Skip to main content
Gut Microbes logoLink to Gut Microbes
. 2024 Aug 20;16(1):2387796. doi: 10.1080/19490976.2024.2387796

Dietary fiber for the prevention of childhood obesity: a focus on the involvement of the gut microbiota

Zhongmin Yang a, Mingyue Yang a, Edward C Deehan b,c, Chenxi Cai d, Karen L Madsen e, Eytan Wine f, Guiling Li a,g, Jian Li a,g, Jingwen Liu a,, Zhengxiao Zhang a,g,
PMCID: PMC11340751  PMID: 39163556

ABSTRACT

Given the worldwide epidemic of overweight and obesity among children, evidence-based dietary recommendations are fundamentally important for obesity prevention. Although the significance of the human gut microbiome in shaping the physiological effects of diet and obesity has been widely recognized, nutritional therapeutics for the mitigation of pediatric obesity globally are only just starting to leverage advancements in the nutritional microbiology field. In this review, we extracted data from PubMed, EMBASE, Scopus, Web of Science, Google Scholar, CNKI, Cochrane Library and Wiley online library that focuses on the characterization of gut microbiota (including bacteria, fungi, viruses, and archaea) in children with obesity. We further review host-microbe interactions as mechanisms mediating the physiological effects of dietary fibers and how fibers alter the gut microbiota in children with obesity. Contemporary nutritional recommendations for the prevention of pediatric obesity are also discussed from a gut microbiological perspective. Finally, we propose an experimental framework for integrating gut microbiota into nutritional interventions for children with obesity and provide recommendations for the design of future studies on precision nutrition for pediatric obesity.

KEYWORDS: Dietary fiber, gut microbiota, childhood, obesity, precision nutrition

1. Introduction

The presence of childhood obesity is a prominent risk factor for the development of obesity-related comorbidities in adolescence, including increased risk of type 2 diabetes1, nonalcoholic fatty liver disease,2 and cardiovascular disease.3 Data released by the World Obesity Federation reported that the increase in the prevalence of obesity is expected to be steepest among children and adolescents aged 5–19 years, rising from 10% to 20% of the world’s boys and from 8% to 18% of the world’s girls between 2020 and 2035.4 Recent research further suggests that having excess body weight in childhood and adolescence was not only found to adversely affect physical health later in life5 but also negatively impact psychosocial wellbeing.6 Nevertheless, current strategies for treating childhood obesity, such as lifestyle changes and pharmacotherapy, have very modest effectiveness,7 concerning side effects,8 or inconsistent outcomes.9 Identifying the causes of childhood obesity and targeting interventions is, therefore, important in the context of preventive health.

Various factors have been suggested as potential causes of obesity, including lifestyle choices and genetic factors,10 with the gut microbiota emerging as an important factor contributing to the pathogenesis of obesity.11 Due to their profound effects on the host, this highly complex group of microbes that reside along the human gastrointestinal tract is referred to as an individual’s “second genome”.12 The gut microbiota is a community of more than 1013 microorganisms that includes bacteria, fungi, viruses, and archaea, which live as a complex ecosystem with the human host.6,13 There is accumulating evidence that the gut microbiota affects all aspects of energy homeostasis through mechanisms involving immune, hormonal and neural systems, and that gut dysbiosis, or an abnormal composition of gut microbial taxa might contribute to a disturbed host metabolism through effects on adipose tissue, muscle, and liver.14

Although growing evidence from animal models that the gut microbiota has a potential causal role in obesity and would be a promising target for precision therapeutics, there is still a lack of convincing evidence for a direct contribution of the gut microbiota to humans with obesity.15 Whether gut microbial alteration exists in children with obesity is yet to be systematically evaluated.16,17 Moreover, the gut microbiome is thought to stabilize and resemble that of an adult after the first 1 to 3 years of life,18 while other research suggests that it continues to develop throughout adolescence and that gaining excess body weight during childhood is likely to play a role in further shaping the gut microbiota.19,20 It is unclear what role the coexistence of gut cross-kingdom community interactions in the gut plays in childhood obesity. Therefore, identifying the gut microbiota characteristics that are conserved in children with obesity and also play a pathogenic role in the development of childhood obesity is a challenge in the field. This has important implications for the development of microbiome-targeted therapies for the prevention and treatment of childhood obesity.

Dietary substances, such as fibers and prebiotics, have been found to induce a positive impact on the children gut microbiota.21 As a result, they hold potential for the management of obesity. Dietary fibers (DFs), carbohydrate polymers that are neither digested nor absorbed in the small intestine, are subject to microbial fermentation in the human colon and, thus, influence the composition of microbial communities as well as their metabolic activities.22 Some DFs can also be classified as prebiotics,23 or substrates selectively used by host microorganisms that confer a health benefit to humans.23 However, currently it is not clear if the beneficial effects of increasing DF intake are due to microbial metabolism and the production of metabolites such as short-chain fatty acids (SCFAs) or due to a DF-altered intestinal transit time, changes in nutrient absorption, fecal bulking and binding of various metabolites, or modulation of the immune system.18,24

It is crucial to keep in mind that the physiological functions of DFs are highly structure-dependent. Subtle variations in the chemical structures of DFs affect their utilization by gut microbes, as bacteria have different abilities to cleave the linkages in the structure of these complex molecules to obtain simple sugars.25 Simultaneously, recent reviews have revealed that intrinsic and isolated fibers have functional specificity as well.26 However, there is insufficient literature evidence to support the targeted modulatory effects of DFs on the gut microbiota of children with obesity. Furthermore, adults are encouraged to consume at least 25–38 g of DFs per day, but there is currently a lack of data to support DF guidelines for children in most countries. As a result, more research is needed to thoroughly investigate the relationship between DFs and childhood obesity from a gut microbiota perspective.

In studies of children with obesity, examination of DF-microbiota-host interactions has been limited, despite recent increases in nutritional microbiology research in adults. In this perspective, we first systematically evaluated whether special microbial alterations exist in children with obesity, and then reviewed the effects of DFs and DFs-related patterns on the gut microbiota in children with obesity. We further apply this information to confront an ongoing debate in the field of childhood obesity research. Finally, we propose a microbiome-targeted research approach for evaluating treatments for the reduction of childhood obesity and outline an experimental framework for systematically incorporating the gut microbiome into future nutrition research.

2. Systematic review: characterization of the gut microbiota in children with obesity

We conducted a systematic literature review in July 2022 (with an updated literature search in June 2024) to assess the pertinent gut microbiota traits of childhood obesity (see Box 1 for search strategy). Of the 63 studies that met the inclusion criteria, some of the gut microbes in children with obesity showed consistent trends (e.g., Firmicutes/Bacteroidetes (F/B) ratios, Akkermansia muciniphila, Lactobacillus spp., Dialister, etc). However, there is considerable heterogeneity among the findings of most published studies investigating gut microbiota in childhood obesity. Understanding the compositional characteristics of the gut microbiota has the potential to help determine their role in the onset or development of obesity in the host, laying the groundwork for microbiome-based therapies for childhood obesity to improve the quality of life.

Box 1
To identify gut microbiota signatures that are characteristics of childhood obesity, we conducted a systematic review. We included all retrieved studies that reported on the gut microbiota of children with obesity, which included cross-sectional and case-control studies. In total, 63 studies (all were observational studies) that assessed changes in the gut microbiome of children with obesity were included in this review for analysis: 67% of studies (n = 42) used 16S rRNA sequencing for microbial analysis,27–68 nine studies used Real-time PCR (RT-PCR),69–77 six studies used shotgun metagenomic sequencing,78–83 and the remaining six studies used other methods to assess microbial differences.84–89 Information on each study in this review is included in Table S1 in the supplementary materials.
Box 1 Figure. Flow Diagram of Article Search and Selection Process.
Inline graphic
The literature search was conducted in July 2022, and an updated literature search was conducted in June 2024.

Summarized in Figure S1 are the main findings of the included studies regarding the relationship between microorganisms and childhood obesity. Only microbial features reported to correlate with obesity in three or more of the publications reviewed were included. The figure summarizes the percentage of included studies reporting the gut microbiota α-diversity, bacteria (7 phyla, 30 genera, 12 species), and archaea (1 species) in children with obesity in agreement with the indicated relationship with adiposity. The proceeding detailed analysis is grouped by gut bacterial diversity and compositional characteristics of bacteria, fungi, viruses, and archaea characteristics in children with obesity.

2.1. Microbial diversity

Of the 63 studies included, 65% (41/63) reported α-diversity in children with and without obesity (Table S1). Eight indices were used to assess α-diversity, and the most widely applied were Chao1, Shannon, Simpson and Observed species (Figure S1). There was no evidence of publication bias in any of the analyses (Figure S2). We conducted a meta-analysis based on the available α-diversity index data (Figure 1 and Figure S3). The results revealed a statistically significant difference in Phylogenetic diversity, Chao1 index, and Shannon index of gut microbiota α-diversity index between children with obesity and control individuals. Compared to control, Phylogenetic diversity (6 studies with 3 estimates provided data, SMD = 1.12, 95% CI 0.67, 1.57, I2 = 33%, p < 0.00001), Shannon index (32 studies with 25 estimates provided data, SMD = 0.28, 95% CI 0.10, 0.45, I2 = 59%, p = 0.002), Chao1 index (22 studies with 17 estimates provided data, SMD = 0.33, 95% CI 0.10, 0.56, I2 = 71%, p = 0.004) and were significantly decreased in children with obesity. Considering the heterogeneity, sensitivity analyses were performed by omitting each study in turn, which showed stability for the Chao1 index, and a decrease in heterogeneity for the Shannon index by omitting Méndez-Salazar 201843 (p = 0.002, I2 = 42%) (Figure S4). Based on the results of regional subgroup analyses, there were statistically significant differences for the Shannon index in Asian (p = 0.0002, I2 = 41%) and European (p = 0.01, I2 = 0%), and Simpson index (p = 0.001, I2 = 15%) in North America, with low heterogeneity. No significant differences or high heterogeneity were observed in any of the results based on age and BMI subgroup analyses (Figure S5).

Figure 1.

Figure 1.

Meta-analysis of gut microbiota differences and α-diversity between obese children and controls. SMD, standardized mean difference. CI, confidence interval. The “p” indicates the level of significance. The “I2” indicates the results of the heterogeneity test. The “n” indicates the number of studies that provided usable data.

Strong microbiota diversity is critical to the ability of the gut microbiota to adapt to stress and is a key indicator of good health. Meta-analysis showed a significant decrease in the Shannon index, Chao1 index and Phylogenetic diversity in obese children, aligning with the prevailing hypothesis. It is important to emphasize that different α-diversity indices exhibit different microbiota characteristics. For example, while the Chao 1 index is based on the total number of bacteria within a community, the Shannon index and the phylogenetic diversity index also consider bacterial evenness and phylogenetic abundance, respectively. Therefore, despite heterogeneity between studies, including differences in region, gender and age, statistically significant differences between alpha diversity indices suggest that ecological disorder in obese children is expressed through disruption of phylogenetic abundance, bacterial homogeneity, and changes in bacterial numbers. However, the common assumption that “more diversity is better” may oversimplify the intricate complex mechanisms.90 Measurement of diversity in children with obesity should be used as a starting point for further research of gut micro-ecological mechanisms in obesity and will help to explore causal relationships between the gut microbiome and its host. Furthermore, the variation between the diversity of gut microbiota in obese children and adults deserves further exploration.91,92

Beta diversity comparison between childhood obesity and controls was reported in 43% (27/63) studies (Table S1). 17 studies reported consistent significant differences of obese versus non-obese children, while 10 studies reported conflicting results. The results of these studies suggest differences in the common phylogenetic structure of obese children compared to controls. However, the method of measurement, geographic region, age, and degree of obesity may influence the outcomes.

2.2. Variations in bacterial composition

Meta-analysis based on relative or absolute abundance data at the phyla level between childhood obesity and controls revealed statistically significant differences for Bacteroidetes, Proteobacteria and F/B ratios (Figure 1 and Figure S6). Compared to control individuals, F/B ratios (23 studies with 11 estimates provided data, SMD = −0.46, 95% CI −0.87, −0.04, I2 = 79%, p = 0.03) and relative abundance of Proteobacteria (21 studies with 14 estimates provided data, SMD = −0.67, 95% CI −1.18, −0.16, I2 = 91%, p = 0.009) were significantly increased, and absolute abundance of Bacteroidetes (36 studies with 5 estimates provided data, SMD = 0.27, 95% CI 0.01, 0.53, I2 = 42%, p = 0.04) was significantly decreased in obese children. Considering the high heterogeneity, sensitivity analyses were performed by omitting each study in turn, with stable results (Figure S7). No publication bias in F/B ratios, relative abundance of Firmicutes and Proteobacteria (Figure S8). Based on the results of region, age and BMI subgroup analyses, there was a significant difference in F/B ratios (p < 0.00001, I2 = 0%) in European, and no significant difference between relative abundance of Firmicutes (p = 0.89, I2 = 0%) in school-aged children and absolute abundance of Firmicutes (p = 0.63, I2 = 0%) in overweight children, with high heterogeneity of the results of the analyses of the other subgroups (Figure S9).

At the genera level, the absolute abundance of Akkermansia (6 studies providing 2 estimates, SMD = 0.38, 95% CI 0.03, 0.73, I2 = 0%, p = 0.04) was significantly decreased in obese children only compared with controls (Figure 1 and Figure S10). Sensitivity analysis showed a significant increase relative abundance of Faecalibacterium by omitting Tian 202365(p = 0.0004, I2 = 0%) (Figure S11). Region subgroup analyses showed low heterogeneity and significant increase relative abundance of Faecalibacterium in North America (p = 0.02, I2 = 21%) (Figure S12).

At the species level, Akkermansia muciniphila (4 studies) was consistently decreased in children with obesity (Figure S1). Meta-analysis results showed that relative abundance of Akkermansia muciniphila (4 studies providing 3 estimates, SMD = 0.50, 95% CI 0.14, 0.86, I2 = 52%, p = 0.007) was significantly decreased and absolute abundance of Lactobacillus spp. (3 studies providing 3 estimates, SMD = −0.47, 95% CI −0.82, −0.12, I2 = 0%, p = 0.008) was significantly increased in obese children compared to controls (Figure 1 and Figure S13). Sensitivity analyses showed a significant decrease relative abundance of A. muciniphila (p < 0.0001, I2 = 0%) and Bacteroides ovatus (p < 0.0001, I2 = 0%) after omitting López-Contreras 201845 (Figure S14). Region subgroup analyses demonstrated low heterogeneity and a significant decrease in the relative abundance of Faecalibacterium prausnitzii among Asian individuals (p = 0.0002, I2 = 21%) (Figure S15).

