Abstract
Background
Bariatric or weight loss surgery is currently the most effective treatment for obesity and metabolic disease. Unlike dieting and pharmacology, its beneficial effects are sustained over decades in most patients, and mortality is among the lowest for major surgery. Because there are not nearly enough surgeons to implement bariatric surgery on a global scale, intensive research efforts have begun to identify its mechanisms of action on a molecular level in order to replace surgery with targeted behavioral or pharmacological treatments. To date, however, there is no consensus as to the critical mechanisms involved.
Scope of review
The purpose of this non-systematic review is to evaluate the existing evidence for specific molecular and inter-organ signaling pathways that play major roles in bariatric surgery-induced weight loss and metabolic benefits, with a focus on Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG), in both humans and rodents.
Major conclusions
Gut-brain communication and its brain targets of food intake control and energy balance regulation are complex and redundant. Although the relatively young science of bariatric surgery has generated a number of hypotheses, no clear and unique mechanism has yet emerged. It seems increasingly likely that the broad physiological and behavioral effects produced by bariatric surgery do not involve a single mechanism, but rather multiple signaling pathways. Besides a need to improve and better validate surgeries in animals, advanced techniques, including inducible, tissue-specific knockout models, and the use of humanized physiological traits will be necessary. State-of-the-art genetically-guided neural identification techniques should be used to more selectively manipulate function-specific pathways.
Keywords: Gut-brain communication, Neural controls of food intake, Energy balance regulation, Gut hormones, Bile acid signaling, Gut microbiome
Highlights
-
•
Bariatric surgery-induced weight loss is key for its major beneficial effects.
-
•
Partial protection from adaptive responses to weight loss such as increased hunger and reduced metabolism is important.
-
•
Identification of the molecular mechanisms enabling this protection is crucial.
-
•
Multiple parallel, rather than a single mechanism(s) seems responsible for the many beneficial effects.
-
•
Better validated animal models and interventional clinical studies will be crucial to identify the underlying mechanisms.
1. Introduction
The prevalence of obesity is at an alarming level globally (WHO fact sheet on obesity, https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight) and no easy cure is in sight. Behavioral changes and pharmacological therapies can be quite effective, and recent insights into mechanisms that sustain behavior change [1], as well as new drugs currently under evaluation [2], are promising (Figure 1). However, drug treatments often have significant side effects and the beneficial effects of drugs last only as long as they are administered. It is certainly not sensible to expect children or adolescents with obesity to take medication for the rest of their lives.
Figure 1.
Comparative effectiveness of obesity treatments over time. Expected average changes in body weight and BMI of hypothetical male and female obese patients at age 40 over 20 years either untreated or treated with standard behavioral techniques and lifestyle changes (light green) in combination with standard anti-obesity drugs (red), hypocaloric diets (green), glucagon-like peptide receptor agonists (GLP1RAs, purple), combination pharmacotherapy (blue), or bariatric surgery (black). Based on non-systematically selected, mostly RCT studies. Note that except for bariatric surgery, most studies were limited to less than 3 years follow-up and typically show weight regain after cessation of interventions.
Bariatric surgery is an attractive alternative, as the procedure is fast and relatively simple with low mortality, and substantial weight loss is typically sustained for many years, if not decades. While bariatric surgery clearly benefits adults with severe obesity, there are potential risks of serious complications and side effects, and some patients regain most or all of the lost weight [[3], [4], [5]]. Furthermore, there are not nearly enough bariatric surgeons to implement it on a global scale. Therefore, it is imperative to understand the mechanisms behind the beneficial effects of bariatric surgery, with an eye towards non-operative treatments that recapitulate these beneficial effects. Although great progress has been made during the last 15 years towards this goal, there is no “smoking gun” mechanism and we still do not have a clear picture of “how” and “why” surgery works so well for obesity when other therapies do not.
The purpose of this non-systematic review is to evaluate the existing evidence for specific molecular and inter-organ signaling pathways that play major roles in bariatric surgery-induced weight loss and metabolic benefits. Such mechanistic insights should provide new impulses towards a more comprehensive and integrative view of body weight regulation and hopefully inform the future development of alternative pharmacological and behavioral therapies. We will explore the many facets and aspects of the controls of food intake and regulation of body weight that are primarily centered on gut-brain communication. The review will mainly focus on Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG), in both humans and rodents.
2. Bariatric surgery types and their effects on body weight in humans and rodent models
Among the common bariatric operations, RYGB is most effective in producing a sustained lowering of body weight and adiposity (Table 1). Many, but not all, human studies find VSG to be only slightly less effective [6]. However, mouse models clearly demonstrate the superiority of RYGB over VSG, with VSG mice re-gaining significant amounts of body weight over the first 10 weeks after surgery (the equivalent of about 5 years of human life) [7,8]. In addition, after a nadir at approximately 1 year after surgery, body weight curves in humans with VSG also show a slow but steady trend towards regain [6]. Because the VSG is a relatively newer bariatric operation compared to the RYGB, it remains to be seen what degree of weight regain humans will experience with VSG since mouse VSG models typically demonstrate complete regain of lost weight over time.
Table 1.
Percent total weight loss after RYGB or VSG in humans and diet-induced obese rodents. For human studies, only representative RCTs in male and female patients with obesity, lasting at least 5 years, and reporting total percent weight loss are included. Standard deviation or 95% confidence intervals are reported depending on availability. For rodents, only studies in diet-induced (either 45% or 60% high-fat diet exposure for various time periods) obese male rats or mice and lasting at least 8 weeks are included. Percent weight loss at specific time points in rodent studies was extrapolated from published body weight curves and are estimates only. For some rodent studies difference of final body weight after surgery vs sham surgery is indicated in square brackets. Numbers in parentheses depict the number of subjects. Note that in rodent studies, different starting body weights, housing methods (e.g. single housing), immediate postsurgical regimens, and experimental loads (e.g. frequent blood sampling) may have influenced surgery-induced weight loss. Based on average life expectancy, 2 and 10 weeks of rodent life can be considered as the approximate equivalent of 1 and 5 years of human life.
RYGB | VSG | ||||
---|---|---|---|---|---|
Human | 1 year | 5–12 years | 1 year | 5–10 years | |
Schauer et al. [6] | −29.4 ± 8.9 (50) −10.2 BMI |
−23.2 ± 9.6 (50) 5 y −8.1 BMI |
−25.1 ± 8.5 (50) −8.9 BMI |
−18.6 ± 7.5 (50) −6.7 BMI 5 y |
|
Salminen et al. [279] | −30.6 (120) | −26.9 (120) 5 y | −27.3 (120) | −22.9 (120) 5 y | |
Arterburn et al. [3] | −31.2 (32,208) | −25.5 (32,208) 5 y | −25.2 (29,693) | −18.8 (29,693) 5 y |
|
Arterburn et al. [280] | −28.4 (17,258) | −21.7 (17,258) 5 y | −23.0 (13,900) | −16.0 (13,900) 10 y |
|
Courcoulas et al. [281] | −33.8 [28.3–39.8] (50) | −28.4 [27.6–29.2] (50) 10 y | |||
Adams et al. [15] | −26.9 [-28.2 to −25.6] (387) 12 y | ||||
Rat | 2 weeks | 10–20 weeks | 2 weeks | 10–20 weeks | |
Stylopoulos et al. [282] | −19.9 (15) | −12.0 (15) 20w [-21.2] | Stefater et al. [119] | −19.0 (9) | +19.8 (9) 17w [-9.8] |
Zheng et al. [27] | −18.2 (13) | −16.5 (10) 20w [-24.5] | Alvarez et al. [283] | −15.0 (13) | ±0.0 (12) 10w [-10.0] |
Abegg et al. [284] | −20.5 (7) | −10.2 (7) 10w [-34.2] | Basso et al. [285] | −14.3 (21) | +18.0 (21) 10w [-1.3] |
Thanos et al. [286] | −15.0 (11) | −19.4 (11) 10w [-24.0} | Mumphrey et al. [37] | −18.5 (8) | −6.0 (8) 10w [-13.6] |
Mouse | |||||
Hatoum et al. [259] | −34.6 (5) | −17.9 (5) 9w [-34.5] | Ryan et al. [243] | −26.3 (8) | ±0.0 (8) 10w [-11.6] |
Uchida et al. [287] | −28.3 (16) | −21.7 (16) 5w [-24.5] | Ding et al. [288] | −26.6 (9) | −14.9 (9) 10w [-21.2] |
Flynn et al. [289] | −28.0 (7) | −29.0 (7) 8w [-41.3] | Du et al. [290] | −24.5 (5) | −10.2 (5) 10w [-20.0] |
Stevenson et al. [8] | −24.4 (6) | −25.6 (6) 10w [-41.6] | −21.7 (6) | −15.2 (6) 10w [+5.0] | |
Hao et al. [7] | −22.5 (9) | −22.5 (9) 10w [-39.8] | −5.0 (9) | +10.5 (9) 10w [-18.3] |
One reason for weight regain appears to be the fact that bariatric surgery patients typically remain in the obesogenic environment that is conducive for weight gain in the first place. Given that this environment is the main cause of obesity in a majority of humans, bariatric surgery is only a symptomatic treatment but does not attack the root cause of obesity, which would require removal or neutralization of the obesogenic environment. However, the sustained effects on body weight after bariatric surgery are nothing but remarkable, considering that similar weight loss induced by dieting provokes very strong adaptive and counter-regulatory responses such as increased hunger and reduced metabolism [9,10]. Such counter-regulatory responses are by and large absent in bariatric surgery patients. These patients are simply not as hungry as before, are satiated by much less than prior to surgery, and eat less [[11], [12], [13]], and there is evidence from rodent models that the expected hypo-metabolic response is abrogated [7,14]. It could thus be said that bariatric surgery makes it easier for patients to cope with the hostile obesogenic environment. As indicated by the large variability in weight loss (e.g. for RYGB see Figure 2 in [15]), surgery is not effective in every individual. While the reasons for suboptimal outcomes of bariatric surgery are not clear, the degree of weight loss can be influenced by pre-surgical physiology and behavior as well as genetic background of the patient, skills and techniques of the surgeon, intensity of peri-operative coaching, and the post-surgical environment.
Figure 2.
Schematic diagram depicting the principal pathways and mechanisms potentially involved in the effects of bariatric surgery on body weight and diabetes resolution. 1) Changes in energy metabolism and expenditure are determined by fecal energy loss, gut metabolic demands, and energy used by muscle, brown fat, and other tissues. 2) The altered gut generates changes in circulating hormones and other molecules as well as changes in neural signals carried by primary afferent neurons of vagal and dorsal root ganglia origin that affect all other organs including the brain. 3) In turn, the brain orchestrates changes in behavioral output including changes in energy intake and food choice, which affect both energy balance and gut luminal environment. 4) Changes in autonomic and endocrine outflow from the brain can affect all organs including the gut.
3. Bariatric surgery changes gut-brain communication
Gut-brain communication (or the gut-brain axis) is increasingly implicated not only in functional gastrointestinal disorders [16], but also in obesity and metabolic diseases [17,18], and a number of neuro-psychiatric disorders (for recent reviews see: [[19], [20], [21]]). Besides absorbed nutrients, metabolites, and hormones, signals generated by gut microbes have attracted much of the recent attention to the gut-brain axis. How surgery changes gut-brain communication is poorly described, despite intensive investigation into how differences in calorie intake, nutrient exposure and nutrient absorption are altered by surgery.
Excessive calorie intake has been recognized as the main cause of obesity for decades [22,23], and therefore bariatric surgery was initially conceived by surgeons to both restrict calories consumed and decrease calories absorbed [[24], [25], [26]]. During millions of years in a rather restrictive environment, the mammalian gut has evolved to very efficiently process and absorb nutrients, including a very large absorptive surface. Reducing ingestive and absorptive capacity to offset the modern environment of plenty seemed like a plausible approach.
After years of intensive research, it is clear that both of these objectives are achieved at least to some extent. Most modern surgery types are restrictive in the sense that large meals are avoided [12,27], malabsorptive, or both [11,28,29]. Biliopancreatic diversion, the most effective operation for weight reduction, produces the greatest degree of malabsorption [30]. In addition, malabsorption of micronutrients such as iron often necessitate lifelong supplementation [31,32]. However, at least for RYGB and VSG, the effects on macronutrient malabsorption are generally small and cannot fully explain the large and sustained weight loss. It is clear that these bariatric operations go far beyond mechanical restriction and malabsorption and likely impinge on normal physiological mechanisms affecting energy intake, expenditure, and metabolic regulation.
Of the approximately 50,000 studies with bariatric surgery as the search term, the overwhelming majority is strictly descriptive, reporting on changes of structure or function of almost every organ and tissue in the body over time after surgery. They span the entire range from molecular changes in the gut itself to changes in brain and behavior. Many of these changes are either non-specific effects of invasive surgery or secondary to the drastic reduction of food intake and body weight and may not reveal potential mechanisms. The more interesting changes are unique to surgery, in that they are not replicated in controls subjected to similar levels of acute hypophagia and chronic weight loss. The fact that some changes appear rapidly (within a few days) after surgery is often taken as evidence that they are not dependent on weight loss, but the hypocaloric state early after surgery greatly improves many metabolic derangements in obesity before significant weight loss occurs (for a more thorough discussion see section 5), and it is therefore important to control for both hypo-nutrition and weight loss. It is also important to realize that appropriate controls such as sham surgery are not possible in human studies. Even in rodents, true sham surgery controls are not feasible, as they would require careful re-anastomosis of all cuts made, including functional reconnection of cut nerve fibers, an impossible demand. It is also clear that the changes after bariatric surgery are dynamic, with some occurring within a few hours and others becoming obvious much later, and it is therefore important to clearly state the time after surgery when a change occurs.
The key inter-organ pathways and fundamental energy-balance mechanisms likely involved in the beneficial effects of bariatric surgery on body weight and diabetes resolution are shown in Figure 2. As for any other successful obesity-treatment, bariatric surgery must either decrease energy intake and/or increase total energy expenditure (including any energy lost in the stool). For changes in food intake, food choice, and other behaviors, the brain is the final arbitrator, while for changes in energy expenditure all metabolically active tissues and organs are responsible, although the brain may modulate this capacity via endocrine and autonomic outflow. Therefore, understanding how bariatric surgery affects gut-brain communication is likely to hold the key to unravel its mechanisms.
3.1. Changes within the gut
3.1.1. Gut hypertrophy and L-cell hyperplasia
In humans and rats, RYGB leads to hypertrophy of the nutrient perfused limbs (Roux and common) of the small intestine [[33], [34], [35], [36], [37], [38], [39], [40]], with increased thickness and mass of all layers and a concomitant increase in the number of L- and other enteroendocrine cells [34,35,37]. In rats, hypertrophy is observed as soon as 2 weeks after RYGB [34,40], and after 10 months embraces both the mucosa and external muscle layers [38]. Similarly, total gut weight and length are significantly increased 20 weeks after RYGB in male mice [41], and 40 weeks after RYGB in juvenile mice that prevented weight gain on a high-fat diet in both male and female mice [42]. In contrast to RYGB, none of these changes were observed after VSG in humans and rats [34,37], except for increased numbers of L-cells and increased mRNA expression of GLP-1 [34].
Intestinal adaptation and regrowth are commonly observed after intestinal resection due to Crohn's disease or necrotizing enterocolitis, and considerable efforts have been put into research aimed at finding ways to accelerate regrowth [[43], [44], [45]]. GLP-2, insulin-like growth factor-1 (IGF-1), and epidermal growth factor (EGF) have been found to be part of intestinal growth mechanisms in rodent models of short bowel syndrome (SBS) [[46], [47], [48], [49], [50]] and RYGB [40]. Furthermore, GLP2R-signaling is pivotal for the adaptive response after chemotherapy-induced intestinal injury [51], and long-acting GLP-2 receptor agonists are currently used to treat SBS in humans [52]. However, the critical triggers for intestinal regrowth, such as the key ‘ingredients’ of the changed luminal contents after bariatric surgery and the respective roles of hormonal and paracrine signaling are not known. It is not surprising that VSG was effective in GLP2R-deficient mice [53], as there is little evidence of gut hypertrophy after VSG. It should be interesting, however, to study the effects of intestine-specific GLP-2 receptor deletion on gut hypertrophy and weight loss following RYGB and whether the different effectiveness between RYGB and VSG can be explained by differential gut hypertrophy.
It is also unclear what factors are responsible for the greatly increased circulating levels of GLP-1, GLP-2, and PYY which are present as early as a few days after both RYGB and VSG; and last for months [33,34,40,[54], [55], [56], [57], [58]]. The increased number of L-cells may be important for RYGB, and increased stimulation may be important after VSG [34].
3.1.2. Metabolic reprogramming
In rats and humans, Roux limb blood glucose uptake is increased after RYGB as measured with 2-deoxyglucose PET imaging [34,39,59], and this increased uptake is associated with increased expression of glucose-transporter-1 (GLUT1) and hexokinase-II (HK-II), the rate-limiting enzyme for glycolysis [39]. Interestingly, this altered GLUT1 not only occurs on the basolateral membrane but the apical membrane as well [34]. This metabolic adaptation or reprogramming appears to be triggered by the sudden exposure to undigested food, as demonstrated by surgically inserting a piece of jejunum between the esophagus and stomach with otherwise unchanged nutrient flow [39], and likely energetically supports the hypertrophic response described above. However, consistent with the absence of a hypertrophic response, there are no adaptive changes in glucose uptake after VSG [34,37]. Furthermore, ex vivo transport of luminal glucose was decreased after VSG [34], suggesting that metabolic reprogramming is different for RYGB and VSG.
Besides glucose metabolism, changes in lipid sensing, transport, and metabolism have also been reported. Twelve weeks after RYGB in high-fat diet-induced obese mice, mRNA expression of several fatty acid sensors in the colon including the GPR40, GPR41, GPR84, and GPR119, known to stimulate GLP-1 release, was selectively increased compared to both sham-operated and calorie-restricted mice [60]. In a rat model of RYGB with a lymph sampling fistula [61], expression of genes in enterocytes implicated in lipid transport and absorption during fasting, and postprandial triglyceride concentrations both in plasma and intestinal lymph were reduced two weeks after surgery [61].
Metabolic reprogramming is not limited to the gut but also occurs in other organs and tissues, as shown in a longitudinal study in rats undergoing RYGB or similar weight loss induced by caloric restriction as a control condition [14]. Notably, the beiging of subcutaneous adipose tissue, increased oxidative metabolism in slow twitch skeletal muscle, and an enhancement of amino acid catabolic processes in the liver occurs in a dynamic fashion and is associated with hypoxia-inducible factor-1 activation and repression of growth hormone receptor signaling [14].
As pointed out by some investigators, all this reprogramming may potentially explain much of the beneficial effects of bariatric surgery [14,39,60,61]. However, the requirement of any of the hypothesized mechanisms and pathways has not been directly tested in mechanistic studies.
