LPS + B. vulgatus 5K1
LPS + B. vulgatus 7K1
LPS + B. vulgatus 11B4
LPS + B. vulgatus 51K1
5 daysNumber of Treg cells
Intestinal epithelial integrity
IL-6
IL-10
TNF
SCFAs
This study highlights B. vulgatus FTJS7K1 as a potential agent for mitigating acute inflammation and intestinal injury through microbial community modulation and cytokine regulation.[67]
The results presented in the table demonstrate statistical significance. SCFAs: Short-chain fatty acids; IL-6: Interleukin-6; TNF: Tumor necrosis factor α; IL-10: Interleukin-10; IFNγ: Interferon-γ; FITC-Dextran: Fluorescein isothiocyanate; NF-κB: Nuclear factor κB; TLR2: Toll-like receptor 2; CCR5: C-C chemokine receptor 5; LPS: Lipopolysaccharide; PSA: Polysaccharide A; WT: Wild type.

3.6. Quality Assessment and Scores

After a quality assessment of the selected studies using the modified Downs and Black checklist, only 9 of the 17 investigational trials on humans scored a superior grade [28,29,31,32,33,34,38,40,44]. Six studies were assessed as high quality [30,35,39,41,42,43]. Eight experiments provided sufficient details on withdrawal and exit [28,31,32,33,34,38,40,44]. We also considered participant empowerment, and all experiments mentioned having gone through an ethical approval process and obtained informed consent from the participants (Table 6). We present our quality assessment for all included studies. This assessment highlights that all selected articles achieved 100% compliance with reporting standards, and 50% of these studies met the criteria for adequate statistical power (Figure 2).
Table 6. Quality assessment and scores of included studies using Downs and Black quality checklist.
Table 6. Quality assessment and scores of included studies using Downs and Black quality checklist.
Larsen et al. [41]Simoes et al. [43]Han et al. [28]Song et al. [29]Gonzalez-Sarrias et al. [30]Chambers et al. [42]Ejtahed et al. [31]Henning et al. [32]Ozkul et al. [45]Kassaian et al. [33]Kahleova et al. [34]Pisanu et al. [35]Seong et al. [44]Vijay et al. [37]Zhang et al. [38]Luo et al. [39]Han et al. [40]
Q1Hypothesis/aim/objective clearly described11111111111111111
Q2Main outcomes in Introduction or Methods11111111111111111
Q3Patient characteristics clearly described11111111111111111
Q4Interventions of interest clearly described11111111111111111
Q5Principal confounders clearly described11111111111111111
Q6Main findings clearly described11111111111111111
Q7Estimates of random variability provided for main outcomes11111111-11111111
Q8All adverse events of intervention reported00111111011110111
Q9Characteristics of patients lost to follow-up described0-1-0-11-11-10101
Q10Probability values reported for main outcomes11111111111111111
Q11Subjects Study participants asked to participate were representative of the source population11111111111111111
Q12Subjects Study participants prepared to participate were representative of the source population11111111111111111
Q13Location and delivery of study treatment were representative of the source population11111111111111111
Q14Study participants blinded to treatment1--11111-11-10111
Q15Blinded outcome assessment1--11111-11-11111
Q16Any data dredging clearly described11111111111111111
Q17Analyses adjusted for differing lengths of follow-up00111111111111101
Q18Appropriate statistical tests performed11111111111111111
Q19Compliance with interventions was reliable11111111111111111
Q20Outcome measures were reliable and valid11111111111111111
Q21All participants recruited from the same source population11111111111111111
Q22All participants recruited over the same time period11111111111111111
Q23Participants randomized to treatment(s)1-111111-1111-111
Q24Allocation of treatment concealed from investigators and participants1-111111-1101-101
Q25Adequate adjustment for confounding10111111111011111
Q26Losses to follow-up taken into account00110011011110111
Q27Sufficient power to detect treatment effect at a significance level of 0.0500100001011111011
TOTAL 2220262524242627192726232619262427
The ‘-’ symbol indicates that this clinical laboratory course can be used without analyzing this question. Count one point.
Figure 2. Quality assessment. (A) presents quality assessment summary: review authors’ judgments about each risk of bias item for each included studies low risk (+: green), high risk (-: red), or unclear risk (?: yellow); (B) indicates quality assessment graph: review authors’ judgments about each assessment item presented as percentages across all included studies.
Figure 2. Quality assessment. (A) presents quality assessment summary: review authors’ judgments about each risk of bias item for each included studies low risk (+: green), high risk (-: red), or unclear risk (?: yellow); (B) indicates quality assessment graph: review authors’ judgments about each assessment item presented as percentages across all included studies.
Ijms 25 00438 g002aIjms 25 00438 g002b