Clinical studies have demonstrated notable benefits in overweight/obese individuals supplementing with pasteurizing A. muciniphila bacteria to significantly improve insulin resistance, blood cholesterol and inflammatory factor levels.93 It is estimated that F. prausnitzii makes up 1% to 6% of the total microbiota of healthy individuals,94 and has been shown to impart anti-inflammatory characteristics in part by the production of butyrate.94,95 However, these anti-inflammatory benefits are not observed in children with obesity. Despite presumably sharing a core microbiome architecture, recent research has indicated that the gut microbiota of children between the ages of 3 and 18 is distinct in terms of both function and taxonomy compared to that of adults.20 This suggests that the establishment of the gut microbiota may require a somewhat lengthy period. A meta-analysis of data from adults with obesity revealed that the abundance of Dorea, Dialister, Fusobacterium, Sutterella, Streptococcus, and Prevotella was significantly increased in obesity. Conversely, Bifidobacterium exhibited a lower abundance.92 In contrast, in children with obesity, Dialister, Streptococcus, and Prevotella did not observe a similar pattern of changes as in adults with obesity (Figure 1), indicating a disparity between the gut microbiota of children and adults with obesity.

2.3. Variations in fungi composition

Human gut fungi are receiving increased research attention96,97 due to their potential involvement in the etiology of numerous gut-associated diseases.98 Candida is a diploid fungus that causes disease by invading the host’s mucous membranes and tissues, triggering local and systemic inflammatory responses.99 There are many species of fungi in the genera Candida, but only a few are pathogenic to humans, mainly Candida albicans, Candida tropicalis, Candida parapsilosis, and Candida dubliniensis.100 Only 5% (3/63) of the systematically reviewed studies investigated the differences in fungi composition between children with and without obesity. As there were fewer than three studies for the same fungi, these studies were not included in Figure S1. The study by Grigorova et al.51 reported that Candida was not detected in any of the tested feces specimens from children with obesity, while in the control group three Candida species were detected: C. albicans, Candida glabrata, and Candida krusei. Borgo et al.88 also found a relatively low abundance of C. albicans and Saccharomyces cerevisiae in children with obesity compared to the normoweight children. Furthermore, C. parapsilosis and C. glabratawere detected in normoweight children but not children with obesity. These findings are contrary to research in mice that show C. parapsilosis stimulates the formation of fatty acids by producing a fungal lipase that causes obesity.101 Peng et al.38 report that Basidiomycota sp. was more abundant in children with obesity. Reporting gut mycobiota in human fecal specimens and its presence and relative abundance is currently nascent without a consensus on what is considered an “ideal mycobiome”.102

Previously, in vitro culturing was the major method used by researchers to study fungi from the intestinal environment. Despite recent breakthroughs in molecular approaches that eliminate the need to cultivate bacteria, this research has mostly concentrated on the non-fungi component of this ecosystem. Consequently, few studies have included the gut mycobiome and even fewer in the pediatric population. It is noteworthy to emphasize that the gut is a diverse and intricate ecosystem within the human body. Research findings indicate that the most prevalent genera in the mycobiome of a healthy adult gut include Candida, Saccharomyces, and Cladosporium.103 Candida spp. continues to be identified as the prevailing fungi species among individuals aged 65 years and older.104 Nevertheless, in diseases such as obesity105 and inflammatory bowel disease,106 an increase in Candida spp. and its inverse correlation with bacterial microbiome diversity have been repeatedly observed. Some studies suggest that Candida spp. may break down starch in foods, releasing simple sugars that are fermented by other bacteria.107 Furthermore, C. albicans was observed to decrease the levels of dissolved oxygen in its immediate environment, creating favorable conditions for the proliferation of anaerobic bacteria, such as Clostridioides difficile.108,109 Overall, observational and exploratory investigations of gut fungi in children with obesity have highlighted the significant role of dysbiosis of the gut microbes in the pathogenesis of obesity. Understanding cross-kingdom microbial interactions between gut microorganisms in pediatric obesity is important for elucidating the relationship between obesity and microbiota.

2.4. Variations in viruses composition

Human feces contain approximately 108 to 109 viruses per gram of feces with DNA bacteriophages predominating and RNA viruses being only a minor fraction of the gut.110,111 Maya-Lucas et al.78 noted that the relative abundance of human herpesvirus 4 (NC-007605.1) increased from 0.08% in normoweight children to 1.53% in children with obesity, while Torque teno midi virus 1 (NC-009225.1) decreased from 2.21% in normoweight children to 0.72% in children with obesity. Furthermore, specific virus particles, notably bacteriophages, might play an essential role in the maintenance of certain strains of gut bacteria associated with the development of obesity.112 Shirley et al.113 used metagenomic sequencing to evaluate virus-like particles (VLPs) in the feces of children with obesity. The results indicate an increase in the prevalence and variety of all phage categories in children with obesity and metabolic syndrome. In the same cohort of children, it was observed that the virome of children with obesity was predominantly dominated by Caudovirales, with only Inoviridae showing a significant increase in the number of viruses.114 Intriguingly, further analysis of the enteroviruses of the same cohort of children revealed that loss of CrAssphage stability was associated with obesity, with the effect being more pronounced in cases of obesity accompanied by metabolic syndrome.115 However, additional research is required to comprehend the underlying reasons behind the decrease in CrAssphage variety and abundance in individuals with obesity and metabolic syndrome, which contradicts the observed patterns in all phagosomes.

The enteric viruses and phage-bacteria interaction are also crucial for health effects. According to a study by Dutilh et al.,116 “CrAss-like” phages can infect Bacteroidetes taxa such as Prevotella intermedia and Bacteroides spp. and persist inside these hosts. Phages can coexist with gut bacteria for long periods and have the potential to modulate the gut microbiota.117 Furthermore, it has been suggested that successful treatments against Clostridioides difficile utilizing bacteria-free fecal filtrate imply that phagosome modification may be a useful therapeutic technique to stabilize bacterial eubiotics in the microbiome.118 As in other niches, phages play a crucial part in childhood obesity and the disorders that accompany it. Although the nature of all viruses and their role in the health or dysbiosis states of the intestinal microbiome are not currently known, the evidence indicates that their role may be fundamental for bacterial dynamics, pointing to the need to perform deep gut virome studies to elucidate their ecological role in this niche.

2.5. Variations in archaea composition

Methanobacteriales are the most abundant order of archaea and their members can produce methane, reduced CO2 or methanol with H2 as the main electron donor. The main methanogens in the gut microbiota are Methanobrevibacter smithii,119 Methanosphaera stadtmanae,120 and Methanomassilicoccus luminyesis.121 Animal studies have suggested that archaea, specifically M. smithii, may play a role in the development of obesity. Samuel and Gordon122 observed that M. smithii playeda critical role in facilitating an increased capacity of B. thetaiotaomicron to digest polyfructose-containing glycans leading to increased production of SCFAs and total liver triglycerides in mice.

However, meta-analysis showed no statistically significant difference in the absolute abundance of Methanobrevibacter smithii (3 studies providing 2 estimates, SMD = 0.15, 95% CI −0.30, 0.60, I2 = 0%, p = 0.52) between obese children and controls (Figure 1). In addition, there was an increase in the relative abundance of a different unclassified species of Methanobrevibacter spp. in children with obesity.78 Members of this genera have productive saccharolytic activity, allowing polysaccharide digestion in the gut.123 Although the reviewed studies have not yielded similar findings as observed in animals, it should be noted that this does not negate the potential significance of methanogens in metabolic dysregulation among children with obesity. However, it is important to acknowledge that the current data available are limited and further research is warranted to elucidate their precise role in obesity.

2.6. Childhood obesity and gut microbiota: outstanding questions

In this review report, although we identified some gut microbiota characteristics of pediatric obesity, it is important to note that our target population is in broad and diversified in terms of physiological developmental stages. The reported results exhibit a high degree of heterogeneity, with confounding factors such as diet, geography, age, sample size, and testing methods often influencing the composition of gut microbiota. Furthermore, whether changes in the gut microbiota precede the development of obesity in children or if they reflect the obese phenotype as a whole remains to be answered. The interrelationship between nutrition, microbiota, genetic factors, immunity, host life behaviors, and the development of childhood obesity is complex, making it challenging to provide definitive conclusions on this subject. Furthermore, the primary succession of the gut microbiota in the human life cycle ends with the formation of the climacteric community, which is thought to be achieved during adolescence and largely maintained into adulthood; this community is characterized by relative stability.18,124 Therefore, this remains a very challenging area of research, not only to determine the causal relationship between gut microbiota and obesity but also to propose potential effective interventions that can host a healthier microbiota from childhood and thus promote a better metabolic balance of the organism.

3. Effects of dietary fibers on the gut microbiota of children with obesity

3.1. Microbial metabolism of dietary fibers and obesity implications

DFs serve as a crucial energy source for microorganisms residing in the gastrointestinal tract. Under specific intestinal conditions, anaerobic bacteria ferment complex carbohydrates by activating mechanisms consisting of critical enzymes and metabolic pathways that produce metabolites such as SCFAs (Figure 2).

Figure 2.

Figure 2.

A summary of the mechanisms by which DFs initiate metabolic changes to combat the development of childhood obesity. These mechanisms include the production of microbial metabolic products, noting SCFAs, a decrease in luminal pH and O2, regulation of the immune system (modulating cytokine production), improving gut barrier function, promoting satiety through gut-brain signaling, and enhancing oxidative metabolism.

SCFAs are organic compounds composed predominantly of acetate, propionate, and butyrate, and one of their mechanisms of action in obesity is to regulate intestinal endocrine signals, influencing glucose and lipid metabolism. Acetate, butyrate, and propionate can interact with G protein-coupled receptors (GPCR41, GPCR43)125–127 leading to the activation of anorexigenic hormones such as glucagon-like peptide 1 (GLP-1) and peptide secretion YY (PYY).128,129 These hormones play a crucial role in regulating appetite, reducing energy intake and helping to promote glucose metabolism.130 SCFAs further improve oxidative metabolism and insulin sensitivity in the liver and adipose tissue by interacting with the G protein-coupled receptor protein FFAR3 and promoting uncoupling protein 2 (UCP2) activity.131 Another mechanism of action of SCFA in improving obesity is the reduction of metabolic endotoxemia and inflammation. Obesity is thought to lead to a “leaky gut” and subsequent metabolic endotoxemia (increased serum Lipopolysaccharides [LPS] levels) in animal models and to some extent in humans.132 LPS appears to bridge the gap between the gut microbiota and inflammation, and once it is translocated into systemic circulation, it binds to Toll-like receptor 4 and initiates the production of pro-inflammatory cytokines.133 SCFAs also play an anti-inflammatory role by regulating the size and function of the colonic regulatory T cells, leading to a decrease in pro-inflammatory factors (e.g., IL-6, IL-8) in intestinal epithelial cells and metabolic tissues (e.g., adipose tissue).134 Furthermore, SCFAs play a crucial role in preserving the integrity of the intestinal mucus barrier and creating a favorable milieu that supports the proliferation of putatively beneficial gut microbes. Bacteroides thetaiotaomicron produces acetate and propionate, which promote differentiation of goblet cells and stimulate mucin secretion. In contrast, F. prausnitzii utilizes acetate to generate butyrate, which attenuates the effect of B. thetaiotaomicron on mucus and helps the epithelial cells to maintain the proper ratio of different types of cells in the secretory lineage, thereby reducing mucus overproduction and protecting the intestinal mucus barrier.135 Butyrate stimulates β-oxidation in mitochondria by activating the nuclear receptor peroxisome proliferator-activated receptor (PPAR) in colonocytes, which maintains anaerobic conditions in the intestinal lumen.136 SCFAs also decrease the pH of the intestinal lumen and prevent the growth of pathobionts.

Collectively, these acids serve as an additional energy source and concurrently regulate various systemic processes by activating signaling cascades within the host. However, it is important to note that observational studies conducted on human obesity have often have frequently yielded disparate findings regarding the quantification of SCFAs in feces or blood. For instance, one study indicated that obese children and adolescents harbor a higher abundance of carbohydrate-fermenting bacteria in their microbiota compared to normoweight counterparts, thereby augmenting SCFAs biosynthesis rates and providing the host with an additional energy source, ultimately stored as lipids or glucose.74 This apparent paradoxical observation implies that in addition to the SCFAs levels made by some members of the gut microbiota, there might be a direct recognition of specific bacterial components by some GPCRs in the host, which in turn modulate other metabolic processes that may contribute to obesity. Furthermore, it is important to note that the physiological mechanisms of SCFAs have primarily been studied in animal models and require further validation through clinical trials.

DFs may also further influence clinical outcomes in obesity by modulating the gut microbiota, thereby affecting the metabolism of the bile acid pool in the gut. Bile acids (BAs) are cholesterol metabolites produced in hepatocytes and specific gut microbes, such as Lactobacillus, Bifidobacterium, and Bacteroides, that can metabolize into secondary BAs through processes of depolymerization, dehydroxylation, and repolymerization.137 Disturbances in the gut microbial population prevent the conversion of primary bile acids, leading to their accumulation and a decrease in secondary bile acids. Primary bile acids act more specifically on G protein-coupled receptors (TGR5), whereas secondary bile acids primarily act on the Farnesoid X receptor (FXR).138 The activation of these receptors has been shown in animal models to cause changes in lipid and carbohydrate metabolism, energy expenditure, and inflammation.137

3.2. Role of dietary fibers in the management of the gut microbiome in children with obesity

As mentioned above, there is evidence that children with obesity have unique gut microbiota signatures that differ from that of healthy children.21,139 Therefore, whether nutritional interventions can redress microbial imbalances in pediatric obesity has garnered increasing attention in the field of nutrition. It is commonly recognized that a healthy diet is a critical environmental factor mediating the composition and metabolic function of the gut microbiota. DFs modulate the gut microbiome primarily including three strategies: 1) with more complex dietary patterns that provide rich sources of intrinsic DFs; 2) with a symbiotic mixture of live microorganisms and selective prebiotics fiber(s); 3) with the isolated fiber(s). The impact of diet on the composition of the gut microbiota is summarized in Table 1.

Table 1.

Studies in which the effects of different DFs on gut microbiota in children with obesity was assessed.

