Skip to main content
Erschienen in: Obesity Surgery 2/2016

Open Access 01.02.2016 | Review Article

Mechanism Underlying the Weight Loss and Complications of Roux-en-Y Gastric Bypass. Review

verfasst von: G Abdeen, CW le Roux

Erschienen in: Obesity Surgery | Ausgabe 2/2016

Abstract

Various bariatric surgical procedures are effective at improving health in patients with obesity associated co-morbidities, but the aim of this review is to specifically describe the mechanisms through which Roux-en-Y gastric bypass (RYGB) surgery enables weight loss for obese patients using observations from both human and animal studies. Perhaps most but not all clinicians would agree that the beneficial effects outweigh the harm of RYGB; however, the mechanisms for both the beneficial and deleterious (for example postprandial hypoglycaemia, vitamin deficiency and bone loss) effects are ill understood. The exaggerated release of the satiety gut hormones, such as GLP-1 and PYY, with their central and peripheral effects on food intake has given new insight into the physiological changes that happen after surgery. The initial enthusiasm after the discovery of the role of the gut hormones following RYGB may need to be tempered as the magnitude of the effects of these hormonal responses on weight loss may have been overestimated. The physiological changes after RYGB are unlikely to be due to a single hormone, or single mechanism, but most likely involve complex gut-brain signalling. Understanding the mechanisms involved with the beneficial and deleterious effects of RYGB will speed up the development of effective, cheaper and safer surgical and non-surgical treatments for obesity.

Introduction

The Roux-en-Y gastric bypass (RYGB) includes a small gastric pouch (15–30 mL) on the lesser gastric curvature [1, 2] which is completely divided from the gastric remnant and then anastomosed to the jejunum (leaving an alimentary or Roux limb of typically 100–150 cm). The size of the gastro-jejunal anastomosis is controversial as initially it was thought that an element of restriction may be helpful in slowing the progress of food from the oesophagus into the jejunum, but more recently the aim has been rapid transit of food into the jejunum to generate the gut signals to reduce meal size [3]. Bowel continuity is restored by an entero-entero anastomosis between the excluded biliopancreatic limb (BPL) and the alimentary limb. This anastomosis is usually performed 100–150 cm distal to the gastro-jejunostomy, although it has also been performed up to 250 cm distally in an attempt to create calorie malabsorption [1]. Usually, the BPL is approximately 50 cm, but since other operations such as the biliopancreatic diversion or mini-gastric bypass with much longer BPL have greater reduction in insulin resistance, renewed interest in the length of the biliopancreatic limb BPL has developed [4].
Operative times vary between 45 and 90 min and the average hospital stay is 1–3 days, although same-day discharge following RYGB procedure have been successful [5]. Early complications, within 30 days after surgery, do occur in approximately 4 % of patients and include bleeding, perforation or leakage, which need immediate surgical re-intervention [6]. Late complications such as significant abdominal pain, small bowel obstruction, anastomotic stenosis or marginal ulceration can occur in 15–20 % of patients after 30 days from surgery to over 10 years, and surgery or endoscopic therapy is often used for both diagnosis and/or treatment [7].
Even though RYGB does not treat some of the aetiological factors of morbid obesity, such as the obesogenic environment we live in, it does successfully result in 20–30 % long term, over 2 years of weight loss and maintenance [810], in addition to an improvement or remission of many obesity-related co-morbidities [1115] such as hypertension, type 2 diabetes mellitus, obstructive sleep apnoea and musculoskeletal pain. Approximately 40 % of obese patients with type 2 diabetes go into remission within days or weeks after RYGB [16].
The RYGB is the best studied procedure regarding underlying mechanisms. The aim of this review is to describe the mechanisms through which RYGB surgery enables weight loss for obese patients and helps in understanding its complications by using observations from both human and animal studies.

Food Intake

Research Studies

Hunger and Fullness

Lifestyle changes with a lower calorie diet can be effective at initiating weight loss; however, most of the results from randomised controlled trials (RCT) are disappointing regarding long-term weight loss maintenance [17, 18]. Approximately 70–80 % of patients fail to maintain their initial lifestyle-induced weight loss thought to be due to physiologically compensatory responses that defend the previous weight ‘set point’ [19]. Whilst on a long-term low-calorie diet, patients usually report an increase in hunger, a decrease in satiety and pre-occupation with energy-dense fatty and sweet food [20, 21]. This may be part of a normal physiological response and not due to lack of motivation.
Reduced calorie intake after RYGB is usually a consequence of significantly smaller meal sizes, and reduced calorie content of food eaten [22] compensated only partially by increased meal frequency [23]. Enhanced satiety is the dominant contributing factor [24]. A dramatic decrease in daily energy intake, 600–700 kcal [22, 25], during the first month post-surgery increases to 1000–1800 kcal during the first year [22, 2632]. An average reduction of 1800 kcal per day from pre-operative intake can be sustained for several years [32, 33]. Protein intake during the first year after surgery is often lower than recommended at 0.5 g/kg, rather than the recommendation of at least 1.5 g/kg/day [27, 34]. The mechanisms are unclear, but may be due to temporary intolerance of higher protein diet and dairy foods [22, 25, 27, 3537]. Relative intake of fat and carbohydrates decrease during the first year post-surgery, but return to the baseline after 1 year [22], although the contribution of high and low glycaemic index carbohydrates may change. Many patients reduce their intake of high glycaemic index carbohydrates and increase their intake of lower glycaemic index carbohydrates. Changes in behaviour associated with eating after RYGB were reported in the 1970s using structured interviews that suggested that patients reached satiety more quickly, with the most common reason given as a ‘lack of desire’ for food [38].

Potential Mediators

Increased Transit of Food into the Midgut Through the Gastric Pouch

Whether the size of the gastric pouch and stoma in RYGB surgery affects food intake and body weight is contested. It remains controversial in both the human and animal literature whether a larger gastric pouch and stoma causes less weight loss [3943]. The stoma becomes more ‘compliant’ with time, allowing food to transit more easily from the pouch into the alimentary limb, but may also result in food being ‘stored’ in the pouch and not emptying rapidly enough. Thus, the initial diameter of the anastomosis may not affect weight loss in the long term [44]. To study a RYGB technique that created a very small pouch, a high-pressure manometer was used, but a large stoma demonstrated that the pressure in the pouch (immediately proximal to the gastroenteral anastomosis) was lower than in the alimentary limb [45]. This suggests there was no restriction at the level of the stoma because of the absence of a high-pressure zone proximal to the pouch. Insertion of a gastric balloon into the alimentary limb and inflation of the balloon to a pressure of 20 cm water demonstrated that patients with the highest pressure generated by the alimentary limb had the smallest meal volume during an ad libitum meal. In contrast, those with the lowest pressure in the alimentary limb took longer to terminate their meal. Mechanoreceptors within the alimentary limb may be important determinants of meal size if food rapidly transits through the pouch to reach the alimentary limb in a less digested state than usual. The component that determines caloric intake may be the alimentary limb and not the pouch size or stoma diameter.

Hormonal

RYGB alters endogenous gut hormone responses to a meal. Glucagon-like peptide-1 (GLP-1), peptide YY (PYY) and ghrelin have been the best studied candidates in the context of reduced food intake and sustained weight loss after RYGB. GLP-1 and PYY responses to mixed meals or oral glucose have been at the centre of interest of several studies investigating patients 6 weeks to 10 years after RYGB [4651]. Significantly elevated responses are seen in GLP-1 and PYY as early as 2 days after RYGB [52] and may remain elevated for more than a decade after RYGB [53]. Patients who lost the most weight after RYGB also had the highest levels of these postprandial satiety gut hormones [54, 55]. Blocking the release of these hormones in humans and rats with octreotide increased food intake after RYGB, but not after adjustable gastric banding (AGB) surgery in humans [51] or sham operations in rats [56].
Mechanistic studies in rodents have suggested the physiological significance of PYY because weight loss in PYY-knockout mice after a RYGB variant was lower than in wild-type mice [57]. Exogenous PYY specific antibodies also increased food intake in rats after bypass type procedures [51]. Physiologically, PYY has been shown to delay gut transit time, but probably does not increase energy expenditure in human [58]. GLP-1 responses are very similar to those of PYY after RYGB, but have additionally been linked with increases in insulin secretion [59, 60]. Postprandial responses of GLP-1 before surgery do not correlate with change in weight loss after surgery, suggesting that pre-operative gut hormone responses are not prognostic [61]. Enhanced GLP-1 signalling on its own is also not sufficient to reduce body weight after RYGB, suggesting that it is multiple gut hormone responses that mediate the increased satiation after a meal [62].
Reduced ghrelin was the first proposed hormonal mechanism to explain weight loss after RYGB. At first, ghrelin levels were thought to be lower compared to diet-induced weight loss which increased ghrelin in a control group of subjects [63]. It was postulated that this decrease was partially responsible for reduced hunger after RYGB. Subsequent studies in patients after RYGB were more controversial reporting a reduction in fasting and postprandial ghrelin levels [50, 6470], no alteration in fasting and postprandial levels [51, 52, 7179] and a rise in fasting ghrelin levels [8084]. Considering all the data and variability, it is likely that RYGB results in a comparative ghrelin deficiency considering that ghrelin normally increases after diet-induced weight loss, but the magnitude of this contribution is unclear [85, 86].

Neural

The vagal afferent fibres in the gastric and proximal small bowel mucosa are known to be sensitive to mechanical stretch in order to detect the volume of ingested food [87]. The vagus nerve with both the ventral and dorsal gastric branches on the large gastric remnant is transected during the formation of the gastric pouch. The vagal fibres to the gastric pouch are thus intact, and these could mediate satiety as food passes through the pouch. The vagal denervation more distally may attenuate signalling. Taken together, this may play a role in satiation [88]. Visceral sensory information from the gut is communicated centrally using the afferent (sensory) vagus nerve signalling to the nucleus of the tractus solitarius (NTS). Here, visceral sensory information and hormonal and metabolic inputs are integrated together with neuronal inputs from other brainstem areas [89] and may well be the most important way in which RYGB signals to the brain. Transmission of these signals involving the gut hormones such as ghrelin may be impaired after vagotomy [90]. RYGB appears to have the potential to alter neural responses [91] to reduce hedonic behaviour associated with eating highly palatable and calorie-dense foods. These changes in reward value of food may alter the amount of food consumed [38, 9294].

Change in Bile Acids

Bile acids are agonists for the cell-membrane G protein-coupled receptors, TGR5, which in turn enhances the release of GLP-1 and PYY. Bile acids also bind the farnesoid X receptor (FXR) [95]. The anatomical changes after RYGB result in bile progressing down the biliopancreatic limb to the distal L cells without mixing with food. As a result, the availability of undiluted bile acids in the distal intestine may enhance stimulation of TGR5 receptors on L cells [96]. Serum bile acid concentration is raised after RYGB [97] and is associated with increased energy expenditure possibly through signalling via the cyclic adenosine monophosphate cAMP-dependent thyroid hormone triggering enzyme type 2 iodothyronine deiodinase [98]. Fibroblast growth factor (FGF) 19 is increased and binds to fibroblast growth factor receptor (FGFR4) activating fibroblast growth factor receptor c-kit (FGRR1c) in the presence of co-receptor β Klotho [99]. The result is increased protein synthesis in the liver [100]. FGF19 also plays a role in enhanced mitochondria activity [100]. Activation of the FXR receptor may facilitate the effects of bile acids on energy homeostasis through FGF19 that is released from ileal enterocytes which can lead to increases in metabolic rate and decreases in adiposity [101, 102].
Bile acids, after a mixed test meal in human subjects, was positively correlated with circulating GLP-1 and PYY, but negatively correlated with ghrelin [103]. Pournaras et al. have demonstrated that total plasma bile acids are elevated after RYGB [104] and suggested that they may be partly responsible for the intestinal hypertrophy, anorexigenic hormone secretion and alterations in gut microbiota [105].