4. Discussion

Bacteroides spp. (BAC) constitute a significant proportion of the human gut microbiota and play a pivotal role in maintaining host metabolic homeostasis and immune regulation. These microbes aid in lipid metabolism, potentially influencing obesity outcomes and systemic inflammation [68]. This review represents the first thorough examination of BAC in obesity, detailing their correlation with body weight and prevalence in diverse populations across multiple countries. It also includes an analysis of preclinical studies on obesity and inflammation, shedding light on the potential mechanisms of action.
There is a recent study demonstrating the ability of BAC to degrade a wide range of polysaccharides and summarizing the utilization of BAC and their beneficial effects on intestinal health that supports our review [69]. This study demonstrated the BAC catabolism of polysaccharides to oligosaccharides or short-chain fatty acids (SCFAs). Fermentation of oligosaccharides and monosaccharides by SCFA-producing bacteria increases the luminal concentrations of acetate, butyrate, and propionate, which are beneficial effects of SCFAs on intestinal inflammation and immunity.
Furthermore, our review summarized clinical studies and randomized controlled trials of human anti-obesity treatments and focused on differences and relationships between body weight and BAC. In our review, all included preclinical studies presented positive improved results. In 70% of the clinical trials, weight loss in humans after treatment was accompanied by increased BAC richness. However, some studies have shown opposite results, of which two examples are provided in this review. We speculate that BAC richness is highly susceptible to changes caused by other drugs or foods, and we suspect an inhibitory relationship between probiotics and BAC. However, we still need to shed light on the actual evidence for this conjecture, and more experimental evidence is required. In addition, a study on microbial enterotypes reported that enterotypes may influence an individual’s ability to lose weight when following a specific diet [6]. We previously speculated that the difference in each individual characteristic affects the amount of BAC in the body. This study suggests that stratifying individuals according to two microbial enterotypes (dominance of either Prevotella or BAC) may help predict the response to diet and medication. It is a challenging point that individual differences can influence our judgment and conclusions regarding the results. Although preclinical studies have predominantly indicated a consistent anti-obesity effect of BAC, human trials present more heterogeneous outcomes. There are also studies that show that human dietary preferences in animal husbandry practices may influence domesticated animals [70]. Dietary homogenization among different species may lead to convergence of microbial characteristics. This discrepancy underscores the complexity of translating microbial interventions from animal models to humans, influenced by factors such as individual microbiome composition, diet, and lifestyle.
Because there are no sufficient clinical trials used to demonstrate the anti-obesity effect of BAC, the preclinical studies were reviewed to overcome this lack of studies. After a review of the preclinical studies focused on the mechanism of BAC in obesity was conducted, we concluded that BAC significantly prevents obesity in the animal model, particularly in mice. Our analysis underscores the potential of certain BAC, such as B. uniformis and B. vulgatus, in mitigating obesity-related parameters. For example, B. uniformis has been shown to reduce lipid accumulation and modulate adipose tissue inflammation, whereas B. vulgatus appears to influence body weight regulation, possibly through interactions with gut mucin dynamics. In our analyses, the BAC measure had to be at least 1 × 106 CFU/day and lasted at least 7 weeks to present validity in the body weight results. We also found that BAC improved immune dysfunction, promoted the growth of Akkermansia by utilizing metabolites from mucin degradation, increased serum insulin levels, and prevented obesity by promoting the catabolism of circulating branched-chain amino acids in the brown adipose tissue. Notably, Agustí et al., administered B. uniformis CECT 7771 to rats for 18 days and found that it affected the brain’s reward response, improved overeating, and reduced anxiety behavior. These effects are produced, at least in part, by altering the levels of dopamine, 5-hydroxytryptamine, and norepinephrine in the nucleus ambiguous, as well as the expression of dopamine D1 and D2 receptors in the prefrontal cortex and gut [57]. Furthermore, certain BAC can modulate systemic and local inflammation. B. vulgatus FTJS7K1 have been observed to downregulate pro-inflammatory cytokines and potentially contribute to maintaining gut barrier integrity, thereby potentially reducing low-grade systemic inflammation often observed in obesity scenarios [67]. The mechanisms underlying these effects are not entirely clear and may involve complex interactions between gut microbiota, host metabolism, and systemic inflammation. Further research is needed to clarify these mechanisms and determine whether similar effects occur in humans.
This study had some limitations. First, this review was constrained by the scarcity of long-term human trials and inconsistencies in the study design, particularly concerning probiotic strains, dosages, and participant demographics. These variations complicate the interpretation of the therapeutic potential of BAC in obesity management. Second, attempts were made to identify all randomized controlled trials on the increase in BAC following treatment efficacy, but potential incompleteness of citation tracking may have resulted in some relevant randomized controlled studies being overlooked. In addition, despite clear eligibility criteria, these studies need to be more consistent regarding this study’s design, probiotic strain, form and duration of application, sample population, and outcomes. Data are often not reported because many studies have not reported positive results for BAC after pharmacological interventions.
The quality assessment of the current studies indicated variability in the quality of the available research. While many studies were conducted, some lacked sufficient rigor, highlighting the need for more high-quality, consistent, and replicable research in this field. Most studies have been short-term, ranging from 4 to 24 weeks. Considering that obesity-related inflammation is a chronic disease, long-term evaluation of treatment is recommended. An apparent strength of this study is that our search included combinations of obesity and BAC and combinations of BAC alone or combinations where it was determined that BAC produced an effect, if available.
This review provides direct and indirect evidence of the effectiveness of BAC supplementation in treating obesity. In conclusion, BAC offers a promising avenue for treating obesity and inflammation, with certain BAC strains having specific therapeutic potential. However, the existing body of research is characterized by variability, necessitating more comprehensive, standardized, and long-term clinical trials. Future investigations are needed to elucidate the mechanistic pathways underlying the anti-obesity effects of BAC, assess the feasibility and acceptance of interventions, such as fecal microbiota transplant (FMT), and establish protocols for overcoming challenges in post-administration microbial colonization. Through this, we can progress toward targeted obesity therapies that harness the beneficial capacities of the gut microbiota.