  Trial design Study
Population
Dose Age n Duration Methodology Microbiota Microbial
Diversity
Metabolic Effects References
Intrinsic DFs and high-fiber diet pattern                      
Low fruit and rich in meat Cross-sectional study Patients with Prader – Willi syndrome (overweight or obesity) 23.1 ± 13.4 g/day 5-8 31 4-day food diary 16S rRNA Klebsiella↑
Eubacterium↑
Lactobacillus↑
Lachnoclostridium↓ Murimonas
Alistipes
Prevotella↓
Shannon index Klebsiella are significantly positively correlated with total and LDL-cholesterol, Alistipes
are significantly negatively correlated with body fat mass, total and LDL-cholesterol levels, fasting insulin levels and HOMA-IR.
Garcia-Ribera et al140
51.6% total carbohydrates, 21.9% total sugar, 5.1% fiber, 31.8% total fat, 16.5% total protein Cross-sectional study Overweight or Obesity 16.1 ± 6.7 g/day 12-19 52 24-hour diet recalls 16S rRNA Bacteroides↑
Prevotella↑
Ruminococcus↑
Blautia↑
none stated Fructose intake is negatively associated with the genera E. eligens and Streptococcus thermophilus Jones et al141
High carbohydrate/high fat diet or high protein/high fat diet Prospective study Overweight or Obesity None stated 2-9 70 7-day diet recalls 16s rRNA Bacteroides
Roseburia
Lachnospira
Slackia
Prevotella
Oscillospira
none stated Inflammatory
(IL-15,TNF-α,
IP-10,IL6,IL-8)↑
Rampelli et al142
Higher fiber and nonstarch polysaccharides diet Cross-sectional study Overweight or Obesity 18 g/day 5 319 one months food diary 16S rRNA Faecalibacterium↑ Eubacterium↑
Roseburia↑
Shannon index None stated Leong et al143
High traditional vegetable-based, low fat, sugar diets Cross-sectional study 28 rural children (Buriram) None stated 9-11 45 7-day dietary records 16S rRNA Lachnospiraceae
Peptostreptococcaceae↑
ACE index
Chao1 index
Parabacteroides↑
Butyrate↑
Kisuse et al144
Whole grains, traditional Chinese medicinal foods, and prebiotics diet Dietary intervention trial Prader – Willi syndrome and obesity 50 g/day 3-16 36 0,30,60,90 days Metagenomic sequencing Faecalibacterium↑ Bifidobacterium↑
Clostridium↑
Collinsella↑
Bifidobacterium catenulatum↑
B.longum↑
B.breve↑
Bacteroides↓
Ruminococcus↓
Blautia
Escherichia↓
None stated None stated Li et al145
38 Bifidobacterium spp.↑
B. pseudocatenulatum↑
Chao1 index
Observed OTU Phylogenetic diversity Shannon index
gene richness
Body weight↓
BMI↓ALT↓
AST ↓
Zhang et al146
Proteins and complex carbohydrate patterns Cross-sectional study Overweight
or Obesity
None stated 6-12 46 \ Metagenomic sequencing Holdemania spp.↑
Coprococcus catus↓
none stated z-BMI↓
waist circumference↓
hip circumference↓
Orbe-Orihuela et al147
Whole grains, traditional Chinese medicinal foods, and prebiotics diet Dietary intervention trial Overweight or Obesity 50 g/day 3-18 87 30, 60, 90 days 16S rRNA Bifidobacterium↑
Lactobacillus↑
none stated None stated Hou et al68
Synbiotic                      
Synbiotic
(FOS)
Randomized triple-masked controlled trial Obesity one capsule 6-18 70 8 weeks surface-cultured of plates Lactobacillus
Bifidobacteria↑
none stated Weight↓
Body mass index↓
Z-score↓
IL-6↓
TNFα↓
Hs-CRP↔
Adiponectin↑
Kelishadi et al140
Synbiotic
(FOS)
Single-center, prospective, randomized, double-blind, placebo-controlled clinical study Obesity 1 sachet each day 8-17 54 12 weeks 16S rRNA Compared to baseline:
Bacteroidetes↑
PrevotellaDialister
F/B ratio↓
Compared to placebo groups: Collinsella stercoris species were dominant in the synbiotic group
Chao1 index↔
Observed ASV↔
Simpson index ↔
Shannon index↔
None stated Kilic Yildirim et al148
Isolated fiber                      
Inulin Double-Blind, RCT Obesity 8 g/day 7-12 42 16 weeks 16S rRNA
qPCR
Actinobacteria↑
Bifidobacterium
Collinsella
B. adolescentis
B.longum
Ruminococcus↓
F.prausnitzii↓
E. eligens↓
B.vulgatus↓
R.gauvreauii↓
Shannon index↑
Simpson index ↔
observed OTUs↑
beta-diversity
(p > 0.05)
Body Weight↓
Body Fat (%)↓
trunk fat ↓
IL-6↓
TAG↓
primary bile acids↔
Nicolucci et al149

3.2.1. Effects of intrinsic dietary fibers and high-fibers diet pattern on the gut microbiota of children with obesity

As depicted in Figure 1, Actinobacteria, Prevotella, Bifidobacterium, Dialister, Escherichia, Anaerostipes, Akkermansia, Bacteroides and α-diversity metrics other than the Simpson index showed a tendency to be less prevalent in children with obesity, and although there was no significance, inclusion in the Meta-analysis reported low heterogeneity. In high DFs dietary patterns 11% (1/9) of studies reported an increase in the Shannon index, ACE index and Chao1 index and 11% of studies (1/9) reported a decrease in Chao1 index, Observed OTUs, Phylogenetic diversity and Shannon index. An increase in Prevotella is associated with long-term dietary patterns, complex carbohydrates, or plant-based foods.150 Many of the species that respond to changes in traditional diet intake (high plant-based and low-fat foods) seem to belong to Actinobacteria, which are nutritionally specialized.151,152 Actinobacteria (such as Bifidobacterium, Bifidobacterium spp.) few genera increase in response to high-fiber diets.143,145–147 In response to the Western diet, the abundance of Bacteroides and Klebsiella increased, which was accompanied by a decrease in Prevotella,141 ,142 ,153

Recently, there has been an increasing recognition that consuming DFs naturally present in food (in contrast to consuming isolated DFs as supplements) may be most effective at regulating gut bacteria and enhancing overall health.154 Regarding dietary interventions for childhood obesity, it is important to consider not only whether the gut microbiota is being modulated, but also the improvement of metabolism in children. SCFAs, as key metabolites of fermentation of DFs by the gut microbiota, have a broad impact on various aspects of host physiology,155 Table 1 shows that only one cross-sectional study investigating high-fiber diets provided evidence of increased SCFAs.144 A traditional grain diet improved weight, BMI, and liver function metabolism in children with obesity, according to another study.146 It is important to note that while the effects of high-fiber diets on the body’s metabolism should be examined in relation to the microbiota, these diets may also affect the metabolism directly, independent of the microbiota. Foods such as those replaced by DFs (if they replace monosaccharides) produce additional benefits.

It is tempting to target ecological markers similar to those found in children with healthy diets (higher richness and diversity, stimulation of beneficial microbes), but it is unclear which microbiota response in children should be supported. In conclusion, most studies have shown a strong association between dietary patterns, gut microbiome, and childhood obesity.

3.2.2. Effects of synbiotics mixture (live microorganisms and selective prebiotics fibers) on the gut microbiota of children with obesity

Synbiotic mixtures are a combination of prebiotics and probiotics that may be used to moderate dysfunctional gut microbiomes.156 The effect of synbiotics on α-diversity was not observed in the included studies (only 1 of the 2 studies reported relevant results). Kelishadi140 and colleagues used synbiotics capsules (fructooligosaccharides [FOS] plus Lactobacillus and Bifidobacteria) to intervene in children with obesity for eight weeks and found significant increases in Lactobacillus, as well as significant reductions BMI, IL-6, and tumor necrosis factor-α (TNFα). Another study used a Synbiotics formulation (probiotics mixture including Lactobacillus acidophilus, Lacticaseibacillus rhamnosus, Bifidobacterium bifidum, Bifidobacterium longum, Enterococcus faecium (total 2.5 × 109 CFU/sachet) and FOS (625 mg/sachet) in children with obesity over a 12-week period; compared to baseline, Bacteroidetes, Prevotella, and Dialister significantly increased, F/B ratio and BMI significantly decreased.148 Interestingly, Bacteroidetes was significantly decreased and Dialister, Lactobacillus showed a trend of reduction in children with obesity. These data suggest that synbiotics can drive changes in the gut microbiota of obese children, with the potential to increase the diversity of the gut microbiota. However, synbiotics formulations often require higher than expected dosages of prebiotics for practical use to ensure that substrates are available for both the resident microbiota and the co-administered microbes in a highly competitive microenvironment within the gut.157–159 Therefore, Precise understanding of the synergistic impact of probiotics and prebiotics in synbiotics, as well as the mechanism by which they regulate the gut microbiota in children with obesity, remains challenging within the realm of precision nutrition.

3.2.3. Effects of isolated dietary fibers on the gut microbiota of children with obesity

Isolated DFs can have a significant impact on the composition, diversity and abundance of the microbiome, providing a wealth of substrates for fermentation reactions carried out by specific microbial species.160 Microorganisms possess the necessary enzymes to hydrolyze a wide variety of complex carbohydrates, including glycoside hydrolases, glycosyltransferases, polysaccharide lyases, and carbohydrate esterases.161 As a result, having a variety of DFs (e.g., cellulose, inulin, fructans, resistant starches) in a diet that contains a range of monosaccharide units and α and β linkages is more supportive of a diverse gut microbial community than a diet with a less diverse substrate load (e.g., refined diets).162 Despite the growing body of literature on the ability of isolated DFs to positively improve childhood obesity,163,164 there is limited information on their impact on microbial diversity and abundance. Several studies have demonstrated that the intake of isolated DFs shapes the gut microbiota of children with obesity.149,165 In children with overweight or obesity, 16 weeks of oligofructose-enriched inulin (8 g/day) increased α-diversity (Shannon index, Observed OTUs), associated with an increase in Actinobacteria (such as Bifidobacterium) and a decrease in Firmicutes (such as Ruminococcus).149 Meanwhile, at the level of OTUs, F. prausnitzii decreased significantly. These changes in microbiota outcomes suggest that DFs are better at improving gut microbiota in obese children (Figure 2). Moreover, this prebiotics intervention improved children’s body weight, body fat, IL-6 levels, and triglyceride content. These beneficial effects of isolated DFs on childhood obesity aspects are thought to be at least partially mediated by the microbiota induced changes but more direct evidence still has to be provided.

In summary, intrinsic dietary, synbiotics, and isolated fibers exert specific modulatory effects on the gut microbiota composition and α-diversity in children with obesity. Current evidence does not support that DFs can modulate the bacteria (e.g., Akkermansia, A. muciniphila) found to be significantly decreased in children with obesity in the systematic review. A. muciniphila, a mucin-degrading bacterium, has a negative association with obesity, diabetes, and colitis.166 Supplementation with oligofructose has been shown to increase the number of goblet cells in animals, which subsequently exerts a prebiotic function on A. muciniphila.167 High-fermentable (pectin) fiber also modulates the gut immune environment in mice, which in turn promotes Akkermansia enrichment,168 and the findings suggest that the fiber effect on Akkermansia may be independent of fermentation but rather by improving the gut microecological environment. Furthermore, low-fermentable fibers augment crypt length, goblet cell maturation, and mucin secretion via mechanical stimulation caused by increased stool bulk.169 Cellulose (an insoluble fiber with low-fermentable) supplementation significantly increased Akkermansia abundance in mice with colitis, potentially by promoting mucin secretion from goblet cells.170 Low-fermented fibers effectively accelerate gut transit time, which may have implications for regulating the structure of the gut microbiota.171 However, there is a lack of clinical evidence investigating the impact of DF supplementation on Akkermansia in children with obesity, particularly the effect of isolated low-fermentable fiber on gut microbiota in this population.

3.2.4. Dietary fibers and gut microbiota in children with obesity: remaining unsolved questions

The identification and implementation of suitable nutritional intervention techniques for modulating the gut microbiota in children with obesity is a prominent subject of interest for forthcoming scientific investigations. For many years, the study of DF-microbiota interactions using a reductionist approach has elucidated DFs dependent and independent effects on specific microbiota.172 Currently, dietary patterns with high DFs are an important strategy in nutritional intervention programs. As shown in Table 1, the more complex dietary interventions assessing the effect of DFs on the gut microbiota in children with obesity provide relatively more evidence, but substantial evidence is still lacking in general (such as randomized clinical trials). Further investigation is required to ascertain the fundamental mechanisms responsible for the distinct impacts of isolated fibers and intrinsic fibers on the gut microbiota of children with obesity.173 In addition, DFs have a significant impact on the composition, diversity, abundance, and function of the gut microbiota in children with obesity, but the strength of the evidence remains insufficient. As shown in Figure 1 and Figure S1, there is a clear downward trend in Bacteroidetes, Prevotella and Bacteroidetes among children with obesity. Not surprisingly, following a high DF intake, all of these bacteria in children with obesity moved in a favorable direction (Table 1). There is still a large degree of uncertainty about the extent to which the effects attributed to DFs on obesity are mediated by the gut microbiota in children and the key species involved in the effects. However, current evidence still does not support the extent to which the effects of DFs on obesity are mediated by the gut microbiota in children and the key species involved in the effects.

However, there is no unanimous agreement on the recommended amounts of fiber for children with obesity, and there is a lack of evidence regarding practical advice on the appropriate type and quantity of DFs to be consumed. It is imperative to acknowledge that existing guidelines for DF consumption in children pertain to the overall intake of fibers and fail to sufficiently account for the origin and type of fibers.174 This is important because various types and sources of fibers exhibit distinct physiological effects. For instance, the source (natural or synthetic, etc.), chemical composition (chain length, lignification, etc.), and physicochemical properties (solubility, viscosity, fermentability, etc.) of DFs affect not only the gut microbiota but also health outcomes.175,176 Hence, in the formulation of a nutritional intervention program targeting children with obesity, it is imperative to take into account not only the overall daily consumption of DFs but also to acknowledge the origin and type of fiber duly. Furthermore, additional research is required to investigate the physiological characteristics of the association between DFs and the health of children with obesity, as well as the optimal ratios of the different fiber sources.

Furthermore, the mechanism of action of DFs in regulating the gut microbiota to ameliorate childhood obesity has not been fully explored. We should be aware that the gut microbiota is an extraordinarily mutually dependent community in which waste products from one organism become nutrients for other organisms via substrate and metabolite cross-feeding. The cross-feeding behaviors of primary degraders and cross-feeders result in a wide range of substrates and metabolites to support microbiota diversity, allowing the gut to maintain a stable community.177 For example, in vitro analysis revealed that the relationship between bifidobacteria and F. prausnitzii, in the presence of inulin-type fructans, could be commensal or competitive, and this relationship was dependent on the bifidobacterial strain and its capacity for prebiotics degradation.178 More importantly, the gut microbiome profile of children with obesity is complex, with changes in the fungi, viruses, and archaea organisms in addition to bacterial homeostasis. To date, no studies have systematically reported the effects of DFs on the fungi, viruses and archaea organisms in the gut of children with obesity. Therefore, we should further our understanding of the complex relationships between fibers, gut microbiota, and childhood obesity, which can lead to the development of low-cost, safe, and efficacious “microbially-directed” dietary interventions.

