Next Article in Journal
A Scoping Review of Food Literacy Interventions
Previous Article in Journal
Effect of the Mediterranean Diet (MeDi) on the Progression of Retinal Disease: A Narrative Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Yoyo Dieting, Post-Obesity Weight Loss, and Their Relationship with Gut Health

by
Kate Phuong-Nguyen
1,*,
Sean L. McGee
1,
Kathryn Aston-Mourney
1,
Bryony A. Mcneill
1,
Malik Q. Mahmood
2 and
Leni R. Rivera
1,*
1
Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
2
School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
*
Authors to whom correspondence should be addressed.
Nutrients 2024, 16(18), 3170; https://doi.org/10.3390/nu16183170
Submission received: 21 August 2024 / Revised: 13 September 2024 / Accepted: 18 September 2024 / Published: 19 September 2024
(This article belongs to the Section Nutrition and Metabolism)

Abstract

:
Excessive body weight is associated with many chronic metabolic diseases and weight loss, so far, remains the gold standard treatment. However, despite tremendous efforts exploring optimal treatments for obesity, many individuals find losing weight and maintaining a healthy body weight difficult. Weight loss is often not sustainable resulting in weight regain and subsequent efforts to lose weight. This cyclic pattern of weight loss and regain is termed “yoyo dieting” and predisposes individuals to obesity and metabolic comorbidities. How yoyo dieting might worsen obesity complications during the weight recurrence phase remains unclear. In particular, there is limited data on the role of the gut microbiome in yoyo dieting. Gut health distress, especially gut inflammation and microbiome perturbation, is strongly associated with metabolic dysfunction and disturbance of energy homeostasis in obesity. In this review, we summarise current evidence of the crosstalk between the gastrointestinal system and energy balance, and the effects of yoyo dieting on gut inflammation and gut microbiota reshaping. Finally, we focus on the potential effects of post-dieting weight loss in improving gut health and identify current knowledge gaps within the field, including gut-derived peptide hormones and their potential suitability as targets to combat weight regain, and how yoyo dieting and associated changes in the microbiome affect the gut barrier and the enteric nervous system, which largely remain to be determined.

1. Introduction

For decades, we have witnessed the increasingly deleterious effects of obesity, which constitutes a major health and economic crisis in the modern world [1]. Obesity and overweight have become a growing problem affecting 52% of the world’s adult population [2,3,4,5], a figure that is expected to continue to rise [1,6]. Obesity presents as an emerging global epidemic leading to more than 2.8 million deaths annually [7] from diseases such as cardiovascular diseases [8,9], type 2 diabetes [10], fatty liver disease [11,12] and various systemic metabolic syndromes [13,14,15,16,17]. Despite much research and clinical effort to identify optimal solutions to control body weight, sustained weight loss is virtually impossible for most individuals with obesity, as most or all of the lost weight often returns within 5 years [18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35]. While weight loss remains a key therapeutic approach for many metabolic diseases [36,37], many individuals experience multiple cycles of recurrent obesity increasing their risk of developing significant obesity-related comorbidities [28,38].

1.1. Yoyo Dieting: Cyclic Weight Regain after Weight Loss

The weight loss journey of every individual is unique and not everyone is successful in losing weight. However, a common pattern for most individuals attempting to lose weight is an initial period of rapid weight loss, which gradually slows, before subsequent weight regain. This occurs despite every effort to adhere to weight management strategies [39]. This phenomenon is called weight cycling or “yoyo dieting” [40]. For many individuals who are medically advised to lose weight, yoyo dieting makes the weight loss journey even more challenging as the initial weight loss often returns and the weight loss process must be restarted. After multiple failed attempts to achieve sustained weight loss, many people refuse to continue their weight loss journey believing that weight loss is an impossible mission [41].
The likelihood of weight regain is greatest in the period following initial weight loss [42,43]. Yoyo dieting does not only affect people with excessive weight, as studies suggest that frequent on–off dieting also triggers weight regain in people who are not overweight or obese [44,45,46]. Dieting has become a popular norm for many people, including individuals with a healthy body weight (body mass index [BMI] between 18.5–24.9 kg/m2), and those underweight (BMI < 18.5 kg/m2) [47,48], who feel the urge to lose weight to achieve their desired body image [49]. However, as is usual with yoyo dieting, these individuals end up gaining all or even more weight after each cycle of weight loss [50,51,52]. Therefore, while the prevalence of obesity has been rising, yoyo dieting requires consideration because it places individuals at greater risk of the progression to obesity [53], and the development of associated chronic complications [54,55,56,57,58].
Taken together, although the link between yoyo dieting and eventual weight gain is well established, a complete picture of the mechanisms driving this response and the strategies to prevent it are still being explored. Weight gain following yoyo dieting is complex and involves changes in peripheral hormones regulating energy balance and metabolic adaptation to weight loss. The gut also plays an important role in body weight regulation and could be a key factor in weight regain after yoyo dieting.

1.2. The Relationship between Obesity, the Gut, and the Susceptibility to Weight Regain Is Potentially Due to Changes in the Gut Microbiota

There is established evidence from both human [59,60] and animal studies [61,62] implicating the gut in obesity development, largely due to an increase in the potentially harmful microbiota (termed ‘gut dysbiosis’ [63]). Gut dysbiosis (e.g., enriched Fusobacterium and reduced Oscillospira, Ruminococcaceae, Coriobacteriaceae, Odoribacter splanchnicus, Akkermansia muciniphila and Bifidobacterium longum) is strongly associated with negative metabolic outcomes in individuals with overweight and obesity [60,64] compared to controls (e.g., enriched Akkermnansia muciniphila, Alistipes indistinctus, Odoribacter splanchnicus, Clostridium sp. CAG:413, Intestinimonas butyriciproducens, and Bifidobacterium longum) [64]. Studies using germ-free mice provide evidence for a direct role of the microbiota in the metabolic alterations typically seen in individuals with obesity. For example, germ-free mice colonised with microbiota from obese mice (enriched Bacteroides uniformis, Parabacteroides merdae, Alistipes putredinis, Eubacterium rectale and reduced Bacteroides thetaiotaomicron) extracted more calories from their food and had reduced energy expenditure, leading to increased adiposity when compared with germ-free mice colonised with microbiota from lean mice [59,65]. These data indicate that the microbiota influences the efficiency of both nutrient absorption and calorie expenditure [17], strongly implicating a causative role of the gut microbiota in obesity pathogenesis [66].
The relationship between yoyo dieting, post-obesity weight loss, and gut health has been largely overlooked. To date, only a small number of studies have explored the effect of weight regain after weight loss on the gut microbiome [61,62]. These studies have indicated that yoyo dieting results not only in enhanced weight gain after dieting but also in potential long-term alterations in gut microbiota composition [61]. Given that most people who lose weight end up regaining all or even more weight than was previously lost, it is evident that this is an area that warrants further investigation. This review will focus on the influence of the gut on body weight changes during yoyo dieting and the relationship between fluctuating body weight, gastrointestinal inflammation, and gut microbiome alterations. Moreover, this review also explores the potential benefits of weight loss in influencing positive changes in the gut and identifies current knowledge gaps that need to be investigated.

2. The Influence of the Gut in Weight Regain after Weight Loss

While there is not yet a complete picture of all the mechanisms behind weight recurrence after loss, evidence suggests it is due to alterations in peripheral peptides that regulate energy balance and metabolic adaptation to weight loss. These changes might contribute to reduced energy expenditure and an increased drive to eat to counteract the energy gap created during dieting [42], hence placing individuals at a higher risk of obesity.

2.1. Gut Peptide Hormones That Regulate Energy Balance

Regulation of energy balance involves adjustment of energy intake, energy expenditure and energy storage [67] and is the main driver of body weight change. Energy balance is also the key concept describing the pathogenesis and prevalence of obesity and its comorbidities and is an important consideration in the development of possible treatments for weight control [48]. Energy balance is regulated not only by the central nervous system but is also strongly influenced by various peripheral signals arising from the gastrointestinal tract, pancreas, and adipose tissue, collectively acting to either stimulate or restrict energy intake [68,69]. These peripheral signals, many of which are peptide hormones, regulate energy balance by directly or indirectly controlling the activity of orexin and anorexic neurons within integrating brain regions such as the hypothalamus [70].
Peptide hormones secreted by the gut act via several pathways to modify energy status. These gut peptide hormones can act locally on gut peptide receptors expressed on vagal or spinal afferent nerve terminals innervating the gut to activate gut-brain neuronal signalling [71]. Moreover, they also act indirectly via receptors on intrinsic neurons of the enteric nervous system to relay neuronal signalling to afferent nerves [72]. The gut-derived hormones peptide tyrosine-tyrosine (PYY) and glucagon-like peptide 1 (GLP-1) delay gastric emptying [73], cholecystokinin (CCK) promotes bile production, pancreatic and gastric secretions [74], and oxyntomodulin (OXM) increases acid secretion in response to food intake [75]. These peptide hormones are also known to regulate feeding behaviour [76], specifically promoting appetite suppression and meal termination [77]. In contrast, the hunger gut-derived peptide hormone, ghrelin, increases gastric emptying, promotes appetite stimulation and lipogenesis, reduces lipid oxidation, and accelerates the use of carbohydrates as a source of energy while sparing fat [78]. Collectively, gut-derived peptide hormones are essential in the direct regulation of hunger/satiety states and energy consumption in humans. Increased appetite that drives energy intake to surpass energy expenditure will increase fat accumulation and result in body weight gain [79]. This is thought to be partly but critically associated with a gut peptide hormone profile that increases energy intake and reduces energy expenditure [80] although it is not yet clear if they are the trigger or consequence of obesity [81].
In addition to these signals from the gut, peptide hormones secreted by adipose tissue act via neuroendocrine mechanisms to convey information on energy status from the periphery to the brain. One such peptide hormone, leptin, is responsible for relaying information regarding hunger and satiety to the hypothalamus [82]. Circulating leptin levels increase in proportion to adipose tissue mass, which can result in a state of leptin resistance in obesity [83,84,85,86]. Adiponectin is a hormone synthesised and secreted exclusively by adipose tissue that suppresses appetite [87] and affects thermogenesis, with its receptors expressed in multiple peripheral tissues (such as liver and muscle) and centrally, in the hypothalamus [88]. In contrast to leptin, adiponectin circulating concentrations are reduced in obesity [89]. Furthermore, the pancreas also plays a crucial role in secreting hormones that affect energy homeostasis and appetite control, namely insulin, amylin, GLP-1, pancreatic polypeptide (PP), and glucose-dependent insulinotropic polypeptide (GIP). These feeding-relevant neuroendocrine signals bind to receptors in the hypothalamus and hindbrain to simultaneously regulate energy homeostasis, normalise the internal milieu during nutrient influx [90], regulate the metabolism of macronutrients [91], suppress appetite [92], and improve energy storage due to adipogenesis [93].
The regulation of appetite, body weight, and fat mass involves a complex coordination of various peripheral peptides and an imbalance of these peptide hormones might contribute to weight regain after loss. Results from several studies indicate that peripheral responses to calorie deficit promote increased hunger and a lower metabolic rate thereby enhancing susceptibility to weight relapse [77,94]. This involves decreases in gut-derived-satiety peptide hormones GLP-1 [77,95], PYY [77,96], and CCK [77,94,97], adipose-derived leptin, and the pancreas peptide hormones insulin and amylin [77,94]; in contrast with increased levels of the hunger peptide hormones ghrelin and PP [77,94]. There is evidence that these changes persist as individuals attempt to maintain reduced body weight, even after the cessation of active weight loss [94]. The continued low energy intake and minimal fat mass indicate energy unavailability, leading to a homeostatic endocrine response aimed at conserving energy and promoting energy intake [42,61,62,98,99,100]. These represent a critical area of ongoing research for obesity pathogenesis and a major challenge to developing strategies for the treatment of this relapsing disorder [68].
While specific mechanisms underlying the reduction in gut-derived satiety hormones following calorie deficit-derived weight loss are not fully understood, increasing evidence suggests it is possibly associated with various alterations related to the gut. Of note, fat is an essential macronutrient stimulant in the small intestine for PYY and CCK release [101]. However, most calorie-deficit diets reduce fat intake, therefore potentially reducing circulating PYY and CCK. Additionally, the gut epithelial cells that produce the incretin peptides, enteroendocrine cells (EECs) [102] located throughout the gastrointestinal tract, undergo constant cellular renewal, being replenished with new EECs formed every 5–7 days [103]. One of the important factors impacting intestinal epithelial cell differentiation and especially EEC renewal is the expression of Inhibitor of differentiation/DNA-binding protein 2 (Id2) transcription factor expressed mainly in the EECs themselves [104] and adipose tissue [105]. Dramatic loss of fat mass is often associated with a reduced expression of Id2 [106]; this potentially leads to decreased EEC renewal, hence reducing satiety hormones commonly seen shortly after weight loss. The reduction of Id2 in the EEC is associated with significantly reduced levels of the satiety peptide hormone PYY and the elevated hunger peptide hormone Ghrelin [104]. A study by Wölnerhanssen et al. [107] indicated that the total number of EECs expressing satiety hormones was significantly reduced during obesity but returned to control levels 3 months post-obesity. This suggests that the number of EECs expressing satiety hormones could be reduced in obesity but eventually restored in the long term if weight loss is substantial. Moreover, it is also worth noting that these findings were taken from a small sample size (n = 14) of individuals with morbid obesity [107]. These indicate that further longitudinal research with a larger study population is warranted to further verify these data. Another factor that is associated with reduced satiety hormone production after a calorie-restrictive diet is the reduced relative abundance of short-chain fatty acids (SCFAs)-producing microbiome. SCFAs are metabolites produced by specific colonic microbes [108] and are potent stimuli for the secretion of PYY [109], GLP-1 [110], and CCK [111]. After weight loss, the relative abundance of several SCFA-producing microbiota was found to be reduced, such as Roseburia intestinalis and Eubacterium rectale [112,113], Faecalibacterium prausnitzii [113], and Agathobacter rectalis [114]. This might help to explain how the gut microbiome influences the imbalance in satiety hormone production that contributes to the yoyo effect, but more studies are required to fully elucidate the mechanisms involved.
The potential benefits of targeting peripheral hormones as an approach for weight management are best highlighted by bariatric surgery. Bariatric surgery alters gastrointestinal anatomy impacting food intake and/or nutrient absorption [115], and is so far the most effective treatment for substantial weight loss. One common finding after bariatric surgery is a sharp rise of many satiety peptide hormones after meals, such as CCK and GLP-1, leading to increased satiety and decreased food reward [116,117]. Evidence suggests that this is due to the accelerated delivery of nutrients down the gastrointestinal tract, where the majority of EECs are situated [116]. Persistent exposure to rapid nutrient entry can lead to enhanced nutrient sensing within the gut and an increased number of satiety hormone-containing ECCs [118] leading to an increased secretion of CCK [116], GLP-1 [118]. Collectively, gut hormones have been demonstrated as key players regulating weight loss in anti-obesity surgeries [119,120,121]. Post-obesity weight loss increases both hunger and food reward, in part due to an imbalance in satiety-signalling hormones. This hormonal imbalance likely contributes to the yoyo effect, as a result of the body’s attempt to re-establish a body weight set point after a period of negative energy balance [122].
New weight loss therapeutics, including GLP-1 receptor agonists (slowing down gut transit [123]—Liraglutide [124] and Semaglutide [125]), dual GIP and GLP-1 receptor agonist (Tirzepatide [126]), and pancreatic and gastric lipase inhibitors (inhibiting dietary triglycerides digestion and absorption in the intestine [127,128]—Orlistat [129]) either signal through peripheral hormone mechanisms or have effects on peripheral hormones that regulate energy balance. To date, these medications are approved for long-term usage [130,131]. However, they are often only used on a short-term basis [131] mainly due to high costs [132], adverse side effects [131,132,133] that most commonly involve gastrointestinal symptoms [132,134], or insufficient response [131,132,133]. This highlights that gut hormones remain a critical focus in weight management [119,120,121].

2.2. Sympathetic Nervous System Regulates Energy Expenditure and Thermogenesis as a Metabolic Adaptation to Weight Loss

A decrease in energy intake causes a decrease in energy expenditure and vice versa. This is mainly influenced by non-adaptive modifications in the metabolically active component of the body known as fat-free mass. However, weight change does not always precisely follow predictions based on the calculation of energy imbalance. This is explained by alterations in the sympathetic nervous system (SNS) activity resulting in adaptive metabolism that is associated with weight gain in obesity [135]. Further, reduced activation of β-adrenergic signalling (stimulating lipolysis and thermogenesis [136]) leads to the promotion of weight gain after loss [137]. In particular, the SNS plays a key role in regulating metabolic homeostasis and is central in monitoring daily energy expenditure via the control of resting metabolic rate and induction of thermogenesis in response to physiologically related stimuli (hyperinsulinemia, energy states, food intake, dietary nutrients and cold exposure). Activation of the SNS in the liver, pancreas, and adipose tissue can also provoke acute catabolic responses (lipolysis and glycogenolysis) [138] that play an essential role in determining adaptive thermogenesis. Thermogenesis (metabolic heat production, mainly in brown adipose tissue [139]) plays a key role in energy expenditure [140,141]. Adaptive thermogenesis restricts modifications in energy stores in response to changing energy balance. It creates an ideal physiological environment to store energy and encourage weight regain by reducing energy expenditure beyond what can be predicted by the loss of fat mass and fat-free mass, hence preventing changes in body composition [142,143]. Changes in adaptive thermogenesis, which are known to be associated with metabolic efficiency [144] and weight change [145], could play an important role in metabolic adaptation to weight loss leading to the yoyo effect.
Taken together, weight loss is associated with changes in the regulation of the SNS, adaptive thermogenesis, and adaptive alterations in satiety gut hormones. These may partially explain insufficient weight loss outcomes and the high risk of weight regain in individuals with obesity and yoyo dieting [142,143].