5. Conclusions

This review rigorously evaluated the effect of BAC supplementation on obesity-related markers, revealing a nuanced relationship between specific species and strains. The analysis delineates the effects of several BAC strains, notably B. uniformis (CECT 7771 and CBA 7346), B. fragilis (HCK-B3, ATCC25285, and NCTC9343), B. vulgatus (SNUG 40005, ATCC 8482, and FTJS7K1), B. ovatus (ELH- B2, JCM5824, and ATCC 8483), B. acidifaciens JCM 10556, B. dorei DSM 17855, and B. thetaiotaomicron DSM 2079, on the modulation of BW, waist circumference (WC), adiposity, and systemic levels of pro-inflammatory cytokines.
Intriguingly, these anti-obesity properties appear to be species- and strain-specific, underscoring the biological complexity and the necessity for precision when considering probiotic interventions. Despite promising indications from preclinical studies, there is a lack of human clinical trials that directly examine the effects of BAC supplementation on obesity. This gap highlights an unresolved dichotomy in the field, wherein empirical support is burgeoning, yet a definitive clinical consensus remains elusive.
Therefore, the therapeutic potential of BAC in managing obesity requires further targeted longitudinal human studies. Future research should prioritize identifying and validating the most efficient BAC strains for obesity mitigation and investigate their mechanistic pathways, optimal dosages, and administration protocols. Moreover, exploring synergistic or antagonistic interactions between different BAC strains and broader microbiome-host dynamics will be instrumental in harnessing the full therapeutic arsenal of these microbes.

Author Contributions

Conceptualization, H.K.; methodology, S.-H.P.; validation, S.-H.P. and H.K.; formal analysis, L.W.; investigation, L.W.; data curation, L.W.; writing—original draft preparation, L.W.; writing—review and editing, S.-H.P.; visualization, L.W.; supervision, H.K.; project administration, H.K.; funding acquisition, H.K. All authors have read and agreed to the published version of the manuscript.