4. Future direction

Information on fiber-microbiome-obesity interactions in children needs to be further validated and refined. To be actionable, integration of the gut microbiome into the prevention of childhood obesity requires evidence of the causal contributions of the microbiome to obesity and the mechanistic physiological effects of fibers. Establishing the causal role of the gut microbiome in susceptibility to chronic diseases such as childhood obesity remains a challenge, which is complicated in nutrition research because the interactions between fibers, the gut microbiome, and human health are complex and multidirectional. These complexities must be considered in the design of future nutrition research to elucidate what factors, including the gut microbiome, mediate the effects of fibers in childhood obesity. We suggest extending future studies through an experimental framework using three pillars that integrate the gut microbiome into stages of fiber nutrition research on children with obesity (Figure 3).

Figure 3.

Figure 3.

Experimental framework of the fiber-microbiota-obesity study in children. (a) microbiome epidemiology can elucidate associations between the gut microbiome, environmental factors (e.g., diet, age, sex, etc.), and physiologic effects caused by environmental factors in children with obesity. This information does not establish causality, but can help to uncover gut microbiome characteristics in children with obesity and contribute to the evidence base for precision nutrition. (b) RCT can determine the causal relationship between DFs and childhood obesity and provide strong evidence of this. N of 1 employs the “individual” as the subject of the study, allowing for the development of personalized and precise nutritional requirements. Combined with microbiome-based multi-omics analysis, microbial biomarkers for precision nutrition strategies can be identified. However, highly personalized microbiomes and causal inference of their role in DF’s effects still pose challenges. (c) causal relationships and potential mechanisms can be further established in in vivo (animal models) and in vitro studies. Causal and mechanistic information provides additional evidence for the development of dietary guidelines and targeted nutritional strategies for DFs. BONCAT, bio-orthogonal non-canonical amino acid tagging; CRISPR, clustered, regularly interspaced, short palindromic repeats; DFs, dietary fibers; FACS, fluorescence-activated cell sorting; RCT, randomized controlled trial.

4.1. Discovery of microbiome in children with obesity generates hypothesis for precision nutrition

Characterization of the gut microbiome may help in the prevention of childhood obesity and related metabolic diseases in the future. Furthermore, the application of multi-omics techniques (e.g., metagenomics, proteomics, metabolomics) and advanced analytical methods, such as machine learning-mediated analyses, has laid the groundwork for elucidating the underlying mechanisms and cause-and-effect relationships of biological pathways.179,180 Recent studies have expanded on this framework by combining the microbiome and the epidemiology of childhood obesity to elucidate the microbiological profile of children with obesity.181 Although such observational studies cannot establish causality, high-quality, large-scale cohort studies can help provide an evidence basis for the causes of childhood obesity. Integrating childhood obesity with microbiome epidemiology will facilitate the identification of links between the microbiome and childhood obesity and the characterization of the gut microbiota in children with obesity. The underlying mechanisms and biological plausibility of these interactions, as well as their value as diagnostic markers, can then be confirmed in experimental systems to confirm the system.

4.2. Integrating the microbiome into nutritional intervention trials in childhood obesity

More clinical research using fiber interventions in childhood obesity, aimed at addressing the gut microbial perspective is needed, as the number of studies identified thus far is small, as there is a paucity of data available. Notably, although numerous epidemiological studies have demonstrated an association between DF intake and childhood obesity, there have been some inconsistent reports. In contrast, a clinical trial by Liber and Szajewska182 showed that consumption of 8 g/d of oligofructose in otherwise healthy overweight or children with obesity aged 7–11 years and 15 g/d in children aged 12–18 years did not result in improvements in BMI or Z-score after 12 weeks compared with placebo control. There were also no improvements in the secondary outcomes of change in body weight and difference in absolute body fat or other metabolic outcomes. A systematic review and meta-analysis of the relationship between DFs and diabetes management found similar results.183 These findings point to variation in the response of DFs to obesity-related metabolic dysregulation in children, which is thought to be due to the high degree of individualization of gut microbes in children. The research of Deehan et al. supports this theory.184 A DF intervention is likely to have no effect on an individual whose microbiome does not include keystone species or other genera that encode the enzymatic machinery to degrade those specific fiber types. This should thus be considered when designing future studies.

Randomized controlled trials are the gold standard for establishing causal effects. Optimally, feeding studies with single ingredient modifications using crossover designs with sufficient washout periods are ideal to assess not only the impact of DFs on gastrointestinal bacteria taxa, but also microbial metabolites and other physiological measures of health such as body composition, blood cholesterol, glycemia, and inflammation. It is critical to include controls, which can eliminate inter-individual differences in individual-specific factors (e.g., microbiome, genetics, metabolite profiles, and baseline clinical measures).185 Furthermore, for the highly individualized gut microbiome of children with obesity, a precision nutrition research design (N of 1 trial) can be used to screen for nutritional interventions that are appropriate for them, using themselves as a control, to achieve true, albeit difficult, precision nutrition.186

Studies should account for variation in other confounding variables such as demographics (age187 and gender188) and lifestyle factors (such as habitual diet).189 Obviously, in view of the complexity of the confounding variables, we need advanced statistical and modeling methods to carry out data analysis. For example, regression and correlation analyses can be applied to determine associations between fibers-induced changes in microbiome composition/functionality and clinical and mechanistic endpoints.190 Machine-learning models can determine if fibers-induced physiological changes can be predicted by effects on the microbiome or biological processes in the host impacted by the microbiome.191 Integration of the microbiome into human intervention trials could provide putative mechanistic explanations for the role of the microbiome in the effects of fibers on childhood obesity, as well as diagnostic microbiome-based biomarkers for precision nutrition strategies.

4.3. Mechanistic insight the microbiomes role in the effects of fibers

Human studies can be paired with animal models to determine whether the microbiome plays a causal role in the physiological effect, identify the causal components within the microbiome, and determine underlying mechanisms192 (Figure 3 C1). The complex and dynamic transcriptional changes of the gut microbiota throughout time, transit, and perturbation in the gut of gnotobiotic animals are recorded by integrating RNA-derived spacers from the transcriptome of gut bacteria into plasmid DNA-encoded Clustered, regularly interspaced, short palindromic repeats (CRISPR) arrays using FsRT-Cas1-Cas2193 (Figure 3C1). This scalable, noninvasive system for assessing intestinal function in vivo archives characteristics of microbial signatures of physiological or pathological states.177 Transcriptome-scale recordings elucidate microbial responses to alterations in the intraluminal environment across nutrition and microbe-microbe interactions.177 Furthermore, a method for tracing isotopes into bacterial-specific protein sequences has been developed that enables the mapping of nutrient routes in vivo and reveals how diet affects the makeup of the microbiome (Figure 3 C1). However, mechanistic studies in animals can be confounded as it is not possible to focus solely on the gut microbiota due to the presence of the host.194 Noteworthy, in vitro gut fermentation models can be used to supplement human and animal studies and overcome some of the limitations of in vivo models195 (Figure 3 C2). Researchers can use a variety of in vitro models in which single or multiple vessels are inoculated with fresh human feces or a defined microbial community. These vessels are operated under anaerobic conditions, and microbial communities are grown with temperature, pH, growth medium, and transit time set to mimic a specific intestinal segment. This tool can be used in combination with new technologies (e.g., Bio-orthogonal non-canonical amino acid tagging-Fluorescence-activated cell sorting [BONCAT-FACS],196 Figure 3 C2) to test the direct effects of prebiotics and DFs on the gut microbiota of children with obesity in the absence of confounding factors. While both animal and in vitro models can mimic some aspects of the human gut microbiome, neither model can fully replicate all of the functions of the human gut. Despite their limitations, their microbiomes, if well controlled and combined with a multi-omics approach, have the capacity to complement human studies as they establish the mechanistic basis for the impact of fibers on children with obesity.

5. Conclusion

The current evidences suggest that a higher intake of DFs may have a beneficial impact on metabolic health through modifications to the gut microbiota.175 Personalized fiber intake recommendations and improved obesity outcomes ought to be feasible with the integration of individual-level genetic and metabolic trait data, along with the development of microbial sequencing technologies and analytical methodologies. Microbiome research possesses the opportunity to furnish exhaustive insights into facets of nutrition strategy. However, the gut microbial signatures observed in children with obesity relative to normoweight children remain unclear and require further research. Overall, in order to improve the convincing evidence supporting dietary guidelines for children, it is imperative that facets of childhood obesity prevention research incorporate gut microbial features.

Supplementary Material

Supplemental Material

Acknowledgments

Z.Y., J.L. and Z.Z. initiated the idea, scope, and outline of this review paper. Z.Y., C.C., and M.Y. conducted the systematic review and data extraction. Z.Y. and Z.Z. drafted the manuscript. Z.Y., J.L. and Z.Z. were responsible for conceptualizing the content of the figure. E.C.D., C.C., K.L.M., E.W., G.L., J.L. J.L. and Z.Z. critically reviewed and edited drafts of the manuscript. All authors approved the final version.

Funding Statement

This work was supported by the National Natural Science Foundation of China [No. 32202068] and the Natural Science Foundation of Fujian Province [No. 2022J01332].

Abbreviations

BAs

Bile acids

BONCAT-FACS

Bio-orthogonal non-canonical amino acid tagging-Fluorescence-activated cell sorting

CRISPR

Clustered, regularly interspaced, short palindromic repeats

DFs

Dietary fibers

FOS

fructooligosaccharides

FXR

Farnesoid X receptor

GPCR

G protein-coupled receptors

GLP-1

glucagon-like peptide 1

HDL

high-density lipoprotein

LPS

Lipopolysaccharides

PPAR

peroxisome proliferator-activated receptor

PYY

peptide secretion YY

RT-PCR

Real-time PCR

SCFAs

short-chain fatty acids

TNFα

tumor necrosis factor-α

UCP2

uncoupling protein 2

VLPs

virus-like particles

BMI

body mass index

Disclosure statement

No potential conflict of interest was reported by the author(s).

Supplementary material

Supplemental data for this article can be accessed online at https://doi.org/10.1080/19490976.2024.2387796