3. Yoyo Dieting and Gut Inflammation

3.1. Links between Yoyo Dieting and Gut Inflammation

It is well established that chronic low-grade systemic inflammation is a hallmark of obesity [146,147,148,149,150]. For example, circulating adipose-derived cytokines, such as tumour necrosis factor-α (TNF-α) or interleukin-6 (IL-6) are elevated in obesity [151,152,153,154,155]. Yoyo dieting also promotes inflammatory responses [156,157] upon weight regain due to increased M1 proinflammatory macrophage activation and monocyte chemotactic protein 1 (MCP-1) proinflammatory cytokine production [156]. This suggests a potential contribution to an obesogenic memory phenotype [156]. Elevated levels of a number of cytokines have been reported after the 2nd weight gain of yoyo dieting, including IL-6 and TNF-α in the cell culture media primed with bone marrow-derived macrophage of yoyo dieting mice [158], interferon-gamma (IFN-γ) in epididymal adipose tissue, and signal transducer and activator of transcription 3 (STAT3) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) in liver [156]. With respect to inflammation specifically within the gut, upregulated intestinal immune control leading to increased gut inflammation and permeability are known characteristics of obesity [150,159,160,161,162,163,164,165,166,167,168,169,170]. Although there are limited studies specifically looking at the direct effect of yoyo dieting on gut inflammation, findings from several animal studies provide insights into potential links between yoyo dieting and gut inflammation. The effects of yoyo dieting have been tested experimentally by frequently switching from high-fat to low-fat diets, and examining the associated effects on gastrointestinal structure, function, and inflammation. For example, a study by Li et al. [171] demonstrated that mice fed a yoyo diet showed significantly lower C1q/tumour necrosis factor-related protein-3 (CTRP3) expression in epididymal adipose tissue in comparison to mice fed an HFD only. Reduced expression of CTRP3 is associated with a greater risk of intestinal inflammation [172,173]. Recently, another study observed a significant increase in CD11c (M1 proinflammatory macrophage marker) and MCP-1 in the epidydimal adipose tissue of mice fed a yoyo diet (HFD, Control, and then HFD) compared to those fed only one cycle of HFD (Control, and then HFD) [156]. MCP-1, one of the key chemokines of inflammation, is predominantly present in the gastric mucosal epithelium [174,175] and is known to be significantly enhanced in inflamed intestinal biopsies of IBD individuals and Caco-2 colon carcinoma cells [174,176,177]. Of note, increased M1 macrophages and MCP-1 expressions are strongly associated with elevated chronic intestinal inflammation in IBD [174,178,179,180,181]. Collectively, these data suggest that weight regain after loss in yoyo dieting contributes to increased intestinal inflammation in animals. However, there is a lack of human studies on yoyo dieting and gut inflammation, highlighting the need for further human studies in order to better translate these findings.

3.2. Post-Obesity Weight Loss as a Potential Approach to Reduce Gut Inflammation in Humans

There are currently limited studies looking at the direct effects of yoyo dieting on gut inflammation. However, the current literature suggests that diet-induced weight loss is a potential approach to reduce gut inflammation in humans. Weight loss after obesity results in significant anti-inflammatory effects in the gut. One of the early studies was conducted by Pendyala et al. [182] who assessed the levels of proinflammatory cytokines and gene expression profiles in rectosigmoid mucosal biopsies from a small human cohort of female individuals with obesity who consumed a low-calorie diet to achieve ≥8% loss of baseline weight [182]. Weight loss was correlated with significant declines in rectosigmoid mucosal TNF-α, IL-1β, IL-8 and MCP-1, suggesting reduced colonic inflammation. Moreover, transcriptomics of the colon reported that the calorie-restricted diet and weight loss significantly downregulated expression of the proinflammatory IL-8, implying a protective effect on the mucosal barrier [177,183,184]. Additionally, genes encoding for circulating peripheral peptides associated with weight loss were downregulated, including PYY and vasoactive intestinal peptide (VIP). The reduction in VIP levels is likely to be beneficial because elevated VIP is considered to be a biomarker for an aggravated state of colonic inflammatory conditions, as seen in inflammatory bowel disease (IBD) [185,186]. However, reduced PYY is associated with faster gastric emptying [187] and reduced satiety, which promotes food intake [188] and reduces energy expenditure [189] together driving positive energy balance. This is characteristic of energy balance hormone dysregulation that attempts to restore body weight upon weight loss [42]. Additionally, a human study by Ott et al. [114] investigated the effects of a 28-day caloric restriction for weight loss in female individuals with obesity and demonstrated decreased high-sensitivity CRP (inflammation biomarker) and paracellular gut permeability markers (polyethylene glycol 1500 and zonulin) in urine, and reduced chemerin (inflammation marker) in plasma. These results signify that weight loss after obesity reduces intestinal inflammation and improves barrier function [190,191]. Collectively, the current literature suggests that weight loss after obesity improves gut health. However, these findings were taken from a small sample size (n = 20) of women participants, indicating that further research is warranted to explore potential sex differences in responses to yoyo dieting.
Taken together, current findings suggest an association between post-obesity weight loss and anti-inflammatory effects on the gut. Animal studies suggest the health benefit of reduced inflammation in the gut during the weight loss phase (of yoyo dieting), but this might not persist once the weight rebounds [156,171]. Moreover, while the majority of human studies also indicate the potential benefits of post-obesity weight loss in improving gut inflammation, no clinical investigation has been conducted to examine changes in gut inflammation once the lost weight is regained. Therefore, more studies, especially in humans, are needed to have a better understanding of the potentially proinflammatory effects of yoyo dieting on gut health.

4. Yoyo Dieting and Gut Microbiome

The gastrointestinal tract is host to an extraordinarily abundant and diverse community of microorganisms, including bacteria, archaea, viruses, fungi, and protozoa, collectively known as the gut microbiota. It is widely recognised that the gut microbiota exerts a vital role in influencing the host health [192,193] and there is a strong association between gut microbiota dysbiosis and obesity, which has been reported in animal and human studies [59,61,62,65,194]. There is also emerging evidence suggesting an important role for the gut microbiome in influencing weight recurrence after dieting. While there have been conflicting findings, most studies indicate that yoyo dieting reduces alpha-diversity, and alters microbiota composition, such as the Firmicutes:Bacteroidetes ratio. Alterations in gut microbiota diversity, abundance, and composition are closely linked to changes in metabolism, as well as the development of obese/lean phenotypes. Several studies have investigated yoyo dieting and potential interventions, aiming not only for weight loss but also for the restoration of normal gut microbiota. These investigations have sought to understand how to recalibrate the microbiota of individuals struggling with obesity as a method to alleviate associated metabolic complications.

4.1. Different Gut Microbiota Profiles in Yoyo Dieting

Several animal studies have indicated an association between yoyo dieting and lower alpha diversity (distribution of microbiota richness and evenness [195]) and alterations in gut microbiota composition and relative abundance [61,62,196]. One of the earliest animal studies investigating the influences of yoyo dieting on the microbiome was conducted by Thaiss et al. [61]. In their study, mice in the yoyo group were exposed to two HFD and LFD cycles with each diet phase continued until these mice were a similar weight to the obese (HFD) and lean (LFD) control mice. In either the 1st or 2nd yoyo cycle the microbiota composition of the mice during obesity (HFD feeding period) significantly differed from that of the LFD control mice. However, even after achieving successful weight loss with similar body weights to lean control mice, the microbiota composition of yoyo mice remained in an intermediate state that was distinct from control mice. In fact, it took 21 weeks after successful weight loss (5 times longer than the last dieting period) for the microbiota composition of the yoyo dieting mice to return to the control state. Therefore, although short-term weight loss after obesity is associated with immediate metabolic health improvements, the gut microbiota requires longer-term maintenance of weight loss to return to a non-obese state [61]. This suggests that gut microbiota could increase the susceptibility to weight regain immediately after weight loss. The specific changes in the microbiome during obesity and after initial weight loss included consistently lower alpha diversity and relative abundance of Christensenella spp. and Lactobacillus reuteri. Christensenella spp. is associated with leanness in both animal and human studies [197,198,199,200,201] and is also associated with a reduced risk of IBD [202,203]. Hence, the reduction of Christensenella spp. could have direct negative effects on body weight and inflammation [201,204]. Additionally, Lactobacillus reuteri is a well-known commensal gut symbiont commonly used as a probiotic [205,206] due to its ability to inhibit pathogenic microbiota colonisation [207], ameliorate inflammation [208,209,210], and improve intestinal barrier function [211]. Therefore, the dramatic loss in the relative abundance of Lactobacillus reuteri could have deleterious effects on these functions.
Another study by Kawashima et al. [196] showed that mice fed a yoyo diet had significantly lower alpha diversity and a higher relative abundance of Firmicutes (genus Ruminococcus) and Proteobacteria (genus Desulfovibrio) compared to mice fed a control diet. Enrichment of Ruminococcus is likely unfavourable because this anaerobic Gram-positive bacteria genus [212] has been associated with an increased risk of Crohn’s disease [213,214] and metabolic syndrome [215]. Moreover, the increased relative abundance of Desulfovibrio is also not likely to be beneficial because this anaerobic Gram-negative rod/sulphate-reducing bacteria [216] has been associated with ulcerative colitis [217,218] and Parkinson’s disease [219].
In another recent mouse study by Humblot et al. [62], the long-term effects of yoyo diets on gut microbiota composition were investigated. Mice fed a yoyo diet ending with LFD had higher alpha diversity than those ending with HFD. There were no significant differences in alpha and beta diversity between obese mice fed HFD only and yoyo mice ending with HFD, and between control mice fed LFD only and yoyo mice ending on LFD. Overall, their results indicated that obese mice and yoyo mice ending with HFD had a higher proportion of Firmicutes (classes Erysipelotrichia and Bacilli) and upregulated functional microbiota genome dedicated to amino acid synthesis, while control mice and yoyo mice ending with LFD had a higher proportion of phylum Bacteroidetes and genus Lactobacillus, and a higher proportion of genes coding for carbohydrate metabolism and biosynthesis of B group vitamins. An increased relative abundance of Erysipelotrichia and Bacilli is associated with increased intestinal inflammation and is considered a biomarker of IBD [220], suggesting a putative role in the obesity phenotype. Conversely, a greater relative abundance of Lactobacillus is strongly associated with improved intestinal barrier function [221], reduced intestinal lipid absorption [222,223], and a representative probiotic for IBD [224] and liver diseases [225]. Their results suggest that the microbiota function and composition of mice fed yoyo diets do not have impacts beyond the diet period and that yoyo dieting would not have any long-term consequences.
A summary of yoyo dieting studies affecting the gut microbiota using animal models is listed in Table 1.
Taken together, current evidence from animal studies suggests that yoyo dieting results in potential impacts on the gut microbiome. However, conflicting results on the residual changes of the gut microbiome during and after yoyo dieting warrants further research, in particular determining whether long-term gut dysbiosis is a result of yoyo dieting and possibly a key factor for weight regain susceptibility.

4.2. Will the Gut Microbiome Be the Next Target to Prevent Weight Regain?

There are limited studies that have investigated yoyo dieting in humans. However, some clues can be taken from animal studies exploring the effect of dietary interventions targeting weight loss and clinical studies exploring the effects of post-obesity weight loss, which indicate significant changes in gut microbiota composition, leading to potentially positive signs of improving gut and metabolic health.

4.2.1. Dietary Interventions Enhance Weight Loss and Alter Gut Microbiota in Animal Studies

Several animal studies investigating dietary interventions for weight loss during yoyo dieting suggest notable alterations in gut microbiota composition, hinting at potential improvements in gut and metabolic health. There has been growing evidence in both in vivo [226] and in vitro studies [227,228,229] suggesting that flavonoids, a type of bioactive compound derived from beverages, vegetables and fruits [230], could improve weight management. For example, a study by Thaiss et al. [61] showed that flavonoid treatment resulted in less weight regain compared to vehicle-treated mice during yoyo dieting. Moreover, the administration of flavonoids was associated with a dramatically elevated expression of the major thermogenic factor uncoupling protein 1 (UCP1) in brown adipose tissue. Apart from the weight control effect, the increase in intestinal flavonoids is likely to be beneficial for gut health because these dietary polyphenols are mostly processed by the gut microbiota to exert effective roles with anti-oxidant and anti-inflammatory factors [231,232]. Furthermore, increased intestinal flavonoid is also associated with amelioration of gut microbiota dysbiosis [233,234,235,236], reduced risk of colorectal cancer [237,238,239,240,241,242] and inhibition of Helicobacter pylori infection-associated gastric cancer [243].
Another study by Kawashima et al. [196] explored the effect of yoyo dieting and the use of daisaikoto (a Japanese traditional herb-based medicine used as a treatment for menstrual pain [244] and fatty liver disease [245,246,247]) in preventing weight regain and reversing gut dysbiosis. In this study, mice were fed a cyclic HFD interspaced with LFD, with and without supplementation of 3% daisaikoto during the LFD feeding period only. Mice fed a yoyo diet supplemented with daisaikoto had significantly lower body weight, visceral fat, and weight regain upon a subsequent HFD feeding period in comparison with mice fed a yoyo diet only. Additionally, yoyo mice fed daisaikoto had a significantly higher relative abundance of Bacteroidetes and lower Firmicutes than those without daisaikoto. Although there are conflicting findings, a Firmicutes:Bacteroidetes ratio greater than 1 is often associated with the obese phenotype, while a ratio lower than 1 is associated with a lean phenotype [248,249,250,251]. This might imply that daisaikoto supplementation in yoyo dieting mice is beneficial for weight loss potentially due to positive changes in the microbiome. Yoyo mice fed daisaikoto had similar relative abundances of Ruminococcus and Desulfovibrio compared to control mice and lower than yoyo mice fed no daisaikoto. Reduced relative abundances of Ruminococcus and Desulfovibrio are likely to be beneficial because these are associated with a reduced risk of IBD development [217,252,253].
Additionally, a recent animal study by Zhong et al. [254] suggested that, following weight loss, a high protein diet during weight regain reduced lipid absorption in the intestine and reduced fat accumulation compared to a standard protein diet during weight regain. Mice fed a high protein diet had higher alpha diversity and a lower relative abundance of Lactobacillus murinus Lam-1 than those fed with a normal protein diet. Remarkably, their finding confirmed that enrichment of Lactobacillus murinus Lam-1 is associated with enhanced intestinal lipid absorption and fat accumulation. These data imply that weight loss potentially endorses the enrichment of intestinal Lactobacillus murinus Lam-1 leading to dramatic weight regain and that the weight recurrent issue might be tackled by a diet high in protein.

4.2.2. Post-Obesity Weight Loss May Be Beneficial in Improving Gut Microbiota in Humans

Clinical studies have shown that post-obesity weight loss is associated with significant changes in gut microbiota composition, leading to potentially positive signs of improving gut and metabolic health. For example, a human study by Ott et al. [114] investigated the effects of a one-month caloric restriction in female individuals with obesity. The low-calorie diet did not significantly alter the microbial diversity and richness, or result in substantial shifts in the microbiota composition after successful weight loss. However, there were dramatic changes in the relative abundances of several microbiota, particularly reduced Proteobacteria and increased Ruminococcus faeces and Bifidobacterium sp. The reduction in Proteobacteria is likely advantageous because this group of Gram-negative bacteria is believed to play a role in proinflammatory conditions in the gut and pathogenesis of IBD [255,256]. Additionally, the elevated relative abundances of Ruminococcus faeces and Bifidobacterium sp. are also likely beneficial because these Gram-positive species are shown to be associated with reduced severity and fibrogenesis of non-alcoholic fatty liver disease [257] and colorectal cancer [258,259,260,261], respectively.
Another clinical study, conducted by Dong et al. [262], examined a 16-week macronutrient standardised diet for weight loss for individuals with overweight or obesity. Microbiota results indicated no significant differences in microbiota richness and composition after weight loss. However, the group that successfully lost at least 5% of their body weight was associated with a reduction in the proportion of Enterococcus, while the group that did not have significant body weight loss had a significantly decreased abundance of Klebsiella and an increase of Coprococcus and Collinsella. Reduced Klebsiella is likely to be beneficial because it is associated with reduced risks of antimicrobial resistance [263,264,265] and pneumonia [266,267]. Increased Coprococcus is also likely to be favourable because its abundance is negatively correlated with depression [268,269] and Parkinson’s disease [270]. However, an enrichment of Collinsella is likely not favourable because it is associated with an increased risk of atherosclerosis [271], and type 2 diabetes [272].
Recently, a randomised controlled trial by Jian et al. [273] showed marked alterations in the gut microbiota composition of individuals with pre-diabetic overweight/obesity after weight loss. The 8-week low-energy diet resulted in significant increases in microbiota richness and alpha diversity, as well as Bray–Curtis values, and a significant reduction in the Firmicutes:Bacteroidetes ratio and capacity for butyrate production in the gut microbiota. Moreover, this weight loss was also associated with significantly reduced Pseudobutyrivibrio and Bifidobacterium and enriched Akkermansia. A reduced abundance of Pseudobutyrivibrio is associated with an increased risk of psoriatic arthritis [274]. However, an increased abundance of Bifidobacterium is likely to be beneficial because it is known as a probiotic agent that can reduce the risk of colorectal carcinoma [258,259,260,261] and IBD [275,276,277,278,279]. Moreover, the increased abundance of Akkermansia is likely to be beneficial because it is an important mucin-utiliser bacterium [280] associated with enhanced gut barrier function [281,282], improved insulin resistance [283], reduced metabolic endotoxemia [284] and cardiometabolic risk factor [285], and has been considered a biomarker for longevity [286].
A summary of human studies exploring the effect of post-obesity weight loss on gut microbiota is listed in Table 2.
Collectively, current evidence from animal models indicates that targeting weight loss via dietary interventions is associated with significant gut microbiota alterations and improves adipogenesis, fat deposition, and susceptibility to weight regain. Moreover, while there has not been any human study exploring the effects of yoyo dieting and gut microbiome, studies have shown that individuals with obesity after weight loss have significantly different microbiota signatures compared to those during the obesity state (Figure 1). Although human studies are important, the complexity of confounding factors in human data, especially from diet and lifestyle studies, often presents constant challenges. Animal models, on the other hand, offer a valuable research model that provides clear mechanistic insights into the intricate interactions between the gut microbiome and host physiology within a controlled experimental design. However, it is worth noting that there are differences between animal and human models [287], especially between mouse and human gut microbiota signatures [288]. This remains a key limitation in translating promising results from animal models to humans, particularly if these microbiome changes have host-specific physiological dependence.
Given the relationship between gut health, obesity, and its comorbidities, modulating the gut microbiota emerges to be a promising strategy for managing weight and improving human health. However, it remains to be determined whether changes in gut microbiota directly or indirectly influence or are influenced by weight loss and the mechanisms behind repeated bouts of weight regain after loss. This highlights the importance of elucidating this complex relationship which remains underexplored in the existing literature. Future longitudinal studies, especially in humans, are needed to understand the potentially distinct effects of (1) continuous on–off diets, (2) weight fluctuations (the two components of yoyo dieting) and (3) gut-derived peptide hormonal imbalance on the gut microbiome. This will provide valuable insights into how to better manage weight regain susceptibility via gut microbiome modulation which can potentially pave the way to ameliorating current adverse metabolic issues and complications related to obesity.