Change in Gut Microbiota

Obesity is associated with low-grade inflammation, increased Firmicutes and decreased Bacteroidetes in animals [106] and humans [107109]. Intestinal microbiota has also been shown to utilise energy from food and thus increase the host’s energy-harvesting capacity [110]. Proteobacteria (gammaproteobacteria) has been shown to increase after RYGB in humans [111] with the major contributor being Enterobacter hormaechei. The significant improvement of weight, inflammation and metabolic status after surgery was associated with increased bacterial variety. An association was observed between adipose tissue gene expression and bacterial genes at baseline with a 10-fold increase 3 months after surgery, and this may suggest a restored crosstalk between both the gut microbiota and the host [112].
After RYGB, acidity was reduced in the alimentary limb leading to a decrease of hydrochloric acid flux in the gut, while bile acids were increased in the biliopancreatic limb. Bacteroidetes growth was attenuated at lower pH, whereas Escherichia coli increased at a higher pH. Gut microbiota quickly adapt in a ‘starvation-like state’ created by RYGB and rapidly and sustainably increase. Changes in microbiota in mice after RYGB were independent of weight alteration and caloric restriction [113]. Transfer of the gut microbiota from RYGB-treated mice to non-operated, germ-free mice resulted in weight loss and reduced fat mass in the recipient animals. The altered microbial production of short-chain fatty acids that increases may partly be an explanation [113]. Although RYGB did not change gut microbiota from the ‘obese state’ to the ‘lean state’, it did create a ‘third state’ which on balance appear to be associated with many of the beneficial characteristics of RYGB.

Food Preferences

Observations

Weight gain has been linked to a preference for both sweet and/or high-fat foods [114, 115], which may partly explain why obese people regain body weight frequently after ‘dieting’ [116, 117]. The common view summarised earlier by Pangborn and Simone is: “In the mind of a normal person, sugar and sweets are ‘fattening’ and most overweight people have a ‘sweet tooth’” [118]. Hedonism associated with palatable foods is considered a significant factor which increases the prevalence of obesity. A motivational factor that is referred to as ‘hedonic hunger’ [119] may be a trigger for overeating [120].
Patients after RYGB tend to increase the intake of fruit and vegetables as well as low-fat food [121, 122]. The dumping syndrome was thought to induce these changes in food preference [123], as initially it was considered as a useful characteristic of the RYGB to ‘teach’ patients to avoid calorie-dense foods and thus consume fewer calories [124]. However, patients after RYGB appear to make healthier food choices and adopt a more balanced diet (even when they do not experience dumping) [121, 125] and have considerable reduction in energy intake (EI) and energy density. A comparison on food groups was done for a group of patients after RYGB and total number of servings from fat, grains and sweetened beverages was reduced and remained reduced in the longer term. However, meats, dairy products, fruits and sweets were reduced in the short term, but then returned to baseline by 12 months [22]. When energy intake was reduced to 1300 kcal, 60 and 25 % of patients ,respectively, were consuming less than one serving per day from both fruits and vegetables. Whole grains intake increased from 25 to 40 % within the first 3 months, but then returned to baseline at 12 months [22]. The association between reduced diet energy density and weight loss is controversial as some studies describe no association [126], while others show that shifts in food preferences are partially responsible for the decreased calorie intake and weight loss after RYGB [127].
RYGB in humans appears to alter taste through unconditional and conditional mechanisms [24, 128130] leading to the concept of ‘behaviour surgery’ [123]. In 1987, Sugerman et al. reported that ‘sweet-eaters’ did particularly well after RYGB [131, 132]. Some of the initial findings were confounded by intolerance to sweets related to symptoms of the dumping syndrome [38, 131133]. Conditioned taste aversion may thus be a factor in some patients. These initial assumptions resulted in many clinicians thinking that the RYGB works by ‘punishing’ the ‘poor behaviours’ of obese patients. The notion that RYGB becomes an external enforcer that goes against the free will of the patient has led to some authors questioning the morality of RYGB as a tool that changes patients’ behaviour against patients’ natural wishes [134]. This misconception may have reduced the wider acceptance of RYGB as a valid physiological treatment for the pathology that results in obesity. Classical conditioned food aversion is, however, an unlikely explanation as most patients with severe dumping still report that they like the taste of sweet foods, but that they have learned to consume only small quantities that do not cause negative visceral symptoms or consume sweets at night before bedtime, suggesting a conditioned food avoidance to be a more likely explanation. Distinguishing between the terms is important because avoidance implies that the palatability of sweet or fat did not change when small quantities are consumed, but that the subject ‘learns’ to stop consuming the food sooner (earlier avoidance) because large quantities may have negative visceral consequences [135137].

Mediators

RYGB could be exerting its effects on food selection and preference through any one of the taste function domains important in normal physiology such as sensory-discriminative (stimulus identification), hedonic (ingestive motivation) and physiological (digestive preparation) [138, 139]. Affective responses to taste stimuli, which can be considered an example of ingestive motivation, can be both conditioned and unconditioned. It remains controversial which of these three domains are involved and what their interactions are to determine food preferences after RYGB surgery. For example, RYGB could have effects directly on the central gustatory pathways related with feeding and reward through gut hormonal mediators. Alternatively, changes in the sensory signals could alter the intensity or the quality of tastants, but also lead to an unconditioned change in palatability. If RYGB causes visceral malaise after ingestion of fat, then it is possible that the palatability of fat could alter through a process of learning (conditioned response) [140].
Although there are suggestions in animal models that the hedonic properties of sweet and fat stimuli may change after RYGB [23, 140144], less work has been done in humans. Miras et al. using the progressive ratio task showed that RYGB resulted in the selective decrease of the reward value of a sweet and fat tastant, but not vegetables [145]. Further support comes from studies of brain reward cognitive systems linked to eating behaviour as studied by functional MRI (fMRI), where brain hedonic responses to calorie-dense food are lower after RYGB compared to patients who have lost similar amounts of weight after adjustable gastric banding [128].

Energy Expenditure

According to the laws of thermodynamics, energy that enters a system (energy intake) must either be stored (body energy gain) or be used (activity, heat or faecal energy loss). Energy expenditure (EE) is usually decreased during food restriction, a phenomenon known as the ‘starvation response’ [146]. Weight loss in rodent models of RYGB is associated with preservation of lean body mass and increased EE [146]. Humans have decreased basal metabolic rate, but increased meal-induced thermogenesis after RYGB [32, 122, 147153]. Evidence is now also emerging to suggest that the metabolic rate of the small bowel is increased after RYGB with more carbohydrate consumption which may explain the changes observed in respiratory quotient after these operations [154]. Reduced resting energy expenditure (REE) or basal metabolic rate after RYGB [122, 147, 155157] may be attenuated due to relative lean mass preservation. Patients who regain the weight they lost 2 years after RYGB have lower REE [149], suggesting that elevating REE after RYGB may enhance weight loss. Physical activity may further help increase activity-related EE and also preserve lean mass, and therefore REE, after RYGB [158].

Calorie Malabsorption

Several bariatric operations were designed to result in malabsorption of calories [159]. The exclusion of the approximately 10 % of the bowel (50 cm of BPL) after RYGB is unlikely to result in calorie malabsorption usually during other small bowel resections. Moreover, the exaggerated gut hormone responses which reduce gut transit have a net result of RYGB not altering oro-caecal transit time or functional enterocyte mass [16]. RYGB may, however, impair pancreatic exocrine function which could contribute to a small amount of fat malabsorption, the magnitude of which is probably too small to contribute substantially to weight loss [160162].

Mechanisms of Complications

The rise in the number of RYGB procedures [163] has also increased the absolute number of complications associated with this procedure even though the percentage of patients with complications has reduced due to better surgical experience [164].
Postprandial hypoglycaemia, even in patients who never had type 2 diabetes, can occur several hours after a meal and is distinct from early dumping syndrome which occurs within minutes after eating [165, 166]. Early dumping is an outcome of rapid emptying of food into the jejunum due to the lack of a pylorus presumably causing neural activation in the proximal alimentary limb [167]. Late dumping, or ‘postprandial hypoglycaemia’, happens 1–3 h after ingesting a meal and is a result of the exaggerated insulin response to high glycaemic index carbohydrates in the meal. The proposed mechanisms involve increased β-cell mass and improved β-cell function and non-β-cell mechanisms, which may include a lack of ghrelin (a counter-regulatory measure to hypoglycaemia) [63, 168]. In addition, the sustained weight loss can reduce insulin resistance which renders the previous insulin responses needed pre-surgery to suddenly become excessive. The aetiology of hypoglycaemia is likely to be different for individual patients and is also probably a mixture of the anatomic, hormonal and metabolic changes after RYGB [169]. Although treatment of this complication can be difficult, pancreatectomies are no longer advised [170], but rather a multimodal medical approach is favoured [171].

Unexplained Abdominal Pain

Up to 10 % of the patients complain of unexplained chronic abdominal pain which can be difficult for both the treating clinician and patient to acknowledge [172, 173]. Mild abdominal pain is reported by up to 95 % of patients at some point after RYGB [172, 174176]. Symptom severity fluctuates between vague discomfort and severe colicky pain [177]. Vomiting and nausea, especially if prolonged, are symptoms of pathology and are not part of the normal postoperative course after RYGB; nonetheless, up to 80 % of patients report the symptoms at some point after surgery [172, 174]. Abdominal pain may be recurrent, and it should be remembered that internal hernias may spontaneously reduce causing intermittent pain. Early investigation when acute symptoms of abdominal pain first presents is mandatory due to the risk of obstruction, volvulus and ischaemia of the herniated bowel [172, 178]. Cross-sectional imaging is often unhelpful and the use of laparoscopy is frequently required for diagnosis. Management protocols for chronic unexplained abdominal pain are not clearly defined, but the jejunal-jejunal anastomosis is currently receiving more attention as a possible cause for these chronic problems.

Anastomotic Stenosis

With the circular stapler technique, this can be a common complication with a reported incidence of up to 27 % and a recurrence rate of up to 33 % [179]. Usually dysphagia occurs within 6 months after surgery. Endoscopy can often be used both as a diagnostic but also an intervention tool.

Vitamin Deficiencies: Iron, Vitamin B12, Folic Acid, Vitamin D and Calcium

Iron deficiency occurs in up to 49 % of patients after RYGB [180]. Reduced acid production in the small stomach pouch decreases iron absorption [181]. For iron to be absorbed, the ferric iron in foods has to be reduced to the ferrous state, but because hydrochloric acid is lower after RYGB, this process is attenuated [182]. Reduced intake of iron-rich foods after RYGB such as red meat may also contribute [183, 184].
In the stomach, both pepsin and hydrochloric acid are required for absorption of vitamin B12. Deficiencies of vitamin B12 occur in up to 70 % of patients after RYGB [184186] because achlorhydria prevents vitamin B12 separation from foods due to reduced ingestion of meat and insufficient secretion of intrinsic factor after surgery [182].
Folic acid deficiency affects up to 35 % of patients after RYGB. Folate absorption is enabled by hydrochloric acid with absorption in the proximal third of the small bowel most important [186]. Vitamin B12 also acts as a coenzyme in converting methyltetrahydrofolate to tetrahydrofolate. Thus, folate deficiency might result from achlorhydria, bypassing of the proximal small bowel, vitamin B12 deficiency and/or decreased folate ingestion [184187].
Hypocalcaemia occurs in up to 10 % and low serum 25-hydroxy vitamin D levels in up to half of RYGB patients [188]. Nevertheless, most obese patients had significantly lower basal 25-hydroxyl vitamin D concentrations and higher parathyroid hormone concentrations as compared to age-matched lean controls [189]. Deficiencies may occur because calcium is typically absorbed in the proximal small bowel which is bypassed after RYGB. Intolerances can develop to calcium-rich sources such as milk especially if the fat content is high. Calcium can be released from bone as evident from the increased bone turnover and subsequent reduced bone mass after RYGB [190, 191]. The higher bone turnover in the RYGB patients could be partly due to the weight loss in these patients [192], but animal studies suggest that bone loss exceeds what would be expected from weight loss alone [193].

Loss of Bone Density

Many patients with obesity have very healthy bone density before surgery due to long-term excessive weight bearing. This may be protective and partly explains the controversy of why the loss of bone density after RYGB does not cause more bone fractures [194, 195] even if the risk for fracturing may be increased. Multiple mechanisms may contribute to RYGB reducing bone density, including physiologically reduced mechanical load related to weight loss after surgery, hyperparathyroidism due to insufficient calcium consumption or reduced intestinal calcium and vitamin D absorption. Humoral factors from adipose tissue (oestradiol, leptin, adiponectin), pancreas (e.g. insulin, amylin) or the gut (ghrelin, glucagon-like peptide-2, glucose-dependent insulinotropic peptide) may also play a role [196, 197] by connecting a web of consistent regulatory pathways [196].