References

  • 1.Bjerregaard LG, Jensen BW, Ängquist L, Osler M, Sørensen TIA, Baker JL.. Change in overweight from childhood to early adulthood and risk of type 2 diabetes. N Engl J Med. 2018;378(14):1302–28. doi: 10.1056/NEJMoa1713231. PMID: 29617589. [DOI] [PubMed] [Google Scholar]
  • 2.Tricò D, Caprio S, Rosaria Umano G, Pierpont B, Nouws J, Galderisi A, Kim G, Mata MM, Santoro N. Metabolic features of nonalcoholic fatty liver (NAFL) in obese adolescents: findings from a multiethnic cohort. Hepatology. 2018;68(4):1376–1390. doi: 10.1002/hep.30035. PMID: 29665034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ortega FB, Lavie CJ, Blair SN. Obesity and cardiovascular disease. Circ Res. 2016;118(11):1752–1770. doi: 10.1161/circresaha.115.306883. PMID: 27230640. [DOI] [PubMed] [Google Scholar]
  • 4.World Obesity Federation, World Obesity Atlas . 2023;https://data.worldobesity.org/publications/?cat=19.
  • 5.Singh AS, Mulder C, Twisk JW, van Mechelen W, Chinapaw MJ. Tracking of childhood overweight into adulthood: a systematic review of the literature. Obes Rev. 2008;9(5):474–488. doi: 10.1111/j.1467-789X.2008.00475.x. PMID: 18331423. [DOI] [PubMed] [Google Scholar]
  • 6.Quek YH, Tam WWS, Zhang MWB, Ho RCM. Exploring the association between childhood and adolescent obesity and depression: a meta-analysis. Obes Rev. 2017;18(7):742–754. doi: 10.1111/obr.12535. PMID: 28401646. [DOI] [PubMed] [Google Scholar]
  • 7.Pereira LJ, Hinnig PF, Matsuo LH, Di Pietro PF, de Assis Maa, Vieira FGK. Association between lifestyle patterns and overweight and obesity in adolescents: a systematic review. Br J Nutr. 2022;1–45. doi: 10.1017/s0007114522000228. PMID: 35086569. [DOI] [PubMed] [Google Scholar]
  • 8.Singh S, Ricardo-Silgado ML, Bielinski SJ, Acosta A. Pharmacogenomics of medication-induced weight gain and antiobesity medications. Obesity (Silver Spring). 2021;29(2):265–273. doi: 10.1002/oby.23068. PMID: 33491309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Tzvetkov MV, Vormfelde SV, Balen D, Meineke I, Schmidt T, Sehrt D, Sabolić I, Koepsell H, Brockmöller J. The effects of genetic polymorphisms in the organic cation transporters OCT1, OCT2, and OCT3 on the renal clearance of metformin. Clin Pharmacol Ther. 2009;86(3):299–306. doi: 10.1038/clpt.2009.92. PMID: 19536068. [DOI] [PubMed] [Google Scholar]
  • 10.Mohammadian Khonsari N, Khashayar P, Shahrestanaki E, Kelishadi R, Mohammadpoor Nami S, Heidari-Beni M, Esmaeili Abdar Z, Tabatabaei-Malazy O, Qorbani M. Normal weight obesity and cardiometabolic risk factors: a systematic review and meta-analysis. Front Endocrinol (Lausanne). 2022;13:857930. doi: 10.3389/fendo.2022.857930. PMID: 35399938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bouter KE, van Raalte Dh, Groen AK, Nieuwdorp M. Role of the gut microbiome in the pathogenesis of obesity and obesity-related metabolic dysfunction. Gastroenterology. 2017;152(7):1671–1678. doi: 10.1053/j.gastro.2016.12.048. PMID: 28192102. [DOI] [PubMed] [Google Scholar]
  • 12.Koboziev I, Reinoso Webb C, Furr KL, Grisham MB. Role of the enteric microbiota in intestinal homeostasis and inflammation. Free Radic Biol Med. 2014;68:122–133. doi: 10.1016/j.freeradbiomed.2013.11.008. PMID: 24275541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 2016;14(8):e1002533. doi: 10.1371/journal.pbio.1002533. PMID: 27541692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Brahe LK, Astrup A, Larsen LH. Can we prevent obesity-related metabolic diseases by dietary modulation of the gut microbiota? Adv Nutr. 2016;7(1):90–101. doi: 10.3945/an.115.010587. PMID: 26773017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.de Wit Df, Hanssen NMJ, Wortelboer K, Herrema H, Rampanelli E, Nieuwdorp M. Evidence for the contribution of the gut microbiome to obesity and its reversal. Sci Transl Med. 2023;15(723):eadg2773. doi: 10.1126/scitranslmed.adg2773. PMID: 37992156. [DOI] [PubMed] [Google Scholar]
  • 16.Duranti S, Ferrario C, van Sinderen D, Ventura M, Turroni F. Obesity and microbiota: an example of an intricate relationship. Genes Nutr. 2017;12(1):18. doi: 10.1186/s12263-017-0566-2. PMID: 28638490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Cunningham AL, Stephens JW, Harris DA. A review on gut microbiota: a central factor in the pathophysiology of obesity. Lipids Health Dis. 2021;20(1):65. doi: 10.1186/s12944-021-01491-z. PMID: 34233682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222–227. doi: 10.1038/nature11053. PMID: 22699611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ronan V, Yeasin R, Claud EC. Childhood development and the microbiome—the intestinal microbiota in maintenance of health and development of disease during childhood development. Gastroenterology. 2021;160(2):495–506. doi: 10.1053/j.gastro.2020.08.065. PMID: 33307032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Derrien M, Alvarez AS, de Vos WM. The gut microbiota in the first decade of life. Trends Microbiol. 2019;27(12):997–1010. doi: 10.1016/j.tim.2019.08.001. PMID: 31474424. [DOI] [PubMed] [Google Scholar]
  • 21.Wang Y, Salonen A, Jian C. Can prebiotics help tackle the childhood obesity epidemic? Front Endocrinol (Lausanne). 2023;14:1178155. doi: 10.3389/fendo.2023.1178155. PMID: 37305030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Korczak R, Slavin JL. Definitions, regulations, and new frontiers for dietary fiber and whole grains. Nutr Rev. 2020;78(Supplement_1):6–12. doi: 10.1093/nutrit/nuz061. PMID: 32728750. [DOI] [PubMed] [Google Scholar]
  • 23.Gibson GR, Hutkins R, Sanders ME, Prescott SL, Reimer RA, Salminen SJ, Scott K, Stanton C, Swanson KS, Cani PD, et al. Expert consensus document: the international scientific association for probiotics and prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat Rev Gastroenterol Hepatol. 2017;14(8):491–502. doi: 10.1038/nrgastro.2017.75. PMID: 28611480. [DOI] [PubMed] [Google Scholar]
  • 24.Cronin P, Joyce SA, O’Toole PW, O’Connor EM. Dietary fibre modulates the gut microbiota. Nutrients. 2021;13(5):1655. doi: 10.3390/nu13051655. PMID: 34068353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Tuncil YE, Thakkar RD, Arioglu-Tuncil S, Hamaker BR, Lindemann SR. Subtle variations in dietary-fiber fine structure differentially influence the composition and metabolic function of gut microbiota. mSphere. 2020;5(3). doi: 10.1128/mSphere.00180-20. PMID: 32376698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Gill SK, Rossi M, Bajka B, Whelan K. Dietary fibre in gastrointestinal health and disease. Nat Rev Gastroenterol Hepatol. 2021;18(2):101–116. doi: 10.1038/s41575-020-00375-4. PMID: 33208922. [DOI] [PubMed] [Google Scholar]
  • 27.Zhu L, Baker SS, Gill C, Liu W, Alkhouri R, Baker RD, Gill SR. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: a connection between endogenous alcohol and NASH. Hepatology. 2013;57(2):601–609. doi: 10.1002/hep.26093. PMID: 23055155. [DOI] [PubMed] [Google Scholar]
  • 28.Yuan X, Zhang Y, Lin X, Yang X, Chen R. Association of gut microbiota and glucose metabolism in children with disparate degrees of adiposity. Pediatr Obes. 2023;18(4):e13009. doi: 10.1111/ijpo.13009. PMID: 36704910. [DOI] [PubMed] [Google Scholar]
  • 29.Yuan X, Chen R, Zhang Y, Lin X, Yang X, McCormick KL. Gut microbiota of Chinese obese children and adolescents with and without insulin resistance. Front Endocrinol (Lausanne). 2021;12:636272. doi: 10.3389/fendo.2021.636272. PMID: 33815293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Yuan X, Chen R, McCormick KL, Zhang Y, Lin X, Yang X. The role of the gut microbiota on the metabolic status of obese children. Microb Cell Fact. 2021;20(1):53. doi: 10.1186/s12934-021-01548-9. PMID: 33639944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wei Y, Liang J, Su Y, Wang J, Amakye WK, Pan J, Chu X, Ma B, Song Y, Li Y, et al. The associations of the gut microbiome composition and short-chain fatty acid concentrations with body fat distribution in children. Clin Nutr. 2021;40(5):3379–3390. doi: 10.1016/j.clnu.2020.11.014. PMID: 33277072. [DOI] [PubMed] [Google Scholar]
  • 32.Wei J, Dai W, Pan X, Zhong Y, Xu N, Ye P, Wang J, Li J, Yang F, Luo J, et al. Identifying the novel gut microbial metabolite contributing to metabolic syndrome in children based on integrative analyses of microbiome-metabolome signatures. Microbiol Spectr. 2023;11(2):e0377122. doi: 10.1128/spectrum.03771-22. PMID: 36794949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Visuthranukul C, Sriswasdi S, Tepaamorndech S, Joyjinda Y, Saengpanit P, Kwanbunbumpen T, Panichsillaphakit E, Uaariyapanichkul J, Chomtho S. Association of human intestinal microbiota with lifestyle activity, adiposity, and metabolic profiles in Thai children with obesity. J Nutr Metab. 2022;2022:1–14. doi: 10.1155/2022/3029582. PMID: 35637874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Vazquez-Moreno M, Perez-Herrera A, Locia-Morales D, Dizzel S, Meyre D, Stearns JC, Cruz M. Association of gut microbiome with fasting triglycerides, fasting insulin and obesity status in Mexican children. Pediatr Obes. 2021;16(5):e12748. doi: 10.1111/ijpo.12748. PMID: 33191616. [DOI] [PubMed] [Google Scholar]
  • 35.Shin S, Cho KY. Altered gut microbiota and shift in bacteroidetes between young obese and normal-weight Korean children: a cross-sectional observational study. Biomed Res Int. 2020;2020:1–19. doi: 10.1155/2020/6587136. PMID: 32908903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.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(1):95–105. doi: 10.1111/1462-2920.13463. PMID: 27450202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Pierri L, Saggese P, Guercio Nuzio S, Troisi J, Di Stasi M, Poeta M, Savastano R, Marchese G, Tarallo R, Massa G, et al. Relations of gut liver axis components and gut microbiota in obese children with fatty liver: a pilot study. Clin Res Hepatol Gastroenterol. 2018;42(4):387–390. doi: 10.1016/j.clinre.2018.03.015. PMID: 29773420. [DOI] [PubMed] [Google Scholar]
  • 38.Peng Y, Tan Q, Afhami S, Deehan EC, Liang S, Gantz M, Triador L, Madsen KL, Walter J, Tun HM, et al. The gut microbiota profile in children with prader–Willi Syndrome. Genes (Basel). 2020;11(8):11. doi: 10.3390/genes11080904. PMID: 32784572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Pan X, Kaminga AC, Liu A, Wen SW, Luo M, Gut Microbiota LJ, Glucose. Gut microbiota, glucose, lipid, and water-electrolyte metabolism in children with nonalcoholic fatty liver disease. Front Cell Infect Microbiol. 2021;11:683743. doi: 10.3389/fcimb.2021.683743. PMID: 34778099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Nirmalkar K, Murugesan S, Pizano-Zárate ML, Villalobos-Flores LE, García-González C, Morales-Hernández RM, Nuñez-Hernández JA, Hernández-Quiroz F, Romero-Figueroa MDS, Hernández-Guerrero C, et al. Gut microbiota and endothelial dysfunction markers in obese Mexican children and adolescents. Nutrients. 2018;10(12):2009. doi: 10.3390/nu10122009. PMID: 30572569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Murugesan S, Ulloa-Martínez M, Martínez-Rojano H, Galván-Rodríguez FM, Miranda-Brito C, Romano MC, Piña-Escobedo A, Pizano-Zárate ML, Hoyo-Vadillo C, García-Mena J. Study of the diversity and short-chain fatty acids production by the bacterial community in overweight and obese Mexican children. Eur J Clin Microbiol Infect Dis. 2015;34(7):1337–1346. doi: 10.1007/s10096-015-2355-4. PMID: 25761741. [DOI] [PubMed] [Google Scholar]
  • 42.Monga Kravetz A, Testerman T, Galuppo B, Graf J, Pierpont B, Siebel S, Feinn R, Santoro N. Effect of gut microbiota and PNPLA3 rs738409 variant on nonalcoholic fatty liver disease (NAFLD) in obese youth. J Clin Endocrinol Metab. 2020;105(10):e3575–85. doi: 10.1210/clinem/dgaa382. PMID: 32561908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Méndez-Salazar EO, Ortiz-López MG, Granados-Silvestre M, Palacios-González B, Menjivar M. Altered gut microbiota and compositional changes in Firmicutes and Proteobacteria in Mexican undernourished and obese children. Front Microbiol. 2018;9:2494. doi: 10.3389/fmicb.2018.02494. PMID: 30386323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ma T, Wu Z, Lin J, Shan C, Abasijiang A, Zhao J. Characterization of the oral and gut microbiome in children with obesity aged 3 to 5 years. Front Cell Infect Microbiol. 2023;13:1102650. doi: 10.3389/fcimb.2023.1102650. PMID: 37065198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.López-Contreras BE, Morán-Ramos S, Villarruel-Vázquez R, Macías-Kauffer L, Villamil-Ramírez H, León-Mimila P, Vega-Badillo J, Sánchez-Muñoz F, Llanos-Moreno LE, Canizalez-Román A, et al. Composition of gut microbiota in obese and normal-weight Mexican school-age children and its association with metabolic traits. Pediatr Obes. 2018;13(6):381–388. doi: 10.1111/ijpo.12262. PMID: 29388394. [DOI] [PubMed] [Google Scholar]
  • 46.Karvonen AM, Sordillo JE, Gold DR, Bacharier LB, O’Connor GT, Zeiger RS, Beigelman A, Weiss ST, Litonjua AA. Gut microbiota and overweight in 3-year old children. Int J Obes (Lond). 2019;43(4):713–723. doi: 10.1038/s41366-018-0290-z. PMID: 30568265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Jiang LF, Wang YY, Peng H, Li R, Zhang F, Wang N, Shao QW, Jiang Q. Association between obesity with the diversity and genus of gut microbiota in school-aged children. Zhonghua Liu Xing Bing Xue Za Zhi. 2022;43(2):260–268. doi: 10.3760/cma.j.cn112338-20210617-00478. PMID: 35184494. [DOI] [PubMed] [Google Scholar]
  • 48.Jaimes JD, A S, Hurych J, Cinek O, Nichols B, Vodolánová L, Černý K, Havlík J, Ishaq SL. Stool metabolome-microbiota evaluation among children and adolescents with obesity, overweight, and normal-weight using 1H NMR and 16S rRNA gene profiling. PLOS ONE. 2021;16(3):e0247378. doi: 10.1371/journal.pone.0247378. PMID: 33765008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hu HJ, Park SG, Jang HB, Choi MK, Park KH, Kang JH, Park SI, Lee HJ, Cho SH. Obesity alters the microbial community profile in Korean adolescents. PLOS ONE. 2015;10(7):e0134333. doi: 10.1371/journal.pone.0134333. PMID: 26230509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Gyarmati P, Song Y, Dotimas J, Yoshiba G, Christison A. Cross-sectional comparisons of gut microbiome and short-chain fatty acid levels among children with varied weight classifications. Pediatr Obes. 2021;16(6):e12750. doi: 10.1111/ijpo.12750. PMID: 33174684. [DOI] [PubMed] [Google Scholar]