Funding

The authors are grateful for funding support from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health and Welfare, Republic of Korea (HF20C0020) and from the National Research Foundation of Korea Grant funded by the Korean Government (NRF-2021R1A2C1011087).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are available upon request to the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Afrizal, A.; Hitch, T.C.; Viehof, A.; Treichel, N.; Riedel, T.; Abt, B.; Buhl, E.M.; Kohlheyer, D.; Overmann, J.; Clavel, T. Anaerobic single-cell dispensing facilitates the cultivation of human gut bacteria. Environ. Microbiol. 2022, 24, 3861–3881. [Google Scholar] [CrossRef] [PubMed]
  2. Shetty, S.A.; Smidt, H.; de Vos, W.M. Reconstructing functional networks in the human intestinal tract using synthetic microbiomes. Curr. Opin. Biotechnol. 2019, 58, 146–154. [Google Scholar] [CrossRef] [PubMed]
  3. Mahowald, M.A.; Rey, F.E.; Seedorf, H.; Turnbaugh, P.J.; Fulton, R.S.; Wollam, A.; Shah, N.; Wang, C.; Magrini, V.; Wilson, R.K.; et al. Characterizing a model human gut microbiota composed of members of its two dominant bacterial phyla. Proc. Natl. Acad. Sci. USA 2009, 106, 5859–5864. [Google Scholar] [CrossRef] [PubMed]
  4. Kulagina, E.V.; Efimov, B.A.; Maximov, P.Y.; Kafarskaia, L.I.; Chaplin, A.V.; Shkoporov, A.N. Species composition of Bacteroidales order bacteria in the feces of healthy people of various ages. Biosci. Biotechnol. Biochem. 2012, 76, 169–171. [Google Scholar] [CrossRef] [PubMed]
  5. Nagy, E.; Maier, T.; Urban, E.; Terhes, G.; Kostrzewa, M.; ESCMID Study Group on Antimicrobial Resistance in Anaerobic Bacteria. Species identification of clinical isolates of Bacteroides by matrix-assisted laser-desorption/ionization time-of-flight mass spectrometry. Clin. Microbiol. Infect. 2009, 15, 796–802. [Google Scholar] [CrossRef]
  6. Wexler, H.M. Bacteroides: The good, the bad, and the nitty-gritty. Clin. Microbiol. Rev. 2007, 20, 593–621. [Google Scholar] [CrossRef]
  7. Sánchez, E.; Donat, E.; Ribes-Koninckx, C.; Calabuig, M.; Sanz, Y. Intestinal Bacteroides species associated with coeliac disease. J. Clin. Pathol. 2010, 63, 1105–1111. [Google Scholar] [CrossRef]
  8. Salyers, A. Bacteroides of the human lower intestinal tract. Annu. Rev. Microbiol. 1984, 38, 293–313. [Google Scholar] [CrossRef]
  9. Erejuwa, O.O.; Sulaiman, S.A.; Ab Wahab, M.S. Modulation of gut microbiota in the management of metabolic disorders: The prospects and challenges. Int. J. Mol. Sci. 2014, 15, 4158–4188. [Google Scholar] [CrossRef]
  10. Jung, S.; Lee, Y.J.; Kim, M.; Kim, M.; Kwak, J.H.; Lee, J.-W.; Ahn, Y.-T.; Sim, J.-H.; Lee, J.H. Supplementation with two probiotic strains, Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032, reduced body adiposity and Lp-PLA2 activity in overweight subjects. J. Funct. Foods 2015, 19, 744–752. [Google Scholar] [CrossRef]
  11. Kadooka, Y.; Sato, M.; Imaizumi, K.; Ogawa, A.; Ikuyama, K.; Akai, Y.; Okano, M.; Kagoshima, M.; Tsuchida, T. Regulation of abdominal adiposity by probiotics (Lactobacillus gasseri SBT2055) in adults with obese tendencies in a randomized controlled trial. Eur. J. Clin. Nutr. 2010, 64, 636–643. [Google Scholar] [CrossRef] [PubMed]
  12. Le, T.K.C.; Hosaka, T.; Nguyen, T.T.; Kassu, A.; Dang, T.O.; Tran, H.B.; Pham, T.P.; Tran, Q.B.; Le, T.H.H.; Da Pham, X. Bifidobacterium species lower serum glucose, increase expressions of insulin signaling proteins, and improve adipokine profile in diabetic mice. Biomed. Res. 2015, 36, 63–70. [Google Scholar] [CrossRef] [PubMed]
  13. Sanchis-Chordà, J.; Del Pulgar, E.M.G.; Carrasco-Luna, J.; Benítez-Páez, A.; Sanz, Y.; Codoñer-Franch, P. Bifidobacterium pseudocatenulatum CECT 7765 supplementation improves inflammatory status in insulin-resistant obese children. Eur. J. Nutr. 2019, 58, 2789–2800. [Google Scholar] [CrossRef] [PubMed]
  14. Yang, J.Y.; Lee, Y.S.; Kim, Y.; Lee, S.H.; Ryu, S.; Fukuda, S.; Hase, K.; Yang, C.S.; Lim, H.S.; Kim, M.S.; et al. Gut commensal Bacteroides acidifaciens prevents obesity and improves insulin sensitivity in mice. Mucosal Immunol. 2017, 10, 104–116. [Google Scholar] [CrossRef] [PubMed]