Kidney Stones

Hyperoxaluria is common after RYGB, but the incidence of renal calculi is much lower than after jejunal-ileal bypass (JIB) [198200]. Comparison with the JIB is important because the incidence as well as the potential mechanisms may be different after RYGB. The lithogenic effects after RYGB may stem from reduced calcium binding to oxalate in the intestinal lumen. The excess oxalate is then cleared by the kidneys resulting in hyperoxaluria and calcium oxalate nephrolithiasis. Almost 21 % of patients after JIB, which causes significant malabsorption, developed kidney stones 5 years after surgery [201], but the incidence of kidney stones after RYGB appears to depend on a combination of other factors such as hydration status and urine volume [200]. Patients in high stone-forming areas of the world have increased number of stones while those in low stone-forming countries may have an incidence similar to the background population [202]. Thus, RYGB alone is not enough to cause kidney stones, but it does potentiate other predisposing factors.

Conclusion

RYGB confers both benefits and complications, the mechanisms of which are still only partially understood. Most, but not all, clinicians would agree that the beneficial effects outweigh the harm that may be caused [203]. The exaggerated release of the satiety gut hormones with their central and peripheral effects on glycaemia and food intake [52, 75, 204] has given new insight into the physiological changes that take place after surgery. The initial enthusiasm after the discovery of the role of the gut hormones may need to be tempered as the magnitude of the effects of these gut hormones on weight loss may have been overestimated. The physiological changes after RYGB are unlikely to be due to a single hormone, or single mechanism, but are more likely to additionally involve complex gut-brain nutrient and neural signalling [205, 206]. Understanding these mechanisms will speed up the development of more effective and safer surgical and non-surgical treatments for obesity.
All studies that are mentioned in the review adhered to the expected high level ethical considerations and were approved by the appropriate institutional review board.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

Die Chirurgie

Print-Titel

Das Abo mit mehr Tiefe

Mit der Zeitschrift Die Chirurgie erhalten Sie zusätzlich Online-Zugriff auf weitere 43 chirurgische Fachzeitschriften, CME-Fortbildungen, Webinare, Vorbereitungskursen zur Facharztprüfung und die digitale Enzyklopädie e.Medpedia.

Bis 30. April 2024 bestellen und im ersten Jahr nur 199 € zahlen!