  • 51.Grigorova EV, Belkova NL, Nemchenko UM, Klimenko ES, Pogodina AV, Romanitsa AI, Novikova EA, Rychkova LV. Metasequencing of V3-V4 variable regions of 16S rRNA gene in opportunistic microbiota and gut biocenosis in obese adolescents. Bull Exp Biol Med. 2021;170(3):321–325. doi: 10.1007/s10517-021-05060-3. PMID: 33452983. [DOI] [PubMed] [Google Scholar]
  • 52.Gao XF, Wu BB, Pan YL, Zhou SM, Zhang M, You YH, Cai YP, Liang Y. Gut microbiome biomarkers in adolescent obesity: a regional study. Health Inf Sci Syst. 2023;11(1):37. doi: 10.1007/s13755-023-00236-9. PMID: 37602197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Gao X, Jia R, Xie L, Kuang L, Feng L, Wan C. A study of the correlation between obesity and intestinal flora in school-age children. Sci Rep. 2018;8(1):14511. doi: 10.1038/s41598-018-32730-6. PMID: 30267022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Gallardo-Becerra L, Cornejo-Granados F, García-López R, Valdez-Lara A, Bikel S, Canizales-Quinteros S, López-Contreras BE, Mendoza-Vargas A, Nielsen H, Ochoa-Leyva A. Metatranscriptomic analysis to define the secrebiome, and 16S rRNA profiling of the gut microbiome in obesity and metabolic syndrome of Mexican children. Microb Cell Fact. 2020;19(1):61. doi: 10.1186/s12934-020-01319-y. PMID: 32143621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ferrer M, Ruiz A, Lanza F, Haange SB, Oberbach A, Till H, Bargiela R, Campoy C, Segura MT, Richter M, et al. Microbiota from the distal guts of lean and obese adolescents exhibit partial functional redundancy besides clear differences in community structure. Environ Microbiol. 2013;15(1):211–226. doi: 10.1111/j.1462-2920.2012.02845.x. PMID: 22891823. [DOI] [PubMed] [Google Scholar]
  • 56.Del Chierico F, Manco M, Gardini S, Guarrasi V, Russo A, Bianchi M, Tortosa V, Quagliariello A, Shashaj B, Fintini D, et al. Fecal microbiota signatures of insulin resistance, inflammation, and metabolic syndrome in youth with obesity: a pilot study. Acta Diabetol. 2021;58(8):1009–1022. doi: 10.1007/s00592-020-01669-4. PMID: 33754165. [DOI] [PubMed] [Google Scholar]
  • 57.Del Chierico F, Abbatini F, Russo A, Quagliariello A, Reddel S, Capoccia D, Caccamo R, Ginanni Corradini S, Nobili V, De Peppo F, et al. Gut microbiota markers in obese adolescent and adult patients: age-dependent differential patterns. Front Microbiol. 2018;9:1210. doi: 10.3389/fmicb.2018.01210. PMID: 29922272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Da Silva CC, Monteil MA, Davis EM. Overweight and obesity in children are associated with an abundance of Firmicutes and reduction of bifidobacterium in their gastrointestinal microbiota. Child Obes. 2020;16(3):204–210. doi: 10.1089/chi.2019.0280. PMID: 31934770. [DOI] [PubMed] [Google Scholar]
  • 59.Cho KY. Lifestyle modifications result in alterations in the gut microbiota in obese children. BMC Microbiol. 2021;21(1):10. doi: 10.1186/s12866-020-02002-3. PMID: 33407104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Chen X, Zhang D, Sun H, Jiang F, Shen Y, Wei P, Shen X. Characterization of the gut microbiota in Chinese children with overweight and obesity using 16S rRNA gene sequencing. PeerJ. 2021;9:e11439. doi: 10.7717/peerj.11439. PMID: 34164233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Chen X, Sun H, Jiang F, Shen Y, Li X, Hu X, Shen X, Wei P. Alteration of the gut microbiota associated with childhood obesity by 16S rRNA gene sequencing. PeerJ. 2020;8:e8317. doi: 10.7717/peerj.8317. PMID: 31976177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Benítez-Páez A, Gómez Del Pugar EM, López-Almela I, Moya-Pérez Á, Codoñer-Franch P, Sanz Y, Turnbaugh PJ. Depletion of Blautia Species in the microbiota of obese children relates to intestinal inflammation and metabolic phenotype worsening. mSystems. 2020;5(2). doi: 10.1128/mSystems.00857-19. PMID: 32209719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Balakrishnan B, Selvaraju V, Chen J, Ayine P, Yang L, Babu JR, Geetha T, Taneja V. Ethnic variability associating gut and oral microbiome with obesity in children. Gut Microbes. 2021;13(1):1–15. doi: 10.1080/19490976.2021.1882926. PMID: 33596768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Jf SM, Qiong W. Analysis of intestinal microecological status and influencing factors in obese children aged 3-6 years. China Maternal And Child Health. 2022;37:2981–2984. doi: 10.19829/j.zgfybj.issn.1001-4411.2022.16.021. [DOI] [Google Scholar]
  • 65.Chunfeng T, Long G, Yufan G, Kai L, Yan B. A multi-omics study of intestinal flora and metabolism in obese Mongolian children. Genomics And Appl Biol. 2023;42:542–551. doi: 10.13417/j.gab.042.000542. [DOI] [Google Scholar]
  • 66.Jing L, Yuanyuan L, Chaonan F, Xu D, Kemin Q. Changes of intestinal flora in preschool obese children. Chinese journal of child health. 2015;23:388–390. [Google Scholar]
  • 67.Alcazar M, Escribano J, Ferré N, Closa-Monasterolo R, Selma-Royo M, Feliu A, Castillejo G, Luque V. Gut microbiota is associated with metabolic health in children with obesity. Clin Nutr. 2022;41(8):1680–1688. doi: 10.1016/j.clnu.2022.06.007. PMID: 35777107. [DOI] [PubMed] [Google Scholar]
  • 68.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. doi: 10.1155/2017/7585989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Gruneck L, Gentekaki E, Kespechara K, Denny J, Sharpton TJ, Marriott LK, Shannon J, Popluechai S. The fecal microbiota of Thai school-aged children associated with demographic factors and diet. PeerJ. 2022;10:e13325. doi: 10.7717/peerj.13325. PMID: 35469202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Xu P, Li M, Zhang J, Zhang T. Correlation of intestinal microbiota with overweight and obesity in Kazakh school children. BMC Microbiol. 2012;12(1):283. doi: 10.1186/1471-2180-12-283. PMID: 23190705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Orbe-Orihuela YC, Lagunas-Martínez A, Bahena-Román M, Madrid-Marina V, Torres-Poveda K, Flores-Alfaro E, Méndez-Padrón A, Díaz-Benítez CE, Peralta-Zaragoza O, Antúnez-Ortiz D, et al. High relative abundance of firmicutes and increased tnf-α levels correlate with obesity in children. Salud Publica Mex. 2018;60(1, ene–feb):5–11. doi: 10.21149/8133. PMID: 29689651. [DOI] [PubMed] [Google Scholar]
  • 72.Mousavi SH, Mehrara S, Barzegari A, Ostadrahimi A. Correlation of gut microbiota profile with body mass index among school age children. Iran Red Crescent Med J. 2018;20(4). doi: 10.5812/ircmj.58049. [DOI] [Google Scholar]
  • 73.Karlsson CL, Onnerfält J, Xu J, Molin G, Ahrné S, Thorngren-Jerneck K. The microbiota of the gut in preschool children with normal and excessive body weight. Obesity (Silver Spring). 2012;20(11):2257–2261. doi: 10.1038/oby.2012.110. PMID: 22546742. [DOI] [PubMed] [Google Scholar]
  • 74.Goffredo M, Mass K, Parks EJ, Wagner DA, McClure EA, Graf J, Savoye M, Pierpont B, Cline G, Santoro N. Role of gut microbiota and short chain fatty acids in modulating energy harvest and fat partitioning in youth. J Clin Endocrinol Metab. 2016;101(11):4367–4376. doi: 10.1210/jc.2016-1797. PMID: 27648960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Bervoets L, Van Hoorenbeeck K, Kortleven I, Van Noten C, Hens N, Vael C, Goossens H, Desager KN, Vankerckhoven V. Differences in gut microbiota composition between obese and lean children: a cross-sectional study. Gut Pathog. 2013;5(1):10. doi: 10.1186/1757-4749-5-10. PMID: 23631345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Balamurugan R, George G, Kabeerdoss J, Hepsiba J, Chandragunasekaran AM, Ramakrishna BS. Quantitative differences in intestinal Faecalibacterium prausnitzii in obese Indian children. Br J Nutr. 2010;103(3):335–338. doi: 10.1017/s0007114509992182. PMID: 19849869. [DOI] [PubMed] [Google Scholar]
  • 77.Jy DZ, Junmei Y. Survey on the current status of overweight and obesity in preschool children and analysis of the results of intestinal flora testing. China Convalescent Med. 2019;28:1226–1228. doi: 10.13517/j.cnki.ccm.2019.11.045. [DOI] [Google Scholar]
  • 78.Maya-Lucas O, Murugesan S, Nirmalkar K, Alcaraz LD, Hoyo-Vadillo C, Pizano-Zárate ML, García-Mena J. The gut microbiome of Mexican children affected by obesity. Anaerobe. 2019;55:11–23. doi: 10.1016/j.anaerobe.2018.10.009. PMID: 30366118. [DOI] [PubMed] [Google Scholar]
  • 79.Zhao Y, Zhou J, Liu J, Wang Z, Chen M, Zhou S. Metagenome of gut microbiota of children with nonalcoholic fatty liver disease. Front Pediatr. 2019;7:518. doi: 10.3389/fped.2019.00518. PMID: 31921729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Testerman T, Li Z, Galuppo B, Graf J, Santoro N. Insights from shotgun metagenomics into bacterial species and metabolic pathways associated with NAFLD in obese youth. Hepatol Commun. 2022;6(8):1962–1974. doi: 10.1002/hep4.1944. PMID: 35344283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Orsso CE, Peng Y, Deehan EC, Tan Q, Field CJ, Madsen KL, Walter J, Prado CM, Tun HM, Haqq AM. Composition and functions of the gut microbiome in pediatric obesity: relationships with markers of insulin resistance. Microorganisms. 2021;9(7):1490. doi: 10.3390/microorganisms9071490. PMID: 34361925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Murga-Garrido SM, Ulloa-Pérez EJ, Díaz-Benítez CE, Orbe-Orihuela YC, Cornejo-Granados F, Ochoa-Leyva A, Sanchez-Flores A, Cruz M, Castañeda-Márquez AC, Plett-Torres T, et al. Virulence factors of the gut microbiome are associated with BMI and Metabolic blood parameters in children with obesity. Microbiol Spectr. 2023;11(2):e0338222. doi: 10.1128/spectrum.03382-22. PMID: 36786619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Michail S, Lin M, Frey MR, Fanter R, Paliy O, Hilbush B, Reo NV. Altered gut microbial energy and metabolism in children with non-alcoholic fatty liver disease. FEMS Microbiol Ecol. 2015;91(2):1–9. doi: 10.1093/femsec/fiu002. PMID: 25764541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Abdallah Ismail N, Ragab SH, Abd Elbaky A, Shoeib AR, Alhosary Y, Fekry D. Frequency of Firmicutes and Bacteroidetes in gut microbiota in obese and normal weight Egyptian children and adults. Arch Med Sci. 2011;7:501–507. doi: 10.5114/aoms.2011.23418. PMID: 22295035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Barczyńska R, Litwin M, Sliżewska K, Szalecki M, Berdowska A, Bandurska K, Libudzisz Z, Kapuśniak J. Bacterial microbiota and fatty acids in the faeces of overweight and obese children. Pol J Microbiol. 2018;67(3):339–345. doi: 10.21307/pjm-2018-041. PMID: 30451451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Nemchenko U, Belkova N, Pogodina A, Romanitsa A, Novikova E, Klimenko E, Grigorova E, Kungurtseva E, Butakova K, Rychkova L. Features of the composition of bifidoflora in the intestinal microbiome of obese adolescents. Mol Genet Microbiol Virol. 2021;36(3):144–151. doi: 10.3103/s0891416821030046. [DOI] [Google Scholar]
  • 87.Ignacio A, Fernandes MR, Rodrigues VA, Groppo FC, Cardoso AL, Avila-Campos MJ, Nakano V. Correlation between body mass index and faecal microbiota from children. Clin Microbiol Infect. 2016;22(3):258.e1–8. doi: 10.1016/j.cmi.2015.10.031. PMID: 26551842. [DOI] [PubMed] [Google Scholar]
  • 88.Borgo F, Verduci E, Riva A, Lassandro C, Riva E, Morace G, Borghi E. Relative abundance in bacterial and fungal gut microbes in obese children: a case control study. Child Obes. 2017;13(1):78–84. doi: 10.1089/chi.2015.0194. PMID: 27007700. [DOI] [PubMed] [Google Scholar]
  • 89.Luoto R, Kalliomäki M, Laitinen K, Delzenne NM, Cani PD, Salminen S, Isolauri E. Initial dietary and microbiological environments deviate in normal-weight compared to overweight children at 10 years of age. J Pediatr Gastroenterol Nutr. 2011;52(1):90–95. doi: 10.1097/MPG.0b013e3181f3457f. PMID: 21150648. [DOI] [PubMed] [Google Scholar]
  • 90.Shade A. Diversity is the question, not the answer. The ISME J. 2017;11(1):1–6. doi: 10.1038/ismej.2016.118. PMID: 27636395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Crovesy L, Masterson D, Rosado EL. Profile of the gut microbiota of adults with obesity: a systematic review. Eur J Clin Nutr. 2020;74(9):1251–1262. doi: 10.1038/s41430-020-0607-6. PMID: 32231226. [DOI] [PubMed] [Google Scholar]
  • 92.Pinart M, Dötsch A, Schlicht K, Laudes M, Bouwman J, Forslund SK, Pischon T, Nimptsch K. Gut microbiome composition in obese and non-obese persons: a systematic review and meta-analysis. Nutrients. 2021;14(1):12. doi: 10.3390/nu14010012. PMID: 35010887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Depommier C, Everard A, Druart C, Plovier H, Van Hul M, Vieira-Silva S, Falony G, Raes J, Maiter D, Delzenne NM, et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nat Med. 2019;25(7):1096–1103. doi: 10.1038/s41591-019-0495-2. PMID: 31263284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Hold GL, Schwiertz A, Aminov RI, Blaut M, Flint HJ. Oligonucleotide probes that detect quantitatively significant groups of butyrate-producing bacteria in human feces. Appl Environ Microbiol. 2003;69(7):4320–4324. doi: 10.1128/aem.69.7.4320-4324.2003. PMID: 12839823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Maioli TU, Borras-Nogues E, Torres L, Barbosa SC, Martins VD, Langella P, Azevedo VA, Chatel JM. Possible benefits of faecalibacterium prausnitzii for obesity-associated gut disorders. Front Pharmacol. 2021;12:740636. doi: 10.3389/fphar.2021.740636. PMID: 34925006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Shuai M, Fu Y, Zhong HL, Gou W, Jiang Z, Liang Y, Miao Z, Xu JJ, Huynh T, Wahlqvist ML, et al. Mapping the human gut mycobiome in middle-aged and elderly adults: multiomics insights and implications for host metabolic health. Gut. 2022;71(9):1812–1820. doi: 10.1136/gutjnl-2021-326298. PMID: 35017200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Kaplan RC, Wang Z, Usyk M, Sotres-Alvarez D, Daviglus ML, Schneiderman N, Talavera GA, Gellman MD, Thyagarajan B, Moon JY, et al. Gut microbiome composition in the Hispanic Community Health Study/Study of Latinos is shaped by geographic relocation, environmental factors, and obesity. Genome Biol. 2019;20(1):219. doi: 10.1186/s13059-019-1831-z. PMID: 31672155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Huseyin CE, O’Toole PW, Cotter PD, Scanlan PD. Forgotten fungi—the gut mycobiome in human health and disease. FEMS Microbiol Rev. 2017;41(4):479–511. doi: 10.1093/femsre/fuw047. PMID: 28430946. [DOI] [PubMed] [Google Scholar]
