Hostname: page-component-8448b6f56d-mp689 Total loading time: 0 Render date: 2024-04-18T16:17:59.077Z Has data issue: false hasContentIssue false

Personal diet–microbiota interactions and weight loss

Published online by Cambridge University Press:  17 February 2022

Henrik M. Roager*
Affiliation:
Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg, Denmark
Lars H. Christensen
Affiliation:
Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg, Denmark
*
*Corresponding author: Henrik M. Roager, email hero@nexs.ku.dk
Rights & Permissions [Opens in a new window]

Abstract

The aim of this review is to provide an overview of how person-specific interactions between diet and the gut microbiota could play a role in affecting diet-induced weight loss responses. The highly person-specific gut microbiota, which is shaped by our diet, secretes digestive enzymes and molecules that affect digestion in the colon. Therefore, weight loss responses could in part depend on personal colonic fermentation responses, which affect energy extraction of food and production of microbial metabolites, such as short-chain fatty acids (SCFAs), which exert various effects on host metabolism. Colonic fermentation is the net result of the complex interplay between availability of dietary substrates, the functional capacity of the gut microbiome and environmental (abiotic) factors in the gut such as pH and transit time. While animal studies have demonstrated that the gut microbiota can causally affect obesity, causal and mechanistic evidence from human studies is still largely lacking. However, recent human studies have proposed that the baseline gut microbiota composition may predict diet-induced weight loss-responses. In particular, individuals characterised by high relative abundance of Prevotella have been found to lose more weight on diets rich in dietary fibre compared to individuals with low Prevotella abundance. Although harnessing of personal diet–microbiota interactions holds promise for more personalised nutrition and obesity management strategies to improve human health, there is currently insufficient evidence to unequivocally link the gut microbiota and weight loss in human subjects. To move the field forward, a greater understanding of the mechanistic underpinnings of personal diet–microbiota interactions is needed.

Type
Conference on ‘Obesity and the brain’
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © The Author(s), 2022. Published by Cambridge University Press

Obesity remains a global health challenge, affecting over 650 million adults(Reference Blüher1). The raised body weight is a major risk factor for non-communicable diseases such as CVD, type 2 diabetes and several cancer forms(Reference Blüher1). Therefore, strategies for treating and preventing excessive weight gain are more needed than ever. The traditional approach has been the prescription of hypoenergetic diets with the aim of reducing energy intake and thereby reducing weight. However, the weight loss success varies from individual to individual(Reference Hjorth, Zohar and Hill2), suggesting that one diet does not fit all equally well. Although the fundamental cause of obesity is an energy imbalance between the energy consumed and energy expended, the aetiology of obesity is multifactorial due to a number of external and individual factors affecting the energy equilibrium. One such individual factor is the gut microbiota (or gut microbiome), a term referring to all microorganisms (i.e. bacteria, archaea, fungi, viruses and protozoa) inhabiting our gut, which 15 years ago was proposed to influence host energy homoeostasis(Reference Bäckhed, Manchester and Semenkovich3Reference Turnbaugh, Ley and Mahowald5). These initial studies fuelled interest in the human gut microbiota, and since then a growing number of mechanisms linking diet, gut microbiota and energy homoeostasis have been discovered(Reference Cani, Van Hul and Lefort6). Here, we first discuss how diet shapes the gut microbiota, then we discuss the role of the gut microbiota in weight loss and the underlying mechanisms linking diet–microbiota interactions with body weight control and finally, we discuss future directions on diet–microbiota interactions in relation to weight loss and obesity management.

Diet shapes the gut microbiota

More than a decade ago, scientists reported that the gut microbiome of mammalian species are strongly dependent on whether they are carnivores (meat eaters), herbivores (plant eaters) or omnivores (both meat and plant eaters)(Reference Ley, Hamady and Lozupone7). Today, we know that the gut microbiome also in human subjects is linked to long-term dietary patterns(Reference Wu, Chen and Hoffmann8Reference De Filippis, Pellegrini and Vannini10). Furthermore, we know that gut microbiomes across populations are strongly associated with lifestyle(Reference Smits, Leach and Sonnenburg9), and that the microbiome is different with lower diversity in industrialised populations compared with ancestral populations(Reference Smits, Leach and Sonnenburg9,Reference Clemente, Pehrsson and Blaser11Reference Yatsunenko, Rey and Manary13) , suggesting a loss of indigenous microbes in industrialised populations(Reference Blaser and Falkow14). In line herewith, migration from a non-western nation to United States was associated with a loss in gut microbiota diversity(Reference Vangay, Johnson and Ward15). Since a low gut microbiota diversity has been associated with diabetes, obesity and inflammatory diseases(Reference Le Chatelier, Nielsen and Qin16Reference Alam, Amos and Murphy18), and poor outcomes of cancer treatments(Reference Gopalakrishnan, Spencer and Nezi19,Reference Sims, El Alam and Karpinets T20) , a high diversity has been suggested as a measure of a healthy gut ecosystem. Indeed, a diverse diet seems to be linked to a diverse gut microbiota(Reference Heiman and Greenway21). When infants transit from a milk-based diet to a solid diet(Reference Laursen, Bahl and Michaelsen22), they gradually increase their gut microbial diversity concurrent with their progression in dietary complexity(Reference Stewart, Ajami and O'Brien23,Reference Laursen, Andersen and Michaelsen24) . Similarly, it has been observed that adults who consume a high variety of plants have higher gut microbial diversity compared with adults who consume a low variety of plants(Reference McDonald, Hyde and Debelius25). However, a high microbiota diversity has also been linked to a firm stool consistency and long colonic transit time(Reference Vandeputte, Falony and Vieira-Silva26Reference Asnicar, Leeming and Dimidi28), which is associated with increased proteolysis(Reference Roager, Hansen and Bahl27,Reference Nestel, Hvass and Bahl29) . Therefore, a high gut microbiota diversity does not per se imply a healthy gut microbial ecosystem if it is merely a reflection of a slow intestinal system trending towards constipation. Therefore, it remains a challenge to define what constitutes a healthy gut microbiome(Reference Shanahan, Ghosh and O'Toole30).

Gut microbiota-targeted diets have been suggested as a novel mean to increase microbiota diversity to combat and prevent diseases(Reference Wastyk, Fragiadakis and Perelman31). From short-term interventions with drastic changes in diets (high-fat/low-fibre vs. low-fat/high-fibre), we know it is possible to modify the gut microbiome composition within 24–48 h(Reference Wu, Chen and Hoffmann8,Reference David, Maurice and Carmody32) . However, these substantial dietary changes did not change the microbiota diversity and the diet-induced effects on the microbiome were transient and disappeared as soon as the dietary change ceased(Reference David, Maurice and Carmody32), emphasising the stability of the gut microbiome(Reference Faith, Guruge and Charbonneau33). Also, a recent 10-week intervention showed that participants consuming a diet rich in fibre increased their microbiome-encoded glycan-degrading carbohydrate active enzymes, but did not increase microbiota diversity(Reference Wastyk, Fragiadakis and Perelman31), indicating that an increased fibre intake alone over a short time period is insufficient to increase microbiota diversity. Alternatively, one could speculate that an increased intake of dietary fibre may accelerate intestinal transit and thereby confound changes in microbiota diversity(Reference Roager, Hansen and Bahl27). Indeed, some studies have found that an increased intake of dietary fibre(Reference Zhao, Zhang and Ding34) and prebiotic inulin-type fructans(Reference Vandeputte, Falony and Vieira-Silva35) reduced gut microbiota richness, a measure of diversity. These results also challenge the current notion that greater overall diversity implies better health. Having said that, a recent study did in fact observe that participants consuming a diet rich in fermented foods steadily increased their microbiota diversity and decreased inflammatory markers(Reference Wastyk, Fragiadakis and Perelman31). Therefore, microbiota diversity is most likely a net result of both intestinal transit time, engraftment of microbes and nutrient availability. The nutrient availability will also depend on the physicochemical characteristics of the dietary fibre, such as whether it dissolves in water (soluble fibre) or not (insoluble fibre), since insoluble fibre (such as cellulose, hemicellulose and lignin) speed up transit time and are generally less available for microbial degradation(Reference Louis, Solvang and Duncan36). The importance of nutrient availability for engraftment of microbes has elegantly been demonstrated in mice. For example, engraftment of an exogenous Bacteroides strain, harbouring a rare gene cluster for marine polysaccharide (porphyrin) utilisation, into the colonic ecosystem was enabled via administration of porphyran from red seaweeds(Reference Shepherd, Deloache and Pruss37). In line herewith, compared to westernised populations the Japanese population has higher abundance of seaweed polysaccharide-degrading bacteria(Reference Hehemann, Correc and Barbeyron38), the Hadza hunter–gatherers of Tanzania have higher microbiome functional capacity for utilisation of plant carbohydrates(Reference Smits, Leach and Sonnenburg9) and vegans have lower circulating levels of trimethylamine, a microbial metabolite derived from conversion of carnitine, an abundant nutrient in red meat(Reference Koeth, Wang and Levison39,Reference Koeth, Lam-Galvez and Kirsop40) . These studies show the links between a habitual diet and microbiome composition and functionality. However, whether a given dietary change is sufficient to modulate the gut microbiome may depend on to what extent a given dietary change is different from a habitual diet. For example, a wholegrain-rich diet did compared to a refined-grain diet not alter the gut microbiome in Danish adults with a high habitual intake of wholegrains(Reference Roager, Vogt and Kristensen41). In contrast, a low-gluten diet, excluding all grains containing gluten from the diet, significantly changed the gut microbiome in a similar group of Danish adults who had a high habitual intake of grains(Reference Hansen, Roager and Søndertoft42). Also in American adults, a change to an animal-based diet, absent in dietary fibre, had a larger impact on the gut microbiome composition compared to a change to a plant-based diet, which reflected a doubling in amount of dietary fibre compared to the habitual intake of the participants(Reference David, Maurice and Carmody32). Thus, adding more of a given food to the diet may not induce changes in the gut microbiome if the particular food already constitutes a significant part of the habitual diet.