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
1.
Zurück zum Zitat Rubino F, Schauer PR, Kaplan LM, Cummings DE. Metabolic surgery to treat type 2 diabetes: clinical outcomes and mechanisms of action. Annu Rev Med. 2010;61:393–411.PubMedCrossRef Rubino F, Schauer PR, Kaplan LM, Cummings DE. Metabolic surgery to treat type 2 diabetes: clinical outcomes and mechanisms of action. Annu Rev Med. 2010;61:393–411.PubMedCrossRef
3.
Zurück zum Zitat Laurenius A, Larsson I, Bueter M, et al. Changes in eating behaviour and meal pattern following Roux-en-Y gastric bypass. Int J Obes. 2012;36:348–55.CrossRef Laurenius A, Larsson I, Bueter M, et al. Changes in eating behaviour and meal pattern following Roux-en-Y gastric bypass. Int J Obes. 2012;36:348–55.CrossRef
4.
Zurück zum Zitat Nora M, Guimarães M, Almeida R, et al. Metabolic laparoscopic gastric bypass for obese patients with type 2 diabetes. Obes Surg. 2011;21:1643–9.PubMedCrossRef Nora M, Guimarães M, Almeida R, et al. Metabolic laparoscopic gastric bypass for obese patients with type 2 diabetes. Obes Surg. 2011;21:1643–9.PubMedCrossRef
5.
Zurück zum Zitat McCarty TM, Arnold DT, Lamont JP, Fisher TL, Kuhn JA. Optimizing outcomes in bariatric surgery: outpatient laparoscopic gastric bypass. Ann Surg. 2005;242:494.PubMedPubMedCentral McCarty TM, Arnold DT, Lamont JP, Fisher TL, Kuhn JA. Optimizing outcomes in bariatric surgery: outpatient laparoscopic gastric bypass. Ann Surg. 2005;242:494.PubMedPubMedCentral
6.
Zurück zum Zitat Sjostrom L, Lindroos AK, Peltonen M, et al. Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery. N Engl J Med. 2004;351:2683–93.PubMedCrossRef Sjostrom L, Lindroos AK, Peltonen M, et al. Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery. N Engl J Med. 2004;351:2683–93.PubMedCrossRef
7.
Zurück zum Zitat Franco J, Ruiz P, Palermo M, Gagner M. A review of studies comparing three laparoscopic procedures in bariatric surgery: sleeve gastrectomy, Roux-en-Y gastric bypass and adjustable gastric banding. OBES SURG. 2011;21:1458–68.PubMedCrossRef Franco J, Ruiz P, Palermo M, Gagner M. A review of studies comparing three laparoscopic procedures in bariatric surgery: sleeve gastrectomy, Roux-en-Y gastric bypass and adjustable gastric banding. OBES SURG. 2011;21:1458–68.PubMedCrossRef
8.
Zurück zum Zitat Wing RR, Phelan S. Long-term weight loss maintenance. Am J Clin Nutr. 2005;82:222S–5S.PubMed Wing RR, Phelan S. Long-term weight loss maintenance. Am J Clin Nutr. 2005;82:222S–5S.PubMed
9.
Zurück zum Zitat Cooper Z, Doll HA, Hawker DM, et al. Testing a new cognitive behavioural treatment for obesity: a randomized controlled trial with three-year follow-up. Behav Res Ther. 2010;48:706–13.PubMedPubMedCentralCrossRef Cooper Z, Doll HA, Hawker DM, et al. Testing a new cognitive behavioural treatment for obesity: a randomized controlled trial with three-year follow-up. Behav Res Ther. 2010;48:706–13.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Olbers T, Gronowitz E, Werling M, et al. Two-year outcome of laparoscopic Roux-en-Y gastric bypass in adolescents with severe obesity: results from a Swedish Nationwide Study (AMOS). Int J Obes. 2012;36:1388–95.CrossRef Olbers T, Gronowitz E, Werling M, et al. Two-year outcome of laparoscopic Roux-en-Y gastric bypass in adolescents with severe obesity: results from a Swedish Nationwide Study (AMOS). Int J Obes. 2012;36:1388–95.CrossRef
11.
Zurück zum Zitat Boza C, Gamboa C, Awruch D, Perez G, Escalona A, Ibañez L, et al. Laparoscopic Roux-en-Y gastric bypass versus laparoscopic adjustable gastric banding: five years of follow-up. Surg Obes Relat Dis. 2010;6:470–5.PubMedCrossRef Boza C, Gamboa C, Awruch D, Perez G, Escalona A, Ibañez L, et al. Laparoscopic Roux-en-Y gastric bypass versus laparoscopic adjustable gastric banding: five years of follow-up. Surg Obes Relat Dis. 2010;6:470–5.PubMedCrossRef
12.
Zurück zum Zitat Wong SK, Kong AP, Mui WL, et al. Laparoscopic bariatric surgery: a five-year review. Hong Kong Med J. 2009;15(2):100–9.PubMed Wong SK, Kong AP, Mui WL, et al. Laparoscopic bariatric surgery: a five-year review. Hong Kong Med J. 2009;15(2):100–9.PubMed
13.
Zurück zum Zitat Kim TH, Daud A, Ude AO, et al. Early U.S. outcomes of laparoscopic gastric bypass versus laparoscopic adjustable silicone gastric banding for morbid obesity. Surg Endosc. 2006;20:202–9.PubMedCrossRef Kim TH, Daud A, Ude AO, et al. Early U.S. outcomes of laparoscopic gastric bypass versus laparoscopic adjustable silicone gastric banding for morbid obesity. Surg Endosc. 2006;20:202–9.PubMedCrossRef
14.
Zurück zum Zitat Lakdawala M, Bhasker A, Mulchandani D, Goel S, Jain S. Comparison between the results of laparoscopic sleeve gastrectomy and laparoscopic Roux-en-Y gastric bypass in the Indian population: a retrospective 1-year study. Obes Surg. 2010;20:1–6.PubMedCrossRef Lakdawala M, Bhasker A, Mulchandani D, Goel S, Jain S. Comparison between the results of laparoscopic sleeve gastrectomy and laparoscopic Roux-en-Y gastric bypass in the Indian population: a retrospective 1-year study. Obes Surg. 2010;20:1–6.PubMedCrossRef
15.
Zurück zum Zitat Weber M, Müller MK, Bucher T, et al. Laparoscopic gastric bypass is superior to laparoscopic gastric banding for treatment of morbid obesity. Ann Surg. 2004;240:975–82.PubMedPubMedCentralCrossRef Weber M, Müller MK, Bucher T, et al. Laparoscopic gastric bypass is superior to laparoscopic gastric banding for treatment of morbid obesity. Ann Surg. 2004;240:975–82.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Pournaras DJ, Osborne A, Hawkins SC, et al. Remission of type 2 diabetes after gastric bypass and banding: mechanisms and 2 year outcomes. Ann Surg. 2010;252:966–71.PubMedCrossRef Pournaras DJ, Osborne A, Hawkins SC, et al. Remission of type 2 diabetes after gastric bypass and banding: mechanisms and 2 year outcomes. Ann Surg. 2010;252:966–71.PubMedCrossRef
17.
18.
Zurück zum Zitat Gerstein HC. Do lifestyle changes reduce serious outcomes in diabetes? N Engl J Med. 2013;369:189–90.PubMedCrossRef Gerstein HC. Do lifestyle changes reduce serious outcomes in diabetes? N Engl J Med. 2013;369:189–90.PubMedCrossRef
19.
Zurück zum Zitat Maclean PS, Bergouignan A, Cornier MA, Jackman MR. Biology’s response to dieting: the impetus for weight regain. Am J Physiol Regul Integr Comp Physiol. 2011;301:R581–600.PubMedPubMedCentralCrossRef Maclean PS, Bergouignan A, Cornier MA, Jackman MR. Biology’s response to dieting: the impetus for weight regain. Am J Physiol Regul Integr Comp Physiol. 2011;301:R581–600.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Sumithran P, Prendergast LA, Delbridge E, et al. Long-term persistence of hormonal adaptations to weight loss. N Engl J Med. 2011;365:1597–604.PubMedCrossRef Sumithran P, Prendergast LA, Delbridge E, et al. Long-term persistence of hormonal adaptations to weight loss. N Engl J Med. 2011;365:1597–604.PubMedCrossRef
21.
Zurück zum Zitat Hofmann W, van Koningsbruggen GM, Stroebe W, Ramanathan S, Aarts H. As pleasure unfolds. Hedonic responses to tempting food. Psychol Sci. 2010;21:1863–70.PubMedCrossRef Hofmann W, van Koningsbruggen GM, Stroebe W, Ramanathan S, Aarts H. As pleasure unfolds. Hedonic responses to tempting food. Psychol Sci. 2010;21:1863–70.PubMedCrossRef
22.
Zurück zum Zitat Miller GD, Norris A, Fernandez A. Changes in nutrients and food groups intake following laparoscopic Roux-en-Y gastric bypass (RYGB). Obes Surg. 2014;24(11):1926–32. 1–7.PubMedPubMedCentralCrossRef Miller GD, Norris A, Fernandez A. Changes in nutrients and food groups intake following laparoscopic Roux-en-Y gastric bypass (RYGB). Obes Surg. 2014;24(11):1926–32. 1–7.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Zheng H, Shin AC, Lenard NR, 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:R1273–82.PubMedPubMedCentralCrossRef Zheng H, Shin AC, Lenard NR, 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:R1273–82.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Miras AD, Le Roux CW. Mechanisms underlying weight loss after bariatric surgery. Nat Rev Gastroenterol Hepatol. 2013;10:575–84.PubMedCrossRef Miras AD, Le Roux CW. Mechanisms underlying weight loss after bariatric surgery. Nat Rev Gastroenterol Hepatol. 2013;10:575–84.PubMedCrossRef
25.
Zurück zum Zitat Xanthakos SA. Nutritional deficiencies in obesity and after bariatric surgery. Pediatr Clin N Am. 2009;56:1105–21.CrossRef Xanthakos SA. Nutritional deficiencies in obesity and after bariatric surgery. Pediatr Clin N Am. 2009;56:1105–21.CrossRef
26.
Zurück zum Zitat Lindroos AK, Lissner L, Sjostrom L. Weight change in relation to intake of sugar and sweet foods before and after weight reducing gastric surgery. Int J Obes Relat Metab Dis J Int Assoc Study Obes. 1996;20:634–43. Lindroos AK, Lissner L, Sjostrom L. Weight change in relation to intake of sugar and sweet foods before and after weight reducing gastric surgery. Int J Obes Relat Metab Dis J Int Assoc Study Obes. 1996;20:634–43.
27.
Zurück zum Zitat Moize V, Geliebter A, Gluck ME, et al. Obese patients have inadequate protein intake related to protein intolerance up to 1 year following Roux-en-Y gastric bypass. Obes Surg. 2003;13:23–8.PubMedCrossRef Moize V, Geliebter A, Gluck ME, et al. Obese patients have inadequate protein intake related to protein intolerance up to 1 year following Roux-en-Y gastric bypass. Obes Surg. 2003;13:23–8.PubMedCrossRef
28.
Zurück zum Zitat Bobbioni-Harsch E, Huber O, Morel PH, et al. Factors influencing energy intake and body weight loss after gastric bypass. Eur J Clin Nutr. 2002;56:551–6.PubMedCrossRef Bobbioni-Harsch E, Huber O, Morel PH, et al. Factors influencing energy intake and body weight loss after gastric bypass. Eur J Clin Nutr. 2002;56:551–6.PubMedCrossRef
29.
Zurück zum Zitat Brolin RL, Robertson LB, Kenler HA, Cody RP. Weight loss and dietary intake after vertical banded gastroplasty and Roux-en-Y gastric bypass. Ann Surg. 1994;220:782.PubMedPubMedCentralCrossRef Brolin RL, Robertson LB, Kenler HA, Cody RP. Weight loss and dietary intake after vertical banded gastroplasty and Roux-en-Y gastric bypass. Ann Surg. 1994;220:782.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Naslund I, Jarnmark I, Andersson H. Dietary intake before and after gastric bypass and gastroplasty for morbid obesity in women. Int J Obes. 1987;12:503–13. Naslund I, Jarnmark I, Andersson H. Dietary intake before and after gastric bypass and gastroplasty for morbid obesity in women. Int J Obes. 1987;12:503–13.
31.
Zurück zum Zitat Coughlin K, Bell RM, Bivins BA, Wrobel S, Griffen WO. Preoperative and postoperative assessment of nutrient intakes in patients who have undergone gastric bypass surgery. Arch Surg. 1983;118:813–6.PubMedCrossRef Coughlin K, Bell RM, Bivins BA, Wrobel S, Griffen WO. Preoperative and postoperative assessment of nutrient intakes in patients who have undergone gastric bypass surgery. Arch Surg. 1983;118:813–6.PubMedCrossRef
32.
Zurück zum Zitat Flancbaum L, Choban P, Bradley LR, BURGE JC. Changes in measured resting energy expenditure after Roux-en-Y gastric bypass for clinically severe obesity. Surgery. 1997;122:943–9.PubMedCrossRef Flancbaum L, Choban P, Bradley LR, BURGE JC. Changes in measured resting energy expenditure after Roux-en-Y gastric bypass for clinically severe obesity. Surgery. 1997;122:943–9.PubMedCrossRef
33.
Zurück zum Zitat Warde-Kamar J, Rogers M, Flancbaum L, Laferrère B. Calorie intake and meal patterns up to 4 years after Roux-en-Y gastric bypass surgery. Obes Surg. 2004;14:1070–9.PubMedCrossRef Warde-Kamar J, Rogers M, Flancbaum L, Laferrère B. Calorie intake and meal patterns up to 4 years after Roux-en-Y gastric bypass surgery. Obes Surg. 2004;14:1070–9.PubMedCrossRef
34.
Zurück zum Zitat Bavaresco M, Paganini S, Lima TP, et al. Nutritional course of patients submitted to bariatric surgery. Obes Surg. 2010;20:716–21.PubMedCrossRef Bavaresco M, Paganini S, Lima TP, et al. Nutritional course of patients submitted to bariatric surgery. Obes Surg. 2010;20:716–21.PubMedCrossRef
35.
Zurück zum Zitat Schinkel ER, Pettine SM, Adams E, Harris M. Impact of varying levels of protein intake on protein status indicators after gastric bypass in patients with multiple complications requiring nutritional support. Obes Surg. 2006;16:24–30.CrossRef Schinkel ER, Pettine SM, Adams E, Harris M. Impact of varying levels of protein intake on protein status indicators after gastric bypass in patients with multiple complications requiring nutritional support. Obes Surg. 2006;16:24–30.CrossRef