5. Conclusions

There is emerging evidence indicating a relationship between yoyo dieting and gut microbiota. Current evidence suggests that an altered gut microbiome profile occurs with weight gain and persists long-term, even after successful weight loss. This might contribute to the greater susceptibility for weight relapse and potentially enhanced rate of weight gain after every cycle of weight loss and regain. Weight loss may be beneficial in ameliorating dysbiosis and intestinal inflammation. However, a full picture regarding yoyo dieting and gut health, including changes in the microbiota metabolites, remains to be determined. Moreover, the crosstalk between yoyo dieting and the gut signifies vital impacts in local and systemic effects on overall human health. There is little known about the molecular mechanism of how the gut influences the yoyo effect, such as how gut-derived peptides are impacted and their potential suitability as targets to combat cyclic weight regain. Furthermore, how yoyo dieting and associated changes in the microbiome affect the gut barrier and the enteric nervous system (intrinsic nervous system in the gut) has also been largely overlooked. Therefore, further studies, especially on humans, are needed to assess changes in peripheral peptides, epithelial lining, intestinal permeability, enteric neurons, and microbiome metabolites to crucially elucidate the effects of yoyo dieting on gut health and further explore the gastrointestinal system as a promising target for long-term weight maintenance.

Author Contributions

Conceptualisation, K.P.-N. and L.R.R.; writing—original draft preparation, K.P.-N.; writing—review and editing, K.P.-N., L.R.R., S.L.M., K.A.-M., B.A.M. and M.Q.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analysed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Malik, V.S.; Willet, W.C.; Hu, F.B. Nearly a decade on—Trends, risk factors and policy implications in global obesity. Nat. Rev. Endocrinol. 2020, 16, 615–616. [Google Scholar] [CrossRef] [PubMed]
  2. World Health Organisation. Obesity and Overweight; World Health Organisation: Geneva, Switzerland, 2021. [Google Scholar]
  3. Barber, T.M. Is obesity a disease? Expert Rev. Endocrinol. Metab. 2018, 13, 59–61. [Google Scholar] [CrossRef] [PubMed]
  4. Sharma, A.M.; Campbell-Scherer, D.L. Redefining obesity: Beyond the numbers. Obesity 2017, 25, 660–661. [Google Scholar] [CrossRef] [PubMed]
  5. Ng, M.; Fleming, T.; Robinson, M.; Thomson, B.; Graetz, N.; Margono, C.; Mullany, E.C.; Biryukov, S.; Abbafati, C.; Abera, S.F.; et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980–2013: A systematic analysis for the Global Burden of Disease Study 2013. Lancet 2014, 384, 766–781. [Google Scholar] [CrossRef]
  6. Bixby, H.; Bentham, J.; Zhou, B.; Di Cesare, M.; Paciorek, C.J.; Bennett, J.E.; Taddei, C.; Stevens, G.A.; Rodriguez-Martinez, A.; Carrillo-Larco, R.M.; et al. Rising rural body-mass index is the main driver of the global obesity epidemic in adults. Nature 2019, 569, 260–264. [Google Scholar]
  7. World Health Organization. Obesity; World Health Organisation: Geneva, Switzerland, 2021. [Google Scholar]
  8. Guh, D.P.; Zhang, W.; Bansback, N.; Amarsi, Z.; Birmingham, C.L.; Anis, A.H. The incidence of co-morbidities related to obesity and overweight: A systematic review and meta-analysis. BMC Public Health 2009, 9, 88. [Google Scholar] [CrossRef]
  9. Petrie, J.R.; Guzik, T.J.; Touyz, R.M. Diabetes, Hypertension, and Cardiovascular Disease: Clinical Insights and Vascular Mechanisms. Can. J. Cardiol. 2017, 34, 575–584. [Google Scholar] [CrossRef]
  10. Al-Goblan, A.S.; Al-Alfi, M.A.; Khan, M.Z. Mechanism linking diabetes mellitus and obesity. Diabetes Metab. Syndr. Obes. 2014, 7, 587–591. [Google Scholar] [CrossRef]
  11. Fabbrini, E.; Sullivan, S.; Klein, S. Obesity and nonalcoholic fatty liver disease: Biochemical, metabolic, and clinical implications. Hepatology 2010, 51, 679–689. [Google Scholar] [CrossRef]
  12. Sarwar, R.; Pierce, N.; Koppe, S. Obesity and nonalcoholic fatty liver disease: Current perspectives. Diabetes Metab. Syndr. Obes. 2018, 11, 533–542. [Google Scholar] [CrossRef]
  13. World Health Organization. Obesity: Preventing and Managing The Global Epidemic; World Health Organisation: Geneva, Switzerland, 2000. [Google Scholar]
  14. O’Brien, P.D.; Hinder, L.M.; Callaghan, B.C.; Feldman, E.L. Neurological consequences of obesity. Lancet Neurol. 2017, 16, 465–477. [Google Scholar] [CrossRef] [PubMed]
  15. Blüher, M. Obesity: Global epidemiology and pathogenesis. Nat. Rev. Endocrinol. 2019, 15, 288–298. [Google Scholar] [CrossRef] [PubMed]
  16. Godoy-Matos, A.F.; Júnior, W.S.S.; Valerio, C.M. NAFLD as a continuum: From obesity to metabolic syndrome and diabetes. Diabetol. Metab. Syndr. 2020, 12, 60. [Google Scholar] [CrossRef] [PubMed]
  17. Lin, K.; Zhu, L.; Yang, L. Gut and obesity/metabolic disease: Focus on microbiota metabolites. Medcomm 2022, 3, e171. [Google Scholar] [CrossRef]
  18. Velapati, S.R.; Shah, M.; Kuchkuntla, A.R.; Abu-Dayyeh, B.; Grothe, K.; Hurt, R.T.; Mundi, M.S. Weight Regain After Bariatric Surgery: Prevalence, Etiology, and Treatment. Curr. Nutr. Rep. 2018, 7, 329–334. [Google Scholar] [CrossRef]
  19. Kraschnewski, J.L.; Boan, J.; Esposito, J.; E Sherwood, N.; Lehman, E.B.; Kephart, D.K.; Sciamanna, C.N. Long-term weight loss maintenance in the United States. Int. J. Obes. 2010, 34, 1644–1654. [Google Scholar] [CrossRef] [PubMed]
  20. Neumark-Sztainer, D.; Rock, C.L.; Thornquist, M.D.; Cheskin, L.J.; Neuhouser, M.L.; Barnett, M.J. Weight-Control Behaviors among Adults and Adolescents: Associations with Dietary Intake. Prev. Med. 2000, 30, 381–391. [Google Scholar] [CrossRef]
  21. Jeffery, R.W.; Adlis, S.A.; Forster, J.L. Prevalence of dieting among working men and women: The healthy worker project. Health Psychol. 1991, 10, 274. [Google Scholar] [CrossRef]
  22. Anderson, J.W.; Konz, E.C.; Frederich, R.C.; Wood, C.L. Long-term weight-loss maintenance: A meta-analysis of US studies. Am. J. Clin. Nutr. 2001, 74, 579–584. [Google Scholar] [CrossRef]
  23. Weiss, E.C.; Galuska, D.A.; Khan, L.K.; Gillespie, C.; Serdula, M.K. Weight regain in US adults who experienced substantial weight loss, 1999–2002. Am. J. Prev. Med. 2007, 33, 34–40. [Google Scholar] [CrossRef]
  24. Bacon, L.; Aphramor, L. Weight Science: Evaluating the Evidence for a Paradigm Shift. Nutr. J. 2011, 10, 9. [Google Scholar] [CrossRef] [PubMed]
  25. Mann, T.; Tomiyama, A.J.; Westling, E.; Lew, A.-M.; Samuels, B.; Chatman, J. Medicare’s search for effective obesity treatments: Diets are not the answer. Am. Psychol. 2007, 62, 220. [Google Scholar] [CrossRef] [PubMed]
  26. Anastasiou, C.A.; Karfopoulou, E.; Yannakoulia, M. Weight regaining: From statistics and behaviors to physiology and metabolism. Metabolism 2015, 64, 1395–1407. [Google Scholar] [CrossRef] [PubMed]
  27. Franz, M.J.; VanWormer, J.J.; Crain, A.L.; Boucher, J.L.; Histon, T.; Caplan, W.; Bowman, J.D.; Pronk, N.P. Weight-loss outcomes: A systematic review and meta-analysis of weight-loss clinical trials with a minimum 1-year follow-up. J. Am. Diet. Assoc. 2007, 107, 1755–1767. [Google Scholar] [CrossRef]
  28. Jimenez, L.S.; Chaim, F.H.M.; Chaim, F.D.M.; Utrini, M.P.; Gestic, M.A.; Chaim, E.A.; Cazzo, E. Impact of Weight Regain on the Evolution of Non-alcoholic Fatty Liver Disease After Roux-en-Y Gastric Bypass: A 3-Year Follow-up. Obes. Surg. 2018, 28, 3131–3135. [Google Scholar] [CrossRef]
  29. Crujeiras, A.B.; Zulet, M.A.; Lopez-Legarrea, P.; de la Iglesia, R.; Pardo, M.; Carreira, M.C.; Martínez, J.A.; Casanueva, F.F. Association between circulating irisin levels and the promotion of insulin resistance during the weight maintenance period after a dietary weight-lowering program in obese patients. Metabolism 2014, 63, 520–531. [Google Scholar] [CrossRef]
  30. Lien, L.F.; Haqq, A.M.; Arlotto, M.; Slentz, C.A.; Muehlbauer, M.J.; McMahon, R.L.; Rochon, J.; Gallup, D.; Bain, J.R.; Ilkayeva, O.; et al. The STEDMAN project: Biophysical, biochemical and metabolic effects of a behavioral weight loss intervention during weight loss, maintenance, and regain. OMICS J. Integr. Biol. 2009, 13, 21–35. [Google Scholar] [CrossRef]
  31. Wang, P.; Holst, C.; Wodzig, W.K.W.H.; Andersen, M.R.; Astrup, A.; van Baak, M.A.; Larsen, T.M.; Jebb, S.A.; Kafatos, A.; Pfeiffer, A.F.H.; et al. Circulating ACE is a predictor of weight loss maintenance not only in overweight and obese women, but also in men. Int. J. Obes. 2012, 36, 1545–1551. [Google Scholar] [CrossRef]
  32. Linna, M.S.; Borg, P.; Kukkonen-Harjula, K.; Fogelholm, M.; Nenonen, A.; Ahotupa, M.; Vasankari, T.J. Successful weight maintenance preserves lower levels of oxidized LDL achieved by weight reduction in obese men. Int. J. Obes. 2006, 31, 245–253. [Google Scholar] [CrossRef]
  33. Thomas, T.R.; Warner, S.O.; Dellsperger, K.C.; Hinton, P.S.; Whaley-Connell, A.T.; Rector, R.S.; Liu, Y.; Linden, M.A.; Chockalingam, A.; Thyfault, J.P.; et al. Exercise and the metabolic syndrome with weight regain. J. Appl. Physiol. 2010, 109, 3–10. [Google Scholar] [CrossRef]
  34. Delbridge, E.A.; Prendergast, L.A.; Pritchard, J.E.; Proietto, J. One-year weight maintenance after significant weight loss in healthy overweight and obese subjects: Does diet composition matter? Am. J. Clin. Nutr. 2009, 90, 1203–1214. [Google Scholar] [CrossRef] [PubMed]
  35. Matsuo, T.; Kato, Y.; Murotake, Y.; Kim, M.-K.; Unno, H.; Tanaka, K. An increase in high-density lipoprotein cholesterol after weight loss intervention is associated with long-term maintenance of reduced visceral abdominal fat. Int. J. Obes. 2010, 34, 1742–1751. [Google Scholar] [CrossRef]
  36. Brunner, K.T.; Henneberg, C.J.; Wilechansky, R.M.; Long, M.T. Nonalcoholic Fatty Liver Disease and Obesity Treatment. Curr. Obes. Rep. 2019, 8, 220–228. [Google Scholar] [CrossRef] [PubMed]
  37. Hallberg, S.J.; Gershuni, V.M.; Hazbun, T.L.; Athinarayanan, S.J. Reversing Type 2 Diabetes: A Narrative Review of the Evidence. Nutrients 2019, 11, 766. [Google Scholar] [CrossRef]
  38. Jeong, S.; Choi, S.; Chang, J.; Kim, K.; Kim, S.M.; Hwang, S.Y.; Son, J.S.; Lee, G.; Park, S.M. Association of weight fluctuation with cardiovascular disease risk among initially obese adults. Sci. Rep. 2021, 11, 10152. [Google Scholar] [CrossRef] [PubMed]
  39. Müller, M.J.; Enderle, J.; Bosy-Westphal, A. Changes in Energy Expenditure with Weight Gain and Weight Loss in Humans. Curr. Obes. Rep. 2016, 5, 413–423. [Google Scholar] [CrossRef] [PubMed]
  40. Brownell, K.D.; Rodin, J. Medical, Metabolic, and Psychological Effects of Weight Cycling. Arch. Intern. Med. 1994, 154, 1325–1330. [Google Scholar] [CrossRef]
  41. Hall, K.D.; Kahan, S. Maintenance of Lost Weight and Long-Term Management of Obesity. Med. Clin. N. Am. 2017, 102, 183–197. [Google Scholar] [CrossRef]
  42. MacLean, P.S.; Higgins, J.A.; Johnson, G.C.; Fleming-Elder, B.K.; Peters, J.C.; Hill, J.O. Metabolic adjustments with the development, treatment, and recurrence of obesity in obesity-prone rats. Am. J. Physiol. Integr. Comp. Physiol. 2004, 287, R288–R297. [Google Scholar] [CrossRef]
  43. Field, A.; Wing, R.; Manson, J.; Spiegelman, D.; Willett, W. Relationship of a large weight loss to long-term weight change among young and middle-aged US women. Int. J. Obes. 2001, 25, 1113–1121. [Google Scholar] [CrossRef]
  44. Sarlio-Lähteenkorva, S.; Rissanen, A.; Kaprio, J. A descriptive study of weight loss maintenance: 6 and 15 year follow-up of initially overweight adults. Int. J. Obes. 2000, 24, 116–125. [Google Scholar] [CrossRef] [PubMed]
  45. Strohacker, K.; Carpenter, K.C.; McFarlin, B.K. Consequences of Weight Cycling: An Increase in Disease Risk? Int. J. Exerc. Sci. 2009, 2, 191–201. [Google Scholar] [CrossRef]
  46. Simonds, S.E.; Pryor, J.T.; Cowley, M.A. Repeated weight cycling in obese mice causes increased appetite and glucose intolerance. Physiol. Behav. 2018, 194, 184–190. [Google Scholar] [CrossRef] [PubMed]
  47. Nuttall, F.Q. Body Mass Index: Obesity, BMI, and Health: A Critical Review. Nutr. Today 2015, 50, 117–128. [Google Scholar] [CrossRef] [PubMed]
  48. Kivimäki, M.; Strandberg, T.; Pentti, J.; Nyberg, S.T.; Frank, P.; Jokela, M.; Ervasti, J.; Suominen, S.B.; Vahtera, J.; Sipilä, P.N.; et al. Body-mass index and risk of obesity-related complex multimorbidity: An observational multicohort study. Lancet Diabetes Endocrinol. 2022, 10, 253–263. [Google Scholar] [CrossRef]
  49. Mikolajczyk, R.T.; E Maxwell, A.; El Ansari, W.; Stock, C.; Petkeviciene, J.; Guillen-Grima, F. Relationship between perceived body weight and body mass index based on self- reported height and weight among university students: A cross-sectional study in seven European countries. BMC Public Health 2010, 10, 40. [Google Scholar] [CrossRef]
  50. Haakstad, L.A.H.; Stensrud, T.; Rugseth, G.; Gjestvang, C. Weight Cycling and Dieting Behavior in Fitness Club Members. Front. Endocrinol. 2022, 13, 851887. [Google Scholar] [CrossRef] [PubMed]
  51. Lakicevic, N.; Mani, D.; Paoli, A.; Roklicer, R.; Bianco, A.; Drid, P. Weight cycling in combat sports: Revisiting 25 years of scientific evidence. BMC Sports Sci. Med. Rehabil. 2021, 13, 154. [Google Scholar] [CrossRef] [PubMed]