  15. Sun, F.; Zhang, Q.; Zhao, J.; Zhang, H.; Zhai, Q.; Chen, W. A potential species of next-generation probiotics? The dark and light sides of Bacteroides fragilis in health. Food Res. Int. 2019, 126, 108590. [Google Scholar] [CrossRef] [PubMed]
  16. Zhang, H.; Duan, Y.; Cai, F.; Cao, D.; Wang, L.; Qiao, Z.; Hong, Q.; Li, N.; Zheng, Y.; Su, M.; et al. Next-Generation Probiotics: Microflora Intervention to Human Diseases. BioMed Res. Int. 2022, 2022, 5633403. [Google Scholar] [CrossRef] [PubMed]
  17. Surana, N.K.; Kasper, D.L. The yin yang of bacterial polysaccharides: Lessons learned from B. fragilis PSA. Immunol. Rev. 2012, 245, 13–26. [Google Scholar] [CrossRef]
  18. Schwalm, N.D.; Groisman, E.A. Navigating the gut buffet: Control of polysaccharide utilization in Bacteroides spp. Trends Microbiol. 2017, 25, 1005–1015. [Google Scholar] [CrossRef]
  19. Cuskin, F.; Lowe, E.C.; Temple, M.J.; Zhu, Y.; Cameron, E.A.; Pudlo, N.A.; Porter, N.T.; Urs, K.; Thompson, A.J.; Cartmell, A.; et al. Human gut Bacteroidetes can utilize yeast mannan through a selfish mechanism. Nature 2015, 517, 165–169. [Google Scholar] [CrossRef]
  20. Ambesh, P.; Stroud, S.; Franzova, E.; Gotesman, J.; Sharma, K.; Wolf, L.; Kamholz, S. Recurrent Lactobacillus bacteremia in a patient with leukemia. J. Investig. Med. High Impact Case Rep. 2017, 5, 2324709617744233. [Google Scholar] [CrossRef]
  21. Mikucka, A.; Deptuła, A.; Bogiel, T.; Chmielarczyk, A.; Nurczyńska, E.; Gospodarek-Komkowska, E. Bacteraemia Caused by Probiotic Strains of Lacticaseibacillus rhamnosus—Case Studies Highlighting the Need for Careful Thought before Using Microbes for Health Benefits. Pathogens 2022, 11, 977. [Google Scholar] [CrossRef] [PubMed]
  22. Widyastuti, Y.; Febrisiantosa, A.; Tidona, F. Health-promoting properties of lactobacilli in fermented dairy products. Front. Microbiol. 2021, 12, 673890. [Google Scholar] [CrossRef] [PubMed]
  23. King, S.J.; McCole, D.F. Epithelial-microbial diplomacy: Escalating border tensions drive inflammation in inflammatory bowel disease. Intest. Res. 2019, 17, 177–191. [Google Scholar] [CrossRef] [PubMed]
  24. Carrow, H.C.; Batachari, L.E.; Chu, H. Strain diversity in the microbiome: Lessons from Bacteroides fragilis. PLoS Pathog. 2020, 16, e1009056. [Google Scholar] [CrossRef] [PubMed]
  25. Hiippala, K.; Kainulainen, V.; Suutarinen, M.; Heini, T.; Bowers, J.R.; Jasso-Selles, D.; Lemmer, D.; Valentine, M.; Barnes, R.; Engelthaler, D.M. Isolation of anti-inflammatory and epithelium reinforcing Bacteroides and Parabacteroides spp. from a healthy fecal donor. Nutrients 2020, 12, 935. [Google Scholar] [CrossRef] [PubMed]
  26. Zatterale, F.; Longo, M.; Naderi, J.; Raciti, G.A.; Desiderio, A.; Miele, C.; Beguinot, F. Chronic adipose tissue inflammation linking obesity to insulin resistance and type 2 diabetes. Front. Physiol. 2020, 10, 1607. [Google Scholar] [CrossRef] [PubMed]
  27. Downs, S.H.; Black, N. The feasibility of creating a checklist for the assessment of the methodological quality both of randomised and non-randomised studies of health care interventions. J. Epidemiol. Community Health 1998, 52, 377–384. [Google Scholar] [CrossRef] [PubMed]
  28. Han, K.; Bose, S.; Kim, Y.M.; Chin, Y.W.; Kim, B.S.; Wang, J.H.; Lee, J.H.; Kim, H. Rehmannia glutinosa reduced waist circumferences of Korean obese women possibly through modulation of gut microbiota. Food Funct. 2015, 6, 2684–2692. [Google Scholar] [CrossRef]
  29. Song, M.Y.; Wang, J.H.; Eom, T.; Kim, H. Schisandra chinensis fruit modulates the gut microbiota composition in association with metabolic markers in obese women: A randomized, double-blind placebo-controlled study. Nutr. Res. 2015, 35, 655–663. [Google Scholar] [CrossRef]
  30. Gonzalez-Sarrias, A.; Romo-Vaquero, M.; Garcia-Villalba, R.; Cortes-Martin, A.; Selma, M.V.; Espin, J.C. The Endotoxemia Marker Lipopolysaccharide-Binding Protein is Reduced in Overweight-Obese Subjects Consuming Pomegranate Extract by Modulating the Gut Microbiota: A Randomized Clinical Trial. Mol. Nutr. Food Res. 2018, 62, e1800160. [Google Scholar] [CrossRef]