Another significant challenge within the field of diet–microbiota interactions is the fact that individuals' gut microbiome respond differently to similar foods(Reference Johnson, Vangay and Al-Ghalith43). Indeed, personal microbiome-dependent responses to dietary fibres(Reference Venkataraman, Sieber and Schmidt44Reference Walker, Ince and Duncan46), artificial sweeteners(Reference Suez, Korem and Zeevi47) and breads(Reference Korem, Zeevi and Zmora48) have been observed. While this complicates the field, it also provides an opportunity for better understanding why people benefit differently in terms of health when adhering to the same diet. For example, the highly individualised gut microbiota compositions have been found to improve predictive models of postprandial plasma glucose (6⋅4 % variation explained), insulin (5⋅8 % variation explained) and TAG (7⋅5 % variation explained) responses in healthy adults(Reference Berry, Valdes and Drew49Reference Mendes-Soares, Raveh-Sadka and Azulay51).

Stratification of subjects according to the gut microbiome was introduced a decade ago with the concept of microbial enterotypes, which were defined according to microbiome variations in the abundance of the genera Bacteroides, Prevotella and Ruminococcus, respectively(Reference Arumugam, Raes and Pelletier52). These genera have consistently been found to explain a large proportion of the microbiome composition variations between populations(Reference Wu, Chen and Hoffmann8,Reference Smits, Leach and Sonnenburg9,Reference Costea, Hildebrand and Manimozhiyan53) , and enterotypes represent a way of capturing preferred microbial community structures in the human gut(Reference Costea, Hildebrand and Manimozhiyan53). Although enterotype establishment has been suggested to occur already between the age of 9 and 36 months(Reference Bergström, Skov and Bahl54), the underlying factors diversifying the gut microbiome compositions into enterotypes remain largely unknown. However, diet is likely to be one of the determinant factors shaping the gut microbiome composition. The dominant genus in industrialised populations, Bacteroides, has been associated with diets rich in protein and animal fat, whereas Prevotella, dominant in traditional populations across Asia, Africa and South America, has been linked to carbohydrates(Reference Wu, Chen and Hoffmann8,Reference Smits, Leach and Sonnenburg9) . Despite the links between enterotypes and these dietary patterns, short-term dietary interventions over 10 d including high-fat/low-fibre or low-fat/high-fibre diets(Reference Wu, Chen and Hoffmann8), as well as a 6-month dietary intervention on a new Nordic diet(Reference Roager, Licht and Poulsen55), were insufficient to change the participants' enterotypes, emphasising that they are mainly associated with long-term diets(Reference Wu, Chen and Hoffmann8). Given that enterotypes appear remarkably stable, enterotypes have been proposed as a biomarker, which could be relevant when assessing personal weight-loss responses to a given dietary change(Reference Christensen, Roager and Astrup56), as discussed further below.

Role of gut microbiota in body weight regulation

The idea that the gut microbiota could causally affect obesity and host energy homeostasis came with the ground-breaking study by Turnbaugh et al. in 2006(Reference Turnbaugh, Ley and Mahowald5). By transplanting obese- and lean-associated microbes, respectively, into germ-free mice (completely devoid of microorganisms), they demonstrated that mice receiving microbes from obese donors gained more weight compared to mice receiving microbes from lean donors, despite consuming the same amount of chow diet(Reference Turnbaugh, Ley and Mahowald5). This study was followed by another landmark study demonstrating that faecal microbiota transplantation (FMT) from human twins with or without obesity into germ-free mice transfers the phenotype of the human donor to the recipient animal(Reference Ridaura, Faith and Rey57). It was suggested that the obese microbiome is more efficient in harvesting energy from the diet(Reference Napolitano and Covasa58). In agreement herewith, another study transplanted stool from sixteen lean and sixteen obese children into germ-free mice and found that weight grain of the mice was negatively associated with faecal gross energy(Reference Zhang, Bahl and Roager59). However, this study also noted that faecal gross energy correlated positively with the sum of caecal SCFAs, which indicated that higher excretion of energy in the faeces is not necessarily due to an inefficient bacterial fermentation(Reference Zhang, Bahl and Roager59). More recently, the gut microbiota has also been suggested to play a role in weight regain following weight loss, which is a central challenge in obesity management following weight loss(Reference Blüher1). Using mouse models of weight loss and recurrent obesity, Thais et al. found that high-fat diet-induced alterations to the microbiome persist over long periods of time and enhance the rate of weight regain during the post-dieting phase(Reference Thaiss, Itav and Rothschild60). FMT confirmed that the weight-regain phenotype could be transferred to germ-free mice and the weight gain magnitude could be predicted by the microbiome composition(Reference Thaiss, Itav and Rothschild60). Today, almost two decades since the initial microbiota-weight findings in mice, similar convincing findings in human studies with respect to energy harvest and FMT are still lacking(Reference Zhang, Mocanu and Cai61). Epidemiological studies offer no clear consensus on associations between the gut microbial composition and adiposity(Reference Vander Wyst, Ortega-Santos and Toffoli62,Reference Crovesy, Masterson and Rosado63) . A few case studies have reported increased weight gain upon FMT in human subjects(Reference Alang and Kelly64,Reference De Clercq, Frissen and Davids65) , but larger FMT studies have not found consistent effects. For example, one study with weekly FMT administration for 3 months in adults with obesity resulted in microbiota changes, but no effects on body weight were observed(Reference Yu, Gao and Stastka66). Similarly, a 6-month FMT intervention did not result in weight loss among obese adults; however, it led to reductions in the android:gynoid fat ratio, indicating improvement of visceral fat distribution(Reference Leong, Jayasinghe and Wilson67). More recently, a trial(Reference Rinott, Youngster and Yaskolka Meir68) evaluated in ninety participants whether diet-modulated autologous FMT, collected during a weight loss period and administrated in a weight regain period, could affect weight regain after the weight loss period. No significant differences in weight regain were observed between the autologous FMT group and placebo. However, a subgroup of the autologous FMT group adhering to a green-Mediterranean diet, enriched with plants and polyphenols, significantly attenuated weight regain(Reference Rinott, Youngster and Yaskolka Meir68).

Another gut microbiota-centred approach to modulate body weight includes the supplementation of live bacteria, often referred to as probiotics. However, only a few probiotic interventions in humans have given promises in regard to promoting fat loss(Reference Guazzelli Marques, de Piano Ganen and Zaccaro de Barros69). In 2013, consumption of fermented milk containing the probiotic strain, Lactobacillus gasseri SBT2055, was found to lower abdominal adiposity, which was not found for the control milk after 12-week consumption(Reference Kadooka, Sato and Ogawa70). Another study also found comparably abdominal adipose tissue-lowering effects following 12-week L. gasseri, as a probiotic intervention in overweight subjects(Reference Kim, Yun and Kim71). In contrast to Lactobacillus strains, which are historically on of the primary bacterial groups applied as probiotics, Akkermansia muciniphila is a novel candidate with great interest as this species consistently has been linked to metabolic health in epidemiological studies(Reference Dao, Everard and Aron-Wisnewsky72,Reference Cani and de Vos73) . Depommier et al. recently demonstrated that 3-month oral supplementation of pasteurised A. muciniphila led to improved insulin sensitivity, reduced plasma total cholesterol and tended to decrease body weight compared to placebo in overweight adults. Notably, these metabolic changes occurred independent of detectable changes in the microbiome composition(Reference Depommier, Everard and Druart74). Altogether, animal experiments have provided compelling evidence suggesting a causal role of the gut microbiota in relation to weight gain and re-gain following weight loss, respectively. However, there is a lack of evidence from human clinical trials to indicate an effect of gut microbiota on weight loss and weight gain, and both FMT and probiotic interventions have shown inconsistent results.