3.1.3. Changes in the gut microbiome
There is an enormous interest in the role of gut microbiota in the pathogenesis of obesity and therapeutic modification of the microbiome (for recent critical reviews see [[62], [63], [64]], but randomized interventional studies are rare. The most persuasive studies with fecal matter transplantation (FMT) in subjects with obesity have shown very modest and mixed outcomes [[65], [66], [67], [68]] and the same is true for studies manipulating the gut microbiome with dietary pre, pro, and synbiotics [[69], [70], [71], [72], [73]].
Not surprisingly, bariatric surgery changes the gut microbiome [[74], [75], [76], [77], [78], [79], [80], [81], [82]]. In a direct comparison of weight loss induced by RYGB vs caloric restriction, the gut microbiome changes were much greater in persons in the surgery group [82]. The more interesting question is whether this change is to any degree causal in the beneficial effects of bariatric surgery [[83], [84], [85], [86]], and the available evidence for a mechanistic role of microbiota is discussed below in section 4.1.3.
3.2. Changes in gut-brain signaling pathways
3.2.1. Circulating factors
As already elaborated above, a number of circulating factors are significantly changed after bariatric surgery. These prominently include the L-cell gut hormones GLP-1, GLP-2, and PYY, which are greatly elevated, but may have limited effects past the liver because of rapid degradation by dipeptidyl peptidase-IV. In addition, serum global metabolomics analyses have identified a great number of metabolites that are changed after bariatric surgery [74,87]. These changes are dynamic [[88], [89], [90]], they vary for different types of surgery [80,91,92], are different for hepatic portal vs. peripheral venous blood [93], are partly independent of weight loss [94], and some of the changed metabolites are associated with the degree of post-surgical metabolic improvements [74,95]. The most prominent among these are changes in the serum bile acid profile and concentrations (see below for a more detailed discussion), lipids, amino acids and their metabolites, microbial metabolites, and circulating markers of inflammation [74,88,89,91]. These circulating molecules have the potential to act in all other organs, including the brain, and contribute to the beneficial effects of bariatric surgery. However, humoral communication is generally non-discrete and relatively slow.
It is important to note that artificially increasing the rate of intestinal nutrient delivery in non-surgical patients can mimic some of these post-operative changes in GLP-1, glucose, and insulin [96]. Even though increased gastric emptying and intestinal nutrient delivery have been posited to explain at least some of the gut peptide effects following bariatric surgery, matched rates of intestinal nutrient delivery show augmented GLP-1 secretion in VSG mice compared to sham [97]. Thus, early and late changes in GLP-1 or other gut peptide secretion may change over time as the intestine adapts, revealing that the timing of measurements in experimental studies is critical. Rates of nutrient delivery may help to explain some of the gut hormonal changes apparent initially after bariatric surgery, though surgery is clearly associated with additional adaptations contributing to altered gut peptide responses that are likely important to driving weight loss and other metabolic effects of surgery.
3.2.2. Sensory and motor neurons of the vagus nerve
The vagus nerve is the principal pathway for discrete and rapid gut-brain communication. It should be noted that both VSG and RYGB operations by themselves are cutting many vagal afferent and efferent fibers (see Figure 4 in [98]. In the case of VSG, this damage is limited to the greater curvature of the stomach. The typical RYGB with a small gastric pouch eliminates most vagal fibers innervating the stomach, and in addition vagal afferent and efferent fibers innervating the pyloric sphincter, proximal duodenum, and parts of the pancreas, as some vagal branches gain access to these targets via stomach and pylorus [99]. However, with normal surgical care, vagal afferents and efferents innervating the distal small intestine and colon, including the Roux-limb, should remain intact in both human and rodent models of RYGB and VSG [98]. Therefore, vagal afferents are in a privileged position to mediate signals emanating from the drastically changed luminal, hormonal, and metabolic environment of the Roux and common limbs. This could be accomplished through the numerous receptors for gut hormones, nutrient metabolites, bile acids, microbial-derived molecules, and other factors [[100], [101], [102], [103]].
Figure 4.
Mechanisms of bile acid metabolism and signaling potentially involved in the effects of bariatric surgery. Left: Schematic diagram showing the major components of enterohepatic bile acid homeostasis. After synthesis from cholesterol in the liver and transport via the bile duct and gall bladder to the intestinal lumen, where they are mainly involved in lipid absorption, taurine and glycine-conjugated primary bile acids such as TCA and TCDCA are modified to secondary bile acids such as DCA and LCA by microbiota. Ninety percent of bile acids are then re-absorbed by enterocytes in the distal intestine via the apical sodium bile acid co-transporter (ASBT) and a small fraction is lost in the stool. Re-absorbed bile acids are then transported back to the liver to provide feedback control over bile acid synthesis together with the gut hormone FGF-19/15 which is stimulated via the nuclear farnesoid bile acid receptor (FXR) in enterocytes. Activation of the Takeda G-protein coupled receptor (TGR5) by bile acids stimulates the release of GLP-1 from enteroendocrine cells and activates vagal afferents. From the hepato-portal circulation, bile acids also gain access to the general circulation, where they interact with FXR and TGR5 in other organs and tissues, notably in brown fat tissue to generate heat, and in the brain, where they can affect food intake and autonomic/endocrine outflow. Right: After RYGB and following bile diversion to the distal ileum, total bile acids are increased and bile acid composition changes both intraluminal and in the plasma, due to changes in the luminal environment and microbiome. Increased re-uptake of bile acids such as β-muricholic acid (βMA), a potent FXR agonist, leads to increased GLP-1 release and improvement in insulin secretion. Increased bile acid concentrations in the lamina propria lead to increased activation of vagal afferents that suppress food intake. The effects of changes in systemic bile acid signaling after RYGB and bile diversion are not known.
High-fat diet-induced obesity has been shown to alter the structure and function of vagal afferents and efferents in rodents resulting in reduced satiety generation and decreased excitability of vagal motor neurons [[104], [105], [106], [107]], it is thus plausible that bariatric surgery may be effective because it reverses some of these impairments. In rats, VSG increased, but RYGB decreased the density of vagal afferent terminals in the nucleus tractus solitarius, and RYGB triggered rapid DNA fragmentation in vagal afferent neurons of the nodose ganglia [108] as well as microglia activation in the dorsal vagal complex [109], indicating neuronal damage. However, RYGB reversed the effects of obesity on the excitability of vagal motor neurons in the dorsal motor nucleus, without normalizing the morphological alterations [104].
3.2.3. Other pathways
Besides communication via vagal afferents, dorsal root afferents via spinal pathways could also play a role in the effects of bariatric surgery but this has not yet been addressed in the literature. Also, humoral communication between the gut and brain could be indirect via crosstalk with other organs (Figure 2).
3.3. Changes in brain function and behavior
We have previously reviewed in great detail the potential recruitment by bariatric surgery of the complex central neural mechanisms responsible for the control of food intake and energy balance regulation in both humans and animal models [110]. Much of the conclusions remain the same, and here we recapitulate a few important points and discuss newer literature.
Data from both humans and rodents show greatly decreased energy intake immediately after surgery, although some of this reduction is typically prescribed by special postsurgical diets. In humans after RYGB, energy intake is still strongly reduced by 20–50% after 2–3 years and by 20% at 10 years [12,13]. In rodents, energy intake returns much more quickly to pre-surgical levels and there is great variability in different studies. In our well-validated rat and mouse models of RYGB with low mortality and complications, energy intake (high-fat diet) returns to near normal 10 days after surgery and remains at that level for up to 10 months in rats [111] and 4 months in mice [112]. However, because up to 20% of ingested energy (mostly fat under high-fat diet conditions) is lost in the feces in both rats [111] and mice [78,113], metabolizable energy intake remains chronically suppressed after RYGB. In addition, rats and mice with RYGB do not exhibit the adaptive hyperphagic response to weight loss that is observed after similar weight loss induced by caloric restriction. These observations strongly suggest that the neural circuits responsible for homeostatic energy balance regulation or their inputs are changed after bariatric surgery in both humans and rodents.
3.3.1. Homeostatic energy balance circuitry
AGRP/NPY and POMC/CART neurons in the medial basal hypothalamus are considered a major hub for assessing fuel availability and regulating homeostatic energy balance and body weight [[114], [115], [116]]. Thus, several investigators have asked whether bariatric surgery impinges on this master regulator by measuring mRNA expression of these peptides in rodents. Given its hunger-reducing and satiety-promoting effects, bariatric surgery could be expected to suppress the activity of AGRP/NPY neurons and/or increase the activity of POMC/CART neurons, and this should be reflected in lower mRNA expression for AGRP/NPY and higher expression for POMC/CART. The outcomes were mixed, with changes in both directions [[117], [118], [119], [120]]. However, in a more recent study in which the control mice were body-weight matched through caloric restriction, we demonstrated that mice after RYGB did not show the strong increase in AGRP and NPY mRNA expression seen in controls [121], suggesting that this surgery effectively re-sets the defended body weight on a high-fat diet.
A brainstem circuit including the dorsal vagal complex, lateral parabrachial nucleus, and central nucleus of the amygdala, is intimately linked to the hypothalamus and considered an essential part of the homeostatic regulatory system [122,123] (Figure 3). Activation of this circuit has been demonstrated to strongly suppress food intake [124], and silencing some of its key neurons prevents the food intake-suppressive and satiety-promoting effects of gut-derived and other internal signals such as CCK, GLP-1, GDF15, and intestinal malaise [[125], [126], [127]]. Importantly, this “anorexia pathway” is strongly activated by the new obesity drug semaglutide [128]. The pathway is also strongly activated when a mouse with RYGB, but not a sham-operated mouse, eats food during the early post-surgical period [129]. We also found that silencing this pathway in mice with RYGB results in a transient increase in food intake and a modest reduction of weight loss (unpublished observations). However, GDF15 which also strongly activates this brainstem anorexia pathway is not necessary for the weight loss induced by VSG [126].
Figure 3.
Major candidate signaling mechanisms underlying weight loss-dependent and weight loss-independent beneficial effects of bariatric surgery. Mechanisms affecting body weight through changes in eating behavior and glucose homeostasis through central pathways are shown in yellow-brown. Mechanisms leading to metabolic improvements independent of body weight/adiposity are shown in green. Framing of signaling molecules indicates that their role has been directly tested in transgenic mouse models, or with chronic pharmacological or surgical blockade. Yellow frames indicate evidence for at least partial involvement in effects on food intake and body weight/adiposity with either RYGB or VSG (microbiome, bile acids, leptin/LepR, MC4R, celiac branch of vagus nerve, CGRP neurons in LPBN). Green frames indicate evidence for involvement in improved glucose homeostasis (GLP-1/GLP1R). Purple frames indicate controversial outcomes (TGR5, FXR, PYY/Y2R). Gray frames indicate no effect on outcome of bariatric surgery (Ghrelin, GLP-2, GDF-15, FGF21, brain 5HTR2c, brain GLP1R, brain Y2R, common hepatic branch of vagus nerve). Note that many other hormones besides GLP-1 can activate vagal afferent neurons. Abbreviations, Brain: AGRP, Agouti-related protein; AP, area postrema; CeA, central amygdala; LPBN, lateral parabrachial nucleus; ME, median eminence; NTS, nucleus tractus solitarius; POMC, pro-opio-melanocortin. Periphery: CCK, cholecystokinin; FGF19, FGF21, fibroblast growth factors 19 and 21; FXR farnesoid nuclear receptor; GDF15, growth and differentiation factor-15; GIP, gastric inhibitory polypeptide; NT, neurotensin; OEA, oleyl-ethanolamide; SCT, secretin; GLP-1, glucagon-like peptide-1; GLP1R, glucagon-like peptide-1 receptor; GLP-2, glucagon-like peptide-2.
Taken together, these few and disparate observations do not provide a clear picture of how bariatric surgery affects the neural circuits underlying homeostatic body weight regulation. However, striking similarities to the actions of the GLP1R-agonist semaglutide warrant further investigation of the brainstem anorexia pathway and its interface with the hypothalamus and hedonic circuitry.
3.3.2. Hedonic and cognitive mechanisms
Besides a reduction in caloric intake, many clinical studies also report changes in food preferences and food choice, as well as in the ‘liking’ and ‘wanting’ of specific nutrients and foods after bariatric surgery [[130], [131], [132], [133], [134], [135], [136], [137]]. Homeostatic mechanisms centering around the hypothalamus have been dominating research in the 20th century, but there is now much interest in the role of hedonic and cognitive brain functions in the controls of food intake and regulation of energy balance [[138], [139], [140]]. The interrelated concepts of food reward [141,142], cue-induced conditioned eating [143,144], executive/inhibitory control [145,146], and their relationship with homeostatic circuits which are mainly controlled by interoceptive signals [[147], [148], [149]], have been in the center of this effort.
Analyses of fMRI studies based on the bold-signal have also evolved - from task-based approaches simply looking at activation or inhibition of specific brain areas to studies assessing connectivity between brain areas and between task-specific networks in the resting state [150,151]. These efforts have identified increased functional connectivity of the orbitofrontal cortex (OFC) and decreased functional connectivity of the insula as the major culprits for problematic eating and obesity [151], and led to formulating the ‘triadic model’ to explain increased food consumption, consisting of 1) increased sensitivity for, and reactivity to food reward, 2) decreased inhibitory control in executive decision making often combined with increased impulsivity, and 3) numbing of interoceptive homeostatic metabolic signaling [[151], [152], [153], [154]].
Applied to bariatric surgery, most studies demonstrate normalization of impaired activity and/or connectivity patterns in the obese (before surgery) towards patterns observed in lean controls (VSG [[155], [156], [157], [158]], RYGB [155,[159], [160], [161], [162], [163], [164], [165]]). In one study presurgical functional connectivity predicted the magnitude of weight loss induced by VSG [166], and another study found that activation of cortical areas involved in inhibitory control was significantly greater after RYGB compared to VSG [155]. Similarly, in a study looking at sweet taste-related reward and corresponding brain areas, RYGB more effectively reversed the blunted activation in mesolimbic pathways found in patients with obesity compared to VSG [167].
Associations between circulating hormones/metabolites and brain activity/connectivity changes have also been reported in more recent studies with inconsistent outcomes [156,157,165,[168], [169], [170]]. Importantly, a few studies tested the potential role of elevated circulating gut hormones in the effects of RYGB on hedonic and cognitive brain functions by inhibiting either their release or receptor activation [162,163,168]. There was a significant positive correlation between the degree of octreotide-induced plasma PYY suppression (but not GLP-1) and food picture-induced brain reward area activation in RYGB but not in non-obese controls [168]. In ten women with obesity, RYGB reduced food cue-responsiveness of the caudate and reduced activation of the insula to the consumption of palatable food, and blocking endogenous GLP-1 action by infusing the GLP1R-antagonist Exendin 9-39, reversed these (beneficial) effects [162]. Similarly, GLP1R-blockade was found to reverse some of the (beneficial) effects of RYGB on resting state network connectivity thought to be responsible for reducing cue-responsivity and enhancing interoceptive satiety signaling [163].
However, note that systemic infusion of receptor blockers such as octreotide and Exendin 9-39 cannot identify the critical site(s) of inhibition, and that the acute character of these studies does not allow conclusions regarding any longer term effects on food intake and body weight. Furthermore, there are many differences in methodology and the specific brain areas implicated, and one study did not find any significant effects of RYGB or VSG on brain functional connectivity [171].
One significant issue for many of the above studies is the inability to distinguish the direct effects of the surgery from the resulting weight loss. Only one study compared low-calorie diet vs. gastric bypass surgery in order to distinguish between surgery-specific and weight loss-specific effects [161]. In a cohort of mostly female patients with obesity and type-2 diabetes, fMRI scans capturing the response to food cues in the fed versus fasted state were conducted before and 4 weeks after either very low calorie diet (VCLD) or RYGB. Compared to VLCD there was a relative reduction in food cue responsiveness in food reward brain areas, a strengthening of cognitive control regions, and reduced activation in the hypothalamus with increased connectivity to reward areas 4 weeks after RYGB [161]. This may suggest that reduced hunger after RYGB is driving the changes in food reward and that overall RYGB makes it easier to sustain weight loss compared to VLCD.
Effects on food preference and choice are also seen in rodent models after weight loss induced with VSG or RYGB in obese animals [7,27,42,[172], [173], [174], [175], [176]], suggesting differences in brain functions related to taste processing and hedonic and cognitive aspects of food intake. However, changes in taste-guided behaviors such as brief-access licking were not found in an RYGB model that uses lean, chow-fed rats [177,178], suggesting that the prior history of obesity is an important factor in determining behavioral changes after surgery. The potentially underlying neural mechanisms were only investigated in a handful of studies. RYGB in mice increased dopamine turnover in dorsal striatum [179], and RYGB in rats decreases μ-opioid signaling in the central amygdala and hypothalamus independent of diet or caloric restriction [180], and increases GABA-A receptor levels in cortical areas involved in object recognition [181]. In Yucatan mini-pigs exposure to Western-diet resulted in impaired working memory, anxiety-like behaviors, loss of motivation, and snacking-type eating behavior, and although weight loss through caloric restriction reversed most of these aberrant behaviors, RYGB did a better job in normalizing inhibitory control [182]. Finally, Hankir et al. demonstrated that RYGB in rats on a high-fat diet recruited a gut-brain pathway involving intestinal endocannabinoid signaling via oleyl-ethanolamide (OEA), the vagus nerve, and dorsal striatal dopamine receptor-1 (D1R) to reduce fat appetite [183]. However, the relevance of this pathway is not clear, as vagotomy, which abolished the changes in fat appetite, had no effect on RYGB-induced weight loss.
In summary, there is an impressive number of clinical studies demonstrating changes in neural activity patterns after bariatric surgery and their correlation with subjective reports of the patient's attitude towards food items and circulating gut hormones. Most of the findings are consistent with the idea that bariatric surgery reverses the detrimental effects of obesity on hedonic and cognitive controls of eating, making it easier for bariatric surgery patients to reduce calorie intake despite massive weight loss. However, none of the clinical and preclinical studies demonstrates causality for the weight loss and metabolic improvements. Such interventional-type studies will be difficult to implement in patients, but the use of function-specific neural pathway manipulations in animal models should eventually lead to such mechanistic conclusions.
4. Major candidate mechanisms for the effects on body weight
The many changes described in the preceding section could all potentially play a role in the beneficial effects of bariatric surgery. However, few specific hypotheses have been tested with interventional type studies. Of more than 30 studies that directly tested a specific hypothesis by comparing wildtype and knockout mice in their ability to sustain bariatric surgery-induced weight loss, the majority was negative, meaning that there was no evidence for the hypothesized signaling mechanism (ghrelin [184]; GLP-1R [[185], [186], [187]]; GLP-2R [53]; PYY/Y2R [41]; FGF21 [188]; TGR5 [189]; FXR [190]; 5-HTR2c [191]; Clock Delta-19 [192]; NTSR1 [193], GDF15 [126]). Only a handful of studies found at least some consequence of knocking out a specific signaling mechanism on RYGB- or VSG-induced weight loss and or other metabolic improvements (Figure 3). Here we discuss signaling pathways and molecules that have been demonstrated to be at least partially required for the beneficial effects of bariatric surgery in more detail.