  31. Ejtahed, H.S.; Tito, R.Y.; Siadat, S.D.; Hasani-Ranjbar, S.; Hoseini-Tavassol, Z.; Rymenans, L.; Verbeke, K.; Soroush, A.R.; Raes, J.; Larijani, B. Metformin induces weight loss associated with gut microbiota alteration in non-diabetic obese women: A randomized double-blind clinical trial. Eur. J. Endocrinol. 2019, 180, 165–176. [Google Scholar] [CrossRef] [PubMed]
  32. Henning, S.M.; Yang, J.; Woo, S.L.; Lee, R.-P.; Huang, J.; Rasmusen, A.; Carpenter, C.L.; Thames, G.; Gilbuena, I.; Tseng, C.-H.; et al. Hass avocado inclusion in a weight-loss diet supported weight loss and altered gut microbiota: A 12-week randomized, parallel-controlled trial. Curr. Dev. Nutr. 2019, 3, nzz068. [Google Scholar] [CrossRef] [PubMed]
  33. Kassaian, N.; Feizi, A.; Rostami, S.; Aminorroaya, A.; Yaran, M.; Amini, M. The effects of 6 mo of supplementation with probiotics and synbiotics on gut microbiota in the adults with prediabetes: A double blind randomized clinical trial. Nutrition 2020, 79–80, 110854. [Google Scholar] [CrossRef] [PubMed]
  34. Kahleova, H.; Rembert, E.; Alwarith, J.; Yonas, W.N.; Tura, A.; Holubkov, R.; Agnello, M.; Chutkan, R.; Barnard, N.D. Effects of a Low-Fat Vegan Diet on Gut Microbiota in Overweight Individuals and Relationships with Body Weight, Body Composition, and Insulin Sensitivity. A Randomized Clinical Trial. Nutrients 2020, 12, 2917. [Google Scholar] [CrossRef] [PubMed]
  35. Pisanu, S.; Palmas, V.; Madau, V.; Casula, E.; Deledda, A.; Cusano, R.; Uva, P.; Vascellari, S.; Boi, F.; Loviselli, A.; et al. Impact of a Moderately Hypocaloric Mediterranean Diet on the Gut Microbiota Composition of Italian Obese Patients. Nutrients 2020, 12, 2707. [Google Scholar] [CrossRef] [PubMed]
  36. Jin, J.; Cheng, R.; Ren, Y.; Shen, X.; Wang, J.; Xue, Y.; Zhang, H.; Jia, X.; Li, T.; He, F.; et al. Distinctive Gut Microbiota in Patients with Overweight and Obesity with Dyslipidemia and its Responses to Long-term Orlistat and Ezetimibe Intervention: A Randomized Controlled Open-label Trial. Front. Pharmacol. 2021, 12, 732541. [Google Scholar] [CrossRef] [PubMed]
  37. Vijay, A.; Astbury, S.; Le Roy, C.; Spector, T.D.; Valdes, A.M. The prebiotic effects of omega-3 fatty acid supplementation: A six-week randomised intervention trial. Gut Microbes 2021, 13, 1863133. [Google Scholar] [CrossRef] [PubMed]
  38. Zhang, S.; Wu, P.; Tian, Y.; Liu, B.; Huang, L.; Liu, Z.; Lin, N.; Xu, N.; Ruan, Y.; Zhang, Z.; et al. Gut Microbiota Serves a Predictable Outcome of Short-Term Low-Carbohydrate Diet (LCD) Intervention for Patients with Obesity. Microbiol. Spectr. 2021, 9, e0022321. [Google Scholar] [CrossRef]
  39. Luo, W.; Zhou, J.; Yang, X.; Wu, R.; Liu, H.; Shao, H.; Huang, B.; Kang, X.; Yang, L.; Liu, D. A Chinese medical nutrition therapy diet accompanied by intermittent energy restriction alleviates type 2 diabetes by enhancing pancreatic islet function and regulating gut microbiota composition. Food Res. Int. 2022, 161, 111744. [Google Scholar] [CrossRef]
  40. Han, A.L.; Jeong, S.J.; Ryu, M.S.; Yang, H.J.; Jeong, D.Y.; Park, D.S.; Lee, H.K. Anti-Obesity Effects of Traditional and Commercial Kochujang in Overweight and Obese Adults: A Randomized Controlled Trial. Nutrients 2022, 14, 2783. [Google Scholar] [CrossRef]
  41. Larsen, N.; Vogensen, F.K.; Gobel, R.J.; Michaelsen, K.F.; Forssten, S.D.; Lahtinen, S.J.; Jakobsen, M. Effect of Lactobacillus salivarius Ls-33 on fecal microbiota in obese adolescents. Clin. Nutr. 2013, 32, 935–940. [Google Scholar] [CrossRef] [PubMed]
  42. Chambers, E.S.; Byrne, C.S.; Morrison, D.J.; Murphy, K.G.; Preston, T.; Tedford, C.; Garcia-Perez, I.; Fountana, S.; Serrano-Contreras, J.I.; Holmes, E.; et al. Dietary supplementation with inulin-propionate ester or inulin improves insulin sensitivity in adults with overweight and obesity with distinct effects on the gut microbiota, plasma metabolome and systemic inflammatory responses: A randomised cross-over trial. Gut 2019, 68, 1430–1438. [Google Scholar] [PubMed]