Baseline gut microbiota as a determinant of diet-induced weight loss success

Although FMTs and probiotic-interventions in human trials have shown limited effects with respect to modulating body weight, differences in the intrinsic gut microbiome could potentially play a role in determining weight loss responses to treatments. This could in particular be of importance when evaluating the effects of diets with high amounts of complex polysaccharides that target different species within the gut(Reference Zhao, Zhang and Ding34,Reference Christensen, Roager and Astrup56,Reference Nguyen, Deehan and Zhang75) . Down these lines, several research groups have explored the concept of baseline gut microbiome features as predictors of weight loss success following interventions. One approach has been to apply machine learning on omics-data including intestinal microbiome and urine metabolome features to predict weight loss. For example, one study found that prediction of weight loss when consuming grain-based diets was improved by inclusion of several microbial features including butyrate-producing species(Reference Nielsen, Helenius and Garcia76). Similarly, another group found that microbiota composition outperformed other relevant parameters in predicting weight loss following a 30–50 % energy-restricted diet for 6-months(Reference Jie, Yu and Liu77). More specifically, Blautia wexlerae and Bacteroides dorei abundances were the strongest predictors of weight loss, but only among the participants with increased abundance of these at baseline(Reference Jie, Yu and Liu77). Although such computational approaches are attractive, many of the algorithms are ‘black boxes’, which depend on the nature of the training data set. This limits the applicability of such approaches across populations. In our group, we have instead applied a more simplistic approach and stratified subjects according to microbial enterotypes, inferred by the Prevotella:Bacteroides ratio(Reference Roager, Licht and Poulsen55). In particular, we have focused on weight-loss responses in high-fibre studies, since the Prevotella enterotype has been suggested to be more specialised in degrading fibre compared to the Bacteroides enterotype(Reference Christensen, Roager and Astrup56). Consistently, high-fibre intervention studies with Danish overweight and obese individuals have shown large inter-individual variation in weight loss(Reference Roager, Vogt and Kristensen41,Reference Poulsen, Due and Jordy78Reference Kjølbæk, Benítez-Páez and Gómez del Pulgar81) , and differences in dietary adherence have not explained this variation, even when evaluating intake by quantitative dietary biomarkers(Reference Dent, McPherson and Harper82). Yet, in five independent post-hoc studies, we have found that the Prevotella enterotype is associated with better weight regulation in response to an increased dietary fibre intake(Reference Hjorth, Ritz and Blaak83Reference Christensen, Sørensen C and Wøhlk87). More specifically, in three 6-month intervention studies, a high intake of fibre (mainly from whole grains) was associated with weight loss among participants with a high Prevotella:Bacteroides ratio, but not among individuals with a low Prevotella:Bacteroides ratio(Reference Hjorth, Ritz and Blaak83Reference Hjorth, Christensen and Kjølbæk85). Also, in a 6-week wholegrain study with increased rye and wheat fibre consumption, Prevotella abundance predicted weight loss and participants with high baseline Prevotella abundance lost 2 kg more compared to the individuals with low Prevotella abundance(Reference Christensen, Vuholm and Roager86). Moreover, when reanalysing a 4-week prebiotic intervention with arabinoxylan oligosaccharides (10⋅4 g/d), a fibre type abundant in whole grains, a small, but significant weight change difference was found between the subjects of the Prevotella and Bacteroides enterotypes(Reference Christensen, Sørensen C and Wøhlk87). Here, subjects with a Bacteroides enterotype gained weight, whereas subjects with a Prevotella enterotype remained at stable weight. By analysing the microbiota composition beyond the genus level and Prevotella:Bacteroides groups, we found Bacteroides cellulosilyticus to be the most important predictor of weight gain(Reference Christensen, Sørensen C and Wøhlk87). This species has previously been found to digest arabinoxylan and to affect interspecies competition among Bacteroides species, which have vastly different functionalities(Reference Patnode, Beller and Han88).

Furthermore, we recently discovered that the association between the Prevotella enterotype and weight loss appeared only to be evident for participants characterised by a low copy number of the salivary α–amylase 1 (AMY1) gene(Reference Hjorth, Christensen and Larsen89). AMY1 is one of the genes with largest copy number variation(Reference Morán-Ramos, Villarreal-Molina and Canizales-Quinteros90) and the secretion of amylase is essential for starch digestion in the oral cavity, stomach and duodenum, until starches are met by the pancreatic amylase(Reference Atkinson, Hancock and Petocz91). Our discovery could indicate that not only wholegrain fibre (e.g. arabinoxylans) but also the availability of starch influences microbial functionality and thereby human metabolism(Reference Deehan, Yang and Perez-Muñoz92). Accordingly, we hypothesise that participants with a low AMY1 copy number consuming diets rich in starch may not fully degrade the starch by salivary and pancreatic amylase, and consequently starch will undergo fermentation in the lower gastrointestinal tract(Reference Hjorth, Christensen and Larsen89). While this remains to be further tested, other studies suggest that a low AMY1 copy number results in distinct gut microbial functions and metabolites, as low AMY1 copy number has been associated with increased microbial abundance of enzymes involved in the degradation of complex carbohydrates(Reference Poole, Goodrich and Youngblut93) and methane production(Reference Atkinson, Hancock and Petocz91). The associations observed in these studies suggest that differences in the baseline gut microbiota composition may predict diet-induced weight loss responses, which could also depend on host genetics. But to date, no studies have tested these hypotheses a priori.

Underlying mechanisms linking diet–microbiota interactions with body weight control

Stepping away from correlation to causation may be facilitated by understanding the underlying mechanisms linking personal diet–microbiota interactions and body weight control. We here discuss the factors that determine colonic fermentation and the resulting diet-derived microbial products, which can interact with our host metabolism.

Personal colonic fermentation responses

To link diet–microbiota interactions with host health and body weight regulation, we need to move beyond profiling of the gut microbiota to the assessment of gut microbial activity, and to understand the factors that shape the colonic fermentation(Reference Roager and Dragsted94). In this regard, intestinal transit time, which is the time food takes to travel through the gastrointestinal system, appears as a largely neglected, but a relevant factor. We and others have shown that both intestinal transit time and stool consistency, a proxy of intestinal transit time, are strongly associated with the gut microbiome composition(Reference Vandeputte, Falony and Vieira-Silva26Reference Nestel, Hvass and Bahl29). Indeed, population studies have reported that measures of transit time explain more of the gut microbiome variation than dietary and health markers(Reference Asnicar, Leeming and Dimidi28,Reference Falony, Joossens and Vieira-Silva95) . Given that intestinal transit time varies a lot from individual to individual(Reference Roager, Hansen and Bahl27,Reference Asnicar, Leeming and Dimidi28) , transit time has been suggested as an important driver of inter- and intra-individual variations in the gut microbiome composition and diversity(Reference Falony, Vieira-Silva and Raes96). This could be due to the fact that differences in transit time have been associated with changes in substrate availability and environmental factors (such as pH) in the colon(Reference Lewis and Cochrane97). Loose stools, reflecting faster transit time, have been found to harbour larger fractions of bacteria with a high predicted maximal growth rate(Reference Vieira-Silva, Falony and Darzi98), whereas firm stools, reflecting slow transit time, have been associated with higher abundance of slow-growing species such as methanogens and higher diversity(Reference Roager, Hansen and Bahl27,Reference Lewis and Cochrane97) , suggesting that bacterial ecosystem dynamics and growth are shaped by transit time. Furthermore, differences in intestinal transit time are also coupled to differences in colonic fermentation, probably as it changes the time for digestion. More specifically, a long intestinal transit time is associated with reduced levels of saccharolytic metabolites (e.g. SCFAs, such as butyrate, propionate and acetate) and increased levels of proteolytic metabolites (e.g. branched SCFAs, such as isobutyric acid and isovaleric acid)(Reference Roager, Hansen and Bahl27,Reference Müller, Hermes and Canfora99,Reference Lewis and Heaton100) , suggesting a switch in bacterial fermentation from carbohydrates to proteins in the case of a long transit time. Our habitual diet also shapes the metabolic capacity of the gut microbiota, which could be key for personal colonic fermentation responses. Enterotypes, which are linked to long-term dietary patterns(Reference Wu, Chen and Hoffmann8), have been suggested to differ in metabolic capacity for degradation of carbohydrates, proteins and lipids(Reference Vieira-Silva, Falony and Darzi98), and in vitro studies have suggested that colonic fermentation of dietary fibres into SCFAs varies according to enterotypes(Reference Chen, Long and Zhang101). In agreement, we previously observed that when stratifying subjects into two enterotypes by the relative abundance of Prevotella, higher faecal levels of propionate were observed at baseline in subjects with high Prevotella abundance compared to the group with low Prevotella abundance(Reference Christensen, Vuholm and Roager86). Yet, we did not observe any changes in faecal SCFA levels following 6-week ad-libitum intake of wholegrains according to the two enterotypes(Reference Christensen, Vuholm and Roager86). Thus, it remains largely unknown whether the observed enterotype-dependent weight loss success on fibre-rich diets are linked to differences in microbiota-dependent energy harvest or distinct microbial metabolite profiles(Reference Christensen, Roager and Astrup56).