  52. Alwan, N.; Moss, S.L.; Davies, I.G.; Elliott-Sale, K.J.; Enright, K. Weight loss practices and eating behaviours among female physique athletes: Acquiring the optimal body composition for competition. PLoS ONE 2022, 17, e0262514. [Google Scholar] [CrossRef]
  53. Miles-Chan, J.L.; Isacco, L. Weight cycling practices in sport: A risk factor for later obesity? Obes. Rev. 2021, 22 (Suppl. S2), e13188. [Google Scholar] [CrossRef]
  54. Rhee, E.-J. Weight Cycling and Its Cardiometabolic Impact. J. Obes. Metab. Syndr. 2017, 26, 237–242. [Google Scholar] [CrossRef] [PubMed]
  55. Kakinami, L.; Knäuper, B.; Brunet, J. Weight cycling is associated with adverse cardiometabolic markers in a cross-sectional representative US sample. J. Epidemiol. Community Health 2020, 74, 662–667. [Google Scholar] [CrossRef]
  56. Zou, H.; Yin, P.; Liu, L.; Duan, W.; Li, P.; Yang, Y.; Li, W.; Zong, Q.; Yu, X. Association between weight cycling and risk of developing diabetes in adults: A systematic review and meta-analysis. J. Diabetes Investig. 2021, 12, 625–632. [Google Scholar] [CrossRef] [PubMed]
  57. Park, K.Y.; Park, K.Y.; Hwang, H.S.; Cho, K.H.; Han, K.; Nam, G.E.; Kim, Y.H.; Kwon, Y.; Park, Y.G. Body Weight Fluctuation as a Risk Factor for Type 2 Diabetes: Results from a Nationwide Cohort Study. J. Clin. Med. 2019, 8, 950. [Google Scholar] [CrossRef] [PubMed]
  58. Oh, T.J.; Moon, J.H.; Choi, S.H.; Lim, S.; Park, K.S.; Cho, N.H.; Jang, H.C. Body-Weight Fluctuation and Incident Diabetes Mellitus, Cardiovascular Disease, and Mortality: A 16-Year Prospective Cohort Study. J. Clin. Endocrinol. Metab. 2018, 104, 639–646. [Google Scholar] [CrossRef]
  59. Ridaura, V.K.; Faith, J.J.; Rey, F.E.; Cheng, J.; Duncan, A.E.; Kau, A.L.; Griffin, N.W.; Lombard, V.; Henrissat, B.; Bain, J.R.; et al. Gut Microbiota from Twins Discordant for Obesity Modulate Metabolism in Mice. Science 2013, 341, 1241214. [Google Scholar] [CrossRef]
  60. Kim, M.-H.; Yun, K.E.; Kim, J.; Park, E.; Chang, Y.; Ryu, S.; Kim, H.-L.; Kim, H.-N. Gut microbiota and metabolic health among overweight and obese individuals. Sci. Rep. 2020, 10, 19417. [Google Scholar] [CrossRef]
  61. Thaiss, C.A.; Itav, S.; Rothschild, D.; Meijer, M.T.; Levy, M.; Moresi, C.; Dohnalová, L.; Braverman, S.; Rozin, S.; Malitsky, S.; et al. Persistent microbiome alterations modulate the rate of post-dieting weight regain. Nature 2016, 540, 544–551. [Google Scholar] [CrossRef] [PubMed]
  62. Humblot, C.; Seyoum, Y.; Turpin, W.; Mrabt, R.; List, E.O.; Berryman, D.E.; Jensen, E.A.; Sustarsic, E.G.; Kopchick, J.J.; Ricort, J. Long Term Weight Cycling Affects Fecal Microbiota of Mice. Mol. Nutr. Food Res. 2022, 66, 2200439. [Google Scholar] [CrossRef]
  63. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef]
  64. Chanda, D.; De, D. Meta-analysis reveals obesity associated gut microbial alteration patterns and reproducible contributors of functional shift. Gut Microbes 2024, 16, 2304900. [Google Scholar] [CrossRef] [PubMed]
  65. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef] [PubMed]
  66. Davis, C.D. The Gut Microbiome and Its Role in Obesity. Nutr. Today 2016, 51, 167–174. [Google Scholar] [CrossRef]
  67. Ayogu, R.N.B.; Oshomegie, H.; Udenta, E.A. Energy intake, expenditure and balance, and factors associated with energy balance of young adults (20–39 years): A retrospective cross-sectional community-based cohort study. BMC Nutr. 2022, 8, 142. [Google Scholar] [CrossRef]
  68. Mietlicki-Baase, E.G.; Hayes, M.R. Gut Hormones and Obesity. In Metabolic Syndrome: A Comprehensive Textbook; Ahima, R.S., Ed.; Springer International Publishing: Cham, Switerzland, 2014; pp. 1–28. [Google Scholar]
  69. Hall, K.D.; Farooqi, I.S.; Friedman, J.M.; Klein, S.; Loos, R.J.F.; Mangelsdorf, D.J.; O’rahilly, S.; Ravussin, E.; Redman, L.M.; Ryan, D.H.; et al. The energy balance model of obesity: Beyond calories in, calories out. Am. J. Clin. Nutr. 2022, 115, 1243–1254. [Google Scholar] [CrossRef]
  70. Méquinion, M.; Foldi, C.J.; Andrews, Z.B. The Ghrelin-AgRP Neuron Nexus in Anorexia Nervosa: Implications for Metabolic and Behavioral Adaptations. Front. Nutr. 2019, 6, 190. [Google Scholar] [CrossRef]
  71. Ye, L.; Liddle, R.A. Gastrointestinal hormones and the gut connectome. Curr. Opin. Endocrinol. Diabetes 2017, 24, 9–14. [Google Scholar] [CrossRef] [PubMed]
  72. Daly, D.M.; Park, S.J.; Valinsky, W.C.; Beyak, M.J. Impaired intestinal afferent nerve satiety signalling and vagal afferent excitability in diet induced obesity in the mouse. J. Physiol. 2011, 589, 2857–2870. [Google Scholar] [CrossRef]
  73. Egerod, K.L.; Engelstoft, M.S.; Grunddal, K.V.; Nøhr, M.K.; Secher, A.; Sakata, I.; Pedersen, J.; Windeløv, J.A.; Füchtbauer, E.-M.; Olsen, J.; et al. A major lineage of enteroendocrine cells coexpress CCK, secretin, GIP, GLP-1, PYY, and neurotensin but not somatostatin. Endocrinology 2012, 153, 5782–5795. [Google Scholar] [CrossRef]
  74. Dockray, G.J. Cholecystokinin. Curr. Opin. Endocrinol. Diabetes Obes. 2012, 19, 8–12. [Google Scholar] [CrossRef]
  75. Buchman, A.L.; Katz, S.; Fang, J.C.; Bernstein, C.N.; Abou-Assi, S.G.; Group, T.S. Teduglutide, a novel mucosally active analog of glucagon-like peptide-2 (GLP-2) for the treatment of moderate to severe Crohn’s disease. Inflamm. Bowel Dis. 2010, 16, 962–973. [Google Scholar] [CrossRef] [PubMed]
  76. Tang-Christensen, M.; Vrang, N.; Larsen, P. Glucagon-like peptide containing pathways in the regulation of feeding behaviour. Int. J. Obes. 2001, 25, S42–S47. [Google Scholar] [CrossRef] [PubMed]
  77. Lean, M.E.J.; Malkova, D. Altered gut and adipose tissue hormones in overweight and obese individuals: Cause or consequence? Int. J. Obes. 2016, 40, 622–632. [Google Scholar] [CrossRef] [PubMed]
  78. Akalu, Y.; Molla, M.D.; Dessie, G.; Ayelign, B. Physiological Effect of Ghrelin on Body Systems. Int. J. Endocrinol. 2020, 2020, 1385138. [Google Scholar] [CrossRef]
  79. Romieu, I.; Dossus, L.; Barquera, S.; Blottière, H.M.; Franks, P.W.; Gunter, M.; Hwalla, N.; Hursting, S.D.; Leitzmann, M.; Margetts, B.; et al. Energy balance and obesity: What are the main drivers? Cancer Causes Control 2017, 28, 247–258. [Google Scholar] [CrossRef]
  80. Woodward, O.R.M.; Gribble, F.M.; Reimann, F.; Lewis, J.E. Gut peptide regulation of food intake—Evidence for the modulation of hedonic feeding. J. Physiol. 2022, 600, 1053–1078. [Google Scholar] [CrossRef]
  81. Nauck, M.A.; Meier, J.J. Incretin hormones: Their role in health and disease. Diabetes Obes. Metab. 2018, 20 (Suppl. 1), 5–21. [Google Scholar] [CrossRef]
  82. Friedman, J.M. Leptin and the endocrine control of energy balance. Nat. Metab. 2019, 1, 754–764. [Google Scholar] [CrossRef]
  83. Chan, J.L.; Heist, K.; DePaoli, A.M.; Veldhuis, J.D.; Mantzoros, C.S. The role of falling leptin levels in the neuroendocrine and metabolic adaptation to short-term starvation in healthy men. J. Clin. Investig. 2003, 111, 1409–1421. [Google Scholar] [CrossRef] [PubMed]
  84. Zurita-Cruz, J.N.; Villasís-Keever, M.A.; Manuel-Apolinar, L.; Damasio-Santana, L.; Garrido-Magaña, E.; Rivera-Hernández, A.d.J. Leptin/adiponectin ratio as a prognostic factor for increased weight gain in girls with central precocious puberty. Front. Endocrinol. 2023, 14, 1101399. [Google Scholar] [CrossRef]
  85. Considine, R.V.; Sinha, M.K.; Heiman, M.L.; Kriauciunas, A.; Stephens, T.W.; Nyce, M.R.; Ohannesian, J.P.; Marco, C.C.; McKee, L.J.; Bauer, T.L.; et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N. Engl. J. Med. 1996, 334, 292–295. [Google Scholar] [CrossRef] [PubMed]
  86. Rahmouni, K.; Fath, M.A.; Seo, S.; Thedens, D.R.; Berry, C.J.; Weiss, R.; Nishimura, D.Y.; Sheffield, V.C. Leptin resistance contributes to obesity and hypertension in mouse models of Bardet-Biedl syndrome. J. Clin. Investig. 2008, 118, 1458–1467. [Google Scholar] [CrossRef] [PubMed]
  87. Weyer, C.; Funahashi, T.; Tanaka, S.; Hotta, K.; Matsuzawa, Y.; Pratley, R.E.; Tataranni, P.A. Hypoadiponectinemia in obesity and type 2 diabetes: Close association with insulin resistance and hyperinsulinemia. J. Clin. Endocrinol. Metab. 2001, 86, 1930–1935. [Google Scholar] [CrossRef] [PubMed]
  88. Kos, K.; Harte, A.L.; da Silva, N.F.; Tonchev, A.; Chaldakov, G.; James, S.; Snead, D.R.; Hoggart, B.; O’hare, J.P.; McTernan, P.G.; et al. Adiponectin and resistin in human cerebrospinal fluid and expression of adiponectin receptors in the human hypothalamus. J. Clin. Endocrinol. Metab. 2007, 92, 1129–1136. [Google Scholar] [CrossRef] [PubMed]
  89. Gariballa, S.; Alkaabi, J.; Yasin, J.; Al Essa, A. Total adiponectin in overweight and obese subjects and its response to visceral fat loss. BMC Endocr. Disord. 2019, 19, 55. [Google Scholar] [CrossRef]
  90. Reinehr, T.; de Sousa, G.; Niklowitz, P.; Roth, C.L. Amylin and its relation to insulin and lipids in obese children before and after weight loss. Obesity 2007, 15, 2006–2011. [Google Scholar] [CrossRef]
  91. Strohacker, K.; McCaffery, J.M.; MacLean, P.S.; Wing, R.R. Adaptations of leptin, ghrelin or insulin during weight loss as predictors of weight regain: A review of current literature. Int. J. Obes. 2013, 38, 388–396. [Google Scholar] [CrossRef]
  92. Zhang, Q.; Delessa, C.T.; Wolfrum, C. The glucose-dependent insulinotropic polypeptide (GIP) regulates body weight and food intake via CNS-GIPR signaling. Cell Metab. 2021, 33, 833–844.e5. [Google Scholar] [CrossRef]
  93. Ghaben, A.L.; Scherer, P.E. Adipogenesis and metabolic health. Nat. Rev. Mol. Cell Biol. 2019, 20, 242–258. [Google Scholar]
  94. Sumithran, P.; Prendergast, L.A.; Delbridge, E.; Purcell, K.; Shulkes, A.; Kriketos, A.; Proietto, J. Long-term persistence of hormonal adaptations to weight loss. N. Engl. J. Med. 2011, 365, 1597–1604. [Google Scholar] [CrossRef]
  95. Weiss, E.P.; Albert, S.G.; Reeds, D.N.; Kress, K.S.; Ezekiel, U.R.; McDaniel, J.L.; Patterson, B.W.; Klein, S.; Villareal, D.T. Calorie Restriction and Matched Weight Loss From Exercise: Independent and Additive Effects on Glucoregulation and the Incretin System in Overweight Women and Men. Diabetes Care 2015, 38, 1253–1262. [Google Scholar] [CrossRef] [PubMed]
  96. le Roux, C.W.; Batterham, R.L.; Aylwin, S.J.B.; Patterson, M.; Borg, C.M.; Wynne, K.J.; Kent, A.; Vincent, R.P.; Gardiner, J.; Ghatei, M.A.; et al. Attenuated peptide YY release in obese subjects is associated with reduced satiety. Endocrinology 2006, 147, 3–8. [Google Scholar] [CrossRef] [PubMed]
  97. Chearskul, S.; Delbridge, E.; Shulkes, A.; Proietto, J.; Kriketos, A. Effect of weight loss and ketosis on postprandial cholecystokinin and free fatty acid concentrations. Am. J. Clin. Nutr. 2008, 87, 1238–1246. [Google Scholar] [CrossRef]
  98. Rinninella, E.; Tohumcu, E.; Raoul, P.; Fiorani, M.; Cintoni, M.; Mele, M.C.; Cammarota, G.; Gasbarrini, A.; Ianiro, G. The role of diet in shaping human gut microbiota. Best Pract. Res. Clin. Gastroenterol. 2023, 62–63, 101828. [Google Scholar] [CrossRef]
  99. Ojo, O.; Feng, Q.-Q.; Ojo, O.O.; Wang, X.-H. The Role of Dietary Fibre in Modulating Gut Microbiota Dysbiosis in Patients with Type 2 Diabetes: A Systematic Review and Meta-Analysis of Randomised Controlled Trials. Nutrients 2020, 12, 3239. [Google Scholar] [CrossRef] [PubMed]
  100. Lai, T.-T.; Tsai, Y.-H.; Liou, C.-W.; Fan, C.-H.; Hou, Y.-T.; Yao, T.-H.; Chuang, H.-L.; Wu, W.-L. The gut microbiota modulate locomotion via vagus-dependent glucagon-like peptide-1 signaling. NPJ Biofilms Microbiomes 2024, 10, 2. [Google Scholar] [CrossRef]
  101. Degen, L.; Drewe, J.; Piccoli, F.; Gräni, K.; Oesch, S.; Bunea, R.; D’Amato, M.; Beglinger, C. Effect of CCK-1 receptor blockade on ghrelin and PYY secretion in men. Am. J. Physiol. Integr. Comp. Physiol. 2007, 292, R1391–R1399. [Google Scholar] [CrossRef]
  102. Gribble, F.M.; Reimann, F. Function and mechanisms of enteroendocrine cells and gut hormones in metabolism. Nat. Rev. Endocrinol. 2019, 15, 226–237. [Google Scholar] [CrossRef]
  103. Barker, N.; Van Es, J.H.; Kuipers, J.; Kujala, P.; Van Den Born, M.; Cozijnsen, M.; Haegebarth, A.; Korving, J.; Begthel, H.; Peters, P.J.; et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 2007, 449, 1003–1007. [Google Scholar] [CrossRef]
  104. Zinina, V.V.; Ruehle, F.; Winkler, P.; Rebmann, L.; Lukas, H.; Möckel, S.; Diefenbach, A.; Mendez-Lago, M.; Soshnikova, N. ID2 controls differentiation of enteroendocrine cells in mouse small intestine. Acta Physiol. 2022, 234, e13773. [Google Scholar] [CrossRef]
  105. Frias, A.B.; Hyzny, E.J.; Buechel, H.M.; Beppu, L.Y.; Xie, B.; Jurczak, M.J.; D’cruz, L.M. The Transcriptional Regulator Id2 Is Critical for Adipose-Resident Regulatory T Cell Differentiation, Survival, and Function. J. Immunol. 2019, 203, 658–664. [Google Scholar] [CrossRef]
  106. Hou, T.Y.; Ward, S.M.; Murad, J.M.; Watson, N.P.; Israel, M.A.; Duffield, G.E. ID2 (inhibitor of DNA binding 2) is a rhythmically expressed transcriptional repressor required for circadian clock output in mouse liver. J. Biol. Chem. 2009, 284, 31735–31745. [Google Scholar] [CrossRef] [PubMed]
  107. Wölnerhanssen, B.K.; Moran, A.W.; Burdyga, G.; Meyer-Gerspach, A.C.; Peterli, R.; Manz, M.; Thumshirn, M.; Daly, K.; Beglinger, C.; Shirazi-Beechey, S.P. Deregulation of transcription factors controlling intestinal epithelial cell differentiation; a predisposing factor for reduced enteroendocrine cell number in morbidly obese individuals. Sci. Rep. 2017, 7, 8174. [Google Scholar] [CrossRef] [PubMed]