  43. Simoes, C.D.; Maukonen, J.; Scott, K.P.; Virtanen, K.A.; Pietilainen, K.H.; Saarela, M. Impact of a very low-energy diet on the fecal microbiota of obese individuals. Eur. J. Nutr. 2014, 53, 1421–1429. [Google Scholar] [CrossRef] [PubMed]
  44. Seong, E.; Bose, S.; Han, S.Y.; Song, E.J.; Lee, M.; Nam, Y.D.; Kim, H. Positive influence of gut microbiota on the effects of Korean red ginseng in metabolic syndrome: A randomized, double-blind, placebo-controlled clinical trial. EPMA J. 2021, 12, 177–197. [Google Scholar] [CrossRef] [PubMed]
  45. 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]
  46. 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]
  47. 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] [PubMed]
  48. Turnbaugh, P.J.; Hamady, M.; Yatsunenko, T.; Cantarel, B.L.; Duncan, A.; Ley, R.E.; Sogin, M.L.; Jones, W.J.; Roe, B.A.; Affourtit, J.P.; et al. A core gut microbiome in obese and lean twins. Nature 2009, 457, 480–484. [Google Scholar] [CrossRef]
  49. Kasai, C.; Sugimoto, K.; Moritani, I.; Tanaka, J.; Oya, Y.; Inoue, H.; Tameda, M.; Shiraki, K.; Ito, M.; Takei, Y.; et al. Comparison of the gut microbiota composition between obese and non-obese individuals in a Japanese population, as analyzed by terminal restriction fragment length polymorphism and next-generation sequencing. BMC Gastroenterol. 2015, 15, 100. [Google Scholar] [CrossRef]
  50. Hu, H.J.; Park, S.G.; Jang, H.B.; Choi, M.K.; Park, K.H.; Kang, J.H.; Park, S.I.; Lee, H.J.; Cho, S.H. Obesity Alters the Microbial Community Profile in Korean Adolescents. PLoS ONE 2015, 10, e0134333. [Google Scholar] [CrossRef]
  51. Hou, Y.P.; He, Q.Q.; Ouyang, H.M.; Peng, H.S.; Wang, Q.; Li, J.; Lv, X.F.; Zheng, Y.N.; Li, S.C.; Liu, H.L.; et al. Human Gut Microbiota Associated with Obesity in Chinese Children and Adolescents. Biomed. Res. Int. 2017, 2017, 7585989. [Google Scholar] [CrossRef] [PubMed]
  52. Liu, R.; Hong, J.; Xu, X.; Feng, Q.; Zhang, D.; Gu, Y.; Shi, J.; Zhao, S.; Liu, W.; Wang, X.; et al. Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention. Nat. Med. 2017, 23, 859–868. [Google Scholar] [CrossRef] [PubMed]
  53. Riva, A.; Borgo, F.; Lassandro, C.; Verduci, E.; Morace, G.; Borghi, E.; Berry, D. Pediatric obesity is associated with an altered gut microbiota and discordant shifts in Firmicutes populations. Environ. Microbiol. 2017, 19, 95–105. [Google Scholar] [CrossRef] [PubMed]
  54. Palmas, V.; Pisanu, S.; Madau, V.; Casula, E.; Deledda, A.; Cusano, R.; Uva, P.; Vascellari, S.; Loviselli, A.; Manzin, A.; et al. Gut microbiota markers associated with obesity and overweight in Italian adults. Sci. Rep. 2021, 11, 5532. [Google Scholar] [CrossRef] [PubMed]
  55. Dong, T.S.; Guan, M.; Mayer, E.A.; Stains, J.; Liu, C.; Vora, P.; Jacobs, J.P.; Lagishetty, V.; Chang, L.; Barry, R.L.; et al. Obesity is associated with a distinct brain-gut microbiome signature that connects Prevotella and Bacteroides to the brain’s reward center. Gut Microbes 2022, 14, 2051999. [Google Scholar] [CrossRef] [PubMed]
  56. Gauffin Cano, P.; Santacruz, A.; Moya, A.; Sanz, Y. Bacteroides uniformis CECT 7771 ameliorates metabolic and immunological dysfunction in mice with high-fat-diet induced obesity. PLoS ONE 2012, 7, e41079. [Google Scholar] [CrossRef] [PubMed]
  57. Agusti, A.; Campillo, I.; Balzano, T.; Benitez-Paez, A.; Lopez-Almela, I.; Romani-Perez, M.; Forteza, J.; Felipo, V.; Avena, N.M.; Sanz, Y. Bacteroides uniformis CECT 7771 Modulates the Brain Reward Response to Reduce Binge Eating and Anxiety-Like Behavior in Rat. Mol. Neurobiol. 2021, 58, 4959–4979. [Google Scholar] [CrossRef]
  58. You, H.J.; Si, J.; Kim, J.; Yoon, S.; Cha, K.H.; Yoon, H.S.; Lee, G.; Yu, J.; Choi, J.S.; Jung, M.; et al. Bacteroides vulgatus SNUG 40005 Restores Akkermansia Depletion by Metabolite Modulation. Gastroenterology 2023, 164, 103–116. [Google Scholar] [CrossRef]
  59. Lee, H.B.; Do, M.H.; Jhun, H.; Ha, S.K.; Song, H.S.; Roh, S.W.; Chung, W.H.; Nam, Y.D.; Park, H.Y. Amelioration of Hepatic Steatosis in Mice through Bacteroides uniformis CBA7346-Mediated Regulation of High-Fat Diet-Induced Insulin Resistance and Lipogenesis. Nutrients 2021, 13, 2989. [Google Scholar] [CrossRef]