Manipulating the amounts and types of dietary fibres in the diet often results in changes in several interrelated bacterial species(Reference Zhao, Zhang and Ding34,Reference Hansen, Roager and Søndertoft42) , which based on their co-abundant behaviour can be defined as guilds(Reference Wu, Zhao and Zhang102). Changes in guilds have also been coupled with changes in colonic fermentation products such as SCFAs(Reference Zhao, Zhang and Ding34) and gases(Reference Hansen, Roager and Søndertoft42), suggesting that the concept of guilds could also be used as a way to reduce the dimensionality of the microbiome and to stratify subjects in dietary weight loss interventions. Also specific bacterial taxa, sometimes referred to as keystone species, could be important for understanding personal colonic fermentation responses to specific dietary fibres(Reference Patnode, Beller and Han88). This has been nicely illustrated for resistant starch(Reference Venkataraman, Sieber and Schmidt44,Reference Walker, Ince and Duncan46,Reference Deehan, Yang and Perez-Muñoz92) . An intervention study including twenty healthy adults showed that daily supplementation with unmodified potato-resistant starch (type 2) increased faecal butyrate concentrations depending on the initial abundance of resistant starch-degrading organisms (Bifidobacterium adolescentis and Ruminococcus bromii)(Reference Venkataraman, Sieber and Schmidt44). Another dose–response trial with three resistant starches (all type 4) in healthy volunteers showed that distinct dietary fibre structures direct SCFA output towards either propionate or butyrate, and induce selective enrichments of a few resistant starch-degrading species that possess adaptations to the respective substrates(Reference Deehan, Yang and Perez-Muñoz92). These studies emphasised that specific bacteria can metabolise distinct fibre structures. Therefore, differences in metabolic capacity of the gut microbiome as captured by enterotypes, bacterial guilds, abundance of specific keystone bacterial species and/or specific genes may determine the colonic fermentation as well. Altogether, colonic fermentation is in essence a trade-off between saccharolytic and proteolytic fermentation, which depends on the complex interplay between gut microbiome's composition and metabolic potential, the substrate availability, colonic pH and transit time(Reference Lewis and Heaton100,Reference Macfarlane, Quigley and Hopkins103,Reference Walker, Duncan and Mcwilliam leitch104) . Since these factors vary substantially from individual to individual, personal colonic fermentation responses and the resulting diet-derived microbial metabolites could be key for elucidating the underlying mechanisms of diet–microbiota interactions in weight-loss responses (Fig. 1).

Fig. 1. Personal diet–microbiota interactions and human energy homeostasis. Person-specific colonic fermentation is a trade-off between saccharolytic and proteolytic fermentation, which depends on the complex interplay between the dietary substrates available, the metabolic potential of the gut microbiota and environmental (abiotic) factors, such as pH and transit time; factors which are highly individual. In addition, also differences in host genetics could affect this interplay. For example, differences in the copy number of the salivary α-amylase 1 (AMY1) gene could affect degradation of starch via amylase in the upper-gastrointestinal tract and thereby affect the availability of starch for colonic fermentation. Consequently, personal diet–microbiota interactions may affect human energy metabolism through energy excretion and the generation of microbiota-derived metabolites, such as SCFAs, tryptophan catabolites, secondary bile acids and metabolites mimetic of host hormones. These microbial metabolites could exert different effects on host metabolism – e.g. by serving as energy substrates, by stimulating secretion of appetite-regulating hormones, including glucagon-like peptide 1 (GLP-1) and peptide YY (PYY) in enteroendocrine cells, by regulating energy expenditure in adipose tissue, and by regulating appetite and satiety in the brain. Stratification by gut microbiota community characteristics defined by enterotypes, guilds, keystone species or specific genes, or abiotic factors could potentially be predictive of person-specific diet–microbiota interactions and linked to weight loss responses.

Energy harvest and SCFAs as mediators of host–microbial cross-talk

The pioneering study by Turnbaugh and colleagues, mentioned previously, linked increased microbiota-dependent energy harvest with increased intestinal levels of the microbial-derived SCFAs, acetate and butyrate(Reference Turnbaugh, Ley and Mahowald5). SCFAs are end-products of bacterial fermentation of complex carbohydrates and to some degree of proteins and peptides that have escaped digestion by host enzymes in the upper gut. These findings were corroborated by other studies that reported an increased microbial metabolic capacity for carbohydrate fermentation in obese mice and human subjects(Reference Bäckhed, Ding and Wang4,Reference Turnbaugh, Hamady and Yatsunenko17) , and increased faecal levels of SCFAs in obese individuals(Reference Schwiertz, Taras and Schafer105,Reference Yamamura, Nakamura and Ukawa106) . Despite the compelling theory that increased energy harvest could be linked to obesity, intestinal SCFA concentrations have not consistently been linked to obesity or related metabolic disorders(Reference Ridaura, Faith and Rey57,Reference Murphy, Cotter and Healy107) , and evidence from human subjects are still rather limited. Nonetheless, SCFAs are likely to be key mediators of host–microbial cross-talk and relevant for body weight control(Reference Koh, De Vadder and Kovatcheva-Datchary108). As reviewed elsewhere(Reference Koh, De Vadder and Kovatcheva-Datchary108), SCFAs can facilitate gut–brain axis signalling by activating cell surface G protein-coupled receptors (GPCRs), including GPR41, GPR43 and GPR109A(Reference Husted, Trauelsen and Rudenko109). Butyrate serves as a primary energy source for colonocytes and is estimated to contribute to 5–10 % of the human energy requirement(Reference McNeil110), acetate mediates fat accumulation via GPR43 in adipose tissue(Reference Kimura, Ozawa and Inoue111), whereas propionate is used as a substrate for gluconeogenesis in the intestine(Reference De Vadder, Kovatcheva-Datchary and Goncalves112), as well as in the liver(Reference Cummings, Pomare and Branch113,Reference den Besten, Lange and Havinga114) . Furthermore, SCFAs stimulate secretion of peptide YY (PYY) and glucagon-like peptide-1 (GLP-1) from enteroendocrine cells (L-cells)(Reference Samuel, Shaito and Motoike115,Reference Tolhurst, Heffron and Lam116) , regulate immune cell functions(Reference Macia, Tan and Vieira117,Reference Brown, Goldsworthy and Barnes118) and affect intestinal transit(Reference Wichmann, Allahyar and Greiner119). Both GLP-1 and PYY are gut peptide hormones, which can affect appetite; either by reaching the brain through the circulation or through direct activation of vagal afferents lying in the lamina propria of the gut(Reference Holst120). Mouse studies have shown that supplementation of SCFAs can protect against weight gain(Reference Gao, Yin and Zhang121,Reference Lin H, Frassetto and Kowalik122) . Consistently, rectal infusions of SCFA mixtures into the colon of overweight/obese men, mimicking the SCFA levels reached after high-fibre intake, increased fat oxidation, energy expenditure and PYY, and decreased lipolysis(Reference Canfora, van der Beek and Jocken123). Similarly, infusions of acetate into the distal colon in overweight/obese men promoted whole-body fat oxidation and plasma PYY in the fasting state(Reference van der Beek, Canfora and Lenaerts124), suggesting short-term beneficial effects on host metabolism. Also, 6-month oral administration of propionate (in the form of inulin-propionate ester) in overweight individuals reduced weight gain compared to the control group(Reference Chambers, Viardot and Psichas125). Altogether, these studies suggest that SCFAs exert multiple beneficial effects and may modulate body weight (Fig. 1). However, human studies linking stool SCFAs to body weight have been inconsistent, as eluted to previously, indicating that stool SCFA concentrations might be context-dependent. Also, what complicates the study of SCFAs is the fact that most of the colonic fermentation and formation of SCFAs occur in the caecum and proximal colon(Reference Cummings, Pomare and Branch113); sites which are rarely sampled in human intervention studies. Furthermore, as 95 % of SCFAs are estimated to be absorbed during transit through the colon(Reference Von Engelhardt, Rönnau and Rechkemmer126), the biological meaning of stool SCFA concentrations is difficult to interpret. Therefore, further research is needed with respect to SCFA patterns, dynamics and equilibria along the gastrointestinal tract to elucidate the complex multi-faceted role of SCFAs in the context of obesity and weight loss interventions.

Microbiota-derived molecules beyond SCFAs in weight regulation

Besides SCFAs, also several other microbial-derived metabolites are likely to play a role in regulating host energy homoeostasis (Fig. 1). This includes secondary bile acids, which are formed when the gut microbiota modifies primary bile acids into secondary bile acids and deconjugated bile acids(Reference Ridlon, Kang and Hylemon127). These chemical modifications change the bile acids' reabsorption from the intestine, affecting the circulating bile acid pool and excretion of bile acids in the faeces. Furthermore, the chemical modifications of the bile acids change their affinity for the farnesoid-X receptor and Takeda-G-protein-receptor-5(Reference Wahlström, Sayin and Marschall128). Bile acid-induced activation of these receptors stimulates GLP-1 secretion from L-cells, increases energy expenditure and thermogenesis in adipose tissue, and mediates satiety in the brain(Reference Wahlström, Sayin and Marschall128). Therefore, differences in microbial conversions of bile acids among individuals could potentially contribute to person-specific weight-loss responses to diets. Also microbial-derived tryptophan catabolites, which in recent years have been linked to several diseases(Reference Roager and Licht129), could potentially be involved in appetite regulation. Indole has been shown to modulate GLP-1 secretion from L-cells(Reference Chimerel, Emery and Summers130), whereas tryptamine, indole and indole-3-aldehyde have been shown to stimulate intestinal serotonin release and affect gut motility(Reference Ye, Bae and Cassilly131,Reference Bhattarai, Williams and Battaglioli132) . Yet, evidence from human studies is still very limited. Other microbial molecules might also interfere with ndocrine regulation. A bacterial protein secreted by Escherichia coli, mimetic of the host peptide α-melanocyte-stimulating hormone, the caseinolytic peptidase B protein homologue, affect food intake and body weight in mice(Reference Breton, Tennoune and Lucas133,Reference Tennoune, Chan and Breton134) . Intriguingly, the abundance of gut bacterial caseinolytic peptidase B-like gene function has been associated with a decreased body weight, and detected in lower abundance in subjects with obesity(Reference Arnoriaga-Rodríguez, Mayneris-Perxachs and Burokas135). Furthermore, higher circulating levels of the caseinolytic peptidase B protein have been detected in individuals with eating disorders such as anorexia nervosa compared with healthy individuals(Reference Breton, Legrand and Akkermann136). The human gut microbiota has also been found to encode N-acyl amides that interact with GPCRs. Mouse and cell-based models have demonstrated that the N-acyl amides regulate metabolic hormones and glucose homoeostasis via GPR119 to the same degree as human ligands(Reference Cohen, Esterhazy and Kim137). Finally, A. muciniphila has also been found to produce an 84 kDa protein (P9), which induces GLP-1 in L-cells and reduces food intake and body weight in mice fed with a high-fat diet(Reference Yoon, Cho and Yun138). This could potentially explain why daily oral supplementation with pasteurised A. muciniphila improved insulin sensitivity and slightly decreased body weight in overweight/obese insulin-resistant volunteers(Reference Depommier, Everard and Druart74).