  108. Cong, J.; Zhou, P.; Zhang, R. Intestinal Microbiota-Derived Short Chain Fatty Acids in Host Health and Disease. Nutrients 2022, 14, 1977. [Google Scholar] [CrossRef] [PubMed]
  109. Larraufie, P.; Martin-Gallausiaux, C.; Lapaque, N.; Dore, J.; Gribble, F.M.; Reimann, F.; Blottiere, H.M. SCFAs strongly stimulate PYY production in human enteroendocrine cells. Sci. Rep. 2018, 8, 74. [Google Scholar] [CrossRef]
  110. Ducastel, S.; Touche, V.; Trabelsi, M.-S.; Boulinguiez, A.; Butruille, L.; Nawrot, M.; Peschard, S.; Chávez-Talavera, O.; Dorchies, E.; Vallez, E.; et al. The nuclear receptor FXR inhibits Glucagon-Like Peptide-1 secretion in response to microbiota-derived Short-Chain Fatty Acids. Sci. Rep. 2020, 10, 174. [Google Scholar] [CrossRef]
  111. Cawthon, C.R.; de La Serre, C.B. The critical role of CCK in the regulation of food intake and diet-induced obesity. Peptides 2021, 138, 170492. [Google Scholar] [CrossRef]
  112. Russell, W.R.; Gratz, S.W.; Duncan, S.H.; Holtrop, G.; Ince, J.; Scobbie, L.; Duncan, G.; Johnstone, A.M.; Lobley, G.E.; Wallace, R.J.; et al. High-protein, reduced-carbohydrate weight-loss diets promote metabolite profiles likely to be detrimental to colonic health. Am. J. Clin. Nutr. 2011, 93, 1062–1072. [Google Scholar] [CrossRef]
  113. Anderson, E.M.; Rozowsky, J.M.; Fazzone, B.J.; Schmidt, E.A.; Stevens, B.R.; O’malley, K.A.; Scali, S.T.; Berceli, S.A. Temporal Dynamics of the Intestinal Microbiome Following Short-Term Dietary Restriction. Nutrients 2022, 14, 2785. [Google Scholar] [CrossRef]
  114. Ott, B.; Skurk, T.; Hastreiter, L.; Lagkouvardos, I.; Fischer, S.; Büttner, J.; Kellerer, T.; Clavel, T.; Rychlik, M.; Haller, D.; et al. Effect of caloric restriction on gut permeability, inflammation markers, and fecal microbiota in obese women. Sci. Rep. 2017, 7, 11955. [Google Scholar] [CrossRef]
  115. Valenzano, A.; Tartaglia, N.; Ambrosi, A.; Tafuri, D.; Monda, M.; Messina, A.; Sessa, F.; Campanozzi, A.; Monda, V.; Cibelli, G.; et al. The Metabolic Rearrangements of Bariatric Surgery: Focus on Orexin-A and the Adiponectin System. J. Clin. Med. 2020, 9, 3327. [Google Scholar] [CrossRef] [PubMed]
  116. Goldstone, A.P.; Miras, A.D.; Scholtz, S.; Jackson, S.; Neff, K.J.; Pénicaud, L.; Geoghegan, J.; Chhina, N.; Durighel, G.; Bell, J.D.; et al. Link Between Increased Satiety Gut Hormones and Reduced Food Reward After Gastric Bypass Surgery for Obesity. J. Clin. Endocrinol. Metab. 2016, 101, 599–609. [Google Scholar] [CrossRef] [PubMed]
  117. Laferrère, B.; Pattou, F. Weight-Independent Mechanisms of Glucose Control After Roux-en-Y Gastric Bypass. Front. Endocrinol. 2018, 9, 530. [Google Scholar] [CrossRef]
  118. Kim, K.-S.; Peck, B.C.; Hung, Y.-H.; Koch-Laskowski, K.; Wood, L.; Dedhia, P.H.; Spence, J.R.; Seeley, R.J.; Sethupathy, P.; Sandoval, D.A. Vertical sleeve gastrectomy induces enteroendocrine cell differentiation of intestinal stem cells through bile acid signaling. J. Clin. Investig. 2022, 7, e154302. [Google Scholar] [CrossRef]
  119. Abdeen, G.; le Roux, C. Mechanism Underlying the Weight Loss and Complications of Roux-en-Y Gastric Bypass. Rev. Obes. Surg. 2015, 26, 410–421. [Google Scholar] [CrossRef]
  120. le Roux, C.W.; Welbourn, R.; Werling, M.; Osborne, A.; Kokkinos, A.; Laurenius, A.; Lönroth, H.; Fändriks, L.; Ghatei, M.A.; Bloom, S.R. Gut hormones as mediators of appetite and weight loss after Roux-en-Y gastric bypass. Ann. Surg. 2007, 246, 780–785. [Google Scholar] [CrossRef]
  121. Guida, C.; Stephen, S.D.; Watson, M.; Dempster, N.; Larraufie, P.; Marjot, T.; Cargill, T.; Rickers, L.; Pavlides, M.; Tomlinson, J.; et al. PYY plays a key role in the resolution of diabetes following bariatric surgery in humans. eBioMedicine 2019, 40, 67–76. [Google Scholar] [CrossRef]
  122. Alexiadou, K.; Tan, T.M.-M. Gastrointestinal Peptides as Therapeutic Targets to Mitigate Obesity and Metabolic Syndrome. Curr. Diabetes Rep. 2020, 20, 26. [Google Scholar] [CrossRef]
  123. Alruwaili, H.; Dehestani, B.; le Roux, C.W. Clinical Impact of Liraglutide as a Treatment of Obesity. Clin. Pharmacol. Adv. Appl. 2021, 13, 53–60. [Google Scholar] [CrossRef]
  124. Pi-Sunyer, X.; Astrup, A.; Fujioka, K.; Greenway, F.; Halpern, A.; Krempf, M.; Lau, D.C.W.; Le Roux, C.W.; Ortiz, R.V.; Jensen, C.B.; et al. A Randomized, Controlled Trial of 3.0 mg of Liraglutide in Weight Management. N. Engl. J. Med. 2015, 373, 11–22. [Google Scholar] [CrossRef]
  125. Singh, G.; Krauthamer, M.; Bjalme-Evans, M. Wegovy (Semaglutide): A New Weight Loss Drug for Chronic Weight Management. J. Investig. Med. 2022, 70, 5–13. [Google Scholar] [CrossRef] [PubMed]
  126. Rosenstock, J.; Wysham, C.; Frías, J.P.; Kaneko, S.; Lee, C.J.; Landó, L.F.; Mao, H.; Cui, X.; A Karanikas, C.A.; Thieu, V.T. Efficacy and safety of a novel dual GIP and GLP-1 receptor agonist tirzepatide in patients with type 2 diabetes (SURPASS-1): A double-blind, randomised, phase 3 trial. Lancet 2021, 398, 143–155. [Google Scholar] [CrossRef] [PubMed]
  127. Li, H.W.R.; Lee, C.P.; Lam, K.S.L.; Ho, P.C. 31—Anti-Obesity Drugs for Obese Women Planning Pregnancy. In Obesity; Mahmood, T., Arulkumaran, S., Eds.; Elsevier: Oxford, UK, 2013; pp. 423–430. [Google Scholar]
  128. Kwon, Y.-J.; Kwon, G.E.; Lee, H.S.; Choi, M.H.; Lee, J.-W. The Effect of Orlistat on Sterol Metabolism in Obese Patients. Front. Endocrinol. 2022, 13, 824269. [Google Scholar] [CrossRef] [PubMed]
  129. Hauptman, J. Orlistat in the long-term treatment of obesity in primary care settings. Arch. Fam. Med. 2000, 9, 160–167. [Google Scholar] [CrossRef]
  130. Apovian, C.M.; Aronne, L.J.; Bessesen, D.H.; McDonnell, M.E.; Murad, M.H.; Pagotto, U.; Ryan, D.H.; Still, C.D. Pharmacological management of obesity: An endocrine Society clinical practice guideline. J. Clin. Endocrinol. Metab. 2015, 100, 342–362. [Google Scholar] [CrossRef]
  131. Lee, C.; Dixon, J. Pharmacotherapy for obesity. Aust. Fam. Physician 2017, 46, 472–477. [Google Scholar]
  132. Colbourne, J.R.M.; Fisher, O.M.; Mo, S.; Rigas, G.S.; Talbot, M.L. The role of adjuvant pharmacotherapy with liraglutide for patients with inadequate weight loss following bariatric surgery. Langenbeck’s Arch. Surg. 2023, 408, 115. [Google Scholar] [CrossRef]
  133. Smits, M.M.; Van Raalte, D.H. Safety of Semaglutide. Front. Endocrinol. 2021, 12, 645563. [Google Scholar]
  134. Ghusn, W.; De la Rosa, A.; Sacoto, D.; Cifuentes, L.; Campos, A.; Feris, F.; Hurtado, M.D.; Acosta, A. Weight Loss Outcomes Associated With Semaglutide Treatment for Patients with Overweight or Obesity. JAMA Netw. Open 2022, 5, e2231982. [Google Scholar] [CrossRef]
  135. Landsberg, L. Insulin-mediated sympathetic stimulation: Role in the pathogenesis of obesity-related hypertension (or, how insulin affects blood pressure, and why). J. Hypertens. 2001, 19, 523–528. [Google Scholar] [CrossRef]
  136. Cero, C.; Lea, H.J.; Zhu, K.Y.; Shamsi, F.; Tseng, Y.-H.; Cypess, A.M. β3-Adrenergic receptors regulate human brown/beige adipocyte lipolysis and thermogenesis. J. Clin. Investig. 2021, 6, e139160. [Google Scholar] [CrossRef] [PubMed]
  137. Valentine, J.M.; Ahmadian, M.; Keinan, O.; Abu-Odeh, M.; Zhao, P.; Zhou, X.; Keller, M.P.; Gao, H.; Yu, R.T.; Liddle, C.; et al. β3-Adrenergic receptor downregulation leads to adipocyte catecholamine resistance in obesity. J. Clin. Investig. 2022, 132, e153357. [Google Scholar] [CrossRef] [PubMed]
  138. Thorp, A.A.; Schlaich, M.P. Relevance of Sympathetic Nervous System Activation in Obesity and Metabolic Syndrome. J. Diabetes Res. 2015, 2015, 341583. [Google Scholar] [CrossRef] [PubMed]
  139. Cannon, B.; Nedergaard, J. Nonshivering thermogenesis and its adequate measurement in metabolic studies. J. Exp. Biol. 2011, 214, 242–253. [Google Scholar] [CrossRef]
  140. Carpentier, A.C.; Blondin, D.P.; Virtanen, K.A.; Richard, D.; Haman, F.; Turcotte, É.E. Brown Adipose Tissue Energy Metabolism in Humans. Front. Endocrinol. 2018, 9, 447. [Google Scholar] [CrossRef]
  141. Liu, X.; Wang, S.; You, Y.; Meng, M.; Zheng, Z.; Dong, M.; Lin, J.; Zhao, Q.; Zhang, C.; Yuan, X.; et al. Brown adipose tissue transplantation reverses obesity in Ob/Ob mice. Endocrinology 2015, 156, 2461–2469. [Google Scholar] [CrossRef]
  142. Goele, K.; Bosy-Westphal, A.; Rümcker, B.; Lagerpusch, M.; Müller, M.J. Influence of changes in body composition and adaptive thermogenesis on the difference between measured and predicted weight loss in obese women. Obes. Facts 2009, 2, 6. [Google Scholar] [CrossRef]
  143. Byrne, N.M.; Wood, R.E.; Schutz, Y.; Hills, A.P. Does metabolic compensation explain the majority of less-than-expected weight loss in obese adults during a short-term severe diet and exercise intervention? Int. J. Obes. 2012, 36, 1472–1478. [Google Scholar] [CrossRef] [PubMed]
  144. Lowell, B.B.; Spiegelman, B.M. Towards a molecular understanding of adaptive thermogenesis. Nature 2000, 404, 652–660. [Google Scholar] [CrossRef]
  145. Müller, M.; Bosy-Westphal, A. Adaptive thermogenesis with weight loss in humans. Obesity 2013, 21, 218–228. [Google Scholar] [CrossRef]
  146. Hildebrandt, X.; Ibrahim, M.; Peltzer, N. Cell death and inflammation during obesity: “Know my methods, WAT(son)”. Cell Death Differ. 2022, 30, 279–292. [Google Scholar] [CrossRef] [PubMed]
  147. Lee, D.-E.; Kehlenbrink, S.; Lee, H.; Hawkins, M.; Yudkin, J.S. Getting the message across: Mechanisms of physiological cross talk by adipose tissue. Am. J. Physiol. Metab. 2009, 296, E1210–E1229. [Google Scholar] [CrossRef] [PubMed]
  148. Greenberg, A.S.; Obin, M.S. Obesity and the role of adipose tissue in inflammation and metabolism. Am. J. Clin. Nutr. 2006, 83, 461S–465S. [Google Scholar] [CrossRef]
  149. Trayhurn, P.; Bing, C.; Wood, I.S. Adipose Tissue and Adipokines—Energy Regulation from the Human Perspective. J. Nutr. 2006, 136, 1935S–1939S. [Google Scholar] [CrossRef]
  150. Ding, S.; Chi, M.M.; Scull, B.P.; Rigby, R.; Schwerbrock, N.M.J.; Magness, S.; Jobin, C.; Lund, P.K. High-Fat Diet: Bacteria Interactions Promote Intestinal Inflammation Which Precedes and Correlates with Obesity and Insulin Resistance in Mouse. PLoS ONE 2010, 5, e12191. [Google Scholar] [CrossRef]
  151. Alzamil, H. Elevated Serum TNF-α Is Related to Obesity in Type 2 Diabetes Mellitus and Is Associated with Glycemic Control and Insulin Resistance. J. Obes. 2020, 2020, 5076858. [Google Scholar] [CrossRef]
  152. Popko, K.; Gorska, E.; Stelmaszczyk-Emmel, A.; Plywaczewski, R.; Stoklosa, A.; Gorecka, D.; Pyrzak, B.; Demkow, U. Proinflammatory cytokines IL-6 and TNF-α and the development of inflammation in obese subjects. Eur. J. Med. Res. 2010, 15, 120–122. [Google Scholar] [CrossRef] [PubMed]
  153. Fried, S.K.; Bunkin, D.A.; Greenberg, A.S. Omental and Subcutaneous Adipose Tissues of Obese Subjects Release Interleukin-6: Depot Difference and Regulation by glucocorticoid. J. Clin. Endocrinol. Metab. 1998, 83, 847–850. [Google Scholar] [CrossRef]
  154. El-Mikkawy, D.M.E.; El-Sadek, M.A.; El-Badawy, M.A.; Samaha, D. Circulating level of interleukin-6 in relation to body mass indices and lipid profile in Egyptian adults with overweight and obesity. Egypt. Rheumatol. Rehabil. 2020, 47, 7. [Google Scholar] [CrossRef]
  155. Han, M.S.; White, A.; Perry, R.J.; Camporez, J.-P.; Hidalgo, J.; Shulman, G.I.; Davis, R.J. Regulation of adipose tissue inflammation by interleukin 6. Proc. Natl. Acad. Sci. USA 2020, 117, 2751–2760. [Google Scholar] [CrossRef]
  156. Sougiannis, A.T.; VanderVeen, B.N.; Cranford, T.L.; Enos, R.T.; Velazquez, K.T.; McDonald, S.; Bader, J.E.; Chatzistamou, I.; Fan, D.; Murphy, E.A. Impact of weight loss and partial weight regain on immune cell and inflammatory markers in adipose tissue in male mice. J. Appl. Physiol. 2020, 129, 909–919. [Google Scholar] [CrossRef] [PubMed]
  157. Tareen, S.H.K.; Kutmon, M.; de Kok, T.M.; Mariman, E.C.M.; van Baak, M.A.; Evelo, C.T.; Adriaens, M.E.; Arts, I.C.W. Stratifying cellular metabolism during weight loss: An interplay of metabolism, metabolic flexibility and inflammation. Sci. Rep. 2020, 10, 1651. [Google Scholar] [CrossRef]
  158. Caslin, H.L.; Cottam, M.A.; Piñon, J.M.; Boney, L.Y.; Hasty, A.H. Weight cycling induces innate immune memory in adipose tissue macrophages. Front. Immunol. 2023, 13, 984859. [Google Scholar] [CrossRef] [PubMed]
  159. Ding, S.; Lund, P.K. Role of intestinal inflammation as an early event in obesity and insulin resistance. Curr. Opin. Clin. Nutr. Metab. Care 2011, 14, 328–333. [Google Scholar] [CrossRef] [PubMed]
  160. Rohm, T.V.; Fuchs, R.; Müller, R.L.; Keller, L.; Baumann, Z.; Bosch, A.J.T.; Schneider, R.; Labes, D.; Langer, I.; Pilz, J.B.; et al. Obesity in Humans Is Characterized by Gut Inflammation as Shown by Pro-Inflammatory Intestinal Macrophage Accumulation. Front. Immunol. 2021, 12, 668654. [Google Scholar] [CrossRef]
  161. Khan, S.; Luck, H.; Winer, S.; Winer, D.A. Emerging concepts in intestinal immune control of obesity-related metabolic disease. Nat. Commun. 2021, 12, 2598. [Google Scholar] [CrossRef]
  162. Volpato, E.; Bosio, C.; Previtali, E.; Leone, S.; Armuzzi, A.; Pagnini, F.; Graffigna, G. The evolution of IBD perceived engagement and care needs across the life-cycle: A scoping review. BMC Gastroenterol. 2021, 21, 293. [Google Scholar] [CrossRef] [PubMed]