4.1. Mechanisms primarily changing food intake
4.1.1. GLP-1, PYY, and their signaling pathways
The first and major hypothesized mechanism involving gut-brain communication is altered gut hormone signaling to the brain, specifically GLP-1 and PYY-signaling. Both GLP-1 and PYY-secretion from intestinal L-cells is significantly and lastingly increased after both RYGB and VSG (but not gastric banding) [[54], [55], [56], [57], [58],194], and there is a large literature demonstrating the ability of these gut hormones to suppress appetite and food intake in both humans and animals [[195], [196], [197], [198], [199]]. Furthermore, and as outlined in section 3.3.2 above, human neuroimaging studies clearly demonstrate a role for endogenous GLP-1 and perhaps PYY in the beneficial effects of RYGB on the activity of neural networks responsible for the hedonic and homeostatic controls of food intake [162,163,168]. These studies collectively show that endogenous GLP-1 suppresses anticipatory food reward, strengthens cognitive inhibition, and enhances interoceptive satiety signaling in corresponding functional neural networks, and that RYGB with its elevated circulating GLP-1 levels takes advantage of these effects.
Together with the recent success with stable GLP-1 receptor agonists such as liraglutide and semaglutide to produce amounts of weight loss rivaling those of bariatric surgery [2,[200], [201], [202], [203]], it would seem to confirm the long sought-after mechanism. A recent study in rodents with radiolabeled semaglutide and the c-fos neural activity stain highlighted a well-known brain network as likely substrate for the appetite-suppressing and body weight-lowering effects of this stable GLP-1R agonist. This network consists of the NTS and area postrema in the caudal brainstem, and the hypothalamic arcuate nucleus, both converging on the parabrachial nucleus in the pons [128], the main hub of the brainstem anorexia pathway extending from the NTS via the PBN to the CeA and BNST [125]. This circuitry would seem to be in an ideal position to mediate the body weight and metabolic effects of RYGB and VSG via increased GLP-1 signaling.
However, several observations raise questions for such a conclusion. First, studies in rodents with GLP-1 receptor deficiency [[185], [186], [187]] or pharmacological blockade of GLP-1 receptors [187,204] concluded that peripheral and central GLP1 receptor signaling is not required for the beneficial effects of RYGB and VSG on body weight. It should be taken into consideration that these were whole body and germline knockouts, with considerable potential for developmental compensation. Only one study used a β-cell-specific GLP-1R knockout mouse model, and indeed demonstrated that improvement of glucose tolerance after VSG partly depended on GLP-1R signaling in pancreatic β-cells, even though body weight loss was not differentially affected [205]. It remains to be seen whether tissue-specific and inducible knockout models can provide more positive outcomes.
Second, it is not completely clear how GLP-1 of intestinal origin leads to suppression of food intake. Because GLP-1 has a short half-life in the circulation, it is unlikely to reach the brain in high enough concentrations, even with the much higher secretion rate after surgery. Therefore, the prevailing view is that GLP-1 of enteroendocrine origin acts locally on vagal sensory nerve fibers [[206], [207], [208], [209], [210]] or on vagal nerve fibers innervating the hepatic portal vein [[211], [212], [213]] that in turn change activity in brainstem and other brain areas to suppress food intake [207]. The observation that surgical transection of the paired celiac branches of the subdiaphragmatic vagus nerve, partially abrogates RYGB-induced suppression of food intake and body weight in rats [214] is consistent with this idea. By preferentially innervating the small and large intestines [215], including the Roux and common limbs, the celiac branches are in a position to pick up any signaling originating from the rearranged gut. Because surgical transection of the common hepatic branch, which supplies vagal sensory innervation to the hepatic portal vein [216] does not affect the outcome of RYGB in rats [217], it is unlikely that GLP-1 sensing mechanisms within the hepatic portal vein play an important role. Given that these surgical subdiaphragmatic vagal branch cuts included both sensory and motor fibers, the findings are consistent with, but do not unequivocally prove, a role for vagal afferents. So, if surgery-induced elevation of GLP-1 secretion is still considered as part of the mechanism, it would most likely involve vagal afferent mediation to the brain. This is in contrast to how the new stable GLP-1 receptor agonists work [206,207,218]. They seem to primarily activate GLP-1 receptors in the brainstem and hypothalamus and involve the lateral parabrachial anorexia pathway [128,207].
Surgery-induced increased secretion of other anorexigenic gut hormones such as PYY, Neurotensin (NT) and CCK are also plausible mechanisms [193,219]. The acute effects of modified RYGB on body weight were lost in PYY-KO mice [220]. However, global deletion of the PYY/Y2R in mice [41] and brain-specific pharmacological blockade of the PYY/Y2 receptor in rats [187] did not affect the ability of RYGB to reduce food intake, lower body weight, and improve glycemic control. In neurotensin receptor-1 (NTSR1) deficient mice, the ability of VSG to reduce fat preference was reduced, but long-term weight loss was not affected [193].
Failure to demonstrate an effect of knocking out a single pathway could at least partially be explained by the often observed additivity or even synergism between two or more anorexigenic gut hormones. A specific case has been made for the synergistic effects of GLP-1 and PYY on food intake in rodents and humans [219,221,222]. Such synergism is also relevant for the question of developmental compensation following whole-body germline knockouts, as PYY/Y2R-signaling or GLPR1-signaling may take over each other's function in single knockouts. To address this question, we recently generated a GLP1R/Y2R double knockout mouse model [223]. However, RYGB had the same ability to lower body weight and improve other metabolic and glycemic parameters in such double-knockout compared to wild-type mice [223].
In summary, the profound changes in the secretion of gut hormones, in particular GLP-1 and PYY after RYGB and VSG, together with receptors at all the ‘right places’, would seem to be in a perfect position to mediate the beneficial effects of these weight-loss operations. However, while there is moderately convincing evidence for an important role of GLP-1 signaling in humans, there is quite strong evidence against it in mice, all be it obtained mostly in whole-body germline knockout models with all their limitations. Therefore, to substantiate a critical role for GLP-1 signaling after RYGB and VSG in humans, future clinical studies could include 1) longer-term administration of GLP-1 receptor blockers, 2) intranasal administration of GLP-1 receptor blockers to distinguish brain-specific from peripheral effects, and 3) direct comparisons of altered brain signaling by bariatric surgery and stable GLP-1 analog treatment. Future preclinical studies with inducible and more selective GLP-1R deletion techniques should also be revealing.
4.1.2. Bile acids and their signaling pathways
Bile acids are synthesized within hepatocytes and are critical for the solubilization and absorption of dietary fat and fat-soluble micronutrients. Once synthesized, conjugated bile acids (taurine or glycine conjugates in rodents and humans, respectively) are secreted into the biliary system and transit to the intestinal tract [224]. Upon reaching the terminal ileum, the majority of bile acids are taken up by high-affinity receptors on the ileocytes, returned to the portal circulation, and then cleared by the liver through high-affinity receptors that creates a highly selective and one-way circulation between the liver and gut referred to as the enterohepatic circulation. (Figure 4). To further increase physiologic complexity, circulating bile acids also increase with intestinal nutrient delivery (i.e. feeding) and peak higher with increasing intestinal lipid delivery [225,226]. Even though only a few primary species are synthesized endogenously [227], bacterial transformation in the intestinal tract and other chemical conjugation (e.g. sulfation, dihydroxylation, deamidation) can lead to numerous bile acid species [228]. These chemical species are the endogenous ligands of several nuclear and transmembrane receptors, most notably the farnesoid X receptor (FXR) and the G-protein Coupled Bile Acid Receptor (GPBAR, TGR5), but also the vitamin D receptor and pregnane X Receptor [229].
Unlike the remarkable effects of stable GLP1R-agonists, clinical trials leveraging the physiological effects of bile acids signaling were little effective for changing food intake and body weight, although they can significantly improve glycemic control in patients with obesity and diabetes [230]. Therefore, the premise for mediating the beneficial effects of bariatric surgery is not as robust for bile acid signaling as it is for GLP-1.
Bariatric surgery is associated with markedly elevated circulating bile acid levels in humans [[231], [232], [233], [234], [235], [236], [237]] and rodents [238]. Fasting and prandial concentrations are augmented by bariatric surgery, and the prandial rise that is lost with obesity is restored by RYGB [231,239,240]. Although many reports demonstrate general increases in bile acids following surgery, studies of the chemical composition require more sophisticated methods and are fewer. Also, whether there are differences in bile acid responses between operations (i.e. VSG and RYGB) remains unknown. Timing is another variable that remains poorly studied, as clinical studies demonstrate increased circulating bile acids as early as 4 weeks postoperatively [232], but data is mixed at earlier time points (∼1–4 weeks in humans) [231,241]. Given that bile acid increases may not be present at these early postoperative time points, when there are clear improvements in glucose regulation, suggests that bile acids may not completely explain the early beneficial effects of RYGB on glucose homeostasis [231,233,234]. This delayed rise in circulating bile acids indicates that a signal stimulating synthesis may be required to drive these increases over time. However, the mechanisms for these dramatic increases in bile acids overall remain unclear. Overall, rodent studies are more limited, but VSG is associated with increased fasting and prandial rises in total bile acids [242] by 2 weeks. Prandial and fasting differences, differences in chemical species, as well as earlier time points postoperatively (earlier than 2 weeks) are poorly characterized to date.
Mechanistic studies have been carried out almost exclusively in mouse models with genetic deletion of either FXR or TGR5. Bile acid signaling through the nuclear receptor FXR was found to be required for the full weight loss induced by either VSG [243] or the diversion of bile to the distal ileum [244], but not for RYGB-induced weight loss [190]. However, RYGB-induced improvements in glucose tolerance and HOMA-IR were partially dependent on a functional FXR [190]. Bile acid signaling through the membrane receptor TGR5, while not required for both RYGB [189] and VSG-induced weight loss [53,245], was partially required for improved VSG-induced glucose tolerance [245]. In other studies using interventional approaches, reduced circulating levels of tauro-deoxy-cholic acid (TUDCA) and valine in high-fat diet-induced obese mice were restored after VSG, and systemic treatment with TUDCA and valine induced the same weight loss as after VSG [246]. Similarly, a specific sulfated cholic acid is enriched in the gut of mice following VSG that is associated with GLP-1 and glycemic control [247]. These interventional studies are beginning to identify how bile acids might modulate post-surgical physiology. Even though bile acids are metabolically active hormones with a plethora of metabolic effects, their specific roles in body weight and other metabolic regulation after bariatric surgery remain unclear. However, recent evidence has demonstrated the importance of TGR5 signaling in the hypothalamus to protect from obesity [248], and in the nucleus accumbens to reduce cocaine reward [249].
In summary, the roles of peripheral and/or central bile acid signaling remains unclear and further studies, especially those using RYGB, require interventional and tissue-specific approaches to clarify how these compounds alter physiology in response to bariatric surgery. The driving force behind the marked increases in bile acid synthesis following bariatric surgery remains unknown, though this mediator could be a potent target for altering metabolic physiology by modifying bile acid levels.
4.1.3. Microbiota and their signaling pathways
As discussed in section 3.1.3, there is no doubt that bariatric surgery leads to profound changes in the gut microbiome. However, the outcome of the few attempts with transplantation of bariatric surgery-modified microbiota to mice is rather disappointing. In one of these studies, transplantation of microbiota from two female RYGB patients after weight loss to germ-free mice very modestly, but significantly, reduced fat mass gain over a two week period compared to transplantation of microbiota from patients with obesity [81]. Transplantation of pooled microbiota obtained from mice with RYGB significantly reduced body weight by about 5% in germ-free mice, but microbiota from sham-operated mice had no effect [78]. Finally, transplantation of gut microbiota from patients with poor weight loss after RYGB into antibiotic-treated recipient mice resulted in significantly greater weight gain compared to mice receiving microbiota from patients with successful weight loss [250]. However, there was no significant difference in weight loss between mice receiving transplants from normal control patients vs. patients with successful weight loss [250].
Potential signaling pathways from gut microbiota to the host may include immune signaling, tryptophan and tyrosine metabolites, as well as bile acids and short-chain fatty acids (for recent review see [251]. For example, transplantation of cecal contents from mice with VSG to germ free mice is sufficient to alter bile acid metabolism and increase the potent TGR5 agonist cholic acid-7-sulfate (CA7S) that is associated with augmented GLP-1 secretion and improved glucose homeostasis [247,252]. Furthermore, deletion of the TLR4-receptor or MYD88 in mice reduced the effects of RYGB on glycemic control. Immune-signaling via TLR4 receptors and myeloid-differentiation factor 88 (MyD88) may be one of the underlying signaling pathways, as deletion of their function resulted in a partial loss of beneficial effects of the surgery on metabolism at 5 weeks after surgery [253]. However, these conclusions are tempered by the fact that the TLR4-KO mouse has a lean phenotype that should make it largely resistant to the body weight-lowering effects of RYGB [253].
In summary, surgery-induced changes in microbiota-signaling may contribute to some small extent to the beneficial effects of bariatric surgery, and individual differences in gut microbiota composition may be predictive of some of the beneficial effects of bariatric surgery.
4.1.4. Other signaling pathways with evidence for a mechanistic role
4.1.4.1. Leptin-signaling
The ability of RYGB to reduce body weight was about 50% less in Ob/Ob mice with leptin receptor deficiency compared to wildtype mice [254]. In another study, RYGB was effective in reducing body weight in obese Ob/Ob mice compared to sham surgery, although at 6 weeks after surgery body weight returned to slightly above presurgical levels [255]. Together, these mouse studies suggest that leptin-signaling is required for the full effects of RYGB on body weight. As plasma leptin levels drop drastically after RYGB [254], which normally triggers increased hunger and reduced metabolism, the ability of RYGB to at least partially suppress these counter-regulatory responses is again demonstrated. Given the wide distribution of leptin receptors, tissue and brain site-specific deletion of leptin receptor signaling will have to be used to ferret out the critical sites of leptin action.
4.1.4.2. Melanocortin-signaling
A recent systematic review of the clinical literature concluded that the MC4R gene is inconsistent in its relationship with the amount of weight loss after bariatric surgery [256]. However, one study demonstrated inferior long-term weight outcome of cases with MC4R variants after VSG compared to RYGB [257], and another study found that MC4R mutation carriers with impaired eating control had more major complications than non-carriers independent of weight loss [258].
Studies in whole body MC4R knockout mice [259,260] as well as a study with long-term pharmacological blockade of the brain MC4R in rats [261] demonstrated the requirement of MC4R-signaling for RYGB-induced weight loss. In the latter study, rats that had lost 15% of body weight after RYGB regained all lost weight during a two-week brain infusion of the MC4R-antagonist SHU9119, and then fell back to the lower body weight level after termination of the infusion [261]. As the lateral ventricle infusions most likely acted on forebrain sites, it is plausible that they affected primarily MC4R-signaling of basomedial hypothalamic POMC and AGRP neurons with their widespread forebrain projections. It is interesting to speculate that SHU9119 inhibited MC4R-signaling augmented by endogenously elevated levels of GLP-1, as GLP1R-bearing POMC neurons respond to both locally administered GLP-1 and systemically administered semaglutide, which in turn activate LPBN neurons directly or via inhibition of inhibitory AGRP neurons [128]. This scenario is also supported by the observation in mice that the weight loss-induced up-regulation of AGRP neurons seen with dietary restriction is not seen with RYGB-induced weight loss [121].
Melanocortin-signaling at the level of autonomic neural outflow may also be involved as shown by rescuing MC4R expression selectively in autonomic neurons in mice with whole body knockouts [260]. Most of the RYGB effects on weight loss and glycemic control were abrogated in whole body MC4R knockout mice. However, MC4R-expression restricted to cholinergic preganglionic autonomic neurons (both vagal and sympathetic) largely rescued these effects [260]. Because the RYGB model used by these investigators was characterized by major changes in energy expenditure (although not corrected for body weight) rather than energy intake, the findings point to MC4Rs on preganglionic sympathetic neurons innervating adipose tissue [262].
4.1.4.3. Insulin-like growth factor binding protein-2 (IGFBP2)
Deletion of insulin-like growth factor binding protein-2 (IGFBP2), which is significantly increased after RYGB in humans, rats and mice, led to approximately 30% less RYGB-induced weight loss compared to wild type mice [194]. This suggests that IGFBP2-signaling is required for the full beneficial effects of RYGB in mice. Interestingly, insulin-like growth factor-1 (IGF-1) is also significantly increased in all limbs of the GI-tract 2 weeks after RYGB in rats [40], and IGF-1-induced intestinal proliferation is inhibited by insulin-like growth factor binding protein-4 (IGFBP4) [263]. Whether IGFBP2 has similar inhibitory effects on IGF-1 induced intestinal proliferation remains to be determined, but the fact that IGFBP2-deficiency did not prevent the RYGB-induced increase in total intestinal wet weight 20 weeks after surgery [194] argues against it.
4.2. Mechanisms affecting primarily energy expenditure and feed efficiency
Some hypothesized mechanisms are restricted to the gut itself, but most of them embrace changes in gut-brain communication and resulting changes in eating behavior and autonomic control of critical organs and tissues.
The major non-neural mechanism purported is the re-programming of gut metabolism towards energetically demanding processes, thereby drastically reducing feed efficiency and body weight [39,112,113]. In addition, changes in peripheral glucose, lipid, and amino acid metabolism could have beneficial effects on plasma glucose and triglyceride levels independent of weight loss [14,39,61]. However, because of the descriptive and associative character of all these studies, direct evidence for the requirement of such mechanisms in the beneficial effects of bariatric surgery is missing. Not knowing the exact nature/molecular identity of the critical steps within these pathways, it has been difficult to bring them to the test. It needs to be demonstrated that interfering with or preventing the hypothesized re-programming abrogates the RYGB or VSG-induced beneficial effects.
Another hypothesized mechanism thought to primarily improve glycemic control through peripheral mechanisms is the bariatric (metabolic) surgery induced suppression of an unknown anti-incretin hormone or factor secreted from the proximal intestine that normally is responsible for the obesity-associated diabetic state [[264], [265], [266]]. However, without knowing the identity of such a hormone or factor, it will be difficult to prove this hypothesis.
5. Weight loss-dependent vs. weight loss-independent effects
Weight loss through dieting and other behavioral modifications is extremely effective in improving metabolic and general health, including diabetes, hepatic steatosis, cardiovascular parameters, cancer susceptibility, sleep, sexual drive, and quality of life. It is now almost 20 years since a randomized controlled trial in subjects with obesity demonstrated that losing just 5–10% of body weight through dieting and lifestyle changes significantly reduces diabetes susceptibility by over 50% [267], and more recent studies have fully confirmed these findings [268]. However, while the highly beneficial effects of dieting in subjects with obesity are uncontested, the problem is sustaining these benefits with continued behavioral changes over the rest of a 20 or 30 year-old's life expectancy of 50–70 more years in an obesogenic environment. It has been estimated that falling into recidivism amounts to 80% or more of subjects dieting. This is where the true value of bariatric surgery is evident, as surgery makes it easier to adhere to behavioral and lifestyle changes over much longer periods of time. Surgery prevents this recidivism by largely preventing the adaptive counter-regulatory responses to weight loss, namely increased hunger and hypometabolism, and current evidence points to key mechanisms acting on food intake and re-setting energy balance in the brain.