These findings suggest that chemical mimicry of eukaryotic signalling molecules may be common among commensal gut bacteria. If proven effective in human trials, microbiota-encoded molecules may provide additional strategies to ameliorate obesity.

Conclusion and future perspectives

Personal microbiota responses and inter-individual variations in weight loss responses to dietary changes are both two well-established concepts. With the fascinating findings on gut microbiota and body weight during the past 15 years, we continue to have good reasons to consider a causal role of the gut microbiota in body weight regulation. This has recently been underlined by a study by Jeffrey Gordon and colleagues showing that a dietary fibre-rich microbiota-directed supplement can improve growth in children with moderate acute malnutrition compared with an existing supplementary food, emphasising that it is possible to direct food towards the gut microbiota and thereby impact body weight(Reference Chen, Mostafa and Hibberd139). Moving forward, human studies with a priori hypotheses are needed to investigate the baseline gut microbiota as a predictor of body weight gain or loss success in dietary interventions. Furthermore, the idea of tailored diets matching the individual's microbiota and genetic makeup with the aim of stimulating weight loss necessitates an enhanced understanding of the mechanistic underpinnings of personal diet–microbiota interactions. To advance the field, a single faecal spot sample to characterise the human microbiota composition may not be adequate in future studies, as significant intra-individual variation exists over time(Reference Vandeputte, De Commer and Tito140), emphasising the need for longitudinal sampling. Furthermore, it is essential to move beyond studying the composition of the gut microbiota to study the gut microbial activity and metabolites(Reference Roager and Dragsted94), the environmental conditions throughout the gut including pH and transit time(Reference Diaz Tartera, Webb and Al-Saffar141), and to sample from different locations throughout the gut. The gut microbiota could play a role in determining nutrient absorption in the small intestine(Reference von Schwartzenberg, Bisanz and Lyalina142) and colonic fermentation in the proximal colon(Reference Cummings, Pomare and Branch113). Yet, these sites remain currently understudied in human diet–microbiota interaction studies.

Recent successful efforts in the development of microbiota-dependent personalised diets regulating blood sugar levels(Reference Zeevi, Korem and Zmora50,Reference Ben-Yacov, Godneva and Rein143) provide hope for future efforts. Similar efforts have not yet been made with respect to weight loss and/or weight gain. Yet, with a better understanding of personal diet–microbiota interactions, stratification according to gut microbiota characteristics at a compositional, functional and/or activity level has the potential to improve personalised nutrition and obesity management strategies.

In conclusion, while animal studies show causal links between the microbiome and body weight regulation, there is currently insufficient evidence to unequivocally show a link between the gut microbiota and weight loss in human subjects. Hence, more human studies are warranted to further investigate interactions between the gut microbiota and diet-induced weight loss responses.

Acknowledgements

The authors thank colleagues and peers for great discussions while putting this review together.

Financial Support

This study was supported by the Novo Nordisk Foundation (NNF19OC0056246; PRIMA – towards Personalised dietary recommendations based on the interaction between diet, microbiome and abiotic conditions in the gut).

Conflict of Interest

None.

Authorship

The authors had sole responsibility for all aspects of preparation of this paper.

Footnotes

Both authors contributed equally to this work.