  163. Serre, C.B.d.L.; Ellis, C.L.; Lee, J.; Hartman, A.L.; Rutledge, J.C.; Raybould, H.E. Propensity to high-fat diet-induced obesity in rats is associated with changes in the gut microbiota and gut inflammation. Am. J. Physiol.-Gastrointest. Liver Physiol. 2010, 299, G440–G448. [Google Scholar] [CrossRef]
  164. Li, X.; Wei, X.; Sun, Y.; Du, J.; Li, X.; Xun, Z.; Li, Y.C. High-fat diet promotes experimental colitis by inducing oxidative stress in the colon. Am. J. Physiol.-Gastrointest. Liver Physiol. 2019, 317, G453–G462. [Google Scholar] [CrossRef]
  165. Pavelock, N.; Masood, U.; Minchenberg, S.; Heisig, D. Effects of obesity on the course of inflammatory bowel disease. Bayl. Univ. Med. Cent. Proc. 2019, 32, 14–17. [Google Scholar] [CrossRef]
  166. Weissman, S.; Patel, K.; Kolli, S.; Lipcsey, M.; Qureshi, N.; Elias, S.; Walfish, A.; Swaminath, A.; Feuerstein, J.D. Obesity in Inflammatory Bowel Disease Is Associated with Early Readmissions Characterised by an Increased Systems and Patient-level Burden. J. Crohn’s Colitis 2021, 15, 1807–1815. [Google Scholar] [CrossRef] [PubMed]
  167. Chan, S.S.M.; Chen, Y.; Casey, K.; Olen, O.; Ludvigsson, J.F.; Carbonnel, F.; Oldenburg, B.; Gunter, M.J.; Tjønneland, A.; Grip, O.; et al. Obesity is Associated With Increased Risk of Crohn’s disease, but not Ulcerative Colitis: A Pooled Analysis of Five Prospective Cohort Studies. Clin. Gastroenterol. Hepatol. 2022, 20, 1048–1058. [Google Scholar] [CrossRef] [PubMed]
  168. Singh, S.; Dulai, P.S.; Zarrinpar, A.; Ramamoorthy, S.; Sandborn, W.J. Obesity in IBD: Epidemiology, pathogenesis, disease course and treatment outcomes. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 110–121. [Google Scholar] [CrossRef] [PubMed]
  169. Flores, A.; Burstein, E.; Cipher, D.J.; Feagins, L.A. Obesity in Inflammatory Bowel Disease: A Marker of Less Severe Disease. Dig. Dis. Sci. 2015, 60, 2436–2445. [Google Scholar] [CrossRef] [PubMed]
  170. Long, M.D.; Crandall, W.V.; Leibowitz, I.H.; Duffy, L.; del Rosario, F.; Kim, S.C.; Integlia, M.J.; Berman, J.; Grunow, J.; Colletti, R.B.; et al. Prevalence and epidemiology of overweight and obesity in children with inflammatory bowel disease. Inflamm. Bowel. Dis. 2011, 17, 2162–2168. [Google Scholar] [CrossRef]
  171. Li, X.; Jiang, L.; Yang, M.; Wu, Y.W.; Sun, J.Z. Impact of weight cycling on CTRP3 expression, adipose tissue inflammation and insulin sensitivity in C57BL/6J mice. Exp. Ther. Med. 2018, 16, 2052–2059. [Google Scholar] [CrossRef]
  172. Yu, H.; Zhang, Z.; Li, G.; Feng, Y.; Xian, L.; Bakhsh, F.; Xu, D.; Xu, C.; Vong, T.; Wu, B.; et al. Adipokine C1q/Tumor Necrosis Factor- Related Protein 3 (CTRP3) Attenuates Intestinal Inflammation via Sirtuin 1/NF-κB Signaling. Cell. Mol. Gastroenterol. Hepatol. 2022, 15, 1000–1015. [Google Scholar] [CrossRef]
  173. Wolf, R.M.; Steele, K.E.; Peterson, L.A.; Magnuson, T.H.; Schweitzer, M.A.; Wong, G.W. Lower Circulating C1q/TNF-Related Protein-3 (CTRP3) Levels Are Associated with Obesity: A Cross-Sectional Study. PLoS ONE 2015, 10, e0133955. [Google Scholar] [CrossRef]
  174. Reinecker, H.-C.; Loh, E.Y.; Ringler, D.J.; Mehta, A.; Rombeau, J.L.; MacDermott, R.P. Monocyte-chemoattractant protein 1 gene expression in intestinal epithelial cells and inflammatory bowel disease mucosa. Gastroenterology 1995, 108, 40–50. [Google Scholar] [CrossRef]
  175. Futagami, S.; Hiratsuka, T.; Tatsuguchi, A.; Suzuki, K.; Kusunoki, M.; Shinji, Y.; Shinoki, K.; Iizumi, T.; Akamatsu, T.; Nishigaki, H.; et al. Monocyte chemoattractant protein 1 (MCP-1) released from Helicobacter pylori stimulated gastric epithelial cells induces cyclooxygenase 2 expression and activation in T cells. Gut 2003, 52, 1257–1264. [Google Scholar] [CrossRef]
  176. Khan, W.I.; Motomura, Y.; Wang, H.; El-Sharkawy, R.T.; Verdu, E.F.; Verma-Gandhu, M.; Rollins, B.J.; Collins, S.M. Critical role of MCP-1 in the pathogenesis of experimental colitis in the context of immune and enterochromaffin cells. Am. J. Physiol. Liver Physiol. 2006, 291, G803–G811. [Google Scholar] [CrossRef]
  177. Grimm, M.C.; Elsbury, S.K.; Pavli, P.; Doe, W.F. Interleukin 8: Cells of origin in inflammatory bowel disease. Gut 1996, 38, 90–98. [Google Scholar] [CrossRef] [PubMed]
  178. Grimm, M.C.; O Elsbury, S.K.; Pavli, P.; Doe, W.F. Enhanced expression and production of monocyte chemoattractant protein-1 in inflammatory bowel disease mucosa. J. Leukoc. Biol. 1996, 59, 804–812. [Google Scholar] [CrossRef] [PubMed]
  179. Lissner, D.; Schumann, M.; Batra, A.; Kredel, L.-I.; Kühl, A.A.; Erben, U.; May, C.; Schulzke, J.-D.; Siegmund, B. Monocyte and M1 Macrophage-induced Barrier Defect Contributes to Chronic Intestinal Inflammation in IBD. Inflamm. Bowel Dis. 2015, 21, 1297–1305. [Google Scholar] [CrossRef] [PubMed]
  180. Zhou, X.; Li, W.; Wang, S.; Zhang, P.; Wang, Q.; Xiao, J.; Zhang, C.; Zheng, X.; Xu, X.; Xue, S.; et al. YAP Aggravates Inflammatory Bowel Disease by Regulating M1/M2 Macrophage Polarization and Gut Microbial Homeostasis. Cell Rep. 2019, 27, 1176–1189.e5. [Google Scholar] [CrossRef]
  181. Pan, X.; Zhu, Q.; Pan, L.-L.; Sun, J. Macrophage immunometabolism in inflammatory bowel diseases: From pathogenesis to therapy. Pharmacol. Ther. 2022, 238, 108176. [Google Scholar] [CrossRef]
  182. Pendyala, S.; Neff, L.M.; Suárez-Fariñas, M.; Holt, P.R. Diet-induced weight loss reduces colorectal inflammation: Implications for colorectal carcinogenesis. Am. J. Clin. Nutr. 2011, 93, 234–242. [Google Scholar] [CrossRef]
  183. Walana, W.; Ye, Y.; Li, M.; Wang, J.; Wang, B.; Cheng, J.-W.; Gordon, J.R.; Li, F. IL-8 antagonist, CXCL8(3-72)K11R/G31P coupled with probiotic exhibit variably enhanced therapeutic potential in ameliorating ulcerative colitis. Biomed. Pharmacother. 2018, 103, 253–261. [Google Scholar] [CrossRef]
  184. Nishitani, Y.; Zhang, L.; Yoshida, M.; Azuma, T.; Kanazawa, K.; Hashimoto, T.; Mizuno, M. Intestinal Anti-Inflammatory Activity of Lentinan: Influence on IL-8 and TNFR1 Expression in Intestinal Epithelial Cells. PLoS ONE 2013, 8, e62441. [Google Scholar] [CrossRef]
  185. Casado-Bedmar, M.; Heil, S.D.S.; Myrelid, P.; Söderholm, J.D.; Keita, V. Upregulation of intestinal mucosal mast cells expressing VPAC1 in close proximity to vasoactive intestinal polypeptide in inflammatory bowel disease and murine colitis. Neurogastroenterol. Motil. 2018, 31, e13503. [Google Scholar] [CrossRef]
  186. Abad, C.; Martinez, C.; Juarranz, M.G.; Arranz, A.; Leceta, J.; Delgado, M.; Gomariz, R.P. Therapeutic effects of vasoactive intestinal peptide in the trinitrobenzene sulfonic acid mice model of Crohn’s disease. Gastroenterology 2003, 124, 961–971. [Google Scholar] [CrossRef] [PubMed]
  187. El-Salhy, M.; Mazzawi, T.; Gundersen, D.; Hatlebakk, J.G.; Hausken, T. The role of peptide YY in gastrointestinal diseases and disorders (review). Int. J. Mol. Med. 2013, 31, 275–282. [Google Scholar] [CrossRef] [PubMed]
  188. Cooper, J.A. Factors affecting circulating levels of peptide YY in humans: A comprehensive review. Nutr. Res. Rev. 2014, 27, 186–197. [Google Scholar] [CrossRef] [PubMed]
  189. Karra, E.; Chandarana, K.; Batterham, R.L. The role of peptide YY in appetite regulation and obesity. J. Physiol. 2009, 587, 19–25. [Google Scholar] [CrossRef] [PubMed]
  190. Inczefi, O.; Bacsur, P.; Resál, T.; Keresztes, C.; Molnár, T. The Influence of Nutrition on Intestinal Permeability and the Microbiome in Health and Disease. Front. Nutr. 2022, 9, 718710. [Google Scholar] [CrossRef]
  191. Ahmad, R.; Sorrell, M.; Batra, S.; Dhawan, P.; Singh, A. Gut permeability and mucosal inflammation: Bad, good or context dependent. Mucosal Immunol. 2017, 10, 307–317. [Google Scholar] [CrossRef]
  192. Clemente, J.C.; Ursell, L.K.; Parfrey, L.W.; Knight, R. The impact of the gut microbiota on human health: An integrative view. Cell 2012, 148, 1258–1270. [Google Scholar] [CrossRef]
  193. Hills, R.D., Jr.; Pontefract, B.A.; Mishcon, H.R.; Black, C.A.; Sutton, S.C.; Theberge, C.R. Gut Microbiome: Profound Implications for Diet and Disease. Nutrients 2019, 11, 1613. [Google Scholar] [CrossRef]
  194. Ley, R.E.; Bäckhed, F.; Turnbaugh, P.; Lozupone, C.A.; Knight, R.D.; Gordon, J.I. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. USA 2005, 102, 11070–11075. [Google Scholar] [CrossRef]
  195. Willis, A.D. Rarefaction, Alpha Diversity, and Statistics. Front. Microbiol. 2019, 10, 2407. [Google Scholar] [CrossRef]
  196. Kawashima, T.; Ogata, M.; Fujita, N.; Takahashi, R. Daisaikoto Prevents Post-dieting Weight Regain by Reversing Dysbiosis and Reducing Serum Corticosterone in Mice. Front. Physiol. 2019, 10, 1483. [Google Scholar] [CrossRef] [PubMed]
  197. Peters, B.A.; Shapiro, J.A.; Church, T.R.; Miller, G.; Trinh-Shevrin, C.; Yuen, E.; Friedlander, C.; Hayes, R.B.; Ahn, J. A taxonomic signature of obesity in a large study of American adults. Sci. Rep. 2018, 8, 9749. [Google Scholar] [CrossRef] [PubMed]
  198. Oki, K.; Toyama, M.; Banno, T.; Chonan, O.; Benno, Y.; Watanabe, K. Comprehensive analysis of the fecal microbiota of healthy Japanese adults reveals a new bacterial lineage associated with a phenotype characterized by a high frequency of bowel movements and a lean body type. BMC Microbiol. 2016, 16, 284. [Google Scholar] [CrossRef]
  199. Fu, J.; Bonder, M.J.; Cenit, M.C.; Tigchelaar, E.F.; Maatman, A.; Dekens, J.A.M.; Brandsma, E.; Marczynska, J.; Imhann, F.; Weersma, R.K.; et al. The gut microbiome contributes to a substantial proportion of the variation in blood lipids. Circ. Res. 2015, 117, 817–824. [Google Scholar] [CrossRef]
  200. Li, Q.; Wang, Q.; Xu, W.; Ma, Y.; Wang, Q.; Eatman, D.; You, S.; Zou, J.; Champion, J.; Zhao, L.; et al. C-Reactive Protein Causes Adult-Onset Obesity Through Chronic Inflammatory Mechanism. Front. Cell Dev. Biol. 2020, 8, 18. [Google Scholar] [CrossRef]
  201. Mazier, W.; Le Corf, K.; Martinez, C.; Tudela, H.; Kissi, D.; Kropp, C.; Coubard, C.; Soto, M.; Elustondo, F.; Rawadi, G.; et al. A New Strain of Christensenella minuta as a Potential Biotherapy for Obesity and Associated Metabolic Diseases. Cells 2021, 10, 823. [Google Scholar] [CrossRef]
  202. Relizani, K.; Le Corf, K.; Kropp, C.; Martin-Rosique, R.; Kissi, D.; Déjean, G.; Bruno, L.; Martinez, C.; Rawadi, G.; Elustondo, F.; et al. Selection of a novel strain of Christensenella minuta as a future biotherapy for Crohn’s disease. Sci. Rep. 2022, 12, 6017. [Google Scholar] [CrossRef] [PubMed]
  203. Waters, J.L.; Ley, R.E. The human gut bacteria Christensenellaceae are widespread, heritable, and associated with health. BMC Biol. 2019, 17, 83. [Google Scholar] [CrossRef]
  204. Kropp, C.; Le Corf, K.; Relizani, K.; Tambosco, K.; Martinez, C.; Chain, F.; Rawadi, G.; Langella, P.; Claus, S.P.; Martin, R. The Keystone commensal bacterium Christensenella minuta DSM 22607 displays anti-inflammatory properties both in vitro and in vivo. Sci. Rep. 2021, 11, 11494. [Google Scholar] [CrossRef]
  205. Jones, S.E.; Versalovic, J. Probiotic Lactobacillus reuteribiofilms produce antimicrobial and anti-inflammatory factors. BMC Microbiol. 2009, 9, 35. [Google Scholar] [CrossRef]
  206. Giraffa, G.; Chanishvili, N.; Widyastuti, Y. Importance of lactobacilli in food and feed biotechnology. Res. Microbiol. 2010, 161, 480–487. [Google Scholar] [CrossRef] [PubMed]
  207. Hu, M.-X.; He, F.; Guo, Y.-X.; Mo, L.-Z.; Zhu, X. Lactobacillus reuteri Biofilms Inhibit Pathogens and Regulate Microbiota in In Vitro Fecal Fermentation. J. Agric. Food Chem. 2022, 70, 11935–11943. [Google Scholar] [CrossRef] [PubMed]
  208. Ahl, D.; Liu, H.; Schreiber, O.; Roos, S.; Phillipson, M.; Holm, L. Lactobacillus reuteri increases mucus thickness and ameliorates dextran sulphate sodium-induced colitis in mice. Acta Physiol. 2016, 217, 300–310. [Google Scholar] [CrossRef] [PubMed]
  209. Wang, G.; Huang, S.; Cai, S.; Yu, H.; Wang, Y.; Zeng, X.; Qiao, S. Lactobacillus reuteri Ameliorates Intestinal Inflammation and Modulates Gut Microbiota and Metabolic Disorders in Dextran Sulfate Sodium-Induced Colitis in Mice. Nutrients 2020, 12, 2298. [Google Scholar] [CrossRef]
  210. Yu, Z.; Chen, J.; Liu, Y.; Meng, Q.; Liu, H.; Yao, Q.; Song, W.; Ren, X.; Chen, X. The role of potential probiotic strains Lactobacillus reuteri in various intestinal diseases: New roles for an old player. Front. Microbiol. 2023, 14, 1095555. [Google Scholar] [CrossRef]
  211. Mu, Q.; Tavella, V.J.; Luo, X.M. Role of Lactobacillus reuteri in Human Health and Diseases. Front. Microbiol. 2018, 9, 757. [Google Scholar] [CrossRef]
  212. La Reau, A.J.; Meier-Kolthoff, J.P.; Suen, G. Sequence-based analysis of the genus Ruminococcus resolves its phylogeny and reveals strong host association. Microb. Genom. 2016, 2, e000099. [Google Scholar] [CrossRef]
  213. Henke, M.T.; Kenny, D.J.; Cassilly, C.D.; Vlamakis, H.; Xavier, R.J.; Clardy, J. Ruminococcus gnavus, a member of the human gut microbiome associated with Crohn’s disease, produces an inflammatory polysaccharide. Proc. Natl. Acad. Sci. USA 2019, 116, 12672–12677. [Google Scholar] [CrossRef]
  214. Hall, A.B.; Yassour, M.; Sauk, J.; Garner, A.; Jiang, X.; Arthur, T.; Lagoudas, G.K.; Vatanen, T.; Fornelos, N.; Wilson, R.; et al. A novel Ruminococcus gnavus clade enriched in inflammatory bowel disease patients. Genome Med. 2017, 9, 103. [Google Scholar] [CrossRef]
  215. Grahnemo, L.; Nethander, M.; Coward, E.; Gabrielsen, M.E.; Sree, S.; Billod, J.-M.; Engstrand, L.; Abrahamsson, S.; Langhammer, A.; Hveem, K.; et al. Cross-sectional associations between the gut microbe Ruminococcus gnavus and features of the metabolic syndrome: The HUNT study. Lancet Diabetes Endocrinol. 2022, 10, 481–483. [Google Scholar] [CrossRef]