When compared head-to-head, it also became clear that using caloric restriction to match the weight loss of bariatric surgery resulted in the same metabolic benefits, specifically improvements in fasting glucose and insulin, glucose tolerance, and insulin sensitivity/resistance. This was demonstrated in several clinical studies varying in length between 10 days and 23 weeks [82,[269], [270], [271], [272], [273]], and in several rodent studies [112,189,194,223,260]. It is important to note that these metabolic benefits are also induced by a hypocaloric state, as extended fasting can rapidly and significantly improve metabolic function (for a recent review and meta-analysis see [274]). In the most comprehensive and technically sophisticated study in patients with obesity randomized for BMI as well as severity and duration of diabetes, the authors concluded that “the metabolic benefits of gastric bypass surgery and diet were similar and were apparently related to weight loss itself, with no evident clinically important effects independent of weight loss” [82]. Clearly, weight loss-independent effects, such as a greater decrease in branched-chain amino acids, an increase in plasma bile acids, and greater changes in the gut microbiome after gastric bypass were confirmed, but these changes were not associated with greater improvements in metabolic function [82].
Nevertheless, some clinical studies provide evidence for weight loss-independent effects of bariatric surgery on glycemic control [see [275] for recent review) and major adverse cardiovascular events based on post-hoc statistical adjustments for weight loss [276]. Some pre-clinical studies also provide evidence for weight loss-independent effects on glycemic control [14,277,278] and hepatic steatosis [242], based on pair-feeding or weight-matching that resulted in similar weight loss. However, the “additional” weight loss-independent effects were generally small compared to the weight loss-dependent effects.
Thus, the term “metabolic surgery” seems premature at this time for several reasons. The major reason is the lack of strong evidence for major weight loss- and hypo-alimentation-independent effects, which is often the result of studies without proper controls. Claims of weight loss-independent beneficial effects are difficult to make when there is no control group in which dietary intake and weight loss are matched to be the same as in surgery patients. This is not to say that weight loss-independent effects do not exist, it merely suggests that they are less powerful and potentially less important than the major effects driven by weight loss overall. Thus, well-spent research funds should have the goal to focus on a better mechanistic insight how bariatric surgery leads to weight loss and to a suppression of counter-regulatory responses that lead to weight regain with dieting or behavioral treatments for obesity. Regardless, it is very likely that bariatric surgery induces novel signaling mechanisms that may be truly independent of weight loss and hypo-alimentation that will be identified in the future.
6. Conclusions
Bariatric surgery is currently the most effective treatment for severe obesity, and has tremendously increased the quality of life for thousands of patients. In contrast to pharmacological treatments the intervention is restricted in time but typically lasts for decades. For logistical reasons, however, it will not be possible to deliver bariatric surgery to the larger population. Therefore, learning from mechanisms that make these operations so effective is a major goal in obesity research. The relatively young science of bariatric surgery has tremendously stimulated metabolic research and generated several lead hypotheses as to their mechanism of action. A healthy level of skepticism by many basic scientists together with the can-do attitude of many surgeons has created a competitive atmosphere and fertile ground for out-of-the-box thinking.
Although no clear and unique mechanism has yet emerged, there is moderately convincing evidence that increased GLP-1 signaling plays a significant role in orchestrating reduced food intake and loss of body weight after RYGB in humans, and in improving glucose homeostasis after RYGB and VSG in rodents and humans. There is also some evidence from transgenic mouse models for the partial involvement of other signaling pathways, including bile acids, gut microbes, leptin, melanocortin, and IGFBP2. However, besides perhaps melanocortin signaling, none of these signaling pathways seems to account for the bulk of the beneficial effects of bariatric surgery. We speculate that, similar to the genetics of obesity, dozens of signaling pathways contribute to these beneficial effects, each with a small size effect and making their experimental demonstration difficult. This was quite unexpected because the initial intervention in bariatric surgery is limited to the gut. If correct, the scenario reveals a much richer cross-communication between the gut and the rest of the body, at least as far as body weight regulation is concerned. As with the genetics of obesity, it is still possible that we are currently missing a signaling pathway(s) with a larger size effect, as methodological advances in clinical and preclinical research become available.
6.1. Lessons learned
-
1.
The complexity and redundancy of gut-brain communication and brain control of food intake and energy balance are more appreciated. It seems unlikely that the elusive mechanism mediating the effects of bariatric surgery involves a single pathway, but instead likely consists of multiple signaling pathways (cascades). This view is supported by the recent push towards multi-pharmacy specifically capitalizing on gut hormones.
-
2.
Gut-brain circuits can be exploited to change brain function and are potential therapeutic targets and gateways into the brain. Administering drugs and manipulating the periphery is far easier than trying to directly target the brain.
-
3.
We should be aware that significant differences may exist between clinical and pre-clinic bariatric surgery models, and that animal models need to be standardized and well-validated.
-
4.
Ask the right question. Not every change observed after a particular surgery leads to a valid hypothesis. Carry out appropriate controls such as pair-feeding and/or weight-matching before formulating a hypothesis.
-
5.
Choose the best animal models for specific operations and hypotheses. Reversal of obesity vs. prevention of obesity. Obese diabetic vs. non-obese-diabetic.
6.2. Future directions
The most promising hypotheses (e.g. the involvement of microbiome, hormone, and bile acid signaling) should be tested with advanced techniques, including inducible, tissue-specific knockout models, both rat and mouse, and with some humanized physiological traits (e.g. bile acid or lipid profile etc.) if possible. Applied to neural gut-brain communication pathways, state-of-the-art neural manipulation techniques should be used. Among these are manipulations (stimulation, inhibition, or silencing) of function-specific vagal or dorsal root afferents and neuron populations in the brain in verified genetic mouse lines. Mouse RYGB surgery should be further developed and practiced in dedicated surgery cores to reduce mortality and replicability.
However, investing in the prevention of obesity and metabolic disease seems unavoidable if we are to stem the tide. Globally, an estimated 39 million children under the age of 5 were overweight or obese in 2020 and knowing the high fidelity progression from childhood overweight to adult obesity is particularly alarming and suggests that prevention should be the first choice in stemming this global crisis. The fact that this insight and recommendation was already available 50 years ago [22] seriously questions the ability of our scientific and political institutions to act (David L. Katz, Time Magazine, May 30, 2017). We need to collectively find the political will to make fundamental changes to the environment, education, food industry, and agricultural policies.
Author contributions
Vance Albaugh: Conceptualization, Data Curation, Validation, Writing – original draft, Writing – review and editing. Yanlin He: Writing – review and editing. Sangho Yu: Writing – review and editing. Heike Munzberg: Writing – review and editing. Christopher Morrison: Writing – review and editing. Hans-Rudolf Berthoud: Conceptualization, Writing – original draft, Writing – review and editing, Visualization.
Funding
Research supported by the National Institutes of Health grants R01DK47348 (HRB); 2R01DK092587, 1R01AT011683 (HM); R01DK123083, R01DK121370 (CDM); P20GM135002 (YH), and the Louisiana Board of Regents Support Fund LEQSF(2021-24-RD-A-12 (SY).
Conflict of interest
None of the other co-authors declare any conflict of interest.
References
- 1.Wood W., Mazar A., Neal D.T. Habits and goals in human behavior: separate but interacting systems. Perspect Psychol Sci. 2021 doi: 10.1177/1745691621994226. 1745691621994226. [DOI] [PubMed] [Google Scholar]
- 2.Wilding J.P.H., Batterham R.L., Calanna S., Davies M., Van Gaal L.F., Lingvay I., et al. Once-Weekly semaglutide in adults with overweight or obesity. N Engl J Med. 2021;384:989. doi: 10.1056/NEJMoa2032183. [DOI] [PubMed] [Google Scholar]
- 3.Arterburn D., Wellman R., Emiliano A., Smith S.R., Odegaard A.O., Murali S., et al. Comparative effectiveness and safety of bariatric procedures for weight loss: a PCORnet cohort study. Ann Intern Med. 2018;169:741–750. doi: 10.7326/M17-2786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Athanasiadis D.I., Martin A., Kapsampelis P., Monfared S., Stefanidis D. Factors associated with weight regain post-bariatric surgery: a systematic review. Surg Endosc. 2021;35:4069–4084. doi: 10.1007/s00464-021-08329-w. [DOI] [PubMed] [Google Scholar]
- 5.Istfan N.W., Lipartia M., Anderson W.A., Hess D.T., Apovian C.M. Approach to the patient: management of the post-bariatric surgery patient with weight regain. J Clin Endocrinol Metab. 2021;106:251–263. doi: 10.1210/clinem/dgaa702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Schauer P.R., Bhatt D.L., Kirwan J.P., Wolski K., Aminian A., Brethauer S.A., et al. Bariatric surgery versus intensive medical therapy for diabetes - 5-year outcomes. N Engl J Med. 2017;376:641–651. doi: 10.1056/NEJMoa1600869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Hao Z., Townsend R.L., Mumphrey M.B., Morrison C.D., Munzberg H., Berthoud H.R. RYGB produces more sustained body weight loss and improvement of glycemic control compared with VSG in the diet-induced obese mouse model. Obes Surg. 2017;27:2424–2433. doi: 10.1007/s11695-017-2660-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Stevenson M., Srivastava A., Lee J., Hall C., Palaia T., Lau R., et al. RYGB is more effective than VSG at protecting mice from prolonged high-fat diet exposure: an occasion to roll up our sleeves? Obes Surg. 2021;31:3227–3241. doi: 10.1007/s11695-021-05389-8. [DOI] [PubMed] [Google Scholar]
- 9.Hall K.D., Guo J. Obesity energetics: body weight regulation and the effects of diet composition. Gastroenterology. 2017;152:1718–1727 e1713. doi: 10.1053/j.gastro.2017.01.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Maclean P.S., Bergouignan A., Cornier M.A., Jackman M.R. Biology's response to dieting: the impetus for weight regain. Am J Physiol Regul Integr Comp Physiol. 2011;301:R581–R600. doi: 10.1152/ajpregu.00755.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kumar R., Lieske J.C., Collazo-Clavell M.L., Sarr M.G., Olson E.R., Vrtiska T.J., et al. Fat malabsorption and increased intestinal oxalate absorption are common after Roux-en-Y gastric bypass surgery. Surgery. 2011;149:654–661. doi: 10.1016/j.surg.2010.11.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Laurenius A., Larsson I., Bueter M., Melanson K.J., Bosaeus I., Forslund H.B., et al. Changes in eating behaviour and meal pattern following Roux-en-Y gastric bypass. Int J Obes. 2012;36:348–355. doi: 10.1038/ijo.2011.217. [DOI] [PubMed] [Google Scholar]
- 13.Sjostrom L., Lindroos A.K., Peltonen M., Torgerson J., Bouchard C., Carlsson B., et al. Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery. N Engl J Med. 2004;351:2683–2693. doi: 10.1056/NEJMoa035622. [DOI] [PubMed] [Google Scholar]
- 14.Ben-Zvi D., Meoli L., Abidi W.M., Nestoridi E., Panciotti C., Castillo E., et al. Time-dependent molecular responses differ between gastric bypass and dieting but are conserved across species. Cell Metabol. 2018;28:310–323 e316. doi: 10.1016/j.cmet.2018.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Adams T.D., Davidson L.E., Litwin S.E., Kim J., Kolotkin R.L., Nanjee M.N., et al. Weight and metabolic outcomes 12 Years after gastric bypass. N Engl J Med. 2017;377:1143–1155. doi: 10.1056/NEJMoa1700459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Keefer L., Ballou S.K., Drossman D.A., Ringstrom G., Elsenbruch S., Ljotsson B. A rome working team report on brain-gut behavior therapies for disorders of gut-brain interaction. Gastroenterology. 2022;162:300–315. doi: 10.1053/j.gastro.2021.09.015. [DOI] [PubMed] [Google Scholar]
- 17.Berthoud H.R. Vagal and hormonal gut-brain communication: from satiation to satisfaction. Neuro Gastroenterol Motil. 2008;20(Suppl 1):64–72. doi: 10.1111/j.1365-2982.2008.01104.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Rebelos E., Iozzo P., Guzzardi M.A., Brunetto M.R., Bonino F. Brain-gut-liver interactions across the spectrum of insulin resistance in metabolic fatty liver disease. World J Gastroenterol. 2021;27:4999–5018. doi: 10.3748/wjg.v27.i30.4999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Foster J.A., Baker G.B., Dursun S.M. The relationship between the gut microbiome-immune system-brain Axis and major depressive disorder. Front Neurol. 2021;12:721126. doi: 10.3389/fneur.2021.721126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Mulak A. Bile acids as key modulators of the brain-gut-microbiota Axis in alzheimer’s disease. J Alzheim Dis. 2021;84:461–477. doi: 10.3233/JAD-210608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Simpson S., McLellan R., Wellmeyer E., Matalon F., George O. Drugs and bugs: the gut-brain Axis and substance use disorders. JJ Neuroimmune Pharmacol. 2021 doi: 10.1007/s11481-021-10022-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Anderson J. Obesity. Br Med J. 1972;1:560–563. doi: 10.1136/bmj.1.5799.560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Sclafani A., Springer D. Dietary obesity in adult rats: similarities to hypothalamic and human obesity syndromes. Physiol Behav. 1976;17:461–471. doi: 10.1016/0031-9384(76)90109-8. [DOI] [PubMed] [Google Scholar]
- 24.Brolin R.E., Robertson L.B., Kenler H.A., Cody R.P. Weight loss and dietary intake after vertical banded gastroplasty and Roux-en-Y gastric bypass. Ann Surg. 1994;220:782–790. doi: 10.1097/00000658-199412000-00012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kral J.G. Malabsorptive procedures in surgical treatment of morbid obesity. Gastroenterol Clin N Am. 1987;16:293–305. [PubMed] [Google Scholar]
- 26.Sugerman H.J., Londrey G.L., Kellum J.M., Wolf L., Liszka T., Engle K.M., et al. Weight loss with vertical banded gastroplasty and Roux-Y gastric bypass for morbid obesity with selective versus random assignment. Am J Surg. 1989;157:93–102. doi: 10.1016/0002-9610(89)90427-3. [DOI] [PubMed] [Google Scholar]
- 27.Zheng H., Shin A.C., Lenard N.R., Townsend R.L., Patterson L.M., Sigalet D.L., et al. Meal patterns, satiety, and food choice in a rat model of Roux-en-Y gastric bypass surgery. Am J Physiol Regul Integr Comp Physiol. 2009;297:R1273–R1282. doi: 10.1152/ajpregu.00343.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Mahawar K.K., Sharples A.J. Contribution of malabsorption to weight loss after roux-en-Y gastric bypass: a systematic review. Obes Surg. 2017;27:2194–2206. doi: 10.1007/s11695-017-2762-y. [DOI] [PubMed] [Google Scholar]
- 29.Odstrcil E.A., Martinez J.G., Santa Ana C.A., Xue B., Schneider R.E., Steffer K.J., et al. The contribution of malabsorption to the reduction in net energy absorption after long-limb Roux-en-Y gastric bypass. Am J Clin Nutr. 2010;92:704–713. doi: 10.3945/ajcn.2010.29870. [DOI] [PubMed] [Google Scholar]
- 30.Li W., Baraboi E.D., Cluny N.L., Roy M.C., Samson P., Biertho L., et al. Malabsorption plays a major role in the effects of the biliopancreatic diversion with duodenal switch on energy metabolism in rats. Surg Obes Relat Dis. 2015;11:356–366. doi: 10.1016/j.soard.2014.07.020. [DOI] [PubMed] [Google Scholar]
- 31.Johansson K., Svensson P.A., Soderling J., Peltonen M., Neovius M., Carlsson L.M.S., et al. Long-term risk of anaemia after bariatric surgery: results from the Swedish Obese Subjects study. Lancet Diabetes Endocrinol. 2021;9:515–524. doi: 10.1016/S2213-8587(21)00141-8. [DOI] [PubMed] [Google Scholar]
- 32.Kwon Y., Ha J., Lee Y.H., Kim D., Lee C.M., Kim J.H., et al. Comparative risk of anemia and related micronutrient deficiencies after Roux-en-Y gastric bypass and sleeve gastrectomy in patients with obesity: an updated meta-analysis of randomized controlled trials. Obes Rev. 2022;23 doi: 10.1111/obr.13419. [DOI] [PubMed] [Google Scholar]
- 33.Borg C.M., le Roux C.W., Ghatei M.A., Bloom S.R., Patel A.G. Biliopancreatic diversion in rats is associated with intestinal hypertrophy and with increased GLP-1, GLP-2 and PYY levels. Obes Surg. 2007;17:1193–1198. doi: 10.1007/s11695-007-9211-2. [DOI] [PubMed] [Google Scholar]
- 34.Cavin J.B., Couvelard A., Lebtahi R., Ducroc R., Arapis K., Voitellier E., et al. Differences in alimentary glucose absorption and intestinal disposal of blood glucose after roux-en-Y gastric bypass vs sleeve gastrectomy. Gastroenterology. 2016;150:454–464 e459. doi: 10.1053/j.gastro.2015.10.009. [DOI] [PubMed] [Google Scholar]