References

Blüher, M (2019) Obesity: global epidemiology and pathogenesis. Nat Rev Endocrinol 15, 288298.CrossRefGoogle ScholarPubMed
Hjorth, MF, Zohar, Y, Hill, JO et al. (2018) Personalized dietary management of obesity based on simple biomarkers. Annu Rev Nutr 38, 119, 28.CrossRefGoogle Scholar
Bäckhed, F, Manchester, JK, Semenkovich, CF et al. (2007) Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci U S A 104, 979984.CrossRefGoogle ScholarPubMed
Bäckhed, F, Ding, H, Wang, T et al. (2004) The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A 101, 1571815723.CrossRefGoogle ScholarPubMed
Turnbaugh, PJ, Ley, RE, Mahowald, MA et al. (2006) An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444, 10271031.CrossRefGoogle ScholarPubMed
Cani, PD, Van Hul, M, Lefort, C et al. (2019) Microbial regulation of organismal energy homeostasis. Nat Metab 1, 3446.CrossRefGoogle ScholarPubMed
Ley, RE, Hamady, M, Lozupone, C et al. (2008) Evolution of mammals and their gut microbes. Science 320, 16471651.CrossRefGoogle ScholarPubMed
Wu, GD, Chen, J, Hoffmann, C et al. (2011) Linking long-term dietary patterns with gut microbial enterotypes. Science 334, 105108.CrossRefGoogle ScholarPubMed
Smits, SA, Leach, J, Sonnenburg, ED et al. (2017) Seasonal cycling in the gut microbiome of the Hadza hunter–gatherers of Tanzania. Science (80). 357, 802805.CrossRefGoogle ScholarPubMed
De Filippis, F, Pellegrini, N, Vannini, L et al. (2016) High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut 65, 18121821.CrossRefGoogle ScholarPubMed
Clemente, JC, Pehrsson, EC, Blaser, MJ et al. (2015) The microbiome of uncontacted Amerindians. Sci Adv 1, 112.CrossRefGoogle ScholarPubMed
Gomez, A, Petrzelkova, KJ, Burns, MB et al. (2016) Gut microbiome of coexisting BaAka pygmies and bantu reflects gradients of traditional subsistence patterns. Cell Rep 14, 21422153.CrossRefGoogle ScholarPubMed
Yatsunenko, T, Rey, FE, Manary, MJ et al. (2012) Human gut microbiome viewed across age and geography. Nature 486, 222227.CrossRefGoogle ScholarPubMed
Blaser, MJ & Falkow, S (2009) What are the consequences of the disappearing human microbiota? Nat Rev Microbiol 7, 887894.CrossRefGoogle ScholarPubMed
Vangay, P, Johnson, AJ, Ward, TL et al. (2018) US immigration westernizes the human gut microbiome. Cell 175, 962972, e10.CrossRefGoogle ScholarPubMed
Le Chatelier, E, Nielsen, T, Qin, J et al. (2013) Richness of human gut microbiome correlates with metabolic markers. Nature 500, 541546.CrossRefGoogle ScholarPubMed
Turnbaugh, PJ, Hamady, M, Yatsunenko, T et al. (2009) A core gut microbiome in obese and lean twins. Nature 457, 480484.Google ScholarPubMed
Alam, MT, Amos, GCA, Murphy, ARJ et al. (2020) Microbial imbalance in inflammatory bowel disease patients at different taxonomic levels. Gut Pathog 12, 18.CrossRefGoogle ScholarPubMed
Gopalakrishnan, V, Spencer, CN, Nezi, L et al. (2018) Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 359, 97103.CrossRefGoogle ScholarPubMed
Sims, TT, El Alam, MB, Karpinets T, V. et al. (2021) Gut microbiome diversity is an independent predictor of survival in cervical cancer patients receiving chemoradiation. Commun Biol. 4, 110.CrossRefGoogle ScholarPubMed
Heiman, ML & Greenway, FL (2016) A healthy gastrointestinal microbiome is dependent on dietary diversity. Mol Metab 5, 317320.CrossRefGoogle ScholarPubMed
Laursen, MF, Bahl, MI, Michaelsen, KF et al. (2017) First foods and gut microbes. Front Microbiol 8, 356.CrossRefGoogle ScholarPubMed
Stewart, CJ, Ajami, NJ, O'Brien, JL et al. (2018) Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature 562, 583588.CrossRefGoogle ScholarPubMed
Laursen, MF, Andersen, LBB, Michaelsen, KF et al. (2016) Infant gut microbiota development is driven by transition to family foods independent of maternal obesity. mSphere 1, 116.CrossRefGoogle ScholarPubMed
McDonald, D, Hyde, E, Debelius, JW et al. (2018) American gut: an open platform for citizen science microbiome research. mSystems 3, 125.CrossRefGoogle ScholarPubMed
Vandeputte, D, Falony, G, Vieira-Silva, S et al. (2015) Stool consistency is strongly associated with gut microbiota richness and composition, enterotypes and bacterial growth rates. Gut 65, 5762.CrossRefGoogle ScholarPubMed
Roager, HM, Hansen, LBS, Bahl, MI et al. (2016) Colonic transit time is related to bacterial metabolism and mucosal turnover in the gut. Nat Microbiol 1, 16093.CrossRefGoogle Scholar
Asnicar, F, Leeming, ER, Dimidi, E et al. (2021) Blue poo: impact of gut transit time on the gut microbiome using a novel marker. Gut 0, 110.Google Scholar
Nestel, N, Hvass, JD, Bahl, MI et al. (2021) The gut microbiome and abiotic factors as potential determinants of postprandial glucose responses: a single-arm meal study. Front Nutr 7, 19.CrossRefGoogle ScholarPubMed
Shanahan, F, Ghosh, TS & O'Toole, PW (2021) The healthy microbiome – what is the definition of a healthy gut microbiome? Gastroenterology 160, 483494.CrossRefGoogle ScholarPubMed
Wastyk, HC, Fragiadakis, GK, Perelman, D et al. (2021) Gut-microbiota-targeted diets modulate human immune status. Cell 184, 41374153, e14.CrossRefGoogle ScholarPubMed
David, LA, Maurice, CF, Carmody, RN et al. (2014) Diet rapidly and reproducibly alters the human gut microbiome. Nature 505, 559563.CrossRefGoogle ScholarPubMed
Faith, JJ, Guruge, JL, Charbonneau, M et al. (2013) The long-term stability of the human gut microbiota. Science (80) 341, 1237439.CrossRefGoogle ScholarPubMed
Zhao, L, Zhang, F, Ding, X et al. (2018) Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science (80) 359, 11511156.CrossRefGoogle ScholarPubMed
Vandeputte, D, Falony, G, Vieira-Silva, S et al. (2017) Prebiotic inulin-type fructans induce specific changes in the human gut microbiota. Gut 66, 19681974.CrossRefGoogle ScholarPubMed
Louis, P, Solvang, M, Duncan, SH et al. (2021) Dietary fibre complexity and its influence on functional groups of the human gut microbiota. Proc Nutr Soc 80, 386397.CrossRefGoogle Scholar
Shepherd, ES, Deloache, WC, Pruss, KM et al. (2018) An exclusive metabolic niche enables strain engraftment in the gut microbiota. Nature 2018(5577705 557), 434438.CrossRefGoogle Scholar
Hehemann, JH, Correc, G, Barbeyron, T et al. (2010) Transfer of carbohydrate-active enzymes from marine bacteria to Japanese gut microbiota. Nature 464, 908912.CrossRefGoogle ScholarPubMed
Koeth, RA, Wang, Z, Levison, BS et al. (2013) Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 19, 576585.CrossRefGoogle ScholarPubMed
Koeth, RA, Lam-Galvez, BR, Kirsop, J et al. (2019) L-Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans. J Clin Invest 129, 373.CrossRefGoogle ScholarPubMed
Roager, HM, Vogt, JK, Kristensen, M et al. (2019) Whole grain-rich diet reduces body weight and systemic low-grade inflammation without inducing major changes of the gut microbiome: a randomised cross-over trial. Gut 68, 8393.CrossRefGoogle ScholarPubMed
Hansen, LBS, Roager, HM, Søndertoft, NB et al. (2018) A low-gluten diet induces changes in the intestinal microbiome of healthy Danish adults. Nat Commun 9, 4630.CrossRefGoogle ScholarPubMed
Johnson, AJ, Vangay, P, Al-Ghalith, GA et al. (2019) Daily sampling reveals personalized diet–microbiome associations in humans. Cell Host Microbe 25, 789802, e5.CrossRefGoogle ScholarPubMed
Venkataraman, A, Sieber, JR, Schmidt, AW et al. (2016) Variable responses of human microbiomes to dietary supplementation with resistant starch. Microbiome 4, 33.CrossRefGoogle ScholarPubMed
Baxter, NT, Schmidt, AW, Venkataraman, A et al. (2019) Dynamics of human gut microbiota and short-chain fatty acids in response to dietary interventions with three fermentable fibers. MBio 10, e02566–18.CrossRefGoogle ScholarPubMed
Walker, AW, Ince, J, Duncan, SH et al. (2011) Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J 5, 220230.CrossRefGoogle ScholarPubMed
Suez, J, Korem, T, Zeevi, D et al. (2014) Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature 514, 181186.CrossRefGoogle ScholarPubMed
Korem, T, Zeevi, D, Zmora, N et al. (2017) Bread affects clinical parameters and induces gut microbiome-associated personal glycemic responses. Cell Metab 25, 12431253, e5.CrossRefGoogle ScholarPubMed
Berry, SE, Valdes, AM, Drew, DA et al. (2020) Human postprandial responses to food and potential for precision nutrition. Nat Med 2020(26), 110.Google Scholar
Zeevi, D, Korem, T, Zmora, N et al. (2015) Personalized nutrition by prediction of glycemic responses. Cell 163, 10791094.CrossRefGoogle ScholarPubMed
Mendes-Soares, H, Raveh-Sadka, T, Azulay, S et al. (2019) Assessment of a personalized approach to predicting postprandial glycemic responses to food Among individuals without diabetes. JAMA Netw Open 2, e188102.CrossRefGoogle ScholarPubMed
Arumugam, M, Raes, J, Pelletier, E et al. (2011) Enterotypes of the human gut microbiome. Nature 473, 174180.CrossRefGoogle ScholarPubMed
Costea, PI, Hildebrand, F, Manimozhiyan, A et al. (2017) Enterotypes in the landscape of gut microbial community composition. Nat Microbiol 3, 816.CrossRefGoogle ScholarPubMed
Bergström, A, Skov, TH, Bahl, MI et al. (2014) Establishment of intestinal microbiota during early life: a longitudinal, explorative study of a large cohort of Danish infants. Appl Environ Microbiol 80, 28892900.CrossRefGoogle ScholarPubMed
Roager, HM, Licht, TR, Poulsen, SK et al. (2014) Microbial enterotypes, inferred by the Prevotella-to-Bacteroides ratio, remained stable during a 6-month randomized controlled diet intervention with the new Nordic diet. Appl Environ Microbiol 80, 11421149.CrossRefGoogle ScholarPubMed