  216. Goldstein, E.J.C.; Citron, D.M.; Peraino, V.A.; Cross, S.A. Desulfovibrio desulfuricans bacteremia and review of human Desulfovibrio infections. J. Clin. Microbiol. 2003, 41, 2752–2754. [Google Scholar] [CrossRef] [PubMed]
  217. Rowan, F.; Docherty, N.G.; Murphy, M.; Murphy, B.; Coffey, J.C.; O’connell, P.R. Desulfovibrio Bacterial Species Are Increased in Ulcerative Colitis. Dis. Colon Rectum 2010, 53, 1530–1536. [Google Scholar] [CrossRef] [PubMed]
  218. Verstreken, I.; Laleman, W.; Wauters, G.; Verhaegen, J. Desulfovibrio desulfuricans bacteremia in an immunocompromised host with a liver graft and ulcerative colitis. J. Clin. Microbiol. 2012, 50, 199–201. [Google Scholar] [CrossRef]
  219. Murros, K.E.; Huynh, V.A.; Takala, T.M.; Saris, P.E.J. Desulfovibrio Bacteria Are Associated With Parkinson’s Disease. Front. Cell. Infect. Microbiol. 2021, 11, 652617. [Google Scholar] [CrossRef] [PubMed]
  220. Lo Presti, A.; Zorzi, F.; Del Chierico, F.; Altomare, A.; Cocca, S.; Avola, A.; De Biasio, F.; Russo, A.; Cella, E.; Reddel, S.; et al. Fecal and Mucosal Microbiota Profiling in Irritable Bowel Syndrome and Inflammatory Bowel Disease. Front. Microbiol. 2019, 10, 1655. [Google Scholar] [CrossRef]
  221. Laval, L.; Martin, R.; Natividad, J.; Chain, F.; Miquel, S.; de Maredsous, C.D.; Capronnier, S.; Sokol, H.; Verdu, E.; Vlieg, J.v.H.; et al. Lactobacillus rhamnosus CNCM I-3690 and the commensal bacterium Faecalibacterium prausnitzii A2-165 exhibit similar protective effects to induced barrier hyper-permeability in mice. Gut Microbes 2015, 6, 1–9. [Google Scholar] [CrossRef]
  222. Jang, H.R.; Park, H.-J.; Kang, D.; Chung, H.; Nam, M.H.; Lee, Y.; Park, J.-H.; Lee, H.-Y. A protective mechanism of probiotic Lactobacillus against hepatic steatosis via reducing host intestinal fatty acid absorption. Exp. Mol. Med. 2019, 51, 1–14. [Google Scholar] [CrossRef]
  223. Chung, H.-J.; Yu, J.G.; Lee, I.-A.; Liu, M.-J.; Shen, Y.-F.; Sharma, S.P.; Jamal, M.A.H.M.; Yoo, J.-H.; Kim, H.-J.; Hong, S.-T. Intestinal removal of free fatty acids from hosts by Lactobacilli for the treatment of obesity. FEBS Open Bio 2016, 6, 64–76. [Google Scholar] [CrossRef]
  224. Curciarello, R.; Canziani, K.E.; Salto, I.; Romero, E.B.; Rocca, A.; Doldan, I.; Peton, E.; Brayer, S.; Sambuelli, A.M.; Goncalves, S.; et al. Probiotic Lactobacilli Isolated from Kefir Promote Down-Regulation of Inflammatory Lamina Propria T Cells from Patients with Active IBD. Front. Pharmacol. 2021, 12, 658026. [Google Scholar] [CrossRef]
  225. Jeong, J.-J.; Park, H.J.; Cha, M.G.; Park, E.; Won, S.-M.; Ganesan, R.; Gupta, H.; Gebru, Y.A.; Sharma, S.P.; Lee, S.B.; et al. The Lactobacillus as a Probiotic: Focusing on Liver Diseases. Microorganisms 2022, 10, 288. [Google Scholar] [CrossRef]
  226. Varshney, R.; Mishra, R.; Das, N.; Sircar, D.; Roy, P. A comparative analysis of various flavonoids in the regulation of obesity and diabetes: An in vitro and in vivo study. J. Funct. Foods 2019, 59, 194–205. [Google Scholar] [CrossRef]
  227. Bertoia, M.L.; Rimm, E.B.; Mukamal, K.J.; Hu, F.B.; Willett, W.C.; Cassidy, A. Dietary flavonoid intake and weight maintenance: Three prospective cohorts of 124 086 US men and women followed for up to 24 years. BMJ 2016, 352, i17. [Google Scholar] [CrossRef]
  228. Vernarelli, J.A.; Lambert, J.D. Flavonoid intake is inversely associated with obesity and C-reactive protein, a marker for inflammation, in US adults. Nutr. Diabetes 2017, 7, e276. [Google Scholar] [CrossRef] [PubMed]
  229. Hoek-van den Hil, E.F.; van Schothorst, E.M.; van der Stelt, I.; Swarts, H.J.M.; van Vliet, M.; Amolo, T.; Vervoort, J.J.M.; Venema, D.; Hollman, P.C.H.; Rietjens, I.M.C.M.; et al. Direct comparison of metabolic health effects of the flavonoids quercetin, hesperetin, epicatechin, apigenin and anthocyanins in high-fat-diet-fed mice. Genes Nutr. 2015, 10, 23. [Google Scholar] [CrossRef] [PubMed]
  230. Hossain, M.K.; Dayem, A.A.; Han, J.; Yin, Y.; Kim, K.; Saha, S.K.; Yang, G.-M.; Choi, H.Y.; Cho, S.-G. Molecular mechanisms of the anti-obesity and anti-diabetic properties of flavonoids. Int. J. Mol. Sci. 2016, 17, 569. [Google Scholar] [CrossRef]
  231. Baky, M.H.; Elshahed, M.S.; Wessjohann, L.A.; Farag, M.A. Interactions between dietary flavonoids and the gut microbiome: A comprehensive review. Br. J. Nutr. 2021, 128, 577–591. [Google Scholar] [CrossRef] [PubMed]
  232. Pei, R.; Liu, X.; Bolling, B. Flavonoids and gut health. Curr. Opin. Biotechnol. 2020, 61, 153–159. [Google Scholar] [CrossRef]
  233. Wang, M.; Yu, F.; Zhang, Y.; Chang, W.; Zhou, M. The Effects and Mechanisms of Flavonoids on Cancer Prevention and Therapy: Focus on Gut Microbiota. Int. J. Biol. Sci. 2022, 18, 1451–1475. [Google Scholar] [CrossRef]
  234. Willing, B.P.; Dicksved, J.; Halfvarson, J.; Andersson, A.F.; Lucio, M.; Zheng, Z.; Järnerot, G.; Tysk, C.; Jansson, J.K.; Engstrand, L. A pyrosequencing study in twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes. Gastroenterology 2010, 139, 1844–1854.e1. [Google Scholar] [CrossRef]
  235. Lin, R.; Piao, M.; Song, Y. Dietary quercetin increases colonic microbial diversity and attenuates colitis severity in Citrobacter rodentium-infected mice. Front. Microbiol. 2019, 10, 1092. [Google Scholar] [CrossRef]
  236. Ju, S.; Ge, Y.; Li, P.; Tian, X.; Wang, H.; Zheng, X.; Ju, S. Dietary quercetin ameliorates experimental colitis in mouse by remodeling the function of colonic macrophages via a heme oxygenase-1-dependent pathway. Cell Cycle 2018, 17, 53–63. [Google Scholar] [CrossRef] [PubMed]
  237. Mudd, A.M.; Gu, T.; Munagala, R.; Jeyabalan, J.; Egilmez, N.K.; Gupta, R.C. Chemoprevention of Colorectal Cancer by Anthocyanidins and Mitigation of Metabolic Shifts Induced by Dysbiosis of the Gut MicrobiomeAnthos Prevent Metabolic Shifts from Microbiome and B [a] P. Cancer Prev. Res. 2020, 13, 41–52. [Google Scholar]
  238. Wu, J.; Tsai, M.; Lai, C.; Lo, C.; Ho, C.; Wang, Y.; Pan, M. Polymethoxyflavones prevent benzo[a]pyrene/dextran sodium sulfate-induced colorectal carcinogenesis through modulating xenobiotic metabolism and ameliorate autophagic defect in ICR mice. Int. J. Cancer 2018, 142, 1689–1701. [Google Scholar] [CrossRef] [PubMed]
  239. Wu, M.; Wu, Y.; Deng, B.; Li, J.; Cao, H.; Qu, Y.; Qian, X.; Zhong, G. Isoliquiritigenin decreases the incidence of colitis-associated colorectal cancer by modulating the intestinal microbiota. Oncotarget 2016, 7, 85318–85331. [Google Scholar] [CrossRef]
  240. Qi, J.; Yu, J.; Li, Y.; Luo, J.; Zhang, C.; Ou, S.; Zhang, G.; Yang, X.; Peng, X. Alternating consumption of β-glucan and quercetin reduces mortality in mice with colorectal cancer. Food Sci. Nutr. 2019, 7, 3273–3285. [Google Scholar] [CrossRef]
  241. Bian, S.; Wan, H.; Liao, X.; Wang, W. Inhibitory effects of apigenin on tumor carcinogenesis by altering the gut microbiota. Mediat. Inflamm. 2020, 2020, 7141970. [Google Scholar] [CrossRef]
  242. Mcfadden, R.M.T.; Larmonier, C.B.; Shehab, K.W.; Midura-Kiela, M.; Ramalingam, R.; Harrison, C.A.; Besselsen, D.G.; Chase, J.H.; Caporaso, J.G.; Jobin, C.; et al. The role of curcumin in modulating colonic microbiota during colitis and colon cancer prevention. Inflamm. Bowel Dis. 2015, 21, 2483–2494. [Google Scholar] [CrossRef] [PubMed]
  243. Chen, L.; Jiang, B.; Zhong, C.; Guo, J.; Zhang, L.; Mu, T.; Zhang, Q.; Bi, X. Chemoprevention of colorectal cancer by black raspberry anthocyanins involved the modulation of gut microbiota and SFRP2 demethylation. Carcinogenesis 2018, 39, 471–481. [Google Scholar] [CrossRef]
  244. Horiba, Y.; Yoshino, T.; Watanabe, K. Daisaikoto for menstrual pain: A lesson from a case with menstrual pain successfully treated with daisaikoto. Case Rep. Med. 2015, 2015, 929514. [Google Scholar] [CrossRef]
  245. Morita, S.; Sakamaki, A.; Koyama, K.; Shibata, O.; Owaki, T.; Oda, C.; Kimura, A.; Nakaya, T.; Ohbuchi, K.; Nahata, M.; et al. Daisaikoto improves fatty liver and obesity in melanocortin-4 receptor gene-deficient mice via the activation of brown adipose tissue. Sci. Rep. 2022, 12, 10105. [Google Scholar] [CrossRef]
  246. Qian, W.; Cai, X.; Zhang, X.; Wang, Y.; Qian, Q.; Hasegawa, J. Effect of daisaikoto on expressions of SIRT1 and NF-kappaB of diabetic fatty liver rats induced by high-fat diet and streptozotocin. Yonago Acta Medica 2016, 59, 149. [Google Scholar] [PubMed]
  247. Ishizawa, S.; Nishi, A.; Kaifuchi, N.; Shimobori, C.; Nahata, M.; Yamada, C.; Iizuka, S.; Ohbuchi, K.; Nishiyama, M.; Fujitsuka, N.; et al. Integrated analysis of effect of daisaikoto, a traditional Japanese medicine, on the metabolome and gut microbiome in a mouse model of nonalcoholic fatty liver disease. Gene 2022, 846, 146856. [Google Scholar] [CrossRef] [PubMed]
  248. Ahmed, N.; Mirshekar-Syahkal, B.; Kennish, L.; Karachalias, N.; Babaei-Jadidi, R.; Thornalley, P.J. Assay of advanced glycation endproducts in selected beverages and food by liquid chromatography with tandem mass spectrometric detection. Mol. Nutr. Food Res. 2005, 49, 691–699. [Google Scholar] [CrossRef] [PubMed]
  249. Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023. [Google Scholar] [CrossRef] [PubMed]
  250. Armougom, F.; Henry, M.; Vialettes, B.; Raccah, D.; Raoult, D. Monitoring bacterial community of human gut microbiota reveals an increase in lactobacillus in obese patients and methanogens in anorexic patients. PLoS ONE 2009, 4, e7125. [Google Scholar] [CrossRef] [PubMed]
  251. Turnbaugh, P.J.; Hamady, M.; Yatsunenko, T.; Cantarel, B.L.; Duncan, A.; Ley, R.E.; Sogin, M.L.; Jones, W.J.; Roe, B.A.; Affourtit, J.P.; et al. A core gut microbiome in obese and lean twins. Nature 2009, 457, 480–484. [Google Scholar] [CrossRef]
  252. Joossens, M.; Huys, G.; Cnockaert, M.; De Preter, V.; Verbeke, K.; Rutgeerts, P.; Vandamme, P.; Vermeire, S. Dysbiosis of the faecal microbiota in patients with Crohn’s disease and their unaffected relatives. Gut 2011, 60, 631–637. [Google Scholar] [CrossRef]
  253. Nishino, K.; Nishida, A.; Inoue, R.; Kawada, Y.; Ohno, M.; Sakai, S.; Inatomi, O.; Bamba, S.; Sugimoto, M.; Kawahara, M.; et al. Analysis of endoscopic brush samples identified mucosa-associated dysbiosis in inflammatory bowel disease. J. Gastroenterol. 2017, 53, 95–106. [Google Scholar] [CrossRef]
  254. Zhong, W.; Wang, H.; Yang, Y.; Zhang, Y.; Lai, H.; Cheng, Y.; Yu, H.; Feng, N.; Huang, R.; Liu, S.; et al. High-protein diet prevents fat mass increase after dieting by counteracting Lactobacillus-enhanced lipid absorption. Nat. Metab. 2022, 4, 1713–1731. [Google Scholar] [CrossRef]
  255. Mukhopadhya, I.; Hansen, R.; El-Omar, E.M.; Hold, G.L. IBD—What role do Proteobacteria play? Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 219–230. [Google Scholar] [CrossRef]
  256. Rizzatti, G.; Lopetuso, L.R.; Gibiino, G.; Binda, C.; Gasbarrini, A. Proteobacteria: A Common Factor in Human Diseases. BioMed. Res. Int. 2017, 2017, 9351507. [Google Scholar] [CrossRef] [PubMed]
  257. Lee, G.; You, H.J.; Bajaj, J.S.; Joo, S.K.; Yu, J.; Park, S.; Kang, H.; Park, J.H.; Kim, J.H.; Lee, D.H.; et al. Distinct signatures of gut microbiome and metabolites associated with significant fibrosis in non-obese NAFLD. Nat. Commun. 2020, 11, 4982. [Google Scholar] [CrossRef] [PubMed]
  258. Le Leu, R.K.; Hu, Y.; Brown, I.L.; Woodman, R.J.; Young, G.P. Synbiotic intervention of Bifidobacterium lactis and resistant starch protects against colorectal cancer development in rats. Carcinogenesis 2010, 31, 246–251. [Google Scholar] [CrossRef]
  259. Sekine, K.; Toida, T.; Saito, M.; Kuboyama, M.; Kawashima, T.; Hashimoto, Y. A new morphologically characterized cell wall preparation (whole peptidoglycan) from Bifidobacterium infantis with a higher efficacy on the regression of an established tumor in mice. Cancer Res. 1985, 45, 1300–1307. [Google Scholar] [PubMed]
  260. Pool-Zobel, B.; Neudecker, C.; Domizlaff, I.; Ji, S.; Schillinger, U.; Rumney, C.; Moretti, M.; Vilarini, I.; Scassellati-Sforzolini, R.; Rowland, I. Lactobacillus-and Bifidobacterium-mediated antigenotoxicity in the colon of rats. Nutr. Cancer 1996, 26, 365–380. [Google Scholar] [CrossRef]
  261. Tavan, E.; Cayuela, C.; Antoine, J.-M.; Cassand, P. Antimutagenic activities of various lactic acid bacteria against food mutagens: Heterocyclic amines. J. Dairy Res. 2002, 69, 335–341. [Google Scholar] [CrossRef]
  262. Dong, T.S.; Luu, K.; Lagishetty, V.; Sedighian, F.; Woo, S.-L.; Dreskin, B.W.; Katzka, W.; Chang, C.; Zhou, Y.; Arias-Jayo, N.; et al. The Intestinal Microbiome Predicts Weight Loss on a Calorie-Restricted Diet and Is Associated With Improved Hepatic Steatosis. Front. Nutr. 2021, 8, 718661. [Google Scholar] [CrossRef]
  263. Holt, K.E.; Wertheim, H.; Zadoks, R.N.; Baker, S.; Whitehouse, C.A.; Dance, D.; Jenney, A.; Connor, T.R.; Hsu, L.Y.; Severin, J. Genomic analysis of diversity, population structure, virulence, and antimicrobial resistance in Klebsiella pneumoniae, an urgent threat to public health. Proc. Natl. Acad. Sci. USA 2015, 112, E3574–E3581. [Google Scholar]
  264. Wand, M.E.; Baker, K.S.; Benthall, G.; McGregor, H.; McCowen, J.W.I.; Deheer-Graham, A.; Sutton, J.M. Characterization of pre-antibiotic era klebsiella pneumoniae isolates with respect to antibiotic/disinfectant susceptibility and virulence in galleria mellonella. Antimicrob. Agents Chemother. 2015, 59, 3966–3972. [Google Scholar] [CrossRef]