36.
Zurück zum Zitat Westerterp-Plantenga MS, Nieuwenhuizen A, Tome D, Soenen S, Westerterp KR. Dietary protein, weight loss, and weight maintenance. Annu Rev Nutr. 2009;29:21–41.PubMedCrossRef Westerterp-Plantenga MS, Nieuwenhuizen A, Tome D, Soenen S, Westerterp KR. Dietary protein, weight loss, and weight maintenance. Annu Rev Nutr. 2009;29:21–41.PubMedCrossRef
37.
Zurück zum Zitat Heber D, Greenway FL, Kaplan LM, Livingston E, Salvador J, Still C. Endocrine and nutritional management of the post-bariatric surgery patient: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metabol. 2010;95:4823–43.CrossRef Heber D, Greenway FL, Kaplan LM, Livingston E, Salvador J, Still C. Endocrine and nutritional management of the post-bariatric surgery patient: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metabol. 2010;95:4823–43.CrossRef
38.
Zurück zum Zitat Halmi KA, Mason E, Falk JR, Stunkard A. Appetitive behavior after gastric bypass for obesity. Int J Obes. 1981;5:457–64.PubMed Halmi KA, Mason E, Falk JR, Stunkard A. Appetitive behavior after gastric bypass for obesity. Int J Obes. 1981;5:457–64.PubMed
39.
Zurück zum Zitat Heneghan HM, Yimcharoen P, Brethauer SA, Kroh M, Chand B. Influence of pouch and stoma size on weight loss after gastric bypass. Surg Obes Relat Dis. 2012;8:408–15.PubMedCrossRef Heneghan HM, Yimcharoen P, Brethauer SA, Kroh M, Chand B. Influence of pouch and stoma size on weight loss after gastric bypass. Surg Obes Relat Dis. 2012;8:408–15.PubMedCrossRef
40.
Zurück zum Zitat Bueter M, Löwenstein C, Ashrafian H, et al. Vagal sparing surgical technique but not stoma size affects body weight loss in rodent model of gastric bypass. Obes Surg. 2010;20:616–22.PubMedPubMedCentralCrossRef Bueter M, Löwenstein C, Ashrafian H, et al. Vagal sparing surgical technique but not stoma size affects body weight loss in rodent model of gastric bypass. Obes Surg. 2010;20:616–22.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Topart P, Becouarn G, Ritz P. Pouch size after gastric bypass does not correlate with weight loss outcome. Obes Surg. 2011;21:1350–4.PubMedCrossRef Topart P, Becouarn G, Ritz P. Pouch size after gastric bypass does not correlate with weight loss outcome. Obes Surg. 2011;21:1350–4.PubMedCrossRef
43.
44.
Zurück zum Zitat Gould J, Garren M, Boll V, Starling J. The impact of circular stapler diameter on the incidence of gastrojejunostomy stenosis and weight loss following laparoscopic Roux-en-Y gastric bypass. Surg Endosc. 2006;20:1017–20.PubMedCrossRef Gould J, Garren M, Boll V, Starling J. The impact of circular stapler diameter on the incidence of gastrojejunostomy stenosis and weight loss following laparoscopic Roux-en-Y gastric bypass. Surg Endosc. 2006;20:1017–20.PubMedCrossRef
45.
Zurück zum Zitat Fandriks L. The role of the smaller stomach. 31-8-2011. Postgraduate course B, XVI World Congress of the International Federation for the Surgery of Obesity and Metabolic Disorders, Hamburg, Germany. Ref Type: Report Fandriks L. The role of the smaller stomach. 31-8-2011. Postgraduate course B, XVI World Congress of the International Federation for the Surgery of Obesity and Metabolic Disorders, Hamburg, Germany. Ref Type: Report
46.
Zurück zum Zitat Laferrere B, Heshka S, Wang K, et al. Incretin levels and effect are markedly enhanced 1 month after Roux-en-Y gastric bypass surgery in obese patients with type 2 diabetes. Diabetes Care. 2007;30:1709–16.PubMedPubMedCentralCrossRef Laferrere B, Heshka S, Wang K, et al. Incretin levels and effect are markedly enhanced 1 month after Roux-en-Y gastric bypass surgery in obese patients with type 2 diabetes. Diabetes Care. 2007;30:1709–16.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Laferrere B, Teixeira J, McGinty J, et al. Effect of weight loss by gastric bypass surgery versus hypocaloric diet on glucose and incretin levels in patients with type 2 diabetes. J Clin Endocrinol Metab. 2008;93:2479–85.PubMedPubMedCentralCrossRef Laferrere B, Teixeira J, McGinty J, et al. Effect of weight loss by gastric bypass surgery versus hypocaloric diet on glucose and incretin levels in patients with type 2 diabetes. J Clin Endocrinol Metab. 2008;93:2479–85.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Morinigo R, Moize V, Musri M, et al. Glucagon-like peptide-1, peptide YY, hunger, and satiety after gastric bypass surgery in morbidly obese subjects. J Clin Endocrinol Metabol. 2006;91:1735–40.CrossRef Morinigo R, Moize V, Musri M, et al. Glucagon-like peptide-1, peptide YY, hunger, and satiety after gastric bypass surgery in morbidly obese subjects. J Clin Endocrinol Metabol. 2006;91:1735–40.CrossRef
49.
Zurück zum Zitat Korner J, Inabnet W, Conwell IM, et al. Differential effects of gastric bypass and banding on circulating gut hormone and leptin levels. Obesity (Silver Spring). 2006;14:1553–61.CrossRef Korner J, Inabnet W, Conwell IM, et al. Differential effects of gastric bypass and banding on circulating gut hormone and leptin levels. Obesity (Silver Spring). 2006;14:1553–61.CrossRef
50.
Zurück zum Zitat Rodieux F, Giusti V, D'Alessio DA, Suter M, Tappy L. Effects of gastric bypass and gastric banding on glucose kinetics and gut hormone release. Obesity (Silver Spring). 2008;16:298–305.CrossRef Rodieux F, Giusti V, D'Alessio DA, Suter M, Tappy L. Effects of gastric bypass and gastric banding on glucose kinetics and gut hormone release. Obesity (Silver Spring). 2008;16:298–305.CrossRef
51.
Zurück zum Zitat le Roux CW, Aylwin SJ, Batterham RL, et al. Gut hormone profiles following bariatric surgery favor an anorectic state, facilitate weight loss, and improve metabolic parameters. Ann Surg. 2006;243:108–14.PubMedPubMedCentralCrossRef le Roux CW, Aylwin SJ, Batterham RL, et al. Gut hormone profiles following bariatric surgery favor an anorectic state, facilitate weight loss, and improve metabolic parameters. Ann Surg. 2006;243:108–14.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Le Roux CW, Welbourn R, Werling M, et al. Gut hormones as mediators of appetite and weight loss after Roux-en-Y gastric bypass. Ann Surg. 2007;246:780–5.PubMedCrossRef Le Roux CW, Welbourn R, Werling M, et al. Gut hormones as mediators of appetite and weight loss after Roux-en-Y gastric bypass. Ann Surg. 2007;246:780–5.PubMedCrossRef
53.
Zurück zum Zitat Dar MS, Chapman III WH, Pender JR, et al. GLP-1 response to a mixed meal: what happens 10 years after Roux-en-Y gastric bypass (RYGB)? Obes Surg. 2012;22:1077–83.PubMedCrossRef Dar MS, Chapman III WH, Pender JR, et al. GLP-1 response to a mixed meal: what happens 10 years after Roux-en-Y gastric bypass (RYGB)? Obes Surg. 2012;22:1077–83.PubMedCrossRef
54.
Zurück zum Zitat Meguid MM, Glade MJ, Middleton FA. Weight regain after Roux-en-Y: a significant 20 % complication related to PYY. Nutrition. 2008;24:832–42.PubMedCrossRef Meguid MM, Glade MJ, Middleton FA. Weight regain after Roux-en-Y: a significant 20 % complication related to PYY. Nutrition. 2008;24:832–42.PubMedCrossRef
55.
Zurück zum Zitat Dirksen C, Jorgensen NB, Bojsen-Moller KN, et al. Gut hormones, early dumping and resting energy expenditure in patients with good and poor weight loss response after Roux-en-Y gastric bypass. Int J Obes. 2013;37(11):1452–9.CrossRef Dirksen C, Jorgensen NB, Bojsen-Moller KN, et al. Gut hormones, early dumping and resting energy expenditure in patients with good and poor weight loss response after Roux-en-Y gastric bypass. Int J Obes. 2013;37(11):1452–9.CrossRef
56.
Zurück zum Zitat Batterham RL, Cohen MA, Ellis SM, et al. Inhibition of food intake in obese subjects by peptide YY3-36. N Engl J Med. 2003;349:941–8.PubMedCrossRef Batterham RL, Cohen MA, Ellis SM, et al. Inhibition of food intake in obese subjects by peptide YY3-36. N Engl J Med. 2003;349:941–8.PubMedCrossRef
57.
Zurück zum Zitat Chandarana K, Gelegen C, Karra E, et al. Diet and gastrointestinal bypass-induced weight loss: the roles of ghrelin and peptide YY. Diabetes. 2011;60:810–8.PubMedPubMedCentralCrossRef Chandarana K, Gelegen C, Karra E, et al. Diet and gastrointestinal bypass-induced weight loss: the roles of ghrelin and peptide YY. Diabetes. 2011;60:810–8.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Sloth B, Holst JJ, Flint A, Gregersen NT, Astrup A. Effects of PYY1-36 and PYY3-36 on appetite, energy intake, energy expenditure, glucose and fat metabolism in obese and lean subjects. Am J Physiol Endocrinol Metabol. 2007;292:E1062–8.CrossRef Sloth B, Holst JJ, Flint A, Gregersen NT, Astrup A. Effects of PYY1-36 and PYY3-36 on appetite, energy intake, energy expenditure, glucose and fat metabolism in obese and lean subjects. Am J Physiol Endocrinol Metabol. 2007;292:E1062–8.CrossRef
59.
Zurück zum Zitat Troy S, Soty M, Ribeiro L, et al. Intestinal gluconeogenesis is a key factor for early metabolic changes after gastric bypass but not after gastric lap-band in mice. Cell Metab. 2008;8:201–11.PubMedCrossRef Troy S, Soty M, Ribeiro L, et al. Intestinal gluconeogenesis is a key factor for early metabolic changes after gastric bypass but not after gastric lap-band in mice. Cell Metab. 2008;8:201–11.PubMedCrossRef
60.
Zurück zum Zitat Peterli R, Wolnerhanssen B, Peters T, et al. Improvement in glucose metabolism after bariatric surgery: comparison of laparoscopic Roux-en-Y gastric bypass and laparoscopic sleeve gastrectomy: a prospective randomized trial. Ann Surg. 2009;250. Peterli R, Wolnerhanssen B, Peters T, et al. Improvement in glucose metabolism after bariatric surgery: comparison of laparoscopic Roux-en-Y gastric bypass and laparoscopic sleeve gastrectomy: a prospective randomized trial. Ann Surg. 2009;250.
61.
Zurück zum Zitat Reinehr T, Roth CL, Schernthaner GH, Kopp HP, Kriwanek S, Schernthaner G. Peptide YY and glucagon-like peptide-1 in morbidly obese patients before and after surgically induced weight loss. Obes Surg. 2007;17:1571–7.PubMedCrossRef Reinehr T, Roth CL, Schernthaner GH, Kopp HP, Kriwanek S, Schernthaner G. Peptide YY and glucagon-like peptide-1 in morbidly obese patients before and after surgically induced weight loss. Obes Surg. 2007;17:1571–7.PubMedCrossRef
62.
Zurück zum Zitat Ye J., Hao Z., Mumphrey M.B. 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 Ye J., Hao Z., Mumphrey M.B. 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
63.
Zurück zum Zitat Cummings DE, Weigle DS, Frayo RS, et al. Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery. N Engl J Med. 2002;346:1623–30.PubMedCrossRef Cummings DE, Weigle DS, Frayo RS, et al. Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery. N Engl J Med. 2002;346:1623–30.PubMedCrossRef
64.
Zurück zum Zitat Foschi D, Corsi F, Colombo F, et al. Different effects of vertical banded gastroplasty and Roux-en-Y gastric bypass on meal inhibition of ghrelin secretion in morbidly obese patients. J Invest Surg. 2008;21:77–81.PubMedCrossRef Foschi D, Corsi F, Colombo F, et al. Different effects of vertical banded gastroplasty and Roux-en-Y gastric bypass on meal inhibition of ghrelin secretion in morbidly obese patients. J Invest Surg. 2008;21:77–81.PubMedCrossRef
65.
Zurück zum Zitat Fruhbeck G, Rotellar F, Hernández-Lizoain JL, et al. Fasting plasma ghrelin concentrations 6 months after gastric bypass are not determined by weight loss or changes in insulinemia. Obes Surg. 2004;14:1208–15.PubMedCrossRef Fruhbeck G, Rotellar F, Hernández-Lizoain JL, et al. Fasting plasma ghrelin concentrations 6 months after gastric bypass are not determined by weight loss or changes in insulinemia. Obes Surg. 2004;14:1208–15.PubMedCrossRef
66.
Zurück zum Zitat Geloneze B, Tambascia MA, Pilla VF, Geloneze SR, Repetto EM, Pareja JC. Ghrelin: a gut-brain hormone: effect of gastric bypass surgery. Obes Surg. 2003;13:17–22.PubMedCrossRef Geloneze B, Tambascia MA, Pilla VF, Geloneze SR, Repetto EM, Pareja JC. Ghrelin: a gut-brain hormone: effect of gastric bypass surgery. Obes Surg. 2003;13:17–22.PubMedCrossRef
67.