Christensen, L, Roager, HM, Astrup, A et al. (2018) Microbial enterotypes in personalized nutrition and obesity management. Am J Clin Nutr 108, 645651.CrossRefGoogle ScholarPubMed
Ridaura, VK, Faith, JJ, Rey, FE et al. (2013) Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 341, 110.CrossRefGoogle ScholarPubMed
Napolitano, M & Covasa, M (2020) Microbiota transplant in the treatment of obesity and diabetes: current and future perspectives. Front Microbiol 11, 116.CrossRefGoogle ScholarPubMed
Zhang, L, Bahl, MI, Roager, HM et al. (2017) Environmental spread of microbes impacts the development of metabolic phenotypes in mice transplanted with microbial communities from humans. ISME J 11, 676690.CrossRefGoogle ScholarPubMed
Thaiss, CA, Itav, S, Rothschild, D et al. (2016) Persistent microbiome alterations modulate the rate of post-dieting weight regain. Nature 540, 544551.CrossRefGoogle ScholarPubMed
Zhang, Z, Mocanu, V, Cai, C et al. (2019) Impact of fecal microbiota transplantation on obesity and metabolic syndrome – a systematic review. Nutrients 11, 116.CrossRefGoogle ScholarPubMed
Vander Wyst, KB, Ortega-Santos, CP, Toffoli, SN et al. (2021) Diet, adiposity, and the gut microbiota from infancy to adolescence: a systematic review. Obes Rev 22, 121.CrossRefGoogle ScholarPubMed
Crovesy, L, Masterson, D & Rosado, EL (2020) Profile of the gut microbiota of adults with obesity: a systematic review. Eur J Clin Nutr 74, 12511262.CrossRefGoogle ScholarPubMed
Alang, N & Kelly, CR (2015) Weight gain after fecal microbiota transplantation. Open Forum Infect Dis 2, 12.CrossRefGoogle ScholarPubMed
De Clercq, NC, Frissen, MN, Davids, M et al. (2019) Weight gain after fecal microbiota transplantation in a patient with recurrent underweight following clinical recovery from anorexia nervosa. Psychother Psychosom 88, 5254.Google Scholar
Yu, EW, Gao, L, Stastka, P et al. (2020) Fecal microbiota transplantation for the improvement of metabolism in obesity: the FMT-trim double-blind placebo-controlled pilot trial. PLoS Med 17, 119.CrossRefGoogle ScholarPubMed
Leong, KSW, Jayasinghe, TN, Wilson, BC et al. (2020) Effects of fecal microbiome transfer in adolescents with obesity: the gut bugs randomized controlled trial. JAMA Netw Open 3, e2030415.CrossRefGoogle ScholarPubMed
Rinott, E, Youngster, I, Yaskolka Meir, A et al. (2021) Effects of diet-modulated autologous fecal microbiota transplantation on weight regain. Gastroenterology 160, 158173, e10.CrossRefGoogle ScholarPubMed
Guazzelli Marques, C, de Piano Ganen, A, Zaccaro de Barros, A et al. (2020) Weight loss probiotic supplementation effect in overweight and obesity subjects: a review. Clin Nutr 39, 694704.CrossRefGoogle ScholarPubMed
Kadooka, Y, Sato, M, Ogawa, A et al. (2013) Effect of Lactobacillus gasseri SBT2055 in fermented milk on abdominal adiposity in adults in a randomised controlled trial. Br J Nutr 110, 16961703.CrossRefGoogle Scholar
Kim, J, Yun, JM, Kim, MK et al. (2018) Lactobacillus gasseri BNR17 supplementation reduces the visceral fat accumulation and waist circumference in obese adults: a randomized, double-blind, placebo-controlled trial. J Med Food 21, 454461.CrossRefGoogle ScholarPubMed
Dao, MC, Everard, A, Aron-Wisnewsky, J et al. (2016) Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology. Gut 65, 426436.CrossRefGoogle ScholarPubMed
Cani, PD & de Vos, WM (2017) Next-generation beneficial microbes: the case of Akkermansia muciniphila. Front Microbiol 8, 18.CrossRefGoogle ScholarPubMed
Depommier, C, Everard, A, Druart, C et al. (2019) Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nat Med 25, 10961103.CrossRefGoogle ScholarPubMed
Nguyen, NK, Deehan, EC, Zhang, Z et al. (2020) 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 8, 121.CrossRefGoogle Scholar
Nielsen, RL, Helenius, M, Garcia, SL et al. (2020) Data integration for prediction of weight loss in randomized controlled dietary trials. Sci Rep 10, 115.CrossRefGoogle ScholarPubMed
Jie, Z, Yu, X, Liu, Y et al. (2021) The baseline gut microbiota directs dieting-induced weight loss trajectories. Gastroenterology 20, 20292042.CrossRefGoogle Scholar
Poulsen, SK, Due, A, Jordy, AB et al. (2014) Health effect of the new Nordic diet in adults with increased waist circumference: a 6-mo randomized controlled trial. Am J Clin Nutr 99, 3545.CrossRefGoogle ScholarPubMed
Hess, AL, Benítez-Páez, A, Blædel, T et al. (2020) The effect of inulin and resistant maltodextrin on weight loss during energy restriction: a randomised, placebo-controlled, double-blinded intervention. Eur J Nutr 59, 25072524.CrossRefGoogle ScholarPubMed
Suhr, J, Vuholm, S, Iversen, KN et al. (2017) Wholegrain rye, but not wholegrain wheat, lowers body weight and fat mass compared with refined wheat: a 6-week randomized study. Eur J Clin Nutr 71, 959967.CrossRefGoogle Scholar
Kjølbæk, L, Benítez-Páez, A, Gómez del Pulgar, EM et al. (2020) Arabinoxylan oligosaccharides and polyunsaturated fatty acid effects on gut microbiota and metabolic markers in overweight individuals with signs of metabolic syndrome: a randomized cross-over trial. Clin Nutr 39, 6779.CrossRefGoogle ScholarPubMed
Dent, R, McPherson, R & Harper, ME (2020) Factors affecting weight loss variability in obesity. Metabolism 113, 154388.CrossRefGoogle ScholarPubMed
Hjorth, MF, Ritz, C, Blaak, EE et al. (2017) Pretreatment fasting plasma glucose and insulin modify dietary weight loss success: results from 3 randomized clinical trials. Am J Clin Nutr 106, 499505.CrossRefGoogle ScholarPubMed
Hjorth, MF, Roager, HM, Larsen, TM et al. (2018) Pre-treatment microbial Prevotella-to-Bacteroides ratio, determines body fat loss success during a 6-month randomized controlled diet intervention. Int J Obes 42, 580583.CrossRefGoogle ScholarPubMed
Hjorth, MF, Christensen, L, Kjølbæk, L et al. (2020) Pretreatment Prevotella-to-Bacteroides ratio and markers of glucose metabolism as prognostic markers for dietary weight loss maintenance. Eur J Clin Nutr 74, 338347.CrossRefGoogle ScholarPubMed
Christensen, L, Vuholm, S, Roager, HM et al. (2019) Prevotella abundance predicts weight loss success in healthy, overweight adults consuming a whole-grain diet ad libitum: a post hoc analysis of a 6-wk randomized controlled trial. J Nutr 149, 18.CrossRefGoogle ScholarPubMed
Christensen, L, Sørensen C, V, Wøhlk, FU et al. (2020) Microbial enterotypes beyond genus level: bacteroides species as a predictive biomarker for weight change upon controlled intervention with arabinoxylan oligosaccharides in overweight subjects. Gut Microbes 12, 116.CrossRefGoogle ScholarPubMed
Patnode, ML, Beller, ZW, Han, ND et al. (2019) Interspecies competition impacts targeted manipulation of human gut bacteria by fiber-derived glycans. Cell 179, 5973, e13.CrossRefGoogle ScholarPubMed
Hjorth, MF, Christensen, L, Larsen, TM et al. (2020) Pretreatment Prevotella-to-Bacteroides ratio and salivary amylase gene copy number as prognostic markers for dietary weight loss. Am J Clin Nutr 111, 10791086.CrossRefGoogle ScholarPubMed
Morán-Ramos, S, Villarreal-Molina, MT & Canizales-Quinteros, S (2019) Host genetics, diet, and microbiome: the role of AMY1. Trends Microbiol 27, 473475.CrossRefGoogle ScholarPubMed
Atkinson, FS, Hancock, D, Petocz, P et al. (2018) The physiologic and phenotypic significance of variation in human amylase gene copy number. Am J Clin Nutr 108, 737748.CrossRefGoogle ScholarPubMed
Deehan, EC, Yang, C, Perez-Muñoz, ME et al. (2020) Precision microbiome modulation with discrete dietary fiber structures directs short-chain fatty acid production. Cell Host Microbe 27, 389404, e6.CrossRefGoogle ScholarPubMed
Poole, AC, Goodrich, JK, Youngblut, ND et al. (2019) Human salivary amylase gene copy number impacts oral and Gut microbiomes article human salivary amylase gene copy number impacts oral and gut microbiomes. Cell Host Microbe 25, 553564, e7.CrossRefGoogle ScholarPubMed
Roager, HM & Dragsted, LO (2019) Diet-derived microbial metabolites in health and disease. Nutr Bull 44, 216227.CrossRefGoogle Scholar
Falony, G, Joossens, M, Vieira-Silva, S et al. (2016) Population-level analysis of gut microbiome variation. Science 352, 560564.CrossRefGoogle ScholarPubMed
Falony, G, Vieira-Silva, S & Raes, J (2018) Richness and ecosystem development across faecal snapshots of the gut microbiota. Nat Microbiol 3, 526528.CrossRefGoogle ScholarPubMed
Lewis, S & Cochrane, S (2007) Alteration of sulfate and hydrogen metabolism in the human colon by changing intestinal transit rate. Am J Gastroenterol 102, 624633.CrossRefGoogle ScholarPubMed
Vieira-Silva, S, Falony, G, Darzi, Y et al. (2016) Species-function relationships shape ecological properties of the human gut microbiome. Nat Microbiol 1, 18.CrossRefGoogle ScholarPubMed
Müller, M, Hermes, GDA, Canfora, EE et al. (2020) Distal colonic transit is linked to gut microbiota diversity and microbial fermentation in humans with slow colonic transit. Am J Physiol: Gastrointest Liver Physiol 318, G361G369.Google ScholarPubMed
Lewis, SJ & Heaton, KW (1997) Increasing butyrate concentration in the distal colon by accelerating intestinal transit. Gut 41, 245251.CrossRefGoogle ScholarPubMed
Chen, T, Long, W, Zhang, C et al. (2017) Fiber-utilizing capacity varies in Prevotella- versus Bacteroides-dominated gut microbiota. Sci Rep 7, 17.Google ScholarPubMed