- 35.Hansen C.F., Bueter M., Theis N., Lutz T., Paulsen S., Dalboge L.S., et al. Hypertrophy dependent doubling of L-cells in roux-en-Y gastric bypass operated rats. PLoS One. 2013;8 doi: 10.1371/journal.pone.0065696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.le Roux C.W., Borg C., Wallis K., Vincent R.P., Bueter M., Goodlad R., et al. Gut hypertrophy after gastric bypass is associated with increased glucagon-like peptide 2 and intestinal crypt cell proliferation. Ann Surg. 2010;252:50–56. doi: 10.1097/SLA.0b013e3181d3d21f. [DOI] [PubMed] [Google Scholar]
- 37.Mumphrey M.B., Hao Z., Townsend R.L., Patterson L.M., Berthoud H.R. Sleeve gastrectomy does not cause hypertrophy and reprogramming of intestinal glucose metabolism in rats. Obes Surg. 2015;25:1468–1473. doi: 10.1007/s11695-014-1547-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Mumphrey M.B., Patterson L.M., Zheng H., Berthoud H.R. Roux-en-Y gastric bypass surgery increases number but not density of CCK-, GLP-1-, 5-HT-, and neurotensin-expressing enteroendocrine cells in rats. Neuro Gastroenterol Motil. 2013;25:e70–e79. doi: 10.1111/nmo.12034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Saeidi N., Meoli L., Nestoridi E., Gupta N.K., Kvas S., Kucharczyk J., et al. Reprogramming of intestinal glucose metabolism and glycemic control in rats after gastric bypass. Science. 2013;341:406–410. doi: 10.1126/science.1235103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Taqi E., Wallace L.E., de Heuvel E., Chelikani P.K., Zheng H., Berthoud H.R., et al. The influence of nutrients, biliary-pancreatic secretions, and systemic trophic hormones on intestinal adaptation in a Roux-en-Y bypass model. J Pediatr Surg. 2010;45:987–995. doi: 10.1016/j.jpedsurg.2010.02.036. [DOI] [PubMed] [Google Scholar]
- 41.Boland B., Mumphrey M.B., Hao Z., Gill B., Townsend R.L., Yu S., et al. The PYY/Y2R-deficient mouse responds normally to high-fat diet and gastric bypass surgery. Nutrients. 2019;11:585. doi: 10.3390/nu11030585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Mumphrey M.B., Hao Z., Leigh Townsend R., Qualls-Creekmore E., Yu S., Lutz T.A., et al. Gastric bypass surgery in lean adolescent mice prevents diet-induced obesity later in life. Sci Rep. 2019;9:7881. doi: 10.1038/s41598-019-44344-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Martin G.R., Beck P.L., Sigalet D.L. Gut hormones, and short bowel syndrome: the enigmatic role of glucagon-like peptide-2 in the regulation of intestinal adaptation. World J Gastroenterol. 2006;12:4117–4129. doi: 10.3748/wjg.v12.i26.4117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Srikrishnaraj A., Jeong H., Brubaker P.L. Complementary and antagonistic effects of combined glucagon-like peptide-2 and glucagon-like peptide-1 receptor agonist administration on parameters relevant to short bowel syndrome. J Parenter Enteral Nutr. 2021 doi: 10.1002/jpen.2307. [DOI] [PubMed] [Google Scholar]
- 45.Sukhotnik I., Shany A., Bashenko Y., Hayari L., Chemodanov E., Mogilner J., et al. Parenteral but not enteral omega-3 fatty acids (Omegaven) modulate intestinal regrowth after massive small bowel resection in rats. J Parenter Enteral Nutr. 2010;34:503–512. doi: 10.1177/0148607110362586. [DOI] [PubMed] [Google Scholar]
- 46.Drucker D.J., Erlich P., Asa S.L., Brubaker P.L. Induction of intestinal epithelial proliferation by glucagon-like peptide 2. Proc Natl Acad Sci USA. 1996;93:7911–7916. doi: 10.1073/pnas.93.15.7911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Dube P.E., Forse C.L., Bahrami J., Brubaker P.L. The essential role of insulin-like growth factor-1 in the intestinal tropic effects of glucagon-like peptide-2 in mice. Gastroenterology. 2006;131:589–605. doi: 10.1053/j.gastro.2006.05.055. [DOI] [PubMed] [Google Scholar]
- 48.Fesler Z., Mitova E., Brubaker P.L. GLP-2, EGF, and the intestinal epithelial IGF-1 receptor interactions in the regulation of crypt cell proliferation. Endocrinology. 2020;161 doi: 10.1210/endocr/bqaa040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Martin G.R., Wallace L.E., Sigalet D.L. Glucagon-like peptide-2 induces intestinal adaptation in parenterally fed rats with short bowel syndrome. Am J Physiol Gastrointest Liver Physiol. 2004;286:G964–G972. doi: 10.1152/ajpgi.00509.2003. [DOI] [PubMed] [Google Scholar]
- 50.Rowland K.J., Trivedi S., Lee D., Wan K., Kulkarni R.N., Holzenberger M., et al. Loss of glucagon-like peptide-2-induced proliferation following intestinal epithelial insulin-like growth factor-1-receptor deletion. Gastroenterology. 2011;141:2166–2175. doi: 10.1053/j.gastro.2011.09.014. e2167. [DOI] [PubMed] [Google Scholar]
- 51.Billeschou A., Hunt J.E., Ghimire A., Holst J.J., Kissow H. Intestinal adaptation upon chemotherapy-induced intestinal injury in mice depends on GLP-2 receptor activation. Biomedicines. 2021;9 doi: 10.3390/biomedicines9010046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Hargrove D.M., Alagarsamy S., Croston G., Laporte R., Qi S., Srinivasan K., et al. Pharmacological characterization of apraglutide, a novel long-acting peptidic glucagon-like peptide-2 agonist, for the treatment of short bowel syndrome. J Pharmacol Exp Therapeut. 2020;373:193–203. doi: 10.1124/jpet.119.262238. [DOI] [PubMed] [Google Scholar]
- 53.Patel A., Yusta B., Matthews D., Charron M.J., Seeley R.J., Drucker D.J. GLP-2 receptor signaling controls circulating bile acid levels but not glucose homeostasis in Gcgr(-/-) mice and is dispensable for the metabolic benefits ensuing after vertical sleeve gastrectomy. Mol Metabol. 2018;16:45–54. doi: 10.1016/j.molmet.2018.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Korner J., Bessler M., Inabnet W., Taveras C., Holst J.J. Exaggerated glucagon-like peptide-1 and blunted glucose-dependent insulinotropic peptide secretion are associated with Roux-en-Y gastric bypass but not adjustable gastric banding. Surg Obes Relat Dis. 2007;3:597–601. doi: 10.1016/j.soard.2007.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.le Roux C.W., Aylwin S.J., Batterham R.L., Borg C.M., Coyle F., Prasad V., et al. Gut hormone profiles following bariatric surgery favor an anorectic state, facilitate weight loss, and improve metabolic parameters. Ann Surg. 2006;243:108–114. doi: 10.1097/01.sla.0000183349.16877.84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.le Roux C.W., Welbourn R., Werling M., Osborne A., Kokkinos A., Laurenius A., et al. Gut hormones as mediators of appetite and weight loss after Roux-en-Y gastric bypass. Ann Surg. 2007;246:780–785. doi: 10.1097/SLA.0b013e3180caa3e3. [DOI] [PubMed] [Google Scholar]
- 57.Peterli R., Steinert R.E., Woelnerhanssen B., Peters T., Christoffel-Courtin C., Gass M., et al. Metabolic and hormonal changes after laparoscopic Roux-en-Y gastric bypass and sleeve gastrectomy: a randomized, prospective trial. Obes Surg. 2012;22:740–748. doi: 10.1007/s11695-012-0622-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Shin A.C., Zheng H., Townsend R.L., Sigalet D.L., Berthoud H.R. Meal-induced hormone responses in a rat model of Roux-en-Y gastric bypass surgery. Endocrinology. 2010;151:1588–1597. doi: 10.1210/en.2009-1332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Franquet E., Watts G., Kolodny G.M., Goldfine A.B., Patti M.E. PET-CT reveals increased intestinal glucose uptake after gastric surgery. Surg Obes Relat Dis. 2019;15:643–649. doi: 10.1016/j.soard.2019.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Peiris M., Aktar R., Raynel S., Hao Z., Mumphrey M.B., Berthoud H.R., et al. Effects of obesity and gastric bypass surgery on nutrient sensors, endocrine cells, and mucosal innervation of the mouse colon. Nutrients. 2018;10:1529. doi: 10.3390/nu10101529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kaufman S., Arnold M., Diaz A.A., Neubauer H., Wolfrum S., Kofeler H., et al. Roux-en-Y gastric bypass surgery reprograms enterocyte triglyceride metabolism and postprandial secretion in rats. Mol Metabol. 2019;23:51–59. doi: 10.1016/j.molmet.2019.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Cani P.D., Moens de Hase E., Van Hul M. Gut microbiota and host metabolism: from proof of concept to therapeutic intervention. Microorganisms. 2021;9:1302. doi: 10.3390/microorganisms9061302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Santos-Paulo S., Costello S.P., Forster S.C., Travis S.P., Bryant R.V. The gut microbiota as a therapeutic target for obesity: a scoping review. Nutr Res Rev. 2021:1–14. doi: 10.1017/S0954422421000160. [DOI] [PubMed] [Google Scholar]
- 64.Walter J., Armet A.M., Finlay B.B., Shanahan F. Establishing or exaggerating causality for the gut microbiome: lessons from human microbiota-associated rodents. Cell. 2020;180:221–232. doi: 10.1016/j.cell.2019.12.025. [DOI] [PubMed] [Google Scholar]
- 65.de Groot P., Scheithauer T., Bakker G.J., Prodan A., Levin E., Khan M.T., et al. Donor metabolic characteristics drive effects of faecal microbiota transplantation on recipient insulin sensitivity, energy expenditure and intestinal transit time. Gut. 2020;69:502–512. doi: 10.1136/gutjnl-2019-318320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hartstra A.V., Schuppel V., Imangaliyev S., Schrantee A., Prodan A., Collard D., et al. Infusion of donor feces affects the gut-brain axis in humans with metabolic syndrome. Mol Metabol. 2020;42:101076. doi: 10.1016/j.molmet.2020.101076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Kootte R.S., Levin E., Salojarvi J., Smits L.P., Hartstra A.V., Udayappan S.D., et al. Improvement of insulin sensitivity after lean donor feces in metabolic syndrome is driven by baseline intestinal microbiota composition. Cell Metabol. 2017;26:611–619 e616. doi: 10.1016/j.cmet.2017.09.008. [DOI] [PubMed] [Google Scholar]
- 68.Vrieze A., Van Nood E., Holleman F., Salojarvi J., Kootte R.S., Bartelsman J.F., et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology. 2012;143:913–916 e917. doi: 10.1053/j.gastro.2012.06.031. [DOI] [PubMed] [Google Scholar]
- 69.Borgeraas H., Johnson L.K., Skattebu J., Hertel J.K., Hjelmesaeth J. Effects of probiotics on body weight, body mass index, fat mass and fat percentage in subjects with overweight or obesity: a systematic review and meta-analysis of randomized controlled trials. Obes Rev. 2018;19:219–232. doi: 10.1111/obr.12626. [DOI] [PubMed] [Google Scholar]
- 70.Eales J., Lenoir-Wijnkoop I., King S., Wood H., Kok F.J., Shamir R., et al. Is consuming yoghurt associated with weight management outcomes? Results from a systematic review. Int J Obes. 2016;40:731–746. doi: 10.1038/ijo.2015.202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Kadooka Y., Sato M., Imaizumi K., Ogawa A., Ikuyama K., Akai Y., et al. Regulation of abdominal adiposity by probiotics (Lactobacillus gasseri SBT2055) in adults with obese tendencies in a randomized controlled trial. Eur J Clin Nutr. 2010;64:636–643. doi: 10.1038/ejcn.2010.19. [DOI] [PubMed] [Google Scholar]
- 72.Osterberg K.L., Boutagy N.E., McMillan R.P., Stevens J.R., Frisard M.I., Kavanaugh J.W., et al. Probiotic supplementation attenuates increases in body mass and fat mass during high-fat diet in healthy young adults. Obesity. 2015;23:2364–2370. doi: 10.1002/oby.21230. [DOI] [PubMed] [Google Scholar]
- 73.Sergeev I.N., Aljutaily T., Walton G., Huarte E. Effects of synbiotic supplement on human gut microbiota, body composition and weight loss in obesity. Nutrients. 2020;12 doi: 10.3390/nu12010222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Aron-Wisnewsky J., Prifti E., Belda E., Ichou F., Kayser B.D., Dao M.C., et al. Major microbiota dysbiosis in severe obesity: fate after bariatric surgery. Gut. 2019;68:70–82. doi: 10.1136/gutjnl-2018-316103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Debedat J., Clement K., Aron-Wisnewsky J. Gut microbiota dysbiosis in human obesity: impact of bariatric surgery. Curr Obes Rep. 2019;8:229–242. doi: 10.1007/s13679-019-00351-3. [DOI] [PubMed] [Google Scholar]
- 76.Koulas S.G., Stefanou C.K., Stefanou S.K., Tepelenis K., Zikos N., Tepetes K., et al. Gut microbiota in patients with morbid obesity before and after bariatric surgery: a ten-year review study (2009-2019) Obes Surg. 2021;31:317–326. doi: 10.1007/s11695-020-05074-2. [DOI] [PubMed] [Google Scholar]
- 77.Li J.V., Ashrafian H., Bueter M., Kinross J., Sands C., le Roux C.W., et al. Metabolic surgery profoundly influences gut microbial-host metabolic cross-talk. Gut. 2011;60:1214–1223. doi: 10.1136/gut.2010.234708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Liou A.P., Paziuk M., Luevano J.M., Jr., Machineni S., Turnbaugh P.J., Kaplan L.M. Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity. Sci Transl Med. 2013;5:178ra141. doi: 10.1126/scitranslmed.3005687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Oduro-Donkor D., Turner M.C., Farnaud S., Renshaw D., Kyrou I., Hanson P., et al. Modification of fecal microbiota as a mediator of effective weight loss and metabolic benefits following bariatric surgery. Expet Rev Endocrinol Metabol. 2020;15:363–373. doi: 10.1080/17446651.2020.1801412. [DOI] [PubMed] [Google Scholar]
- 80.Sanchez-Alcoholado L., Gutierrez-Repiso C., Gomez-Perez A.M., Garcia-Fuentes E., Tinahones F.J., Moreno-Indias I. Gut microbiota adaptation after weight loss by Roux-en-Y gastric bypass or sleeve gastrectomy bariatric surgeries. Surg Obes Relat Dis. 2019;15:1888–1895. doi: 10.1016/j.soard.2019.08.551. [DOI] [PubMed] [Google Scholar]
- 81.Tremaroli V., Karlsson F., Werling M., Stahlman M., Kovatcheva-Datchary P., Olbers T., et al. Roux-en-Y gastric bypass and vertical banded gastroplasty induce long-term changes on the human gut microbiome contributing to fat mass regulation. Cell Metabol. 2015;22:228–238. doi: 10.1016/j.cmet.2015.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Yoshino M., Kayser B.D., Yoshino J., Stein R.I., Reeds D., Eagon J.C., et al. Effects of diet versus gastric bypass on metabolic function in diabetes. N Engl J Med. 2020;383:721–732. doi: 10.1056/NEJMoa2003697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Bell D.S. Changes seen in gut bacteria content and distribution with obesity: causation or association? Postgrad Med. 2015;127:863–868. doi: 10.1080/00325481.2015.1098519. [DOI] [PubMed] [Google Scholar]
- 84.Cani P.D. Severe obesity and gut microbiota: does bariatric surgery really reset the system? Gut. 2019;68:5–6. doi: 10.1136/gutjnl-2018-316815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Gutierrez-Repiso C., Moreno-Indias I., Tinahones F.J. Shifts in gut microbiota and their metabolites induced by bariatric surgery. Impact of factors shaping gut microbiota on bariatric surgery outcomes. Rev Endocr Metab Disord. 2021;4:1137–1156. doi: 10.1007/s11154-021-09676-8. [DOI] [PubMed] [Google Scholar]
- 86.Hoozemans J., de Brauw M., Nieuwdorp M., Gerdes V. Gut microbiome and metabolites in patients with NAFLD and after bariatric surgery: a comprehensive review. Metabolites. 2021;11 doi: 10.3390/metabo11060353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Liu R., Hong J., Xu X., Feng Q., Zhang D., Gu Y., et al. Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention. Nat Med. 2017;23:859–868. doi: 10.1038/nm.4358. [DOI] [PubMed] [Google Scholar]
- 88.Narath S.H., Mautner S.I., Svehlikova E., Schultes B., Pieber T.R., Sinner F.M., et al. An untargeted metabolomics approach to characterize short-term and long-term metabolic changes after bariatric surgery. PLoS One. 2016;11 doi: 10.1371/journal.pone.0161425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Shen N., Caixas A., Ahlers M., Patel K., Gao Z., Dutia R., et al. Longitudinal changes of microbiome composition and microbial metabolomics after surgical weight loss in individuals with obesity. Surg Obes Relat Dis. 2019;15:1367–1373. doi: 10.1016/j.soard.2019.05.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Villarreal-Calderon J.R., Cuellar-Tamez R., Castillo E.C., Luna-Ceron E., Garcia-Rivas G., Elizondo-Montemayor L. Metabolic shift precedes the resolution of inflammation in a cohort of patients undergoing bariatric and metabolic surgery. Sci Rep. 2021;11:12127. doi: 10.1038/s41598-021-91393-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Ilhan Z.E., DiBaise J.K., Isern N.G., Hoyt D.W., Marcus A.K., Kang D.W., et al. Distinctive microbiomes and metabolites linked with weight loss after gastric bypass, but not gastric banding. ISME J. 2017;11:2047–2058. doi: 10.1038/ismej.2017.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Pereira S.S., Jarak I., Carvalho R.A., Oliveira P.F., Alves M.G., Guimaraes M., et al. Different malabsorptive obesity surgery interventions result in distinct postprandial amino acid metabolomic signatures. Obes Surg. 2020;30:4019–4028. doi: 10.1007/s11695-020-04774-z. [DOI] [PubMed] [Google Scholar]
- 93.Stefater M.A., Pacheco J.A., Bullock K., Pierce K., Deik A., Liu E., et al. Portal venous metabolite profiling after RYGB in male rats highlights changes in gut-liver Axis. J Endocr Soc. 2020;4:bvaa003. doi: 10.1210/jendso/bvaa003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Palau-Rodriguez M., Tulipani S., Marco-Ramell A., Minarro A., Jauregui O., Sanchez-Pla A., et al. Metabotypes of response to bariatric surgery independent of the magnitude of weight loss. PLoS One. 2018;13 doi: 10.1371/journal.pone.0198214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Ha J., Jang M., Kwon Y., Park Y.S., Park D.J., Lee J.H., et al. Metabolomic profiles predict diabetes remission after bariatric surgery. J Clin Med. 2020;9 doi: 10.3390/jcm9123897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Tamboli R.A., Sidani R.M., Garcia A.E., Antoun J., Isbell J.M., Albaugh V.L., et al. Jejunal administration of glucose enhances acyl ghrelin suppression in obese humans. Am J Physiol Endocrinol Metab. 2016;311:E252–E259. doi: 10.1152/ajpendo.00082.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Chambers A.P., Smith E.P., Begg D.P., Grayson B.E., Sisley S., Greer T., et al. Regulation of gastric emptying rate and its role in nutrient-induced GLP-1 secretion in rats after vertical sleeve gastrectomy. Am J Physiol Endocrinol Metab. 2014;306:E424–E432. doi: 10.1152/ajpendo.00469.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Berthoud H.R. The vagus nerve, food intake and obesity. Regul Pept. 2008;149:15–25. doi: 10.1016/j.regpep.2007.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Berthoud H.R., Neuhuber W.L. Functional and chemical anatomy of the afferent vagal system. Auton Neurosci. 2000;85:1–17. doi: 10.1016/S1566-0702(00)00215-0. [DOI] [PubMed] [Google Scholar]