Zurück zum Zitat Leonetti F, Silecchia G, Iacobellis G, et al. Different plasma ghrelin levels after laparoscopic gastric bypass and adjustable gastric banding in morbid obese subjects. J Clin Endocrinol Metab. 2003;88:4227–31.PubMedCrossRef Leonetti F, Silecchia G, Iacobellis G, et al. Different plasma ghrelin levels after laparoscopic gastric bypass and adjustable gastric banding in morbid obese subjects. J Clin Endocrinol Metab. 2003;88:4227–31.PubMedCrossRef
68.
Zurück zum Zitat Lin E, Gletsu N, Fugate K, et al. The effects of gastric surgery on systemic ghrelin levels in the morbidly obese. Arch Surg. 2004;139:780–4.PubMedCrossRef Lin E, Gletsu N, Fugate K, et al. The effects of gastric surgery on systemic ghrelin levels in the morbidly obese. Arch Surg. 2004;139:780–4.PubMedCrossRef
69.
Zurück zum Zitat Morinigo R, Casamitjana R, Moize V, et al. Short-term effects of gastric bypass surgery on circulating ghrelin levels. Obes Res. 2004;12:1108–16.PubMedCrossRef Morinigo R, Casamitjana R, Moize V, et al. Short-term effects of gastric bypass surgery on circulating ghrelin levels. Obes Res. 2004;12:1108–16.PubMedCrossRef
70.
Zurück zum Zitat Tritos NA, Mun E, Bertkau A, Grayson R, Maratos-Flier E, Goldfine A. Serum ghrelin levels in response to glucose load in obese subjects post-gastric bypass surgery. Obes Res. 2003;11:919–24.PubMedCrossRef Tritos NA, Mun E, Bertkau A, Grayson R, Maratos-Flier E, Goldfine A. Serum ghrelin levels in response to glucose load in obese subjects post-gastric bypass surgery. Obes Res. 2003;11:919–24.PubMedCrossRef
71.
Zurück zum Zitat Couce M, Cottam D, Esplen J, Schauer P, Burguera B. Is ghrelin the culprit for weight loss after gastric bypass surgery? A negative answer. Obes Surg. 2006;16:870–8.PubMedCrossRef Couce M, Cottam D, Esplen J, Schauer P, Burguera B. Is ghrelin the culprit for weight loss after gastric bypass surgery? A negative answer. Obes Surg. 2006;16:870–8.PubMedCrossRef
72.
Zurück zum Zitat Faraj M, Havel PJ, Phélis S, Blank D, Sniderman AD, Cianflone K. Plasma acylation-stimulating protein, adiponectin, leptin, and ghrelin before and after weight loss induced by gastric bypass surgery in morbidly obese subjects. J Clin Endocrinol Metab. 2003;88:1594–602.PubMedCrossRef Faraj M, Havel PJ, Phélis S, Blank D, Sniderman AD, Cianflone K. Plasma acylation-stimulating protein, adiponectin, leptin, and ghrelin before and after weight loss induced by gastric bypass surgery in morbidly obese subjects. J Clin Endocrinol Metab. 2003;88:1594–602.PubMedCrossRef
73.
Zurück zum Zitat Karamanakos SN, Vagenas K, Kalfarentzos F, Alexandrides TK. Weight loss, appetite suppression, and changes in fasting and postprandial ghrelin and peptide-YY levels after Roux-en-Y gastric bypass and sleeve gastrectomy: a prospective, double blind study. Ann Surg. 2008;247:401–7.PubMedCrossRef Karamanakos SN, Vagenas K, Kalfarentzos F, Alexandrides TK. Weight loss, appetite suppression, and changes in fasting and postprandial ghrelin and peptide-YY levels after Roux-en-Y gastric bypass and sleeve gastrectomy: a prospective, double blind study. Ann Surg. 2008;247:401–7.PubMedCrossRef
74.
Zurück zum Zitat Korner J, Bessler M, Cirilo LJ, et al. Effects of Roux-en-Y gastric bypass surgery on fasting and postprandial concentrations of plasma ghrelin, peptide YY, and insulin. J Clin Endocrinol Metab. 2005;90:359–65.PubMedCrossRef Korner J, Bessler M, Cirilo LJ, et al. Effects of Roux-en-Y gastric bypass surgery on fasting and postprandial concentrations of plasma ghrelin, peptide YY, and insulin. J Clin Endocrinol Metab. 2005;90:359–65.PubMedCrossRef
75.
Zurück zum Zitat Korner J, Inabnet W, Febres G, et al. Prospective study of gut hormone and metabolic changes after adjustable gastric banding and Roux-en-Y gastric bypass. Int J Obes (Lond). 2009;33:786–95.CrossRef Korner J, Inabnet W, Febres G, et al. Prospective study of gut hormone and metabolic changes after adjustable gastric banding and Roux-en-Y gastric bypass. Int J Obes (Lond). 2009;33:786–95.CrossRef
76.
Zurück zum Zitat Liou JM, Lin JT, Lee WJ, et al. The serial changes of ghrelin and leptin levels and their relations to weight loss after laparoscopic minigastric bypass surgery. Obes Surg. 2008;18:84–9.PubMedCrossRef Liou JM, Lin JT, Lee WJ, et al. The serial changes of ghrelin and leptin levels and their relations to weight loss after laparoscopic minigastric bypass surgery. Obes Surg. 2008;18:84–9.PubMedCrossRef
77.
Zurück zum Zitat Olivan B, Teixeira J, Bose M, et al. Effect of weight loss by diet or gastric bypass surgery on peptide YY3-36 levels. Ann Surg. 2009;249:948–53.PubMedPubMedCentralCrossRef Olivan B, Teixeira J, Bose M, et al. Effect of weight loss by diet or gastric bypass surgery on peptide YY3-36 levels. Ann Surg. 2009;249:948–53.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Stoeckli R, Chanda R, Langer I, Keller U. Changes of body weight and plasma ghrelin levels after gastric banding and gastric bypass. Obes Res. 2004;12:346–50.PubMedCrossRef Stoeckli R, Chanda R, Langer I, Keller U. Changes of body weight and plasma ghrelin levels after gastric banding and gastric bypass. Obes Res. 2004;12:346–50.PubMedCrossRef
79.
Zurück zum Zitat Whitson BA, Leslie DB, Kellogg TA, et al. Entero-endocrine changes after gastric bypass in diabetic and nondiabetic patients: a preliminary study. J Surg Res. 2007;141:31–9.PubMedCrossRef Whitson BA, Leslie DB, Kellogg TA, et al. Entero-endocrine changes after gastric bypass in diabetic and nondiabetic patients: a preliminary study. J Surg Res. 2007;141:31–9.PubMedCrossRef
80.
Zurück zum Zitat Garcia-Fuentes E, Garrido-Sanchez L, Garcia-Almeida JM, et al. Different effect of laparoscopic Roux-en-Y gastric bypass and open biliopancreatic diversion of Scopinaro on serum PYY and ghrelin levels. Obes Surg. 2008;18:1424–9.PubMedCrossRef Garcia-Fuentes E, Garrido-Sanchez L, Garcia-Almeida JM, et al. Different effect of laparoscopic Roux-en-Y gastric bypass and open biliopancreatic diversion of Scopinaro on serum PYY and ghrelin levels. Obes Surg. 2008;18:1424–9.PubMedCrossRef
81.
Zurück zum Zitat Holdstock C, Engström BE, Ohrvall M, Lind L, Sundbom M, Karlsson FA. Ghrelin and adipose tissue regulatory peptides: effect of gastric bypass surgery in obese humans. J Clin Endocrinol Metab. 2003;88:3177–83.PubMedCrossRef Holdstock C, Engström BE, Ohrvall M, Lind L, Sundbom M, Karlsson FA. Ghrelin and adipose tissue regulatory peptides: effect of gastric bypass surgery in obese humans. J Clin Endocrinol Metab. 2003;88:3177–83.PubMedCrossRef
82.
Zurück zum Zitat Sundbom M, Holdstock C, Engström BE, Karlsson FA. Early changes in ghrelin following Roux-en-Y gastric bypass: influence of vagal nerve functionality? Obes Surg. 2007;17:304–10.PubMedCrossRef Sundbom M, Holdstock C, Engström BE, Karlsson FA. Early changes in ghrelin following Roux-en-Y gastric bypass: influence of vagal nerve functionality? Obes Surg. 2007;17:304–10.PubMedCrossRef
83.
Zurück zum Zitat Vendrell J, Broch M, Vilarrasa N, et al. Resistin, adiponectin, ghrelin, leptin, and proinflammatory cytokines: relationships in obesity. Obes Res. 2004;12:962–71.PubMedCrossRef Vendrell J, Broch M, Vilarrasa N, et al. Resistin, adiponectin, ghrelin, leptin, and proinflammatory cytokines: relationships in obesity. Obes Res. 2004;12:962–71.PubMedCrossRef
84.
Zurück zum Zitat Ybarra J, Bobbioni-Harsch E, Chassot G, et al. Persistent correlation of ghrelin plasma levels with body mass index both in stable weight conditions and during gastric-bypass-induced weight loss. Obes Surg. 2009;19:327–31.PubMedCrossRef Ybarra J, Bobbioni-Harsch E, Chassot G, et al. Persistent correlation of ghrelin plasma levels with body mass index both in stable weight conditions and during gastric-bypass-induced weight loss. Obes Surg. 2009;19:327–31.PubMedCrossRef
85.
Zurück zum Zitat Pournaras DJ, le Roux CW. Ghrelin and metabolic surgery. Int J Pept. 2010;2010:733–43.CrossRef Pournaras DJ, le Roux CW. Ghrelin and metabolic surgery. Int J Pept. 2010;2010:733–43.CrossRef
87.
89.
Zurück zum Zitat Schwartz MW, Woods SC, Porte D, Seeley RJ, Baskin DG. Central nervous system control of food intake. Nature. 2000;404:661–71.PubMed Schwartz MW, Woods SC, Porte D, Seeley RJ, Baskin DG. Central nervous system control of food intake. Nature. 2000;404:661–71.PubMed
90.
Zurück zum Zitat le Roux CW, Neary NM, Halsey TJ, et al. Ghrelin does not stimulate food intake in patients with surgical procedures involving vagotomy. J ClinEndocrinol Metabol. 2005;90:4521–4.CrossRef le Roux CW, Neary NM, Halsey TJ, et al. Ghrelin does not stimulate food intake in patients with surgical procedures involving vagotomy. J ClinEndocrinol Metabol. 2005;90:4521–4.CrossRef
91.
Zurück zum Zitat Ochner CN, Kwok Y, Conceicao E, et al. Selective reduction in neural responses to high calorie foods following gastric bypass surgery. Ann Surg. 2011;253:502–7.PubMedPubMedCentralCrossRef Ochner CN, Kwok Y, Conceicao E, et al. Selective reduction in neural responses to high calorie foods following gastric bypass surgery. Ann Surg. 2011;253:502–7.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Forman EM, Hoffman KL, McGrath KB, Herbert JD, Brandsma LL, Lowe MR. A comparison of acceptance- and control-based strategies for coping with food cravings: an analog study. Behav Res Ther. 2007;45:2372–86.PubMedCrossRef Forman EM, Hoffman KL, McGrath KB, Herbert JD, Brandsma LL, Lowe MR. A comparison of acceptance- and control-based strategies for coping with food cravings: an analog study. Behav Res Ther. 2007;45:2372–86.PubMedCrossRef
93.
Zurück zum Zitat Lowe MR, van Steenburgh J, Ochner CN. Individual differences in brain activation in relation to ingestive behavior and obesity. Physiol Behav. 2009;5:561–71.CrossRef Lowe MR, van Steenburgh J, Ochner CN. Individual differences in brain activation in relation to ingestive behavior and obesity. Physiol Behav. 2009;5:561–71.CrossRef
94.
Zurück zum Zitat Lowe MR, Butryn ML, Didie ER, et al. The Power of Food Scale. A new measure of the psychological influence of the food environment. Appetite. 2009;53:114–8.PubMedCrossRef Lowe MR, Butryn ML, Didie ER, et al. The Power of Food Scale. A new measure of the psychological influence of the food environment. Appetite. 2009;53:114–8.PubMedCrossRef
95.
Zurück zum Zitat Kreymann B, Ghatei MA, Williams G, Bloom SR. Glucagon-like peptide-1 7–36: a physiological incretin in man. Lancet. 1987;330:1300–4.CrossRef Kreymann B, Ghatei MA, Williams G, Bloom SR. Glucagon-like peptide-1 7–36: a physiological incretin in man. Lancet. 1987;330:1300–4.CrossRef
96.
Zurück zum Zitat Katsuma S, Hirasawa A, Tsujimoto G. Bile acids promote glucagon-like peptide-1 secretion through TGR5 in a murine enteroendocrine cell line STC-1. Biochem Biophys Res Commun. 2005;329:386–90.PubMedCrossRef Katsuma S, Hirasawa A, Tsujimoto G. Bile acids promote glucagon-like peptide-1 secretion through TGR5 in a murine enteroendocrine cell line STC-1. Biochem Biophys Res Commun. 2005;329:386–90.PubMedCrossRef
97.
Zurück zum Zitat Patti ME, Houten SM, Bianco AC, et al. Serum bile acids are higher in humans with prior gastric bypass: potential contribution to improved glucose and lipid metabolism. Obesity. 2009;17:1671–7.PubMedPubMedCentralCrossRef Patti ME, Houten SM, Bianco AC, et al. Serum bile acids are higher in humans with prior gastric bypass: potential contribution to improved glucose and lipid metabolism. Obesity. 2009;17:1671–7.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Watanabe M, Houten SM, Mataki C, et al. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature. 2006;439:484–9.PubMedCrossRef Watanabe M, Houten SM, Mataki C, et al. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature. 2006;439:484–9.PubMedCrossRef
99.
Zurück zum Zitat Ge H, Baribault H, Vonderfecht S, et al. Characterization of a FGF19 variant with altered receptor specificity revealed a central role for FGFR1c in the regulation of glucose metabolism. PLoS One. 2012;7:e33603.PubMedPubMedCentralCrossRef Ge H, Baribault H, Vonderfecht S, et al. Characterization of a FGF19 variant with altered receptor specificity revealed a central role for FGFR1c in the regulation of glucose metabolism. PLoS One. 2012;7:e33603.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Morton G.J., Kaiyala K.J., Foster-Schubert K.E., Cummings D.E., Schwartz M.W. Carbohydrate feeding dissociates the postprandial FGF19 response from circulating bile acid levels in humans. The Journal of Clinical Endocrinology & Metabolism 2013 Morton G.J., Kaiyala K.J., Foster-Schubert K.E., Cummings D.E., Schwartz M.W. Carbohydrate feeding dissociates the postprandial FGF19 response from circulating bile acid levels in humans. The Journal of Clinical Endocrinology & Metabolism 2013