Wu, G, Zhao, N, Zhang, C et al. (2021) Guild-based analysis for understanding gut microbiome in human health and diseases. Genome Med 13, 112.CrossRefGoogle ScholarPubMed
Macfarlane, S, Quigley, M, Hopkins, M et al. (1998) Polysaccharide degradation by human intestinal bacteria during growth under multi-substrate limiting conditions in a three-stage continuous culture system. FEMS Microbiol Ecol 26, 231243.CrossRefGoogle Scholar
Walker, AW, Duncan, SH, Mcwilliam leitch, EC et al. (2005) pH and peptide supply can radically alter bacterial populations and short-chain fatty acid ratios within microbial communities from the human colon. Appl Environ Microbiol 71, 36923700.CrossRefGoogle ScholarPubMed
Schwiertz, A, Taras, D, Schafer, K et al. (2010) Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring) 18, 190195.CrossRefGoogle ScholarPubMed
Yamamura, R, Nakamura, K, Ukawa, S et al. (2021) Fecal short-chain fatty acids and obesity in a community-based Japanese population: the DOSANCO health study. Obes Res Clin Pract 15, 345350.CrossRefGoogle Scholar
Murphy, EF, Cotter, PD, Healy, S et al. (2010) Composition and energy harvesting capacity of the gut microbiota: relationship to diet, obesity and time in mouse models. Gut 59, 16351642.CrossRefGoogle ScholarPubMed
Koh, A, De Vadder, F, Kovatcheva-Datchary, P et al. (2016) From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell 165, 13321345.CrossRefGoogle ScholarPubMed
Husted, AS, Trauelsen, M, Rudenko, O et al. (2017) GPCR-mediated signaling of metabolites. Cell Metab 25, 777796.CrossRefGoogle ScholarPubMed
McNeil, NI (1984) The contribution of the large intestine to energy supplies in man. Am J Clin Nutr 39, 338342.CrossRefGoogle ScholarPubMed
Kimura, I, Ozawa, K, Inoue, D et al. (2013) The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat Commun 4, 112.CrossRefGoogle ScholarPubMed
De Vadder, F, Kovatcheva-Datchary, P, Goncalves, D et al. (2014) Microbiota-generated metabolites promote metabolic benefits via gut–brain neural circuits. Cell 156, 8496.CrossRefGoogle ScholarPubMed
Cummings, JH, Pomare, EW, Branch, WJ et al. (1987) Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 28, 12211227.CrossRefGoogle ScholarPubMed
den Besten, G, Lange, K, Havinga, R et al. (2013) Gut-derived short-chain fatty acids are vividly assimilated into host carbohydrates and lipids. Am J Physiol: Gastrointest Liver Physiol 305, 900911.Google ScholarPubMed
Samuel, BS, Shaito, A, Motoike, T et al. (2008) Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, GPR41. Proc Natl Acad Sci USA 105, 1676716772.CrossRefGoogle ScholarPubMed
Tolhurst, G, Heffron, H, Lam, YS et al. (2012) Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 61, 364371.CrossRefGoogle ScholarPubMed
Macia, L, Tan, J, Vieira, AT et al. (2015) Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nat Commun 6, 6734.CrossRefGoogle ScholarPubMed
Brown, AJ, Goldsworthy, SM, Barnes, AA et al. (2003) The orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J Biol Chem 278, 1131211319.CrossRefGoogle ScholarPubMed
Wichmann, A, Allahyar, A, Greiner, TU et al. (2013) Microbial modulation of energy availability in the colon regulates intestinal transit. Cell Host Microbe 14, 582590.CrossRefGoogle ScholarPubMed
Holst, JJ (2007) The physiology of glucagon-like peptide 1. Physiol Rev 87, 14091439.CrossRefGoogle ScholarPubMed
Gao, Z, Yin, J, Zhang, J et al. (2009) Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes 58, 15091517.CrossRefGoogle ScholarPubMed
Lin H, V, Frassetto, A, Kowalik, EJ et al. (2012) Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS One 7, 19.Google ScholarPubMed
Canfora, EE, van der Beek, CM, Jocken, JWE et al. (2017) Colonic infusions of short-chain fatty acid mixtures promote energy metabolism in overweight/obese men: a randomized crossover trial. Sci Rep 7, 2360.CrossRefGoogle ScholarPubMed
van der Beek, CM, Canfora, EE, Lenaerts, K et al. (2016) Distal, not proximal, colonic acetate infusions promote fat oxidation and improve metabolic markers in overweight/obese men. Clin Sci 130, 20732082.CrossRefGoogle Scholar
Chambers, ES, Viardot, A, Psichas, A et al. (2015) Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut 64, 17441754.CrossRefGoogle Scholar
Von Engelhardt, W, Rönnau, K, Rechkemmer, G et al. (1989) Absorption of short-chain fatty acids and their role in the hindgut of monogastric animals. Anim Feed Sci Technol 23, 4353.CrossRefGoogle Scholar
Ridlon, JM, Kang, D-JJ & Hylemon, PB (2006) Bile salt biotransformations by human intestinal bacteria. J Lipid Res 47, 241259.CrossRefGoogle ScholarPubMed
Wahlström, A, Sayin, SI, Marschall, HU et al. (2016) Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab 24, 4150.CrossRefGoogle ScholarPubMed
Roager, HM & Licht, TR (2018) Microbial tryptophan catabolites in health and disease. Nat Commun 9, 3294.CrossRefGoogle ScholarPubMed
Chimerel, C, Emery, E, Summers, DK et al. (2014) Bacterial metabolite indole modulates incretin secretion from intestinal enteroendocrine L cells. Cell Rep 9, 12021208.CrossRefGoogle ScholarPubMed
Ye, L, Bae, M, Cassilly, CD et al. (2021) Enteroendocrine cells sense bacterial tryptophan catabolites to activate enteric and vagal neuronal pathways. Cell Host Microbe 29, 179196, e9.CrossRefGoogle ScholarPubMed
Bhattarai, Y, Williams, BB, Battaglioli, EJ et al. (2018) Gut Microbiota-produced tryptamine activates an epithelial G-protein-coupled receptor to increase colonic secretion. Cell Host Microbe 23, 775785, e5.CrossRefGoogle ScholarPubMed
Breton, J, Tennoune, N, Lucas, N et al. (2016) Gut commensal E. coli proteins activate host satiety pathways following nutrient-induced bacterial growth. Cell Metab 23, 324334.CrossRefGoogle ScholarPubMed
Tennoune, N, Chan, P, Breton, J et al. (2014) Bacterial ClpB heat-shock protein, an antigen-mimetic of the anorexigenic peptide α-MSH, at the origin of eating disorders. Transl Psychiatry 4, e458e458.CrossRefGoogle ScholarPubMed
Arnoriaga-Rodríguez, M, Mayneris-Perxachs, J, Burokas, A et al. (2020) Gut bacterial ClpB-like gene function is associated with decreased body weight and a characteristic microbiota profile. Microbiome 8, 110.CrossRefGoogle Scholar
Breton, J, Legrand, R, Akkermann, K et al. (2016) Elevated plasma concentrations of bacterial ClpB protein in patients with eating disorders. Int J Eat Disord 49, 805808.CrossRefGoogle ScholarPubMed
Cohen, LJ, Esterhazy, D, Kim, S-H et al. (2017) Commensal bacteria make GPCR ligands that mimic human signalling molecules. Nature 549, 120.CrossRefGoogle ScholarPubMed
Yoon, HS, Cho, CH, Yun, MS et al. (2021) Akkermansia muciniphila secretes a glucagon-like peptide-1-inducing protein that improves glucose homeostasis and ameliorates metabolic disease in mice. Nat Microbiol 6, 563573.CrossRefGoogle ScholarPubMed
Chen, RY, Mostafa, I, Hibberd, MC et al. (2021) A microbiota-directed food intervention for undernourished children. N Engl J Med 384, 15171528.CrossRefGoogle ScholarPubMed
Vandeputte, D, De Commer, L, Tito, RY et al. (2021) Temporal variability in quantitative human gut microbiome profiles and implications for clinical research. Nat Commun 12, 113.CrossRefGoogle ScholarPubMed
Diaz Tartera, HO, Webb, DL, Al-Saffar, AK et al. (2017) Validation of SmartPill® wireless motility capsule for gastrointestinal transit time: intra-subject variability, software accuracy and comparison with video capsule endoscopy. Neurogastroenterol Motil 29, 19.CrossRefGoogle ScholarPubMed
von Schwartzenberg, RJ, Bisanz, JE, Lyalina, S et al. (2021) Caloric restriction disrupts the microbiota and colonization resistance. Nature 595, 272277.CrossRefGoogle ScholarPubMed
Ben-Yacov, O, Godneva, A, Rein, M et al. (2021) Personalized postprandial glucose response-targeting diet versus Mediterranean diet for glycemic control in prediabetes. Diabetes Care 44, 19801991.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Personal diet–microbiota interactions and human energy homeostasis. Person-specific colonic fermentation is a trade-off between saccharolytic and proteolytic fermentation, which depends on the complex interplay between the dietary substrates available, the metabolic potential of the gut microbiota and environmental (abiotic) factors, such as pH and transit time; factors which are highly individual. In addition, also differences in host genetics could affect this interplay. For example, differences in the copy number of the salivary α-amylase 1 (AMY1) gene could affect degradation of starch via amylase in the upper-gastrointestinal tract and thereby affect the availability of starch for colonic fermentation. Consequently, personal diet–microbiota interactions may affect human energy metabolism through energy excretion and the generation of microbiota-derived metabolites, such as SCFAs, tryptophan catabolites, secondary bile acids and metabolites mimetic of host hormones. These microbial metabolites could exert different effects on host metabolism – e.g. by serving as energy substrates, by stimulating secretion of appetite-regulating hormones, including glucagon-like peptide 1 (GLP-1) and peptide YY (PYY) in enteroendocrine cells, by regulating energy expenditure in adipose tissue, and by regulating appetite and satiety in the brain. Stratification by gut microbiota community characteristics defined by enterotypes, guilds, keystone species or specific genes, or abiotic factors could potentially be predictive of person-specific diet–microbiota interactions and linked to weight loss responses.