- 100.Bai L., Mesgarzadeh S., Ramesh K.S., Huey E.L., Liu Y., Gray L.A., et al. Genetic identification of vagal sensory neurons that control feeding. Cell. 2019;179:1129–1143. doi: 10.1016/j.cell.2019.10.031. e1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Bob-Manuel J., Gautron L. Detection of G Protein-coupled receptor expression in mouse vagal afferent neurons using multiplex in situ hybridization. JoVE. 2021;175 doi: 10.3791/62945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Raybould H.E., Zumpano D.L. Microbial metabolites and the vagal afferent pathway in the control of food intake. Physiol Behav. 2021;240:113555. doi: 10.1016/j.physbeh.2021.113555. [DOI] [PubMed] [Google Scholar]
- 103.Wu X., Li J.Y., Lee A., Lu Y.X., Zhou S.Y., Owyang C. Satiety induced by bile acids is mediated via vagal afferent pathways. JCI Insight. 2020;5 doi: 10.1172/jci.insight.132400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Browning K.N., Fortna S.R., Hajnal A. Roux-en-Y gastric bypass reverses the effects of diet-induced obesity to inhibit the responsiveness of central vagal motoneurones. J Physiol. 2013;591:2357–2372. doi: 10.1113/jphysiol.2012.249268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Kentish S.J., O'Donnell T.A., Frisby C.L., Li H., Wittert G.A., Page A.J. Altered gastric vagal mechanosensitivity in diet-induced obesity persists on return to normal chow and is accompanied by increased food intake. Int J Obes. 2014;38:636–642. doi: 10.1038/ijo.2013.138. [DOI] [PubMed] [Google Scholar]
- 106.Lee S.J., Krieger J.P., Vergara M., Quinn D., McDougle M., de Araujo A., et al. Blunted vagal cocaine- and amphetamine-regulated transcript promotes hyperphagia and weight gain. Cell Rep. 2020;30:2028–2039 e2024. doi: 10.1016/j.celrep.2020.01.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Page A.J., Kentish S.J. Plasticity of gastrointestinal vagal afferent satiety signals. Neuro Gastroenterol Motil. 2017;29 doi: 10.1111/nmo.12973. [DOI] [PubMed] [Google Scholar]
- 108.Minaya D.M., Di Lorenzo P.M., Hajnal A., Czaja K. RouxenY gastric bypass surgery triggers rapid DNA fragmentation in vagal afferent neurons in rats. Acta Neurobiol Exp. 2019;79:432–444. [PMC free article] [PubMed] [Google Scholar]
- 109.Ballsmider L.A., Vaughn A.C., David M., Hajnal A., Di Lorenzo P.M., Czaja K. Sleeve gastrectomy and Roux-en-Y gastric bypass alter the gut-brain communication. Neural Plast. 2015;2015:601985. doi: 10.1155/2015/601985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Munzberg H., Laque A., Yu S., Rezai-Zadeh K., Berthoud H.R. Appetite and body weight regulation after bariatric surgery. Obes Rev. 2015;16(Suppl 1):77–90. doi: 10.1111/obr.12258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Shin A.C., Zheng H., Townsend R.L., Patterson L.M., Holmes G.M., Berthoud H.R. Longitudinal assessment of food intake, fecal energy loss, and energy expenditure after Roux-en-Y gastric bypass surgery in high-fat-fed obese rats. Obes Surg. 2013;23:531–540. doi: 10.1007/s11695-012-0846-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Hao Z., Mumphrey M.B., Townsend R.L., Morrison C.D., Munzberg H., Ye J., et al. Body composition, food intake, and energy expenditure in a murine model of roux-en-Y gastric bypass surgery. Obes Surg. 2016;26:2173–2182. doi: 10.1007/s11695-016-2062-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Nestoridi E., Kvas S., Kucharczyk J., Stylopoulos N. Resting energy expenditure and energetic cost of feeding are augmented after roux-en-Y gastric bypass in obese mice. Endocrinology. 2012;153:2234–2244. doi: 10.1210/en.2011-2041. [DOI] [PubMed] [Google Scholar]
- 114.Cone R.D. Anatomy and regulation of the central melanocortin system. Nat Neurosci. 2005;8:571–578. doi: 10.1038/nn1455. [DOI] [PubMed] [Google Scholar]
- 115.Schwartz M.W., Woods S.C., Porte D., Jr., Seeley R.J., Baskin D.G. Central nervous system control of food intake. Nature. 2000;404:661–671. doi: 10.1038/35007534. [DOI] [PubMed] [Google Scholar]
- 116.Sternson S.M., Eiselt A.K. Three pillars for the neural control of appetite. Annu Rev Physiol. 2017;79:401–423. doi: 10.1146/annurev-physiol-021115-104948. [DOI] [PubMed] [Google Scholar]
- 117.Nadreau E., Baraboi E.D., Samson P., Blouin A., Hould F.S., Marceau P., et al. Effects of the biliopancreatic diversion on energy balance in the rat. Int J Obes. 2006;30:419–429. doi: 10.1038/sj.ijo.0803166. [DOI] [PubMed] [Google Scholar]
- 118.Romanova I.V., Ramos E.J., Xu Y., Quinn R., Chen C., George Z.M., et al. Neurobiologic changes in the hypothalamus associated with weight loss after gastric bypass. J Am Coll Surg. 2004;199:887–895. doi: 10.1016/j.jamcollsurg.2004.07.013. [DOI] [PubMed] [Google Scholar]
- 119.Stefater M.A., Perez-Tilve D., Chambers A.P., Wilson-Perez H.E., Sandoval D.A., Berger J., et al. Sleeve gastrectomy induces loss of weight and fat mass in obese rats, but does not affect leptin sensitivity. Gastroenterology. 2010;138:2426–2436. doi: 10.1053/j.gastro.2010.02.059. 2436 e2421-2423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Warne J.P., Padilla B.E., Horneman H.F., Ginsberg A.B., Pecoraro N.C., Akana S.F., et al. Metabolic and neuroendocrine consequences of a duodenal-jejunal bypass in rats on a choice diet. Ann Surg. 2009;249:269–276. doi: 10.1097/SLA.0b013e3181961d5d. [DOI] [PubMed] [Google Scholar]
- 121.Patkar P.P., Hao Z., Mumphrey M.B., Townsend R.L., Berthoud H.R., Shin A.C. Unlike calorie restriction, Roux-en-Y gastric bypass surgery does not increase hypothalamic AgRP and NPY in mice on a high-fat diet. Int J Obes. 2019;43:2143–2150. doi: 10.1038/s41366-019-0328-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Campos C.A., Bowen A.J., Schwartz M.W., Palmiter R.D. Parabrachial CGRP neurons control meal termination. Cell Metabol. 2016;23:811–820. doi: 10.1016/j.cmet.2016.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Cheng W., Gonzalez I., Pan W., Tsang A.H., Adams J., Ndoka E., et al. Calcitonin receptor neurons in the mouse nucleus tractus solitarius control energy balance via the non-aversive suppression of feeding. Cell Metabol. 2020;31:301–312 e305. doi: 10.1016/j.cmet.2019.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Wu Q., Boyle M.P., Palmiter R.D. Loss of GABAergic signaling by AgRP neurons to the parabrachial nucleus leads to starvation. Cell. 2009;137:1225–1234. doi: 10.1016/j.cell.2009.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Carter M.E., Soden M.E., Zweifel L.S., Palmiter R.D. Genetic identification of a neural circuit that suppresses appetite. Nature. 2013;503:111–114. doi: 10.1038/nature12596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Frikke-Schmidt H., Hultman K., Galaske J.W., Jorgensen S.B., Myers M.G., Jr., Seeley R.J. GDF15 acts synergistically with liraglutide but is not necessary for the weight loss induced by bariatric surgery in mice. Mol Metabol. 2019;21:13–21. doi: 10.1016/j.molmet.2019.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Sabatini P.V., Frikke-Schmidt H., Arthurs J., Gordian D., Patel A., Rupp A.C., et al. Proc Natl Acad Sci US; 2021. (GFRAL-expressing neurons suppress food intake via aversive pathways). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Gabery S., Salinas C.G., Paulsen S.J., Ahnfelt-Ronne J., Alanentalo T., Baquero A.F., et al. Semaglutide lowers body weight in rodents via distributed neural pathways. JCI Insight. 2020;5 doi: 10.1172/jci.insight.133429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Mumphrey M.B., Hao Z., Townsend R.L., Patterson L.M., Munzberg H., Morrison C.D., et al. Eating in mice with gastric bypass surgery causes exaggerated activation of brainstem anorexia circuit. Int J Obes. 2016;40:921–928. doi: 10.1038/ijo.2016.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Guyot E., Dougkas A., Nazare J.A., Bagot S., Disse E., Iceta S. A systematic review and meta-analyses of food preference modifications after bariatric surgery. Obes Rev. 2021;22 doi: 10.1111/obr.13315. [DOI] [PubMed] [Google Scholar]
- 131.Laurenius A., Larsson I., Melanson K.J., Lindroos A.K., Lonroth H., Bosaeus I., et al. Decreased energy density and changes in food selection following Roux-en-Y gastric bypass. Eur J Clin Nutr. 2013;67:168–173. doi: 10.1038/ejcn.2012.208. [DOI] [PubMed] [Google Scholar]
- 132.Lewis K.H., Ji M., Bai Y., Arterburn D.E., Bhakta B.B., Cornejo M., et al. Bariatric Surgical Alterations in Tolerability, Enjoyment and Cravings in the Diet (BSATED) instrument: a new scale to measure food preferences following bariatric surgery. Appetite. 2021;162:105151. doi: 10.1016/j.appet.2021.105151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Nance K., Acevedo M.B., Pepino M.Y. Changes in taste function and ingestive behavior following bariatric surgery. Appetite. 2020;146:104423. doi: 10.1016/j.appet.2019.104423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Nielsen M.S., Christensen B.J., Ritz C., Holm L., Lunn S., Taekker L., et al. Factors associated with favorable changes in food preferences after bariatric surgery. Obes Surg. 2021;31:3514–3524. doi: 10.1007/s11695-021-05374-1. [DOI] [PubMed] [Google Scholar]
- 135.Pepino M.Y., Bradley D., Eagon J.C., Sullivan S., Abumrad N.A., Klein S. Changes in taste perception and eating behavior after bariatric surgery-induced weight loss in women. Obesity. 2014;22:E13–E20. doi: 10.1002/oby.20649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Perez-Leighton C.E., Hamm J.D., Shechter A., Tamura S., Laferrere B., Xavier P.-S., et al. Preoperative liking and wanting for sweet beverages as predictors of body weight loss after Roux-en-Y gastric bypass and sleeve gastrectomy. Int J Obes. 2020;44:1350–1359. doi: 10.1038/s41366-019-0474-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Primeaux S.D., Tzeng T.H., Allerton T.D., Chiang M.C., Cosentino G., Dubin R.L., et al. Differences in short-term food preferences following vertical sleeve gastrectomy and Roux-en-Y gastric bypass surgery. Obes Res Clin Pract. 2015;9:628–632. doi: 10.1016/j.orcp.2015.09.003. [DOI] [PubMed] [Google Scholar]
- 138.Berthoud H.R. Multiple neural systems controlling food intake and body weight. Neurosci Biobehav Rev. 2002;26:393–428. doi: 10.1016/s0149-7634(02)00014-3. [DOI] [PubMed] [Google Scholar]
- 139.Ferrario C.R., Labouebe G., Liu S., Nieh E.H., Routh V.H., Xu S., et al. Homeostasis meets motivation in the battle to control food intake. J Neurosci. 2016;36:11469–11481. doi: 10.1523/JNEUROSCI.2338-16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Saper C.B., Chou T.C., Elmquist J.K. The need to feed: homeostatic and hedonic control of eating. Neuron. 2002;36:199–211. doi: 10.1016/s0896-6273(02)00969-8. [DOI] [PubMed] [Google Scholar]
- 141.Berridge K.C. Food reward: brain substrates of wanting and liking. Neurosci Biobehav Rev. 1996;20:1–25. doi: 10.1016/0149-7634(95)00033-b. [DOI] [PubMed] [Google Scholar]
- 142.Gearhardt A.N., Schulte E.M. Is food addictive? A review of the science. Annu Rev Nutr. 2021;41:387–410. doi: 10.1146/annurev-nutr-110420-111710. [DOI] [PubMed] [Google Scholar]
- 143.Boswell R.G., Kober H. Food cue reactivity and craving predict eating and weight gain: a meta-analytic review. Obes Rev. 2016;17:159–177. doi: 10.1111/obr.12354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Weingarten H.P. Conditioned cues elicit feeding in sated rats: a role for learning in meal initiation. Science. 1983;220:431–433. doi: 10.1126/science.6836286. [DOI] [PubMed] [Google Scholar]
- 145.Bartholdy S., Dalton B., O'Daly O.G., Campbell I.C., Schmidt U. A systematic review of the relationship between eating, weight and inhibitory control using the stop signal task. Neurosci Biobehav Rev. 2016;64:35–62. doi: 10.1016/j.neubiorev.2016.02.010. [DOI] [PubMed] [Google Scholar]
- 146.Seabrook L.T., Borgland S.L. The orbitofrontal cortex, food intake and obesity. J Psychiatry Neurosci. 2020;45:304–312. doi: 10.1503/jpn.190163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Berthoud H.R. Neural control of appetite: cross-talk between homeostatic and non-homeostatic systems. Appetite. 2004;43:315–317. doi: 10.1016/j.appet.2004.04.009. [DOI] [PubMed] [Google Scholar]
- 148.Berthoud H.R. Interactions between the "cognitive" and "metabolic" brain in the control of food intake. Physiol Behav. 2007;91:486–498. doi: 10.1016/j.physbeh.2006.12.016. [DOI] [PubMed] [Google Scholar]
- 149.Berthoud H.R. Metabolic and hedonic drives in the neural control of appetite: who is the boss? Curr Opin Neurobiol. 2011;21:888–896. doi: 10.1016/j.conb.2011.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Neseliler S., Han J.E., Dagher A. In: Appetite and food intake: central control. Harris R.B.S., editor. 2017. The use of functional magnetic resonance imaging in the study of appetite and obesity; pp. 117–134. Boca Raton (FL) [Google Scholar]
- 151.Parsons N., Steward T., Clohesy R., Almgren H., Duehlmeyer L. A systematic review of resting-state functional connectivity in obesity: refining current neurobiological frameworks and methodological considerations moving forward. Rev Endocr Metab Disord. 2021 doi: 10.1007/s11154-021-09665-x. [DOI] [PubMed] [Google Scholar]
- 152.Chen R., Li D.P., Turel O., Sorensen T.A., Bechara A., Li Y., et al. Decision making deficits in relation to food cues influence obesity: a triadic neural model of problematic eating. Front Psychiatr. 2018;9:264. doi: 10.3389/fpsyt.2018.00264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Weygandt M., Mai K., Dommes E., Ritter K., Leupelt V., Spranger J., et al. Impulse control in the dorsolateral prefrontal cortex counteracts post-diet weight regain in obesity. NeuroImage. 2015;109:318–327. doi: 10.1016/j.neuroimage.2014.12.073. [DOI] [PubMed] [Google Scholar]
- 154.Wijngaarden M.A., Veer I.M., Rombouts S.A., van Buchem M.A., Willems van Dijk K., Pijl H., et al. Obesity is marked by distinct functional connectivity in brain networks involved in food reward and salience. Behav Brain Res. 2015;287:127–134. doi: 10.1016/j.bbr.2015.03.016. [DOI] [PubMed] [Google Scholar]
- 155.Baboumian S., Pantazatos S.P., Kothari S., McGinty J., Holst J., Geliebter A. Functional magnetic resonance imaging (fMRI) of neural responses to visual and auditory food stimuli pre and post roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG) Neuroscience. 2019;409:290–298. doi: 10.1016/j.neuroscience.2019.01.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Dong T.S., Gupta A., Jacobs J.P., Lagishetty V., Gallagher E., Bhatt R.R., et al. Improvement in uncontrolled eating behavior after laparoscopic sleeve gastrectomy is associated with alterations in the brain-gut-microbiome Axis in obese women. Nutrients. 2020;12 doi: 10.3390/nu12102924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Hu Y., Ji G., Li G., Manza P., Zhang W., Wang J., et al. Brain connectivity, and hormonal and behavioral correlates of sustained weight loss in obese patients after laparoscopic sleeve gastrectomy. Cerebr Cortex. 2021;31:1284–1295. doi: 10.1093/cercor/bhaa294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Wiemerslage L., Zhou W., Olivo G., Stark J., Hogenkamp P.S., Larsson E.M., et al. A resting-state fMRI study of obese females between pre- and postprandial states before and after bariatric surgery. Eur J Neurosci. 2017;45:333–341. doi: 10.1111/ejn.13428. [DOI] [PubMed] [Google Scholar]
- 159.Bach P., Grosshans M., Koopmann A., Pfeifer A.M., Vollstadt-Klein S., Otto M., et al. Predictors of weight loss in participants with obesity following bariatric surgery - a prospective longitudinal fMRI study. Appetite. 2021;163:105237. doi: 10.1016/j.appet.2021.105237. [DOI] [PubMed] [Google Scholar]
- 160.Olivo G., Zhou W., Sundbom M., Zhukovsky C., Hogenkamp P., Nikontovic L., et al. Resting-state brain connectivity changes in obese women after Roux-en-Y gastric bypass surgery: a longitudinal study. Sci Rep. 2017;7:6616. doi: 10.1038/s41598-017-06663-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Salem V., Demetriou L., Behary P., Alexiadou K., Scholtz S., Tharakan G., et al. Weight loss by low-calorie diet versus gastric bypass surgery in people with diabetes results in divergent brain activation patterns: a functional MRI study. Diabetes Care. 2021;44:1842–1851. doi: 10.2337/dc20-2641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Ten Kulve J.S., Veltman D.J., Gerdes V.E.A., van Bloemendaal L., Barkhof F., Deacon C.F., et al. Elevated postoperative endogenous GLP-1 levels mediate effects of roux-en-Y gastric bypass on neural responsivity to food cues. Diabetes Care. 2017;40:1522–1529. doi: 10.2337/dc16-2113. [DOI] [PubMed] [Google Scholar]
- 163.van Duinkerken E., Bernardes G., van Bloemendaal L., Veltman D.J., Barkhof F., Mograbi D.C., et al. Cerebral effects of glucagon-like peptide-1 receptor blockade before and after Roux-en-Y gastric bypass surgery in obese women: a proof-of-concept resting-state functional MRI study. Diabetes Obes Metabol. 2021;23:415–424. doi: 10.1111/dom.14233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Zoon H.F.A., de Bruijn S.E.M., Jager G., Smeets P.A.M., de Graaf C., Janssen I.M.C., et al. Altered neural inhibition responses to food cues after Roux-en-Y Gastric Bypass. Biol Psychol. 2018;137:34–41. doi: 10.1016/j.biopsycho.2018.06.005. [DOI] [PubMed] [Google Scholar]