  • 99.Mba IE, Nweze EI. Mechanism of Candida pathogenesis: revisiting the vital drivers. Eur J Clin Microbiol Infect Dis. 2020;39(10):1797–1819. doi: 10.1007/s10096-020-03912-w. PMID: 32372128. [DOI] [PubMed] [Google Scholar]
  • 100.Polke M, Hube B, Jacobsen ID. Candida survival strategies. Adv Appl Microbiol. 2015;91:139–235. doi: 10.1016/bs.aambs.2014.12.002. PMID: 25911234. [DOI] [PubMed] [Google Scholar]
  • 101.Sun S, Sun L, Wang K, Qiao S, Zhao X, Hu X, Chen W, Zhang S, Li H, Dai H, et al. The gut commensal fungus, Candida parapsilosis, promotes high fat-diet induced obesity in mice. Commun Biol. 2021;4(1):1220. doi: 10.1038/s42003-021-02753-3. PMID: 34697386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Mar Rodríguez M, Pérez D, Javier Chaves F, Esteve E, Marin-Garcia P, Xifra G, Vendrell J, Jové M, Pamplona R, Ricart W, et al. Obesity changes the human gut mycobiome. Sci Rep. 2015;5(1):14600. doi: 10.1038/srep14600. PMID: 26455903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Hallen-Adams HE, Kachman SD, Kim J, Legge RM, Martínez I. Fungi inhabiting the healthy human gastrointestinal tract: a diverse and dynamic community. Fungal Ecol. 2015;15:9–17. doi: 10.1016/j.funeco.2015.01.006. [DOI] [Google Scholar]
  • 104.Wu L, Zeng T, Deligios M, Milanesi L, Langille MGI, Zinellu A, Rubino S, Carru C, Kelvin DJ. Age-related variation of bacterial and fungal communities in different body habitats across the young, elderly, and centenarians in Sardinia. mSphere. 2020;5(1). doi: 10.1128/mSphere.00558-19. PMID: 32102941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.García-Gamboa R, Kirchmayr MR, Gradilla-Hernández MS, Pérez-Brocal V, Moya A, González-Avila M. The intestinal mycobiota and its relationship with overweight, obesity and nutritional aspects. J Hum Nutr Diet. 2021;34(4):645–655. doi: 10.1111/jhn.12864. PMID: 33586805. [DOI] [PubMed] [Google Scholar]
  • 106.Sokol H, Leducq V, Aschard H, Pham HP, Jegou S, Landman C, Cohen D, Liguori G, Bourrier A, Nion-Larmurier I, et al. Fungal microbiota dysbiosis in IBD. Gut. 2017;66(6):1039–1048. doi: 10.1136/gutjnl-2015-310746. PMID: 26843508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Hills RD Jr., Pontefract BA, Mishcon HR, Black CA, Sutton SC, Theberge CR. Gut microbiome: profound implications for diet and disease. Nutrients. 2019;11(7):11. doi: 10.3390/nu11071613. PMID: 31315227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Lambooij JM, Hoogenkamp MA, Brandt BW, Janus MM, Krom BP. Fungal mitochondrial oxygen consumption induces the growth of strict anaerobic bacteria. Fungal Genet Biol. 2017;109:1–6. doi: 10.1016/j.fgb.2017.10.001. PMID: 28989089. [DOI] [PubMed] [Google Scholar]
  • 109.van Leeuwen Pt, van der Peet Jm, Bikker FJ, Hoogenkamp MA, Oliveira Paiva AM, Kostidis S, Mayboroda OA, Smits WK, Krom BP. Interspecies interactions between Clostridium difficile and Candida albicans. mSphere. 2016;1(6). doi: 10.1128/mSphere.00187-16. PMID: 27840850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Shabana SS, Irfan U. The gut microbiota and its potential role in obesity. Future Microbiol. 2018;13:589–603. doi: 10.2217/fmb-2017-0179. PMID: 29533087. [DOI] [PubMed] [Google Scholar]
  • 111.Zhang T, Breitbart M, Lee WH, Run JQ, Wei CL, Soh SW, Hibberd ML, Liu ET, Rohwer F, Ruan Y. RNA viral community in human feces: prevalence of plant pathogenic viruses. PLoS Biol. 2006;4(1):e3. doi: 10.1371/journal.pbio.0040003. PMID: 16336043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Yadav H, Jain S, Nagpal R, Marotta F. Increased fecal viral content associated with obesity in mice. World J Diabetes. 2016;7(15):316–320. doi: 10.4239/wjd.v7.i15.316. PMID: 27555892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Bikel S, López-Leal G, Cornejo-Granados F, Gallardo-Becerra L, García-López R, Sánchez F, Equihua-Medina E, Ochoa-Romo JP, López-Contreras BE, Canizales-Quinteros S, et al. Gut dsDNA virome shows diversity and richness alterations associated with childhood obesity and metabolic syndrome. iScience. 2021;24(8):102900. doi: 10.1016/j.isci.2021.102900. PMID: 34409269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Bikel S, López-Leal G, Cornejo-Granados F, Gallardo-Becerra L, Sánchez F, Equihua-Medina E, Ochoa-Romo JP, López-Contreras BE, Canizales-Quinteros S, Leyva AO. Gut phageome analysis reveals disease-specific hallmarks in childhood obesity. bioRxiv. 2020:2020.07. 29.227637. doi: 10.1101/2020.07.29.227637. [DOI] [Google Scholar]
  • 115.Cervantes-Echeverría M, Gallardo-Becerra L, Cornejo-Granados F, Ochoa-Leyva A. The two-faced role of crAssphage subfamilies in obesity and metabolic syndrome: between good and evil. Genes (Basel). 2023;14(1):139. doi: 10.3390/genes14010139. PMID: 36672880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Dutilh BE, Cassman N, McNair K, Sanchez SE, Silva GG, Boling L, Barr JJ, Speth DR, Seguritan V, Aziz RK, et al. A highly abundant bacteriophage discovered in the unknown sequences of human faecal metagenomes. Nat Commun. 2014;5(1):4498. doi: 10.1038/ncomms5498. PMID: 25058116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Shen J, Zhang J, Mo L, Li Y, Li Y, Li C, Kuang X, Tao Z, Qu Z, Wu L, et al. Large-scale phage cultivation for commensal human gut bacteria. Cell Host & Microbe. 2023;31(4):665–77.e7. doi: 10.1016/j.chom.2023.03.013. PMID: 37054680. [DOI] [PubMed] [Google Scholar]
  • 118.Ott SJ, Waetzig GH, Rehman A, Moltzau-Anderson J, Bharti R, Grasis JA, Cassidy L, Tholey A, Fickenscher H, Seegert D, et al. Efficacy of sterile fecal filtrate transfer for treating patients with clostridium difficile infection. Gastroenterology. 2017;152(4):799–811.e7. doi: 10.1053/j.gastro.2016.11.010. PMID: 27866880. [DOI] [PubMed] [Google Scholar]
  • 119.Miller TL, Wolin MJ, Conway de Macario E, Macario AJ. Isolation of methanobrevibacter smithii from human feces. Appl Environ Microbiol. 1982;43(1):227–232. doi: 10.1128/aem.43.1.227-232.1982. PMID: 6798932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Miller TL, Wolin MJ. Methanosphaera stadtmaniae gen. nov. sp. nov.: a species that forms methane by reducing methanol with hydrogen. Arch Microbiol. 1985;141(2):116–122. doi: 10.1007/bf00423270. PMID: 3994486. [DOI] [PubMed] [Google Scholar]
  • 121.Dridi B, Fardeau ML, Ollivier B, Raoult D, Drancourt M. Methanomassiliicoccus luminyensis gen. nov. sp. nov. a methanogenic archaeon isolated from human faeces. Int J Syst Evol Microbiol. 2012;62(Pt_8):1902–1907. doi: 10.1099/ijs.0.033712-0. PMID: 22859731. [DOI] [PubMed] [Google Scholar]
  • 122.Samuel BS, Gordon JI. A humanized gnotobiotic mouse model of host–archaeal–bacterial mutualism. Proc Natl Acad Sci USA. 2006;103(26):10011–10016. doi: 10.1073/pnas.0602187103. PMID: 16782812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Samuel BS, Hansen EE, Manchester JK, Coutinho PM, Henrissat B, Fulton R, Latreille P, Kim K, Wilson RK, Gordon JI. Genomic and metabolic adaptations of methanobrevibacter smithii to the human gut. Proc Natl Acad Sci USA. 2007;104(25):10643–10648. doi: 10.1073/pnas.0704189104. PMID: 17563350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Gregory AC, Zablocki O, Zayed AA, Howell A, Bolduc B, Sullivan MB. The gut virome database reveals age-dependent patterns of virome diversity in the human gut. Cell Host Microbe. 2020;28(5):724–40.e8. doi: 10.1016/j.chom.2020.08.003. PMID: 32841606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Le Poul E, Loison C, Struyf S, Springael JY, Lannoy V, Decobecq ME, Brezillon S, Dupriez V, Vassart G, Van Damme J, et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem. 2003;278(28):25481–25489. doi: 10.1074/jbc.M301403200. PMID: 12711604. [DOI] [PubMed] [Google Scholar]
  • 126.Husted AS, Trauelsen M, Rudenko O, Hjorth SA, Schwartz TW. GPCR-Mediated signaling of metabolites. Cell Metab. 2017;25(4):777–796. doi: 10.1016/j.cmet.2017.03.008. PMID: 28380372. [DOI] [PubMed] [Google Scholar]
  • 127.Ahmed K, Tunaru S, Offermanns S. GPR109A, GPR109B and GPR81, a family of hydroxy-carboxylic acid receptors. Trends Pharmacol Sci. 2009;30(11):557–562. doi: 10.1016/j.tips.2009.09.001. PMID: 19837462. [DOI] [PubMed] [Google Scholar]
  • 128.Lin HV, Frassetto A, Kowalik EJ Jr., Nawrocki AR, Lu MM, Kosinski JR, Hubert JA, Szeto D, Yao X, Forrest G, et al. Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS One. 2012;7(4):e35240. doi: 10.1371/journal.pone.0035240. PMID: 22506074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Coppola S, Avagliano C, Calignano A, Berni Canani R. The protective role of butyrate against obesity and obesity-related diseases. Molecules. 2021;26(3):682. doi: 10.3390/molecules26030682. PMID: 33525625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud DJ, Bakker BM. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res. 2013;54(9):2325–2340. doi: 10.1194/jlr.R036012. PMID: 23821742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Delzenne NM, Neyrinck AM, Bäckhed F, Cani PD. Targeting gut microbiota in obesity: effects of prebiotics and probiotics. Nat Rev Endocrinol. 2011;7(11):639–646. doi: 10.1038/nrendo.2011.126. PMID: 21826100. [DOI] [PubMed] [Google Scholar]
  • 132.Usuda H, Okamoto T, Wada K. Leaky gut: effect of dietary fiber and fats on microbiome and intestinal barrier. Int J Mol Sci. 2021;22(14):7613. doi: 10.3390/ijms22147613. PMID: 34299233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Boutagy NE, McMillan RP, Frisard MI, Hulver MW. Metabolic endotoxemia with obesity: Is it real and is it relevant? Biochimie. 2016;124:11–20. doi: 10.1016/j.biochi.2015.06.020. PMID: 26133659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Akhtar M, Chen Y, Ma Z, Zhang X, Shi D, Khan JA, Liu H. Gut microbiota-derived short chain fatty acids are potential mediators in gut inflammation. Anim Nutr. 2022;8:350–360. doi: 10.1016/j.aninu.2021.11.005. PMID: 35510031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Wrzosek L, Miquel S, Noordine ML, Bouet S, Joncquel Chevalier-Curt M, Robert V, Philippe C, Bridonneau C, Cherbuy C, Robbe-Masselot C, et al. Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii influence the production of mucus glycans and the development of goblet cells in the colonic epithelium of a gnotobiotic model rodent. BMC Biol. 2013;11(1):61. doi: 10.1186/1741-7007-11-61. PMID: 23692866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Byndloss MX, Olsan EE, Rivera-Chávez F, Tiffany CR, Cevallos SA, Lokken KL, Torres TP, Byndloss AJ, Faber F, Gao Y, et al. Microbiota-activated ppar-γ signaling inhibits dysbiotic Enterobacteriaceae expansion. Science. 2017;357(6351):570–575. doi: 10.1126/science.aam9949. PMID: 28798125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Wahlström A, Sayin SI, Marschall HU, Bäckhed F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. 2016;24(1):41–50. doi: 10.1016/j.cmet.2016.05.005. PMID: 27320064. [DOI] [PubMed] [Google Scholar]
  • 138.Grüner N, Mattner J. Bile acids and microbiota: multifaceted and versatile regulators of the liver–gut axis. Int J Mol Sci. 2021;22(3):1397. doi: 10.3390/ijms22031397. PMID: 33573273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Basuray N, Deehan EC, Vieira FT, Avedzi HM, Duke RL, Colín-Ramírez E, Tun HM, Zhang Z, Wine E, Madsen KL, et al. Dichotomous effect of dietary fiber in pediatrics: a narrative review of the health benefits and tolerance of fiber. Eur J Clin Nutr. 2024;78(7):557–568. doi: 10.1038/s41430-024-01429-5. PMID: 38480843. [DOI] [PubMed] [Google Scholar]
  • 140.Kelishadi R, Farajian S, Safavi M, Mirlohi M, Hashemipour M. A randomized triple-masked controlled trial on the effects of synbiotics on inflammation markers in overweight children. J Pediatr (Rio J). 2014;90(2):161–168. doi: 10.1016/j.jped.2013.07.003. PMID: 24184299. [DOI] [PubMed] [Google Scholar]
  • 141.Jones RB, Alderete TL, Kim JS, Millstein J, Gilliland FD, Goran MI. High intake of dietary fructose in overweight/obese teenagers associated with depletion of Eubacterium and streptococcus in gut microbiome. Gut Microbes. 2019;10(6):712–719. doi: 10.1080/19490976.2019.1592420. PMID: 30991877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Rampelli S, Guenther K, Turroni S, Wolters M, Veidebaum T, Kourides Y, Molnár D, Lissner L, Benitez-Paez A, Sanz Y, et al. Pre-obese children’s dysbiotic gut microbiome and unhealthy diets may predict the development of obesity. Commun Biol. 2018;1(1):222. doi: 10.1038/s42003-018-0221-5. PMID: 30534614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Leong C, Haszard JJ, Heath AM, Tannock GW, Lawley B, Cameron SL, Szymlek-Gay EA, Gray AR, Taylor BJ, Galland BC, et al. Using compositional principal component analysis to describe children’s gut microbiota in relation to diet and body composition. Am J Clin Nutr. 2020;111(1):70–78. doi: 10.1093/ajcn/nqz270. PMID: 31711093. [DOI] [PubMed] [Google Scholar]
  • 144.Kisuse J, La-Ongkham O, Nakphaichit M, Therdtatha P, Momoda R, Tanaka M, Fukuda S, Popluechai S, Kespechara K, Sonomoto K, et al. Urban diets linked to gut microbiome and metabolome alterations in children: a comparative cross-sectional study in Thailand. Front Microbiol. 2018;9:1345. doi: 10.3389/fmicb.2018.01345. PMID: 29988433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Li H, Zhao L, Zhang M. Gut microbial SNPs induced by high-fiber diet dominate nutrition metabolism and environmental adaption of faecalibacterium prausnitzii in obese children. Front Microbiol. 2021;12:683714. doi: 10.3389/fmicb.2021.683714. PMID: 34135881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Zhang C, Yin A, Li H, Wang R, Wu G, Shen J, Zhang M, Wang L, Hou Y, Ouyang H, et al. Dietary modulation of gut microbiota contributes to alleviation of both genetic and simple obesity in children. EBioMedicine. 2015;2(8):968–984. doi: 10.1016/j.ebiom.2015.07.007. PMID: 26425705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Orbe-Orihuela YC, Godoy-Lozano EE, Lagunas-Martínez A, Castañeda-Márquez AC, Murga-Garrido S, Díaz-Benítez CE, Ochoa-Leyva A, Cornejo-Granados F, Cruz M, Estrada K, et al. Association of gut microbiota with dietary-dependent childhood obesity. Arch Med Res. 2022;53(4):407–415. doi: 10.1016/j.arcmed.2022.03.007. PMID: 35382951. [DOI] [PubMed] [Google Scholar]