  60. Yoshida, N.; Yamashita, T.; Osone, T.; Hosooka, T.; Shinohara, M.; Kitahama, S.; Sasaki, K.; Sasaki, D.; Yoneshiro, T.; Suzuki, T.; et al. Bacteroides spp. promotes branched-chain amino acid catabolism in brown fat and inhibits obesity. iScience 2021, 24, 103342. [Google Scholar] [CrossRef]
  61. Garg, S.S.; Kushwaha, K.; Dubey, R.; Gupta, J. Association between obesity, inflammation and insulin resistance: Insights into signaling pathways and therapeutic interventions. Diabetes Res. Clin. Pract. 2023, 200, 110691. [Google Scholar] [CrossRef] [PubMed]
  62. Lee, Y.K.; Mehrabian, P.; Boyajian, S.; Wu, W.L.; Selicha, J.; Vonderfecht, S.; Mazmanian, S.K. The Protective Role of Bacteroides fragilis in a Murine Model of Colitis-Associated Colorectal Cancer. mSphere 2018, 3, e00587-18. [Google Scholar] [CrossRef] [PubMed]
  63. Delday, M.; Mulder, I.; Logan, E.T.; Grant, G. Bacteroides thetaiotaomicron Ameliorates Colon Inflammation in Preclinical Models of Crohn’s Disease. Inflamm. Bowel Dis. 2019, 25, 85–96. [Google Scholar] [CrossRef] [PubMed]
  64. Tan, H.; Zhao, J.; Zhang, H.; Zhai, Q.; Chen, W. Novel strains of Bacteroides fragilis and Bacteroides ovatus alleviate the LPS-induced inflammation in mice. Appl. Microbiol. Biotechnol. 2019, 103, 2353–2365. [Google Scholar] [CrossRef] [PubMed]
  65. Ihekweazu, F.D.; Fofanova, T.Y.; Queliza, K.; Nagy-Szakal, D.; Stewart, C.J.; Engevik, M.A.; Hulten, K.G.; Tatevian, N.; Graham, D.Y.; Versalovic, J.; et al. Bacteroides ovatus ATCC 8483 monotherapy is superior to traditional fecal transplant and multi-strain bacteriotherapy in a murine colitis model. Gut Microbes 2019, 10, 504–520. [Google Scholar] [CrossRef] [PubMed]
  66. Ramakrishna, C.; Kujawski, M.; Chu, H.; Li, L.; Mazmanian, S.K.; Cantin, E.M. Bacteroides fragilis polysaccharide A induces IL-10 secreting B and T cells that prevent viral encephalitis. Nat. Commun. 2019, 10, 2153. [Google Scholar] [CrossRef] [PubMed]
  67. Wang, C.; Xiao, Y.; Yu, L.; Tian, F.; Zhao, J.; Zhang, H.; Chen, W.; Zhai, Q. Protective effects of different Bacteroides vulgatus strains against lipopolysaccharide-induced acute intestinal injury, and their underlying functional genes. J. Adv. Res. 2022, 36, 27–37. [Google Scholar] [CrossRef]
  68. Cencic, A.; Chingwaru, W. The role of functional foods, nutraceuticals, and food supplements in intestinal health. Nutrients 2010, 2, 611–625. [Google Scholar] [CrossRef]
  69. Cheng, J.; Hu, J.; Geng, F.; Nie, S. Bacteroides utilization for dietary polysaccharides and their beneficial effects on gut health. Food Sci. Hum. Wellness 2022, 11, 1101–1110. [Google Scholar] [CrossRef]
  70. Reese, A.T.; Chadaideh, K.S.; Diggins, C.E.; Schell, L.D.; Beckel, M.; Callahan, P.; Ryan, R.; Emery Thompson, M.; Carmody, R.N. Effects of domestication on the gut microbiota parallel those of human industrialization. eLife 2021, 10, e60197. [Google Scholar] [CrossRef]
Figure 1. Flowchart of the process for screening articles on systematic reviews that directly and indirectly demonstrate the effects of Bacteroides spp. on human obesity.
Figure 1. Flowchart of the process for screening articles on systematic reviews that directly and indirectly demonstrate the effects of Bacteroides spp. on human obesity.
Ijms 25 00438 g001
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

Wu, L.; Park, S.-H.; Kim, H. Direct and Indirect Evidence of Effects of Bacteroides spp. on Obesity and Inflammation. Int. J. Mol. Sci. 2024, 25, 438. https://doi.org/10.3390/ijms25010438

AMA Style

Wu L, Park S-H, Kim H. Direct and Indirect Evidence of Effects of Bacteroides spp. on Obesity and Inflammation. International Journal of Molecular Sciences. 2024; 25(1):438. https://doi.org/10.3390/ijms25010438

Chicago/Turabian Style

Wu, Liangliang, Seo-Hyun Park, and Hojun Kim. 2024. "Direct and Indirect Evidence of Effects of Bacteroides spp. on Obesity and Inflammation" International Journal of Molecular Sciences 25, no. 1: 438. https://doi.org/10.3390/ijms25010438

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