- 165.Zoon H.F.A., de Bruijn S.E.M., Smeets P.A.M., de Graaf C., Janssen I.M.C., Schijns W., et al. Altered neural responsivity to food cues in relation to food preferences, but not appetite-related hormone concentrations after RYGB-surgery. Behav Brain Res. 2018;353:194–202. doi: 10.1016/j.bbr.2018.07.016. [DOI] [PubMed] [Google Scholar]
- 166.Cerit H., Davidson P., Hye T., Moondra P., Haimovici F., Sogg S., et al. Resting-state brain connectivity predicts weight loss and cognitive control of eating behavior after vertical sleeve gastrectomy. Obesity. 2019;27:1846–1855. doi: 10.1002/oby.22607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Smith K.R., Papantoni A., Veldhuizen M.G., Kamath V., Harris C., Moran T.H., et al. Taste-related reward is associated with weight loss following bariatric surgery. J Clin Invest. 2020;130:4370–4381. doi: 10.1172/JCI137772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Goldstone A.P., Miras A.D., Scholtz S., Jackson S., Neff K.J., Penicaud L., 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. doi: 10.1210/jc.2015-2665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Holsen L.M., Davidson P., Cerit H., Hye T., Moondra P., Haimovici F., et al. Neural predictors of 12-month weight loss outcomes following bariatric surgery. Int J Obes. 2018;42:785–793. doi: 10.1038/ijo.2017.190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Scholtz S., Miras A.D., Chhina N., Prechtl C.G., Sleeth M.L., Daud N.M., et al. Obese patients after gastric bypass surgery have lower brain-hedonic responses to food than after gastric banding. Gut. 2014;63:891–902. doi: 10.1136/gutjnl-2013-305008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Heinrichs H.S., Beyer F., Medawar E., Prehn K., Ordemann J., Floel A., et al. Effects of bariatric surgery on functional connectivity of the reward and default mode network: a pre-registered analysis. Hum Brain Mapp. 2021;42:5357–5373. doi: 10.1002/hbm.25624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Hajnal A., Kovacs P., Ahmed T.A., Meirelles K., Lynch C.J., Cooney R.N. Gastric bypass surgery alters behavioral and neural taste functions for sweet taste in obese rats. Am J Physiol Gastrointest Liver Physiol. 2010;299:G967–G979. doi: 10.1152/ajpgi.00070.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Saeidi N., Nestoridi E., Kucharczyk J., Uygun M.K., Yarmush M.L., Stylopoulos N. Sleeve gastrectomy and Roux-en-Y gastric bypass exhibit differential effects on food preferences, nutrient absorption and energy expenditure in obese rats. Int J Obes. 2012;36:1396–1402. doi: 10.1038/ijo.2012.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Shin A.C., Berthoud H.R. Food reward functions as affected by obesity and bariatric surgery. Int J Obes. 2011;35(Suppl 3):S40–S44. doi: 10.1038/ijo.2011.147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Shin A.C., Townsend R.L., Patterson L.M., Berthoud H.R. "Liking" and "wanting" of sweet and oily food stimuli as affected by high-fat diet-induced obesity, weight loss, leptin, and genetic predisposition. Am J Physiol Regul Integr Comp Physiol. 2011;301:R1267–R1280. doi: 10.1152/ajpregu.00314.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Shin A.C., Zheng H., Pistell P.J., Berthoud H.R. Roux-en-Y gastric bypass surgery changes food reward in rats. Int J Obes. 2011;35:642–651. doi: 10.1038/ijo.2010.174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Hyde K.M., Blonde G.D., Bueter M., le Roux C.W., Spector A.C. Gastric bypass in female rats lowers concentrated sugar solution intake and preference without affecting brief-access licking after long-term sugar exposure. Am J Physiol Regul Integr Comp Physiol. 2020;318:R870–R885. doi: 10.1152/ajpregu.00240.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Mathes C.M., Bohnenkamp R.A., Blonde G.D., Letourneau C., Corteville C., Bueter M., et al. Gastric bypass in rats does not decrease appetitive behavior towards sweet or fatty fluids despite blunting preferential intake of sugar and fat. Physiol Behav. 2015;142:179–188. doi: 10.1016/j.physbeh.2015.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Reddy I.A., Wasserman D.H., Ayala J.E., Hasty A.H., Abumrad N.N., Galli A. Striatal dopamine homeostasis is altered in mice following Roux-en-Y gastric bypass surgery. ACS Chem Neurosci. 2014;5:943–951. doi: 10.1021/cn500137d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.McGregor M., Hamilton J., Hajnal A., Thanos P.K. Roux-en-Y gastric bypass in rat reduces mu-opioid receptor levels in brain regions associated with stress and energy regulation. PLoS One. 2019;14 doi: 10.1371/journal.pone.0218680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.McGregor M., Hamilton J., Hajnal A., Thanos P.K. Roux-en-Y gastric bypass increases GABA-A receptor levels in regions of the rat brain involved in object recognition memory and perceptual acuity. Physiol Behav. 2020;224:113053. doi: 10.1016/j.physbeh.2020.113053. [DOI] [PubMed] [Google Scholar]
- 182.Gautier Y., Bergeat D., Serrand Y., Rethore N., Maherault M., Malbert C.H., et al. Western diet, obesity and bariatric surgery sequentially modulated anxiety, eating patterns and brain responses to sucrose in adult Yucatan minipigs. Sci Rep. 2020;10:20130. doi: 10.1038/s41598-020-76910-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Hankir M.K., Seyfried F., Hintschich C.A., Diep T.A., Kleberg K., Kranz M., et al. Gastric bypass surgery recruits a gut PPAR-alpha-striatal D1R pathway to reduce fat appetite in obese rats. Cell Metabol. 2017;25:335–344. doi: 10.1016/j.cmet.2016.12.006. [DOI] [PubMed] [Google Scholar]
- 184.Chambers A.P., Kirchner H., Wilson-Perez H.E., Willency J.A., Hale J.E., Gaylinn B.D., et al. The effects of vertical sleeve gastrectomy in rodents are ghrelin independent. Gastroenterology. 2013;144:50–52 e55. doi: 10.1053/j.gastro.2012.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Mokadem M., Zechner J.F., Margolskee R.F., Drucker D.J., Aguirre V. Effects of Roux-en-Y gastric bypass on energy and glucose homeostasis are preserved in two mouse models of functional glucagon-like peptide-1 deficiency. Mol Metabol. 2014;3:191–201. doi: 10.1016/j.molmet.2013.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Wilson-Perez H.E., Chambers A.P., Ryan K.K., Li B., Sandoval D.A., Stoffers D., et al. Vertical sleeve gastrectomy is effective in two genetic mouse models of glucagon-like Peptide 1 receptor deficiency. Diabetes. 2013;62:2380–2385. doi: 10.2337/db12-1498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Ye J., Hao Z., Mumphrey M.B., Townsend R.L., Patterson L.M., Stylopoulos N., et al. GLP-1 receptor signaling is not required for reduced body weight after RYGB in rodents. Am J Physiol Regul Integr Comp Physiol. 2014;306:R352–R362. doi: 10.1152/ajpregu.00491.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Morrison C.D., Hao Z., Mumphrey M.B., Townsend R.L., Munzberg H., Ye J., et al. Roux-en-Y gastric bypass surgery is effective in fibroblast growth factor-21 deficient mice. Mol Metabol. 2016;5:1006–1014. doi: 10.1016/j.molmet.2016.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Hao Z., Leigh Townsend R., Mumphrey M.B., Gettys T.W., Yu S., Munzberg H., et al. Roux-en-Y gastric bypass surgery-induced weight loss and metabolic improvements are similar in TGR5-deficient and wildtype mice. Obes Surg. 2018;28:3227–3236. doi: 10.1007/s11695-018-3297-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Li K., Zou J., Li S., Guo J., Shi W., Wang B., et al. Farnesoid X receptor contributes to body weight-independent improvements in glycemic control after Roux-en-Y gastric bypass surgery in diet-induced obese mice. Mol Metabol. 2020;37:100980. doi: 10.1016/j.molmet.2020.100980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Carmody J.S., Ahmad N.N., Machineni S., Lajoie S., Kaplan L.M. Weight loss after RYGB is independent of and complementary to serotonin 2C receptor signaling in male mice. Endocrinology. 2015;156:3183–3191. doi: 10.1210/en.2015-1226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Arble D.M., Sandoval D.A., Turek F.W., Woods S.C., Seeley R.J. Metabolic effects of bariatric surgery in mouse models of circadian disruption. Int J Obes. 2015;39:1310–1318. doi: 10.1038/ijo.2015.54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Ratner C., Shin J.H., Dwibedi C., Tremaroli V., Bjerregaard A., Hartmann B., et al. Anorexia and fat aversion induced by vertical sleeve gastrectomy is attenuated in neurotensin receptor 1-deficient mice. Endocrinology. 2021;162 doi: 10.1210/endocr/bqab130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Faramia J., Hao Z., Mumphrey M.B., Townsend R.L., Miard S., Carreau A.M., et al. IGFBP-2 partly mediates the early metabolic improvements caused by bariatric surgery. Cell Rep Med. 2021;2:100248. doi: 10.1016/j.xcrm.2021.100248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Batterham R.L., Cowley M.A., Small C.J., Herzog H., Cohen M.A., Dakin C.L., et al. Gut hormone PYY(3-36) physiologically inhibits food intake. Nature. 2002;418:650–654. doi: 10.1038/nature00887. [DOI] [PubMed] [Google Scholar]
- 196.Chelikani P.K., Haver A.C., Reidelberger R.D. Intravenous infusion of glucagon-like peptide-1 potently inhibits food intake, sham feeding, and gastric emptying in rats. Am J Physiol Regul Integr Comp Physiol. 2005;288:R1695–R1706. doi: 10.1152/ajpregu.00870.2004. [DOI] [PubMed] [Google Scholar]
- 197.Gutzwiller J.P., Goke B., Drewe J., Hildebrand P., Ketterer S., Handschin D., et al. Glucagon-like peptide-1: a potent regulator of food intake in humans. Gut. 1999;44:81–86. doi: 10.1136/gut.44.1.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Toft-Nielsen M.B., Madsbad S., Holst J.J. Continuous subcutaneous infusion of glucagon-like peptide 1 lowers plasma glucose and reduces appetite in type 2 diabetic patients. Diabetes Care. 1999;22:1137–1143. doi: 10.2337/diacare.22.7.1137. [DOI] [PubMed] [Google Scholar]
- 199.Turton M.D., O'Shea D., Gunn I., Beak S.A., Edwards C.M., Meeran K., et al. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature. 1996;379:69–72. doi: 10.1038/379069a0. [DOI] [PubMed] [Google Scholar]
- 200.Blundell J., Finlayson G., Axelsen M., Flint A., Gibbons C., Kvist T., et al. Effects of once-weekly semaglutide on appetite, energy intake, control of eating, food preference and body weight in subjects with obesity. Diabetes Obes Metabol. 2017;19:1242–1251. doi: 10.1111/dom.12932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Chao A.M., Wadden T.A., Walsh O.A., Gruber K.A., Alamuddin N., Berkowitz R.I., et al. Effects of liraglutide and behavioral weight loss on food cravings, eating behaviors, and eating disorder psychopathology. Obesity. 2019;27:2005–2010. doi: 10.1002/oby.22653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Christou G.A., Katsiki N., Blundell J., Fruhbeck G., Kiortsis D.N. Semaglutide as a promising antiobesity drug. Obes Rev. 2019;20:805–815. doi: 10.1111/obr.12839. [DOI] [PubMed] [Google Scholar]
- 203.Wadden T.A., Walsh O.A., Berkowitz R.I., Chao A.M., Alamuddin N., Gruber K., et al. Intensive behavioral therapy for obesity combined with liraglutide 3.0 mg: a randomized controlled trial. Obesity. 2019;27:75–86. doi: 10.1002/oby.22359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Carmody J.S., Munoz R., Yin H., Kaplan L.M. Peripheral, but not central, GLP-1 receptor signaling is required for improvement in glucose tolerance after Roux-en-Y gastric bypass in mice. Am J Physiol Endocrinol Metabol. 2016;310:E855–E861. doi: 10.1152/ajpendo.00412.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Garibay D., McGavigan A.K., Lee S.A., Ficorilli J.V., Cox A.L., Michael M.D., et al. Beta-cell glucagon-like peptide-1 receptor contributes to improved glucose tolerance after vertical sleeve gastrectomy. Endocrinology. 2016;157:3405–3409. doi: 10.1210/en.2016-1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Brierley D.I., Holt M.K., Singh A., de Araujo A., McDougle M., Vergara M., et al. Central and peripheral GLP-1 systems independently suppress eating. Nat Metabol. 2021;3:258–273. doi: 10.1038/s42255-021-00344-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Drucker D.J. GLP-1 physiology informs the pharmacotherapy of obesity. Mol Metabol. 2021:101351. doi: 10.1016/j.molmet.2021.101351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Krieger J.P. Intestinal glucagon-like peptide-1 effects on food intake: physiological relevance and emerging mechanisms. Peptides. 2020;131:170342. doi: 10.1016/j.peptides.2020.170342. [DOI] [PubMed] [Google Scholar]
- 209.Krieger J.P., Arnold M., Pettersen K.G., Lossel P., Langhans W., Lee S.J. Knockdown of GLP-1 receptors in vagal afferents affects normal food intake and glycemia. Diabetes. 2016;65:34–43. doi: 10.2337/db15-0973. [DOI] [PubMed] [Google Scholar]
- 210.Muller T.D., Finan B., Bloom S.R., D'Alessio D., Drucker D.J., Flatt P.R., et al. Glucagon-like peptide 1 (GLP-1) Mol Metabol. 2019;30:72–130. doi: 10.1016/j.molmet.2019.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Nakabayashi H., Nishizawa M., Nakagawa A., Takeda R., Niijima A. Vagal hepatopancreatic reflex effect evoked by intraportal appearance of tGLP-1. Am J Physiol. 1996;271:E808–E813. doi: 10.1152/ajpendo.1996.271.5.E808. [DOI] [PubMed] [Google Scholar]
- 212.Nishizawa M., Nakabayashi H., Uchida K., Nakagawa A., Niijima A. The hepatic vagal nerve is receptive to incretin hormone glucagon-like peptide-1, but not to glucose-dependent insulinotropic polypeptide, in the portal vein. J Auton Nerv Syst. 1996;61:149–154. doi: 10.1016/s0165-1838(96)00071-9. [DOI] [PubMed] [Google Scholar]
- 213.Vahl T.P., Tauchi M., Durler T.S., Elfers E.E., Fernandes T.M., Bitner R.D., et al. Glucagon-like peptide-1 (GLP-1) receptors expressed on nerve terminals in the portal vein mediate the effects of endogenous GLP-1 on glucose tolerance in rats. Endocrinology. 2007;148:4965–4973. doi: 10.1210/en.2006-0153. [DOI] [PubMed] [Google Scholar]
- 214.Hao Z., Townsend R.L., Mumphrey M.B., Patterson L.M., Ye J., Berthoud H.R. Vagal innervation of intestine contributes to weight loss after Roux-en-Y gastric bypass surgery in rats. Obes Surg. 2014;24:2145–2151. doi: 10.1007/s11695-014-1338-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Berthoud H.R., Carlson N.R., Powley T.L. Topography of efferent vagal innervation of the rat gastrointestinal tract. Am J Physiol. 1991;260:R200–R207. doi: 10.1152/ajpregu.1991.260.1.R200. [DOI] [PubMed] [Google Scholar]
- 216.Berthoud H.R., Kressel M., Neuhuber W.L. An anterograde tracing study of the vagal innervation of rat liver, portal vein and biliary system. Anat Embryol. 1992;186:431–442. doi: 10.1007/BF00185458. [DOI] [PubMed] [Google Scholar]
- 217.Shin A.C., Zheng H., Berthoud H.R. Vagal innervation of the hepatic portal vein and liver is not necessary for Roux-en-Y gastric bypass surgery-induced hypophagia, weight loss, and hypermetabolism. Ann Surg. 2012;255:294–301. doi: 10.1097/SLA.0b013e31823e71b7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Kanoski S.E., Fortin S.M., Arnold M., Grill H.J., Hayes M.R. Peripheral and central GLP-1 receptor populations mediate the anorectic effects of peripherally administered GLP-1 receptor agonists, liraglutide and exendin-4. Endocrinology. 2011;152:3103–3112. doi: 10.1210/en.2011-0174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Talsania T., Anini Y., Siu S., Drucker D.J., Brubaker P.L. Peripheral exendin-4 and peptide YY(3-36) synergistically reduce food intake through different mechanisms in mice. Endocrinology. 2005;146:3748–3756. doi: 10.1210/en.2005-0473. [DOI] [PubMed] [Google Scholar]
- 220.Chandarana K., Gelegen C., Karra E., Choudhury A.I., Drew M.E., Fauveau V., et al. Diet and gastrointestinal bypass-induced weight loss: the roles of ghrelin and peptide YY. Diabetes. 2011;60:810–818. doi: 10.2337/db10-0566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Kjaergaard M., Salinas C.B.G., Rehfeld J.F., Secher A., Raun K., Wulff B.S. PYY(3-36) and exendin-4 reduce food intake and activate neuronal circuits in a synergistic manner in mice. Neuropeptides. 2019;73:89–95. doi: 10.1016/j.npep.2018.11.004. [DOI] [PubMed] [Google Scholar]
- 222.Schmidt J.B., Gregersen N.T., Pedersen S.D., Arentoft J.L., Ritz C., Schwartz T.W., et al. Effects of PYY3-36 and GLP-1 on energy intake, energy expenditure, and appetite in overweight men. Am J Physiol Endocrinol Metabol. 2014;306:E1248–E1256. doi: 10.1152/ajpendo.00569.2013. [DOI] [PubMed] [Google Scholar]
- 223.Boland B.B., Mumphrey M.B., Hao Z., Townsend R.L., Gill B., Oldham S., et al. Combined loss of GLP-1R and Y2R does not alter progression of high-fat diet-induced obesity or response to RYGB surgery in mice. Mol Metabol. 2019;25:64–72. doi: 10.1016/j.molmet.2019.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Alnouti Y. Bile Acid sulfation: a pathway of bile acid elimination and detoxification. Toxicol Sci. 2009;108:225–246. doi: 10.1093/toxsci/kfn268. [DOI] [PubMed] [Google Scholar]
- 225.Fiamoncini J., Yiorkas A.M., Gedrich K., Rundle M., Alsters S.I., Roeselers G., et al. Determinants of postprandial plasma bile acid kinetics in human volunteers. Am J Physiol Gastrointest Liver Physiol. 2017;313:G300–G312. doi: 10.1152/ajpgi.00157.2017. [DOI] [PubMed] [Google Scholar]
- 226.Sonne D.P., van Nierop F.S., Kulik W., Soeters M.R., Vilsboll T., Knop F.K. Postprandial plasma concentrations of individual bile acids and FGF-19 in patients with type 2 diabetes. J Clin Endocrinol Metab. 2016;101:3002–3009. doi: 10.1210/jc.2016-1607. [DOI] [PubMed] [Google Scholar]
- 227.Chiang J.Y. Bile acid metabolism and signaling. Compr Physiol. 2013;3:1191–1212. doi: 10.1002/cphy.c120023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Ridlon J.M., Kang D.J., Hylemon P.B. Bile salt biotransformations by human intestinal bacteria. J Lipid Res. 2006;47:241–259. doi: 10.1194/jlr.R500013-JLR200. [DOI] [PubMed] [Google Scholar]