  • 148.Kilic Yildirim G, Dinleyici M, Vandenplas Y, Dinleyici EC. Effects of synbiotic supplementation on intestinal microbiota composition in children and adolescents with exogenous obesity: (probesity-2 trial). Gut Pathog. 2023;15(1):36. doi: 10.1186/s13099-023-00563-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Nicolucci AC, Hume MP, Martínez I, Mayengbam S, Walter J, Reimer RA. Prebiotics reduce body fat and alter intestinal microbiota in children who are overweight or with obesity. Gastroenterology. 2017;153(3):711–722. doi: 10.1053/j.gastro.2017.05.055. PMID: 28596023. [DOI] [PubMed] [Google Scholar]
  • 150.Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, Bewtra M, Knights D, Walters WA, Knight R, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334(6052):105–108. doi: 10.1126/science.1208344. PMID: 21885731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Flint HJ, Scott KP, Louis P, Duncan SH. The role of the gut microbiota in nutrition and health. Nat Rev Gastroenterol Hepatol. 2012;9(10):577–589. doi: 10.1038/nrgastro.2012.156. PMID: 22945443. [DOI] [PubMed] [Google Scholar]
  • 152.Flint HJ, Scott KP, Duncan SH, Louis P, Forano E. Microbial degradation of complex carbohydrates in the gut. Gut Microbes. 2012;3(4):289–306. doi: 10.4161/gmic.19897. PMID: 22572875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Garcia-Ribera S, Amat-Bou M, Climent E, Llobet M, Chenoll E, Corripio R, Ibáñez L, Ramon-Krauel M, Lerin C. Specific dietary components and gut microbiota composition are associated with obesity in children and adolescents with prader–Willi Syndrome. Nutrients. 2020;12(4):12. doi: 10.3390/nu12041063. PMID: 32290434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Qin W, Sun L, Miao M, Zhang G. Plant-sourced intrinsic dietary fiber: physical structure and health function. Trends Food Sci Technol. 2021;118:341–355. doi: 10.1016/j.tifs.2021.09.022. [DOI] [Google Scholar]
  • 155.Koh A, De Vadder F, Kovatcheva-Datchary P, Bäckhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell. 2016;165(6):1332–1345. doi: 10.1016/j.cell.2016.05.041. PMID: 27259147. [DOI] [PubMed] [Google Scholar]
  • 156.Ferro LE, Crowley LN, Bittinger K, Friedman ES, Decker JE, Russel K, Katz S, Kim JK, Trabulsi JC. Effects of prebiotics, probiotics, and synbiotics on the infant gut microbiota and other health outcomes: a systematic review. Crit Rev Food Sci Nutr. 2023;63(22):5620–5642. doi: 10.1080/10408398.2021.2022595. PMID: 37667870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Walter J, Maldonado-Gómez MX, Martínez I. To engraft or not to engraft: an ecological framework for gut microbiome modulation with live microbes. Curr Opin Biotechnol. 2018;49:129–139. doi: 10.1016/j.copbio.2017.08.008. PMID: 28866242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Reynés B, Palou M, Rodríguez AM, Palou A. Regulation of adaptive thermogenesis and Browning by prebiotics and postbiotics. Front Physiol. 2018;9:1908. doi: 10.3389/fphys.2018.01908. PMID: 30687123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Duque A, Demarqui FM, Santoni MM, Zanelli CF, Adorno MAT, Milenkovic D, Mesa V, Sivieri K. Effect of probiotic, prebiotic, and synbiotic on the gut microbiota of autistic children using an in vitro gut microbiome model. Food Res Int. 2021;149:110657. doi: 10.1016/j.foodres.2021.110657. PMID: 34600659. [DOI] [PubMed] [Google Scholar]
  • 160.Fransen F, Sahasrabudhe NM, Elderman M, Bosveld M, El Aidy S, Hugenholtz F, Borghuis T, Kousemaker B, Winkel S, van der Gaast-de Jongh C, et al. β2→1-fructans modulate the immune system in vivo in a microbiota-dependent and -Independent fashion. Front Immunol. 2017;8:154. doi: 10.3389/fimmu.2017.00154. PMID: 28261212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Lombard V, Golaconda Ramulu H, Drula E, Coutinho PM, Henrissat B. The carbohydrate-active enzymes database (CAZy) in 2013. Nucl Acids Res. 2014;42(D1):D490–5. doi: 10.1093/nar/gkt1178. PMID: 24270786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.El Kaoutari A, Armougom F, Gordon JI, Raoult D, Henrissat B. The abundance and variety of carbohydrate-active enzymes in the human gut microbiota. Nat Rev Microbiol. 2013;11(7):497–504. doi: 10.1038/nrmicro3050. PMID: 23748339. [DOI] [PubMed] [Google Scholar]
  • 163.Fulgoni VL 3rd, Brauchla M, Fleige L, Chu Y. Association of whole-grain and dietary fiber intake with cardiometabolic risk in children and adolescents. Nutr Health. 2020;26(3):243–251. doi: 10.1177/0260106020928664. PMID: 32552292. [DOI] [PubMed] [Google Scholar]
  • 164.Hume MP, Nicolucci AC, Reimer RA. Prebiotic supplementation improves appetite control in children with overweight and obesity: a randomized controlled trial. Am J Clin Nutr. 2017;105(4):790–799. doi: 10.3945/ajcn.116.140947. PMID: 28228425. [DOI] [PubMed] [Google Scholar]
  • 165.Barczynska R, Slizewska K, Litwin M, Szalecki M, Kapusniak J. Effects of dietary fiber preparations made from maize starch on the growth and activity of selected bacteria from the Firmicutes, Bacteroidetes, and Actinobacteria phyla in fecal samples from obese children. Acta Biochim Pol. 2016;63(2):261–266. doi: 10.18388/abp.2015_1068. PMID: 26929930. [DOI] [PubMed] [Google Scholar]
  • 166.Cani PD, de Vos WM. Next-generation beneficial microbes: the case of Akkermansia muciniphila. Front Microbiol. 2017;8:1765. doi: 10.3389/fmicb.2017.01765. PMID: 29018410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, Guiot Y, Derrien M, Muccioli GG, Delzenne NM, et al. Cross-talk between akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci USA. 2013;110(22):9066–9071. doi: 10.1073/pnas.1219451110. PMID: 23671105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Lam KC, Araya RE, Huang A, Chen Q, Di Modica M, Rodrigues RR, Lopès A, Johnson SB, Schwarz B, Bohrnsen E, et al. Microbiota triggers sting-type I ifn-dependent monocyte reprogramming of the tumor microenvironment. Cell. 2021;184(21):5338–56.e21. doi: 10.1016/j.cell.2021.09.019. PMID: 34624222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Schmidt-Wittig U, Enss ML, Coenen M, Gärtner K, Hedrich HJ. Response of rat colonic mucosa to a high fiber diet. Ann Nutr Metab. 1996;40(6):343–350. doi: 10.1159/000177936. PMID: 9087313. [DOI] [PubMed] [Google Scholar]
  • 170.Kim Y, Hwang SW, Kim S, Lee YS, Kim TY, Lee SH, Kim SJ, Yoo HJ, Kim EN, Kweon MN. Dietary cellulose prevents gut inflammation by modulating lipid metabolism and gut microbiota. Gut Microbes. 2020;11(4):944–961. doi: 10.1080/19490976.2020.1730149. PMID: 32138587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Procházková N, Falony G, Dragsted LO, Licht TR, Raes J, Roager HM. Advancing human gut microbiota research by considering gut transit time. Gut. 2023;72(1):180–191. doi: 10.1136/gutjnl-2022-328166. PMID: 36171079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Deehan EC, Yang C, Perez-Muñoz ME, Nguyen NK, Cheng CC, Triador L, Zhang Z, Bakal JA, Walter J. Precision microbiome modulation with discrete dietary fiber structures directs short-chain fatty acid production. Cell Host Microbe. 2020;27(3):389–404.e6. doi: 10.1016/.chom.2020.01.006. [DOI] [PubMed] [Google Scholar]
  • 173.Puhlmann ML, de Vos WM. Intrinsic dietary fibers and the gut microbiome: rediscovering the benefits of the plant cell matrix for human health. Front Immunol. 2022;13:954845. doi: 10.3389/fimmu.2022.954845. PMID: 36059540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Dietary Reference Intakes: Nutrient recommendations and databases [Internet]. National Academies Press; 2005. [cited 2023 Apr].https://ods.od.nih.gov/HealthInformation/nutrientrecommendations.aspx. [Google Scholar]
  • 175.Deehan EC, Mocanu V, Madsen KL. Effects of dietary fibre on metabolic health and obesity. Nat Rev Gastroenterol Hepatol. 2024;21(5):301–318. doi: 10.1038/s41575-023-00891-z. PMID: 38326443. [DOI] [PubMed] [Google Scholar]
  • 176.McRorie JW Jr., McKeown NM. Understanding the physics of functional fibers in the gastrointestinal tract: an evidence-based approach to resolving enduring misconceptions about insoluble and soluble fiber. J Acad Nutr Diet. 2017;117(2):251–264. doi: 10.1016/j.jand.2016.09.021. PMID: 27863994. [DOI] [PubMed] [Google Scholar]
  • 177.Hoek M, Merks RMH. Emergence of microbial diversity due to cross-feeding interactions in a spatial model of gut microbial metabolism. BMC Syst Biol. 2017;11(1):56. doi: 10.1186/s12918-017-0430-4. PMID: 28511646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Moens F, Weckx S, De Vuyst L. Bifidobacterial inulin-type fructan degradation capacity determines cross-feeding interactions between bifidobacteria and faecalibacterium prausnitzii. Int J Food Microbiol. 2016;231:76–85. doi: 10.1016/j.ijfoodmicro.2016.05.015. PMID: 27233082. [DOI] [PubMed] [Google Scholar]
  • 179.Mills RH, Dulai PS, Vázquez-Baeza Y, Sauceda C, Daniel N, Gerner RR, Batachari LE, Malfavon M, Zhu Q, Weldon K, et al. Multi-omics analyses of the ulcerative colitis gut microbiome link bacteroides vulgatus proteases with disease severity. Nat Microbiol. 2022;7(2):262–276. doi: 10.1038/s41564-021-01050-3. PMID: 35087228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Mars RAT, Yang Y, Ward T, Houtti M, Priya S, Lekatz HR, Tang X, Sun Z, Kalari KR, Korem T, et al. Longitudinal multi-omics reveals subset-specific mechanisms underlying irritable bowel syndrome. Cell. 2020;182(6):1460–73.e17. doi: 10.1016/j.cell.2020.08.007. PMID: 32916129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.de Cuevillas B, Milagro FI, Tur JA, Gil-Campos M, de Miguel-Etayo P, Martínez JA, Navas-Carretero S. Fecal microbiota relationships with childhood obesity: a scoping comprehensive review. Obes Rev. 2022;23(Suppl 1):e13394. doi: 10.1111/obr.13394. PMID: 34913242. [DOI] [PubMed] [Google Scholar]
  • 182.Liber A, Szajewska H. Effect of oligofructose supplementation on body weight in overweight and obese children: a randomised, double-blind, placebo-controlled trial. Br J Nutr. 2014;112(12):2068–2074. doi: 10.1017/s0007114514003110. PMID: 25327394. [DOI] [PubMed] [Google Scholar]
  • 183.Reynolds AN, Akerman AP, Mann J. Dietary fibre and whole grains in diabetes management: systematic review and meta-analyses. PLoS Med. 2020;17(3):e1003053. doi: 10.1371/journal.pmed.1003053. PMID: 32142510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Deehan EC, Duar RM, Armet AM, Perez-Muñoz ME, Jin M, Walter J, Britton RA, Cani PD. Modulation of the gastrointestinal microbiome with nondigestible fermentable carbohydrates to improve human health. Microbiol Spectr. 2017;5(5). doi: 10.1128/microbiolspec.BAD-0019-2017. PMID: 28936943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Lichtenstein AH, Petersen K, Barger K, Hansen KE, Anderson CAM, Baer DJ, Lampe JW, Rasmussen H, Matthan NR. Perspective: design and conduct of human nutrition randomized controlled trials. Adv Nutr. 2021;12(1):4–20. doi: 10.1093/advances/nmaa109. PMID: 33200182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Kane PB, Bittlinger M, Kimmelman J. Individualized therapy trials: navigating patient care, research goals and ethics. Nat Med. 2021;27(10):1679–1686. doi: 10.1038/s41591-021-01519-y. PMID: 34642487. [DOI] [PubMed] [Google Scholar]
  • 187.Ghosh TS, Das M, Jeffery IB, O’Toole PW. Adjusting for age improves identification of gut microbiome alterations in multiple diseases. Elife. 2020;9. doi: 10.7554/eLife.50240. PMID: 32159510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.de la Cuesta-Zuluaga J, Kelley ST, Chen Y, Escobar JS, Mueller NT, Ley RE, McDonald D, Huang S, Swafford AD, Knight R, et al. Age- and sex-dependent patterns of gut microbial diversity in human adults. mSystems. 2019;4(4). doi: 10.1128/mSystems.00261-19. PMID: 31098397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Bowyer RCE, Jackson MA, Pallister T, Skinner J, Spector TD, Welch AA, Steves CJ. Use of dietary indices to control for diet in human gut microbiota studies. Microbiome. 2018;6(1):77. doi: 10.1186/s40168-018-0455-y. PMID: 29695307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Meslier V, Laiola M, Roager HM, De Filippis F, Roume H, Quinquis B, Giacco R, Mennella I, Ferracane R, Pons N, et al. Mediterranean diet intervention in overweight and obese subjects lowers plasma cholesterol and causes changes in the gut microbiome and metabolome independently of energy intake. Gut. 2020;69(7):1258–1268. doi: 10.1136/gutjnl-2019-320438. PMID: 32075887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Nguyen NK, Deehan EC, Zhang Z, Jin M, Baskota N, Perez-Muñoz ME, Cole J, Tuncil YE, Seethaler B, Wang T, et al. Gut microbiota modulation with long-chain corn bran arabinoxylan in adults with overweight and obesity is linked to an individualized temporal increase in fecal propionate. Microbiome. 2020;8(1):118. doi: 10.1186/s40168-020-00887-w. PMID: 32814582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Walter J, Armet AM, Finlay BB, Shanahan F. Establishing or exaggerating causality for the gut microbiome: lessons from human microbiota-associated rodents. Cell. 2020;180:221–232. doi: 10.1016/j.cell.2019.12.025. PMID: 31978342. [DOI] [PubMed] [Google Scholar]
  • 193.Schmidt F, Zimmermann J, Tanna T, Farouni R, Conway T, Macpherson AJ, Platt RJ. Noninvasive assessment of gut function using transcriptional recording sentinel cells. Science. 2022;376(6594):eabm6038. doi: 10.1126/science.abm6038. PMID: 35549411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Arrieta MC, Walter J, Finlay BB. Human microbiota-associated mice: a model with challenges. Cell Host Microbe. 2016;19(5):575–578. doi: 10.1016/j.chom.2016.04.014. PMID: 27173924. [DOI] [PubMed] [Google Scholar]
  • 195.Paul W, Marta C, Tom VW. Resolving host-microbe interactions in the gut: the promise of in vitro models to complement in vivo research. Curr Opin Microbiol. 2018;44:28–33. doi: 10.1016/j.mib.2018.07.001. PMID: 30031243. [DOI] [PubMed] [Google Scholar]
  • 196.Deehan EC, Zhang Z, Riva A, Armet AM, Perez-Muñoz ME, Nguyen NK, Krysa JA, Seethaler B, Zhao YY, Cole J, et al. Elucidating the role of the gut microbiota in the physiological effects of dietary fiber. Microbiome. 2022;10(1):77. doi: 10.1186/s40168-022-01248-5. PMID: 35562794. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Material

Articles from Gut Microbes are provided here courtesy of Taylor & Francis

RESOURCES