101.
Zurück zum Zitat Holt JA, Luo G, Billin AN, et al. Definition of a novel growth factor-dependent signal cascade for the suppression of bile acid biosynthesis. Genes Dev. 2003;17:1581–91.PubMedPubMedCentralCrossRef Holt JA, Luo G, Billin AN, et al. Definition of a novel growth factor-dependent signal cascade for the suppression of bile acid biosynthesis. Genes Dev. 2003;17:1581–91.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Inagaki T, Choi M, Moschetta A, et al. Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis. Cell Metab. 2005;2:217–25.PubMedCrossRef Inagaki T, Choi M, Moschetta A, et al. Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis. Cell Metab. 2005;2:217–25.PubMedCrossRef
103.
Zurück zum Zitat Roberts RE, Glicksman C, Alaghband-Zadeh J, Sherwood RA, Akuji N, le Roux CW. The relationship between postprandial bile acid concentration, GLP-1, PYY and ghrelin. Clin Endocrinol (Oxf). 2011;74:67–72.CrossRef Roberts RE, Glicksman C, Alaghband-Zadeh J, Sherwood RA, Akuji N, le Roux CW. The relationship between postprandial bile acid concentration, GLP-1, PYY and ghrelin. Clin Endocrinol (Oxf). 2011;74:67–72.CrossRef
104.
Zurück zum Zitat Pournaras DJ, Glicksman C, Vincent RP, et al. The role of bile after Roux-en-Y gastric bypass in promoting weight loss and improving glycaemic control. Endocrinology. 2012;153:3613–9.PubMedPubMedCentralCrossRef Pournaras DJ, Glicksman C, Vincent RP, et al. The role of bile after Roux-en-Y gastric bypass in promoting weight loss and improving glycaemic control. Endocrinology. 2012;153:3613–9.PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Furet JP, Kong LC, Tap J, et al. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: links with metabolic and low-grade inflammation markers. Diabetes. 2010;59:3049–57.PubMedPubMedCentralCrossRef Furet JP, Kong LC, Tap J, et al. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: links with metabolic and low-grade inflammation markers. Diabetes. 2010;59:3049–57.PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Clarke SF, Murphy EF, Nilaweera K, et al. The gut microbiota and its relationship to diet and obesity: new insights. Gut Microbes. 2012;3:186–202.PubMedPubMedCentralCrossRef Clarke SF, Murphy EF, Nilaweera K, et al. The gut microbiota and its relationship to diet and obesity: new insights. Gut Microbes. 2012;3:186–202.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Armougom F, Henry M, Vialettes B, Raccah D, Raoult D. Monitoring bacterial community of human gut microbiota reveals an increase in Lactobacillus in obese patients and methanogens in anorexic patients. PLoS One. 2009;4:e7125.PubMedPubMedCentralCrossRef Armougom F, Henry M, Vialettes B, Raccah D, Raoult D. Monitoring bacterial community of human gut microbiota reveals an increase in Lactobacillus in obese patients and methanogens in anorexic patients. PLoS One. 2009;4:e7125.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444:1022–3.PubMedCrossRef Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444:1022–3.PubMedCrossRef
110.
Zurück zum Zitat Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–131.PubMedCrossRef Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–131.PubMedCrossRef
111.
112.
Zurück zum Zitat Aron-Wisnewsky J, Clement K. Gut microbiota in obesity and type-2 diabetes: links with diet and weight loss intervention. Intestin Microbiota Health Dis Mod Concepts. 2014;307. Aron-Wisnewsky J, Clement K. Gut microbiota in obesity and type-2 diabetes: links with diet and weight loss intervention. Intestin Microbiota Health Dis Mod Concepts. 2014;307.
113.
Zurück zum Zitat Liou A.P., Paziuk M., Luevano J.M., 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; 5:178ra41. Liou A.P., Paziuk M., Luevano J.M., 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; 5:178ra41.
114.
Zurück zum Zitat Wurtman Judith J. The involvement of brain serotonin in excessive carbohydrate snacking by obese carbohydrate cravers. J Am Diet Assoc. 1984;84(9):1004–7. Wurtman Judith J. The involvement of brain serotonin in excessive carbohydrate snacking by obese carbohydrate cravers. J Am Diet Assoc. 1984;84(9):1004–7.
115.
Zurück zum Zitat Spitzer L, Rodin J. Human eating behavior: a critical review of studies in normal weight and overweight individuals. Appetite. 1981;2:293–329.CrossRef Spitzer L, Rodin J. Human eating behavior: a critical review of studies in normal weight and overweight individuals. Appetite. 1981;2:293–329.CrossRef
116.
Zurück zum Zitat Drewnowski A, Brunzell JD, Sande K, Iverius PH, Greenwood MRC. Sweet tooth reconsidered: taste responsiveness in human obesity. Physiol Behav. 1985;35:617–22.PubMedCrossRef Drewnowski A, Brunzell JD, Sande K, Iverius PH, Greenwood MRC. Sweet tooth reconsidered: taste responsiveness in human obesity. Physiol Behav. 1985;35:617–22.PubMedCrossRef
117.
Zurück zum Zitat Rodin J, Radke-Sharpe N, Rebuffé-Scrive M, Greenwood MR. Weight cycling and fat distribution. Int J Obes. 1990;14:303–10.PubMed Rodin J, Radke-Sharpe N, Rebuffé-Scrive M, Greenwood MR. Weight cycling and fat distribution. Int J Obes. 1990;14:303–10.PubMed
118.
Zurück zum Zitat Pangborn R, Simon M. Body size and sweetness preference. J Am Diet Assoc. 1958;34(9):924–8.PubMed Pangborn R, Simon M. Body size and sweetness preference. J Am Diet Assoc. 1958;34(9):924–8.PubMed
119.
Zurück zum Zitat Lowe MR, Butryn ML. Hedonic hunger: a new dimension of appetite? Physiol Behav. 2007;91:432–9.PubMedCrossRef Lowe MR, Butryn ML. Hedonic hunger: a new dimension of appetite? Physiol Behav. 2007;91:432–9.PubMedCrossRef
120.
Zurück zum Zitat Schultes B, Ernst B, Wilms B, Thurnheer M, Hallschmid M. Hedonic hunger is increased in severely obese patients and is reduced after gastric bypass surgery. Am J Clin Nutr. 2010;92:277–83.PubMedCrossRef Schultes B, Ernst B, Wilms B, Thurnheer M, Hallschmid M. Hedonic hunger is increased in severely obese patients and is reduced after gastric bypass surgery. Am J Clin Nutr. 2010;92:277–83.PubMedCrossRef
121.
Zurück zum Zitat Ernst B, Thurnheer M, Wilms B, Schultes B. Differential changes in dietary habits after gastric bypass versus gastric banding operations. Obes Surg. 2009;19:274–80.PubMedCrossRef Ernst B, Thurnheer M, Wilms B, Schultes B. Differential changes in dietary habits after gastric bypass versus gastric banding operations. Obes Surg. 2009;19:274–80.PubMedCrossRef
122.
Zurück zum Zitat Olbers T, Bjorkman S, Lindroos A, et al. Body composition, dietary intake, and energy expenditure after laparoscopic Roux-en-Y gastric bypass and laparoscopic vertical banded gastroplasty: a randomized clinical trial. Ann Surg. 2006;244:715–22.PubMedPubMedCentralCrossRef Olbers T, Bjorkman S, Lindroos A, et al. Body composition, dietary intake, and energy expenditure after laparoscopic Roux-en-Y gastric bypass and laparoscopic vertical banded gastroplasty: a randomized clinical trial. Ann Surg. 2006;244:715–22.PubMedPubMedCentralCrossRef
123.
Zurück zum Zitat Miras AD, Le Roux CW. Bariatric surgery and taste: novel mechanisms of weight loss. Curr Opin Gastroenterol. 2010;26(2):140–5.PubMedCrossRef Miras AD, Le Roux CW. Bariatric surgery and taste: novel mechanisms of weight loss. Curr Opin Gastroenterol. 2010;26(2):140–5.PubMedCrossRef
124.
125.
Zurück zum Zitat Thomas JR, Gizis F, Marcus E. Food selections of Roux-en-Y gastric bypass patients up to 2.5 years postsurgery. J Am Diet Assoc. 2010;110:608–12.PubMedCrossRef Thomas JR, Gizis F, Marcus E. Food selections of Roux-en-Y gastric bypass patients up to 2.5 years postsurgery. J Am Diet Assoc. 2010;110:608–12.PubMedCrossRef
126.
Zurück zum Zitat Laurenius A, Larsson I, Melanson KJ, et al. Decreased energy density and changes in food selection following Roux-en-Y gastric bypass. Eur J Clin Nutr. 2013;67:168–73.PubMedCrossRef Laurenius A, Larsson I, Melanson KJ, et al. Decreased energy density and changes in food selection following Roux-en-Y gastric bypass. Eur J Clin Nutr. 2013;67:168–73.PubMedCrossRef
127.
Zurück zum Zitat Kenler HA, Brolin RE, Cody RP. Changes in eating behavior after horizontal gastroplasty and Roux-en-Y gastric bypass. Am J Clin Nutr. 1990;52:87–92.PubMed Kenler HA, Brolin RE, Cody RP. Changes in eating behavior after horizontal gastroplasty and Roux-en-Y gastric bypass. Am J Clin Nutr. 1990;52:87–92.PubMed
128.
Zurück zum Zitat Scholtz S., Miras A.D., Chhina N. et al. Obese patients after gastric bypass surgery have lower brain-hedonic responses to food than after gastric banding. Gut, gutjnl-2013 2013. Scholtz S., Miras A.D., Chhina N. et al. Obese patients after gastric bypass surgery have lower brain-hedonic responses to food than after gastric banding. Gut, gutjnl-2013 2013.
129.
Zurück zum Zitat Tam CS, Berthoud HR, Bueter M, et al. Could the mechanisms of bariatric surgery hold the key for novel therapies?: report from a Pennington Scientific Symposium. Obes Rev. 2011;12:984–94.PubMedPubMedCentralCrossRef Tam CS, Berthoud HR, Bueter M, et al. Could the mechanisms of bariatric surgery hold the key for novel therapies?: report from a Pennington Scientific Symposium. Obes Rev. 2011;12:984–94.PubMedPubMedCentralCrossRef
130.
Zurück zum Zitat Meillon S, Miras AD, Roux CW. Gastric bypass surgery alters food preferences through changes in the perception of taste. Clinical Practice. 2013;10:471–9.CrossRef Meillon S, Miras AD, Roux CW. Gastric bypass surgery alters food preferences through changes in the perception of taste. Clinical Practice. 2013;10:471–9.CrossRef
131.
Zurück zum Zitat Sugerman HJ, Londrey GL, Kellum JM, et al. Weight loss with vertical banded gastroplasty and Roux-en-Y gastric bypass: selective vs. random assignment. Am J Surg. 1989;5:457–64. Sugerman HJ, Londrey GL, Kellum JM, et al. Weight loss with vertical banded gastroplasty and Roux-en-Y gastric bypass: selective vs. random assignment. Am J Surg. 1989;5:457–64.
132.
Zurück zum Zitat Sugerman HJ, Starkey JV, Birkenhauer R. A randomized prospective trial of gastric bypass versus vertical banded gastroplasty for morbid obesity and their effects on sweets versus non-sweets eaters. Ann Surg. 1987;205:613–24.PubMedPubMedCentralCrossRef Sugerman HJ, Starkey JV, Birkenhauer R. A randomized prospective trial of gastric bypass versus vertical banded gastroplasty for morbid obesity and their effects on sweets versus non-sweets eaters. Ann Surg. 1987;205:613–24.PubMedPubMedCentralCrossRef
133.
Zurück zum Zitat Linner JH. Comparative effectiveness of gastric bypass and gastroplasty: a clinical study. Arch Surg. 1982;117:695–700.PubMedCrossRef Linner JH. Comparative effectiveness of gastric bypass and gastroplasty: a clinical study. Arch Surg. 1982;117:695–700.PubMedCrossRef
134.
Zurück zum Zitat Hofmann B, Hjelmesaeth J, Sovik TT. Moral challenges with surgical treatment of type 2 diabetes. J Diabetes Complicat. 2013;27:597–603.PubMedCrossRef Hofmann B, Hjelmesaeth J, Sovik TT. Moral challenges with surgical treatment of type 2 diabetes. J Diabetes Complicat. 2013;27:597–603.PubMedCrossRef
135.
Zurück zum Zitat Schafe G.E., Bernstein I.L. Taste aversion learning. 1996 Schafe G.E., Bernstein I.L. Taste aversion learning. 1996
136.
Zurück zum Zitat Garcia J, Koelling RA. Relation of cue to consequence in avoidance learning. Psychon Sci. 1966;4:123–4.CrossRef Garcia J, Koelling RA. Relation of cue to consequence in avoidance learning. Psychon Sci. 1966;4:123–4.CrossRef
139.
Zurück zum Zitat Spector AC. Linking gustatory neurobiology to behavior in vertebrates. Neurosci Biobehav Rev. 2000;24:391–416.PubMedCrossRef Spector AC. Linking gustatory neurobiology to behavior in vertebrates. Neurosci Biobehav Rev. 2000;24:391–416.PubMedCrossRef
140.
Zurück zum Zitat Le Roux CW, Bueter M, Theis N, et al. Gastric bypass reduces fat intake and preference. Am J Physiol Regul Integr Comp Physiol. 2011;301(4):R1057–66.PubMedPubMedCentralCrossRef Le Roux CW, Bueter M, Theis N, et al. Gastric bypass reduces fat intake and preference. Am J Physiol Regul Integr Comp Physiol. 2011;301(4):R1057–66.PubMedPubMedCentralCrossRef
141.