  265. Kot, B.; Piechota, M.; Szweda, P.; Mitrus, J.; Wicha, J.; Grużewska, A.; Witeska, M. Virulence analysis and antibiotic resistance of Klebsiella pneumoniae isolates from hospitalised patients in Poland. Sci. Rep. 2023, 13, 4448. [Google Scholar] [CrossRef]
  266. Chen, J.; Li, J.; Huang, F.; Fang, J.; Cao, Y.; Zhang, K.; Zhou, H.; Cai, J.; Cui, W.; Chen, C.; et al. Clinical characteristics, risk factors and outcomes of Klebsiella pneumoniae pneumonia developing secondary Klebsiella pneumoniae bloodstream infection. BMC Pulm. Med. 2023, 23, 102. [Google Scholar] [CrossRef]
  267. Verani, J.R.; Blau, D.M.; Gurley, E.S.; Akelo, V.; Assefa, N.; Baillie, V.; Bassat, Q.; Berhane, M.; Bunn, J.; Cossa, A.C.A.; et al. Child deaths caused by Klebsiella pneumoniae in sub-Saharan Africa and south Asia: A secondary analysis of Child Health and Mortality Prevention Surveillance (CHAMPS) data. Lancet Microbe 2024, 5, e131–e141. [Google Scholar] [CrossRef] [PubMed]
  268. Reininghaus, E.Z.; Platzer, M.; Kohlhammer-Dohr, A.; Hamm, C.; Mörkl, S.; Bengesser, S.A.; Fellendorf, F.T.; Lahousen-Luxenberger, T.; Leitner-Afschar, B.; Schöggl, H.; et al. PROVIT: Supplementary probiotic treatment and vitamin B7 in depression—A randomized controlled trial. Nutrients 2020, 12, 3422. [Google Scholar] [CrossRef] [PubMed]
  269. Valles-Colomer, M.; Falony, G.; Darzi, Y.; Tigchelaar, E.F.; Wang, J.; Tito, R.Y.; Schiweck, C.; Kurilshikov, A.; Joossens, M.; Wijmenga, C.; et al. The neuroactive potential of the human gut microbiota in quality of life and depression. Nat. Microbiol. 2019, 4, 623–632. [Google Scholar] [CrossRef] [PubMed]
  270. Keshavarzian, A.; Green, S.J.; Engen, P.A.; Voigt, R.M.; Naqib, A.; Forsyth, C.B.; Mutlu, E.; Shannon, K.M. Colonic bacterial composition in Parkinson’s disease. Mov. Disord. 2015, 30, 1351–1360. [Google Scholar] [CrossRef] [PubMed]
  271. Karlsson, C.L.J.; Önnerfält, 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 2012, 20, 2257–2261. [Google Scholar] [CrossRef] [PubMed]
  272. Gotelli, N.J.; Chao, A. Measuring and Estimating Species Richness, Species Diversity, and Biotic Similarity from Sampling Data. In Encyclopedia of Biodiversity; Elsevier: Oxford, UK, 2013; pp. 195–211. [Google Scholar] [CrossRef]
  273. Jian, C.; Silvestre, M.P.; Middleton, D.; Korpela, K.; Jalo, E.; Broderick, D.; de Vos, W.M.; Fogelholm, M.; Taylor, M.W.; Raben, A.; et al. Gut microbiota predicts body fat change following a low-energy diet: A PREVIEW intervention study. Genome Med. 2022, 14, 54. [Google Scholar] [CrossRef]
  274. Bernstein, C.N.; Forbes, J.D. Gut Microbiome in Inflammatory Bowel Disease and Other Chronic Immune-Mediated Inflammatory Diseases. Inflamm. Intest. Dis. 2017, 2, 116–123. [Google Scholar] [CrossRef]
  275. Gionchetti, P.; Rizzello, F.; Venturi, A.; Campieri, M. Probiotics in infective diarrhoea and inflammatory bowel diseases. J. Gastroenterol. Hepatol. 2000, 15, 489–493. [Google Scholar] [CrossRef]
  276. Venturi, A.; Gionchetti, P.; Rizzello, F.; Johansson, R.; Zucconi, E.; Brigidi, P.; Matteuzzi, D.; Campieri, M. Impact on the composition of the faecal flora by a new probiotic preparation: Preliminary data on maintenance treatment of patients with ulcerative colitis. Aliment. Pharmacol. Ther. 1999, 13, 1103–1108. [Google Scholar] [CrossRef]
  277. Singh, S.; Bhatia, R.; Khare, P.; Rajarammohan, S.; Bishnoi, M.; Bhadada, S.K.; Sharma, S.S.; Kaur, J.; Kondepudi, K.K. Anti-inflammatory Bifidobacterium strains prevent dextran sodium sulfate induced colitis and associated gut microbial dysbiosis in mice. Sci. Rep. 2020, 10, 18597. [Google Scholar] [CrossRef] [PubMed]
  278. Yao, S.; Zhao, Z.; Wang, W.; Liu, X. Bifidobacterium Longum: Protection against Inflammatory Bowel Disease. J. Immunol. Res. 2021, 2021, 18597. [Google Scholar] [CrossRef] [PubMed]
  279. Dong, Y.; Liao, W.; Tang, J.; Fei, T.; Gai, Z.; Han, M. Bifidobacterium BLa80 mitigates colitis by altering gut microbiota and alleviating inflammation. AMB Express 2022, 12, 67. [Google Scholar] [CrossRef] [PubMed]
  280. Luis, A.S.; Hansson, G.C. Intestinal mucus and their glycans: A habitat for thriving microbiota. Cell Host Microbe 2023, 31, 1087–1100. [Google Scholar] [CrossRef]
  281. Depommier, C.; Everard, A.; Druart, C.; Plovier, H.; Van Hul, M.; Vieira-Silva, S.; Falony, G.; Raes, J.; Maiter, D.; Delzenne, N.M.; et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study. Nat. Med. 2019, 25, 1096–1103. [Google Scholar] [CrossRef]
  282. Chelakkot, C.; Choi, Y.; Kim, D.-K.; Park, H.T.; Ghim, J.; Kwon, Y.; Jeon, J.; Kim, M.-S.; Jee, Y.-K.; Gho, Y.S.; et al. Akkermansia muciniphila-derived extracellular vesicles influence gut permeability through the regulation of tight junctions. Exp. Mol. Med. 2018, 50, e450. [Google Scholar] [CrossRef]
  283. Dao, M.C.; Everard, A.; Aron-Wisnewsky, J.; Sokolovska, N.; Prifti, E.; Verger, E.O.; Kayser, B.D.; Levenez, F.; Chilloux, J.; Hoyles, L.; et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: Relationship with gut microbiome richness and ecology. Gut 2016, 65, 426–436. [Google Scholar] [CrossRef]
  284. Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Guiot, Y.; Derrien, M.; Muccioli, G.G.; Delzenne, N.M.; et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071. [Google Scholar] [CrossRef]
  285. Alcazar, M.; Escribano, J.; Ferré, N.; Closa-Monasterolo, R.; Selma-Royo, M.; Feliu, A.; Castillejo, G.; Luque, V.; Feliu-Rovira, A.; Muñoz-Hernando, J.; et al. Gut microbiota is associated with metabolic health in children with obesity. Clin. Nutr. 2022, 41, 1680–1688. [Google Scholar] [CrossRef]
  286. Luo, Y.; Lan, C.; Li, H.; Ouyang, Q.; Kong, F.; Wu, A.; Ren, Z.; Tian, G.; Cai, J.; Yu, B.; et al. Rational consideration of Akkermansia muciniphila targeting intestinal health: Advantages and challenges. NPJ Biofilms Microbiomes 2022, 8, 81. [Google Scholar] [CrossRef]
  287. Leenaars, C.H.C.; Kouwenaar, C.; Stafleu, F.R.; Bleich, A.; Ritskes-Hoitinga, M.; De Vries, R.B.M.; Meijboom, F.L.B. Animal to human translation: A systematic scoping review of reported concordance rates. J. Transl. Med. 2019, 17, 223. [Google Scholar] [CrossRef] [PubMed]
  288. Nagpal, R.; Wang, S.; Solberg Woods, L.C.; Seshie, O.; Chung, S.T.; Shively, C.A.; Register, T.C.; Craft, S.; McClain, D.A.; Yadav, H. Comparative Microbiome Signatures and Short-Chain Fatty Acids in Mouse, Rat, Non-human Primate, and Human Feces. Front. Microbiol. 2018, 9, 2897. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of effects of yoyo dieting and post-obesity weight loss on gut health. Yoyo dieting refers to repeated phases of dieting and non-dieting leading to cyclic weight loss and regain. There has been growing evidence from animal studies suggesting that yoyo dieting is associated with increased susceptibility to weight regain, gut dysbiosis, and gut inflammation. The current literature also suggests that diet-induced weight loss is a potential approach to reduce gut inflammation. However, post-obesity weight loss in humans has also been associated with increased gut dysbiosis. ↑: increase; ↓: decrease.
Figure 1. Overview of effects of yoyo dieting and post-obesity weight loss on gut health. Yoyo dieting refers to repeated phases of dieting and non-dieting leading to cyclic weight loss and regain. There has been growing evidence from animal studies suggesting that yoyo dieting is associated with increased susceptibility to weight regain, gut dysbiosis, and gut inflammation. The current literature also suggests that diet-induced weight loss is a potential approach to reduce gut inflammation. However, post-obesity weight loss in humans has also been associated with increased gut dysbiosis. ↑: increase; ↓: decrease.
Nutrients 16 03170 g001
Table 1. Key animal studies exploring how yoyo dieting and weight loss interventions during yoyo dieting affect the gut microbiome. ↑: increase; ↓: decrease.
Table 1. Key animal studies exploring how yoyo dieting and weight loss interventions during yoyo dieting affect the gut microbiome. ↑: increase; ↓: decrease.
Overall AimsModelDiet RegimeMicrobiota Changes and Metabolic OutcomesReference
Yoyo dieting changes microbiota composition and the administration of flavonoids might be beneficial in reduced weight regain Male C57BL/6 miceHFD → LFD → HFD → LFDYoyo mice:
  • Different microbiota composition compared to mice fed a control diet or an HFD.
  • ↓ alpha diversity compared to control mice.
  • ↓ relative abundance of Christensenella spp. and Lactobacillus reuteri during obesity and after obesity.
Yoyo mice treated with flavonoids:
  • No effect on gut microbiota composition
  • ↓ weight regain compared to yoyo mice.
  • ↑ expression of UCP1 in brown adipose tissue.
[61]
The effect of yoyo dieting and the use of daisaikoto Female C57BL/6 miceYoyo dieting only:
HFD → LFD → HFD
Yoyo dieting with daisaikoto supplementation:
HFD → LFD + daisaikoto → HFD
Yoyo mice:
  • ↓ alpha diversity compared to control mice.
  • Similar relative abundances of Bacteroidetes and Firmicutes compared to control mice.
  • ↑ relative abundance of Rminococcus and Desulfovibrio compared to control mice.
Yoyo mice fed daisaikoto:
  • ↓ alpha diversity compared to control mice.
  • ↑ alpha diversity compared to yoyo mice.
  • ↑ relative abundance of Bacteroidetes and ↓ relative abundance of Firmicutes compared to yoyo mice.
  • Similar relative abundance of Ruminococcus and Desulfovibrio compared to control mice.
  • ↓ relative abundance of Ruminococcus and Desulfovibrio compared to yoyo mice.
  • ↓ body weight, visceral fat, and weight regain compared to yoyo mice.
[196]
Long term effect of yoyo dieting on faecal microbiotaMale C57BL/6 mice(1) HFD → LFD → HFD → LFD → HFD → LFD → HFD → LFD → HFD → LFD
or
(2) LFD → HFD → LFD → HFD → LFD → HFD → LFD → HFD → LFD → HFD
Yoyo mice a fed a yoyo diet ending with a LFD:
  • No significant differences in alpha and beta diversity compared to control mice.
  • ↑ relative abundance of Bacteroidetes and Lactobacillus compared to obese mice and yoyo mice ending with a LFD.
  • ↓ body weight at the end of the dietary intervention compared to yoyo mice fed a LFD/HFD cyclic diet.
Yoyo mice fed a yoyo diet ending with a HFD:
  • No significant differences in alpha and beta diversity compared to obese mice.
  • ↑ relative abundance of Firmicutes (classes Erysipelotrichia and Bacilli) compared to control mice and yoyo mice ending with a LFD.
  • ↑ body weight at the end of the dietary intervention compared to yoyo mice fed a HFD/LFD cyclic diet.
[62]
Table 2. Key human studies exploring how post-obesity weight loss affects the gut microbiome. ↑: increase; ↓: decrease.
Table 2. Key human studies exploring how post-obesity weight loss affects the gut microbiome. ↑: increase; ↓: decrease.
Overall AimsModelDiet RegimeMicrobiota ChangesReference
Examining effects of one-month caloric restriction on gut permeability and microbiome in female individuals with obesityFemale individuals with obesity1-month low-calorie diet
  • No change in alpha and beta diversities compared to controls.
  • ↓ relative abundance of Proteobacteria.
  • ↑ relative abundance of Ruminococcus faeces and Bifidobacterium sp.
[114]
The intestinal microbiome predicts weight loss on a calorie-restricted diet in overweight and obesity individualsMale and female individuals with overweight/obesity16-week macronutrient standardised diet for weight lossNo gut microbiota difference at baseline. Individuals successfully lost at least 5% body weight:
  • ↓ relative abundance of Enterococcus.
  • ↓ relative abundance of Klebsiella, Megasphaera, Sellimonas, and Lactobacillus compared to those lost <5% body weight.
Individuals lost <5% body weight:
  • ↓ relative abundance of Klebsiella.
  • ↑ relative abundance of Coprococcus and Collinsella.
[262]
A PREVIEW intervention study investigating the effect of low-energy diets on gut microbiome of individuals with overweight, or obesity and prediabetesMale and female individuals with overweight/obesity and prediabetes8-week low-energy dietIndividuals successfully lost at least 8% body weight
  • ↑ microbiota richness and alpha diversity.
  • ↑ inter-individual Bray-Curtis values.
  • ↓ Firmicutes:Bacteroidetes ratio.
  • ↓ relative abundance of Pseudobutyrivibrio and Bifidobacterium.
  • ↑ relative abundance of Akkermansia.
[273]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Phuong-Nguyen, K.; McGee, S.L.; Aston-Mourney, K.; Mcneill, B.A.; Mahmood, M.Q.; Rivera, L.R. Yoyo Dieting, Post-Obesity Weight Loss, and Their Relationship with Gut Health. Nutrients 2024, 16, 3170. https://doi.org/10.3390/nu16183170

AMA Style

Phuong-Nguyen K, McGee SL, Aston-Mourney K, Mcneill BA, Mahmood MQ, Rivera LR. Yoyo Dieting, Post-Obesity Weight Loss, and Their Relationship with Gut Health. Nutrients. 2024; 16(18):3170. https://doi.org/10.3390/nu16183170

Chicago/Turabian Style

Phuong-Nguyen, Kate, Sean L. McGee, Kathryn Aston-Mourney, Bryony A. Mcneill, Malik Q. Mahmood, and Leni R. Rivera. 2024. "Yoyo Dieting, Post-Obesity Weight Loss, and Their Relationship with Gut Health" Nutrients 16, no. 18: 3170. https://doi.org/10.3390/nu16183170

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

Article Metrics

Back to TopTop