- 229.Ticho A.L., Malhotra P., Dudeja P.K., Gill R.K., Alrefai W.A. Bile acid receptors and gastrointestinal functions. Liver Res. 2019;3:31–39. doi: 10.1016/j.livres.2019.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Calderon G., McRae A., Rievaj J., Davis J., Zandvakili I., Linker-Nord S., et al. Ileo-colonic delivery of conjugated bile acids improves glucose homeostasis via colonic GLP-1-producing enteroendocrine cells in human obesity and diabetes. EBioMedicine. 2020;55:102759. doi: 10.1016/j.ebiom.2020.102759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Ahmad N.N., Pfalzer A., Kaplan L.M. Roux-en-Y gastric bypass normalizes the blunted postprandial bile acid excursion associated with obesity. Int J Obes. 2013;37:1553–1559. doi: 10.1038/ijo.2013.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Albaugh V.L., Flynn C.R., Cai S., Xiao Y., Tamboli R.A., Abumrad N.N. Early increases in bile acids post roux-en-Y gastric bypass are driven by insulin-sensitizing, secondary bile acids. J Clin Endocrinol Metab. 2015;100:E1225–E1233. doi: 10.1210/jc.2015-2467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Jahansouz C., Xu H., Hertzel A.V., Serrot F.J., Kvalheim N., Cole A., et al. Bile acids increase independently from hypocaloric restriction after bariatric surgery. Ann Surg. 2016;264:1022–1028. doi: 10.1097/SLA.0000000000001552. [DOI] [PubMed] [Google Scholar]
- 234.Jorgensen N.B., Dirksen C., Bojsen-Moller K.N., Kristiansen V.B., Wulff B.S., Rainteau D., et al. Improvements in glucose metabolism early after gastric bypass surgery are not explained by increases in total bile acids and fibroblast growth factor 19 concentrations. J Clin Endocrinol Metabol. 2015;100:E396–E406. doi: 10.1210/jc.2014-1658. [DOI] [PubMed] [Google Scholar]
- 235.Nakatani H., Kasama K., Oshiro T., Watanabe M., Hirose H., Itoh H. Serum bile acid along with plasma incretins and serum high-molecular weight adiponectin levels are increased after bariatric surgery. Metab Clin Exp. 2009;58:1400–1407. doi: 10.1016/j.metabol.2009.05.006. [DOI] [PubMed] [Google Scholar]
- 236.Patti M.E., Houten S.M., Bianco A.C., Bernier R., Larsen P.R., Holst J.J., et al. Serum bile acids are higher in humans with prior gastric bypass: potential contribution to improved glucose and lipid metabolism. Obesity. 2009;17:1671–1677. doi: 10.1038/oby.2009.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Spinelli V., Lalloyer F., Baud G., Osto E., Kouach M., Daoudi M., et al. Influence of Roux-en-Y gastric bypass on plasma bile acid profiles: a comparative study between rats, pigs and humans. Int J Obes. 2016;40:1260–1267. doi: 10.1038/ijo.2016.46. [DOI] [PubMed] [Google Scholar]
- 238.Myronovych A., Salazar-Gonzalez R.M., Ryan K.K., Miles L., Zhang W., Jha P., et al. The role of small heterodimer partner in nonalcoholic fatty liver disease improvement after sleeve gastrectomy in mice. Obesity. 2014;22:2301–2311. doi: 10.1002/oby.20890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Kohli R., Bradley D., Setchell K.D., Eagon J.C., Abumrad N., Klein S. Weight loss induced by Roux-en-Y gastric bypass but not laparoscopic adjustable gastric banding increases circulating bile acids. J Clin Endocrinol Metab. 2013;98:E708–E712. doi: 10.1210/jc.2012-3736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Werling M., Vincent R.P., Cross G.F., Marschall H.U., Fandriks L., Lonroth H., et al. Enhanced fasting and post-prandial plasma bile acid responses after Roux-en-Y gastric bypass surgery. Scand J Gastroenterol. 2013;48:1257–1264. doi: 10.3109/00365521.2013.833647. [DOI] [PubMed] [Google Scholar]
- 241.Yang C., Brecht J., Weiss C., Reissfelder C., Otto M., Buchwald J.N., et al. Serum glucagon, bile acids, and FGF-19: metabolic behavior patterns after roux-en-Y gastric bypass and vertical sleeve gastrectomy. Obes Surg. 2021;31:4939–4946. doi: 10.1007/s11695-021-05677-3. [DOI] [PubMed] [Google Scholar]
- 242.Myronovych A., Kirby M., Ryan K.K., Zhang W., Jha P., Setchell K.D., et al. Vertical sleeve gastrectomy reduces hepatic steatosis while increasing serum bile acids in a weight-loss-independent manner. Obesity. 2014;22:390–400. doi: 10.1002/oby.20548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Ryan K.K., Tremaroli V., Clemmensen C., Kovatcheva-Datchary P., Myronovych A., Karns R., et al. FXR is a molecular target for the effects of vertical sleeve gastrectomy. Nature. 2014;509:183–188. doi: 10.1038/nature13135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Albaugh V.L., Banan B., Antoun J., Xiong Y., Guo Y., Ping J., et al. Role of bile acids and GLP-1 in mediating the metabolic improvements of bariatric surgery. Gastroenterology. 2019;156:1041–1051 e1044. doi: 10.1053/j.gastro.2018.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.McGavigan A.K., Garibay D., Henseler Z.M., Chen J., Bettaieb A., Haj F.G., et al. TGR5 contributes to glucoregulatory improvements after vertical sleeve gastrectomy in mice. Gut. 2017;66:226–234. doi: 10.1136/gutjnl-2015-309871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Quante M., Iske J., Heinbokel T., Desai B.N., Cetina Biefer H.R., Nian Y., et al. Restored TDCA and valine levels imitate the effects of bariatric surgery. Elife. 2021;10 doi: 10.7554/eLife.62928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Chaudhari S.N., Harris D.A., Aliakbarian H., Luo J.N., Henke M.T., Subramaniam R., et al. Bariatric surgery reveals a gut-restricted TGR5 agonist with anti-diabetic effects. Nat Chem Biol. 2021;17:20–29. doi: 10.1038/s41589-020-0604-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Castellanos-Jankiewicz A., Guzman-Quevedo O., Fenelon V.S., Zizzari P., Quarta C., Bellocchio L., et al. Hypothalamic bile acid-TGR5 signaling protects from obesity. Cell Metabol. 2021;33:1483–1492 e1410. doi: 10.1016/j.cmet.2021.04.009. [DOI] [PubMed] [Google Scholar]
- 249.Reddy I.A., Smith N.K., Erreger K., Ghose D., Saunders C., Foster D.J., et al. Bile diversion, a bariatric surgery, and bile acid signaling reduce central cocaine reward. PLoS Biology. 2018;16 doi: 10.1371/journal.pbio.2006682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Fouladi F., Brooks A.E., Fodor A.A., Carroll I.M., Bulik-Sullivan E.C., Tsilimigras M.C.B., et al. The role of the gut microbiota in sustained weight loss following roux-en-Y gastric bypass surgery. Obes Surg. 2019;29:1259–1267. doi: 10.1007/s11695-018-03653-y. [DOI] [PubMed] [Google Scholar]
- 251.Wei Y.X., Zheng K.Y., Wang Y.G. Gut microbiota-derived metabolites as key mucosal barrier modulators in obesity. World J Gastroenterol. 2021;27:5555–5565. doi: 10.3748/wjg.v27.i33.5555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Chaudhari S.N., Luo J.N., Harris D.A., Aliakbarian H., Yao L., Paik D., et al. A microbial metabolite remodels the gut-liver axis following bariatric surgery. Cell Host Microbe. 2021;29:408–424 e407. doi: 10.1016/j.chom.2020.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Abu El-Haija M., Ye Y., Chu Y., Herz H., Linden B., Shahi S.K., et al. Toll-like receptor 4 and myeloid differentiation factor 88 are required for gastric bypass-induced metabolic effects. Surg Obes Relat Dis. 2021;17:1996–2006. doi: 10.1016/j.soard.2021.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Hao Z., Munzberg H., Rezai-Zadeh K., Keenan M., Coulon D., Lu H., et al. Leptin deficient ob/ob mice and diet-induced obese mice responded differently to Roux-en-Y bypass surgery. Int J Obes. 2015;39:798–805. doi: 10.1038/ijo.2014.189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Mokadem M., Zechner J.F., Uchida A., Aguirre V. Leptin is required for glucose homeostasis after roux-en-Y gastric bypass in mice. PLoS One. 2015;10 doi: 10.1371/journal.pone.0139960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Gupta S.R., Zhou Y., Wadden T.A., Berkowitz R.I., Chao A.M. A systematic review of genetic correlates of weight loss after bariatric surgery. Obes Surg. 2021;31:4612–4623. doi: 10.1007/s11695-021-05585-6. [DOI] [PubMed] [Google Scholar]
- 257.Cooiman M.I., Alsters S.I.M., Duquesnoy M., Hazebroek E.J., Meijers-Heijboer H.J., Chahal H., et al. Long-term weight outcome after bariatric surgery in patients with melanocortin-4 receptor gene variants: a case-control study of 105 patients. Obes Surg. 2022;32:837–844. doi: 10.1007/s11695-021-05869-x. [DOI] [PubMed] [Google Scholar]
- 258.Bonnefond A., Keller R., Meyre D., Stutzmann F., Thuillier D., Stefanov D.G., et al. Eating behavior, low-frequency functional mutations in the melanocortin-4 receptor (MC4R) gene, and outcomes of bariatric operations: a 6-year prospective study. Diabetes Care. 2016;39:1384–1392. doi: 10.2337/dc16-0115. [DOI] [PubMed] [Google Scholar]
- 259.Hatoum I.J., Stylopoulos N., Vanhoose A.M., Boyd K.L., Yin D.P., Ellacott K.L., et al. Melanocortin-4 receptor signaling is required for weight loss after gastric bypass surgery. J Clin Endocrinol Metab. 2012;97:E1023–E1031. doi: 10.1210/jc.2011-3432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Zechner J.F., Mirshahi U.L., Satapati S., Berglund E.D., Rossi J., Scott M.M., et al. Weight-independent effects of roux-en-Y gastric bypass on glucose homeostasis via melanocortin-4 receptors in mice and humans. Gastroenterology. 2013;144:580–590 e587. doi: 10.1053/j.gastro.2012.11.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Mumphrey M.B., Hao Z., Townsend R.L., Patterson L.M., Morrison C.D., Munzberg H., et al. Reversible hyperphagia and obesity in rats with gastric bypass by central MC3/4R blockade. Obesity. 2014;22:1847–1853. doi: 10.1002/oby.20773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Song C.K., Vaughan C.H., Keen-Rhinehart E., Harris R.B., Richard D., Bartness T.J. Melanocortin-4 receptor mRNA expressed in sympathetic outflow neurons to brown adipose tissue: neuroanatomical and functional evidence. Am J Physiol Regul Integr Comp Physiol. 2008;295:R417–R428. doi: 10.1152/ajpregu.00174.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Austin K., Tsang D., Chalmers J.A., Maalouf M.F., Brubaker P.L. Insulin-like growth factor-binding protein-4 inhibits epithelial growth and proliferation in the rodent intestine. Am J Physiol Gastrointest Liver Physiol. 2018;315:G206–G219. doi: 10.1152/ajpgi.00349.2017. [DOI] [PubMed] [Google Scholar]
- 264.Maffei A., Segal A.M., Alvarez-Perez J.C., Garcia-Ocana A., Harris P.E. Anti-incretin, anti-proliferative action of dopamine on beta-cells. Mol Endocrinol. 2015;29:542–557. doi: 10.1210/me.2014-1273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Mingrone G., Castagneto-Gissey L. Mechanisms of early improvement/resolution of type 2 diabetes after bariatric surgery. Diabetes Metabol. 2009;35:518–523. doi: 10.1016/S1262-3636(09)73459-7. [DOI] [PubMed] [Google Scholar]
- 266.Thomas S., Schauer P. Bariatric surgery and the gut hormone response. Nutr Clin Pract. 2010;25:175–182. doi: 10.1177/0884533610361739. [DOI] [PubMed] [Google Scholar]
- 267.Knowler W.C., Barrett-Connor E., Fowler S.E., Hamman R.F., Lachin J.M., Walker E.A., et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med. 2002;346:393–403. doi: 10.1056/NEJMoa012512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Lean M.E., Leslie W.S., Barnes A.C., Brosnahan N., Thom G., McCombie L., et al. Primary care-led weight management for remission of type 2 diabetes (DiRECT): an open-label, cluster-randomised trial. Lancet. 2018;391:541–551. doi: 10.1016/S0140-6736(17)33102-1. [DOI] [PubMed] [Google Scholar]
- 269.Isbell J.M., Tamboli R.A., Hansen E.N., Saliba J., Dunn J.P., Phillips S.E., et al. The importance of caloric restriction in the early improvements in insulin sensitivity after Roux-en-Y gastric bypass surgery. Diabetes Care. 2010;33:1438–1442. doi: 10.2337/dc09-2107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Jackness C., Karmally W., Febres G., Conwell I.M., Ahmed L., Bessler M., et al. Very low-calorie diet mimics the early beneficial effect of Roux-en-Y gastric bypass on insulin sensitivity and beta-cell Function in type 2 diabetic patients. Diabetes. 2013;62:3027–3032. doi: 10.2337/db12-1762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Lingvay I., Guth E., Islam A., Livingston E. Rapid improvement in diabetes after gastric bypass surgery: is it the diet or surgery? Diabetes Care. 2013;36:2741–2747. doi: 10.2337/dc12-2316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Lips M.A., de Groot G.H., van Klinken J.B., Aarts E., Berends F.J., Janssen I.M., et al. Calorie restriction is a major determinant of the short-term metabolic effects of gastric bypass surgery in obese type 2 diabetic patients. Clin Endocrinol. 2013;80:834–842. doi: 10.1111/cen.12254. [DOI] [PubMed] [Google Scholar]
- 273.Schmidt J.B., Pedersen S.D., Gregersen N.T., Vestergaard L., Nielsen M.S., Ritz C., et al. Effects of RYGB on energy expenditure, appetite and glycaemic control: a randomized controlled clinical trial. Int J Obes. 2016;40:281–290. doi: 10.1038/ijo.2015.162. [DOI] [PubMed] [Google Scholar]
- 274.Yang F., Liu C., Liu X., Pan X., Li X., Tian L., et al. Effect of epidemic intermittent fasting on cardiometabolic risk factors: a systematic review and meta-analysis of randomized controlled trials. Front Nutr. 2021;8:669325. doi: 10.3389/fnut.2021.669325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Mingrone G., Cummings D.E. Changes of insulin sensitivity and secretion after bariatric/metabolic surgery. Surg Obes Relat Dis. 2016;12:1199–1205. doi: 10.1016/j.soard.2016.05.013. [DOI] [PubMed] [Google Scholar]
- 276.Aminian A., Zajichek A., Tu C., Wolski K.E., Brethauer S.A., Schauer P.R., et al. How much weight loss is required for cardiovascular benefits? Insights from a metabolic surgery matched-cohort study. Ann Surg. 2020;272:639–645. doi: 10.1097/SLA.0000000000004369. [DOI] [PubMed] [Google Scholar]
- 277.Abu-Gazala S., Horwitz E., Ben-Haroush Schyr R., Bardugo A., Israeli H., Hija A., et al. Sleeve gastrectomy improves glycemia independent of weight loss by restoring hepatic insulin sensitivity. Diabetes. 2018;67:1079–1085. doi: 10.2337/db17-1028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Chambers A.P., Jessen L., Ryan K.K., Sisley S., Wilson-Perez H.E., Stefater M.A., et al. Weight-independent changes in blood glucose homeostasis after gastric bypass or vertical sleeve gastrectomy in rats. Gastroenterology. 2011;141:950–958. doi: 10.1053/j.gastro.2011.05.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Salminen P., Helmio M., Ovaska J., Juuti A., Leivonen M., Peromaa-Haavisto P., et al. Effect of laparoscopic sleeve gastrectomy vs laparoscopic roux-en-Y gastric bypass on weight loss at 5 Years among patients with morbid obesity: the SLEEVEPASS randomized clinical trial. JAMA. 2018;319:241–254. doi: 10.1001/jama.2017.20313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Arterburn D.E., Johnson E., Coleman K.J., Herrinton L.J., Courcoulas A.P., Fisher D., et al. Weight outcomes of sleeve gastrectomy and gastric bypass compared to nonsurgical treatment. Ann Surg. 2021;274:e1269–e1276. doi: 10.1097/SLA.0000000000003826. [DOI] [PubMed] [Google Scholar]
- 281.Courcoulas A., Abu Dayyeh B.K., Eaton L., Robinson J., Woodman G., Fusco M., et al. Intragastric balloon as an adjunct to lifestyle intervention: a randomized controlled trial. Int J Obes. 2017;41:427–433. doi: 10.1038/ijo.2016.229. [DOI] [PubMed] [Google Scholar]
- 282.Stylopoulos N., Hoppin A.G., Kaplan L.M. Roux-en-Y gastric bypass enhances energy expenditure and extends lifespan in diet-induced obese rats. Obesity. 2009;17:1839–1847. doi: 10.1038/oby.2009.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Alvarez R., Lewis A.G., Toure M.H., Seeley R.J. A comparison of rodent models of vertical sleeve gastrectomy. Surg Obes Relat Dis. 2018;14:1471–1479. doi: 10.1016/j.soard.2018.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284.Abegg K., Corteville C., Bueter M., Lutz T.A. Alterations in energy expenditure in Roux-en-Y gastric bypass rats persist at thermoneutrality. Int J Obes. 2016;40:1215–1221. doi: 10.1038/ijo.2016.55. [DOI] [PubMed] [Google Scholar]
- 285.Basso N., Soricelli E., Castagneto-Gissey L., Casella G., Albanese D., Fava F., et al. Insulin resistance, microbiota, and fat distribution changes by a new model of vertical sleeve gastrectomy in obese rats. Diabetes. 2016;65:2990–3001. doi: 10.2337/db16-0039. [DOI] [PubMed] [Google Scholar]
- 286.Thanos P.K., Subrize M., Delis F., Cooney R.N., Culnan D., Sun M., et al. Gastric bypass increases ethanol and water consumption in diet-induced obese rats. Obes Surg. 2012;22:1884–1892. doi: 10.1007/s11695-012-0749-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Uchida A., Zechner J.F., Mani B.K., Park W.M., Aguirre V., Zigman J.M. Altered ghrelin secretion in mice in response to diet-induced obesity and Roux-en-Y gastric bypass. Mol Metabol. 2014;3:717–730. doi: 10.1016/j.molmet.2014.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288.Ding L., Sousa K.M., Jin L., Dong B., Kim B.W., Ramirez R., et al. Vertical sleeve gastrectomy activates GPBAR-1/TGR5 to sustain weight loss, improve fatty liver, and remit insulin resistance in mice. Hepatology. 2016;64:760–773. doi: 10.1002/hep.28689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289.Flynn C.R., Albaugh V.L., Cai S., Cheung-Flynn J., Williams P.E., Brucker R.M., et al. Bile diversion to the distal small intestine has comparable metabolic benefits to bariatric surgery. Nat Commun. 2015;6:7715. doi: 10.1038/ncomms8715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290.Du J., Tian J., Ding L., Trac C., Xia B., Sun S., et al. Vertical sleeve gastrectomy reverses diet-induced gene-regulatory changes impacting lipid metabolism. Sci Rep. 2017;7:5274. doi: 10.1038/s41598-017-05349-2. [DOI] [PMC free article] [PubMed] [Google Scholar]