Zurück zum Zitat Bueter M, Miras AD, Chichger H, et al. Alterations of sucrose preference after Roux-en-Y gastric bypass. Physiol Behav. 2011;104:709–21.PubMedCrossRef Bueter M, Miras AD, Chichger H, et al. Alterations of sucrose preference after Roux-en-Y gastric bypass. Physiol Behav. 2011;104:709–21.PubMedCrossRef
142.
Zurück zum Zitat Hajnal A, Kovacs P, Ahmed T, Meirelles K, Lynch CJ, Cooney RN. Gastric bypass surgery alters behavioral and neural taste functions for sweet taste in obese rats. Am J Physiol Gastrointest Liver Physiol. 2010;299:G967–79.PubMedPubMedCentralCrossRef Hajnal A, Kovacs P, Ahmed T, Meirelles K, Lynch CJ, Cooney RN. Gastric bypass surgery alters behavioral and neural taste functions for sweet taste in obese rats. Am J Physiol Gastrointest Liver Physiol. 2010;299:G967–79.PubMedPubMedCentralCrossRef
143.
Zurück zum Zitat Shin AC, Zheng H, Pistell PJ, Berthoud HR. Roux-en-Y gastric bypass surgery changes food reward in rats. Int J Obes. 2011;35:642–51.CrossRef Shin AC, Zheng H, Pistell PJ, Berthoud HR. Roux-en-Y gastric bypass surgery changes food reward in rats. Int J Obes. 2011;35:642–51.CrossRef
144.
Zurück zum Zitat Tichansky D, Rebecca GA, Madan A, Harper J, Tokita K, Boughter J. Decrease in sweet taste in rats after gastric bypass surgery. Surg Endosc. 2011;25:1176–81.PubMedCrossRef Tichansky D, Rebecca GA, Madan A, Harper J, Tokita K, Boughter J. Decrease in sweet taste in rats after gastric bypass surgery. Surg Endosc. 2011;25:1176–81.PubMedCrossRef
145.
Zurück zum Zitat Miras AD, Jackson RN, Goldstone AP, Olbers T, Hackenberg T, Spector AC, et al. Gastric bypass surgery for obesity decreases the reward value of a sweet-fat stimulus as assessed in a progressive ratio task. Am J Clin Nutr. 2012;96(3):467–73. Ref Type: Journal (Full).PubMedCrossRef Miras AD, Jackson RN, Goldstone AP, Olbers T, Hackenberg T, Spector AC, et al. Gastric bypass surgery for obesity decreases the reward value of a sweet-fat stimulus as assessed in a progressive ratio task. Am J Clin Nutr. 2012;96(3):467–73. Ref Type: Journal (Full).PubMedCrossRef
146.
Zurück zum Zitat 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;153:2234–44.PubMedCrossRef 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;153:2234–44.PubMedCrossRef
147.
Zurück zum Zitat Carrasco F, Papapietro K, Csendes A, et al. Changes in resting energy expenditure and body composition after weight loss following Roux-en-Y gastric bypass. Obes Surg. 2007;17:608–16.PubMedCrossRef Carrasco F, Papapietro K, Csendes A, et al. Changes in resting energy expenditure and body composition after weight loss following Roux-en-Y gastric bypass. Obes Surg. 2007;17:608–16.PubMedCrossRef
148.
Zurück zum Zitat Flancbaum L, Verducci J, Choban P. Changes in measured resting energy expenditure after Roux-en-Y gastric bypass for clinically severe obesity are not related to bypass limb-length. Obes Surg. 1998;8:437–43.PubMedCrossRef Flancbaum L, Verducci J, Choban P. Changes in measured resting energy expenditure after Roux-en-Y gastric bypass for clinically severe obesity are not related to bypass limb-length. Obes Surg. 1998;8:437–43.PubMedCrossRef
149.
Zurück zum Zitat Faria S, Kelly E, Faria O. Energy expenditure and weight regain in patients submitted to Roux-en-Y gastric bypass. Obes Surg. 2009;19:856–9.PubMedCrossRef Faria S, Kelly E, Faria O. Energy expenditure and weight regain in patients submitted to Roux-en-Y gastric bypass. Obes Surg. 2009;19:856–9.PubMedCrossRef
150.
Zurück zum Zitat Carrasco F, Rojas P, Ruz M, et al. Energy expenditure and body composition in severe and morbid obese women after gastric bypass]. Revista m_Ñdica de Chile. 2008;136:570. Carrasco F, Rojas P, Ruz M, et al. Energy expenditure and body composition in severe and morbid obese women after gastric bypass]. Revista m_Ñdica de Chile. 2008;136:570.
151.
Zurück zum Zitat Adami GF, Campostano A, Bessarione D, et al. Resting energy expenditure in long-term postobese subjects after weight normalization by dieting or biliopancreatic diversion. Obes Surg. 1993;3:397–9.PubMedCrossRef Adami GF, Campostano A, Bessarione D, et al. Resting energy expenditure in long-term postobese subjects after weight normalization by dieting or biliopancreatic diversion. Obes Surg. 1993;3:397–9.PubMedCrossRef
152.
Zurück zum Zitat Castro CM, Lima MM, Rasera Jr I, Oliveira Jr A, Gomes GP, Aparecida CG. Effects of Roux-en-Y gastric bypass on resting energy expenditure in women. Obes Surg. 2008;18:1376–80.CrossRef Castro CM, Lima MM, Rasera Jr I, Oliveira Jr A, Gomes GP, Aparecida CG. Effects of Roux-en-Y gastric bypass on resting energy expenditure in women. Obes Surg. 2008;18:1376–80.CrossRef
153.
Zurück zum Zitat Adami GF, Campostano A, Gandolfo P, Marinari G, Bessarione D, Scopinaro N. Body composition and energy expenditure in obese patients prior to and following biliopancreatic diversion for obesity. Eur Surg Res. 1996;28:295–8.PubMedCrossRef Adami GF, Campostano A, Gandolfo P, Marinari G, Bessarione D, Scopinaro N. Body composition and energy expenditure in obese patients prior to and following biliopancreatic diversion for obesity. Eur Surg Res. 1996;28:295–8.PubMedCrossRef
154.
Zurück zum Zitat Saeidi N, Meoli L, Nestoridi E, et al. Reprogramming of intestinal glucose metabolism and glycemic control in rats after gastric bypass. Science. 2013;341:406–10.PubMedPubMedCentralCrossRef Saeidi N, Meoli L, Nestoridi E, et al. Reprogramming of intestinal glucose metabolism and glycemic control in rats after gastric bypass. Science. 2013;341:406–10.PubMedPubMedCentralCrossRef
155.
Zurück zum Zitat Bobbioni-Harsch E, Morel P, Huber O, et al. Energy economy hampers body weight loss after gastric bypass. J Clin Endocrinol Metab. 2000;85:4695–700.PubMedCrossRef Bobbioni-Harsch E, Morel P, Huber O, et al. Energy economy hampers body weight loss after gastric bypass. J Clin Endocrinol Metab. 2000;85:4695–700.PubMedCrossRef
156.
Zurück zum Zitat Carey D, Pliego G, Raymond R. Body composition and metabolic changes following bariatric surgery: effects on fat mass, lean mass and basal metabolic rate: six months to one-year follow-up. Obes Surg. 2006;16:1602–8.PubMedCrossRef Carey D, Pliego G, Raymond R. Body composition and metabolic changes following bariatric surgery: effects on fat mass, lean mass and basal metabolic rate: six months to one-year follow-up. Obes Surg. 2006;16:1602–8.PubMedCrossRef
157.
Zurück zum Zitat Das SK, Roberts SB, Kehayias JJ, et al. Body composition assessment in extreme obesity and after massive weight loss induced by gastric bypass surgery. Am J Physiol Endocrinol Metab. 2003;284:E1080–8.PubMedCrossRef Das SK, Roberts SB, Kehayias JJ, et al. Body composition assessment in extreme obesity and after massive weight loss induced by gastric bypass surgery. Am J Physiol Endocrinol Metab. 2003;284:E1080–8.PubMedCrossRef
158.
Zurück zum Zitat Metcalf B, Rabkin R, Rabkin J, Metcalf L, Lehman-Becker L. Weight loss composition: the effects of exercise following obesity surgery as measured by bioelectrical impedance analysis. Obes Surg. 2005;15:183–6.PubMedCrossRef Metcalf B, Rabkin R, Rabkin J, Metcalf L, Lehman-Becker L. Weight loss composition: the effects of exercise following obesity surgery as measured by bioelectrical impedance analysis. Obes Surg. 2005;15:183–6.PubMedCrossRef
159.
Zurück zum Zitat O'Brien PE. Bariatric surgery: mechanisms, indications and outcomes. J Gastroenterol Hepatol. 2010;25:1358–65.PubMedCrossRef O'Brien PE. Bariatric surgery: mechanisms, indications and outcomes. J Gastroenterol Hepatol. 2010;25:1358–65.PubMedCrossRef
160.
Zurück zum Zitat Kumar R, Lieske JC, Collazo-Clavell ML, et al. Fat malabsorption and increased intestinal oxalate absorption are common after Roux-en-Y gastric bypass surgery. Surgery. 2011;149:654–61.PubMedPubMedCentralCrossRef Kumar R, Lieske JC, Collazo-Clavell ML, et al. Fat malabsorption and increased intestinal oxalate absorption are common after Roux-en-Y gastric bypass surgery. Surgery. 2011;149:654–61.PubMedPubMedCentralCrossRef
161.
Zurück zum Zitat Odstrcil EA, Martinez JG, Santa Ana CA, 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;92:704–13.PubMedCrossRef Odstrcil EA, Martinez JG, Santa Ana CA, 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;92:704–13.PubMedCrossRef
162.
Zurück zum Zitat Carswell KA, Vincent RP, Belgaumkar AP, et al. The effect of bariatric surgery on intestinal absorption and transit time. Obes Surg. 2014;1–10(5):796–805. 1–10.CrossRef Carswell KA, Vincent RP, Belgaumkar AP, et al. The effect of bariatric surgery on intestinal absorption and transit time. Obes Surg. 2014;1–10(5):796–805. 1–10.CrossRef
163.
Zurück zum Zitat Santry HP, Gillen DL, Lauderdale DS. Trends in bariatric surgical procedures. JAMA. 2005;294:1909–17.PubMedCrossRef Santry HP, Gillen DL, Lauderdale DS. Trends in bariatric surgical procedures. JAMA. 2005;294:1909–17.PubMedCrossRef
164.
Zurück zum Zitat National Bariatric Surgery Registry, First Registry Report to march 2010. 2010. Ref Type: Report National Bariatric Surgery Registry, First Registry Report to march 2010. 2010. Ref Type: Report
165.
Zurück zum Zitat Tack J, Arts J, Caenepeel P, De Wulf D, Bisschops R. Pathophysiology, diagnosis and management of postoperative dumping syndrome. Nat Rev Gastroenterol Hepatol. 2009;6:583–90.PubMedCrossRef Tack J, Arts J, Caenepeel P, De Wulf D, Bisschops R. Pathophysiology, diagnosis and management of postoperative dumping syndrome. Nat Rev Gastroenterol Hepatol. 2009;6:583–90.PubMedCrossRef
166.
Zurück zum Zitat Ukleja A. Dumping syndrome: pathophysiology and treatment. Nutr Clin Pract. 2005;20:517–25.PubMedCrossRef Ukleja A. Dumping syndrome: pathophysiology and treatment. Nutr Clin Pract. 2005;20:517–25.PubMedCrossRef
167.
Zurück zum Zitat Mathews DH. Change in effective circulating volume during experimental dumping syndrome. Surgery. 1960;48(1):185–94.PubMed Mathews DH. Change in effective circulating volume during experimental dumping syndrome. Surgery. 1960;48(1):185–94.PubMed
168.
Zurück zum Zitat Cummings DE, Foster-Schubert KE, Overduin J. Ghrelin and energy balance: focus on current controversies. Curr Drug Targets. 2005;6:153–69.PubMedCrossRef Cummings DE, Foster-Schubert KE, Overduin J. Ghrelin and energy balance: focus on current controversies. Curr Drug Targets. 2005;6:153–69.PubMedCrossRef
169.
Zurück zum Zitat Kapoor RR, James C, Hussain K. Advances in the diagnosis and management of hyperinsulinemic hypoglycemia. Nat Clin Pract End Met. 2009;5:101–12.CrossRef Kapoor RR, James C, Hussain K. Advances in the diagnosis and management of hyperinsulinemic hypoglycemia. Nat Clin Pract End Met. 2009;5:101–12.CrossRef
170.
Zurück zum Zitat Service GJ, Thompson GB, Service FJ, Andrews JC, Collazo-Clavell ML, Lloyd RV. Hyperinsulinemic hypoglycemia with nesidioblastosis after gastric-bypass surgery. N Engl J Med. 2005;353:249–54.PubMedCrossRef Service GJ, Thompson GB, Service FJ, Andrews JC, Collazo-Clavell ML, Lloyd RV. Hyperinsulinemic hypoglycemia with nesidioblastosis after gastric-bypass surgery. N Engl J Med. 2005;353:249–54.PubMedCrossRef
172.
Zurück zum Zitat Comeau E, Gagner M, Inabnet WB, Herron DM, Quinn TM, Pomp A. Symptomatic internal hernias after laparoscopic bariatric surgery. Surg Endosc. 2005;19:34–9.PubMedCrossRef Comeau E, Gagner M, Inabnet WB, Herron DM, Quinn TM, Pomp A. Symptomatic internal hernias after laparoscopic bariatric surgery. Surg Endosc. 2005;19:34–9.PubMedCrossRef
173.
Zurück zum Zitat Husain S, Ahmed AR, Johnson J, Boss T, O'Malley W. Small-bowel obstruction after laparoscopic Roux-en-Y gastric bypass: etiology, diagnosis, and management. Arch Surg. 2007;142:988.PubMedCrossRef Husain S, Ahmed AR, Johnson J, Boss T, O'Malley W. Small-bowel obstruction after laparoscopic Roux-en-Y gastric bypass: etiology, diagnosis, and management. Arch Surg. 2007;142:988.PubMedCrossRef
174.
Zurück zum Zitat Garza Jr E, Kuhn J, Arnold D, Nicholson W, Reddy S, McCarty T. Internal hernias after laparoscopic Roux-en-Y gastric bypass. Am J Surg. 2004;188:796–800.PubMedCrossRef Garza Jr E, Kuhn J, Arnold D, Nicholson W, Reddy S, McCarty T. Internal hernias after laparoscopic Roux-en-Y gastric bypass. Am J Surg. 2004;188:796–800.PubMedCrossRef
175.
Zurück zum Zitat Cho M, Carrodeguas L, Pinto D, et al. Diagnosis and management of partial small bowel obstruction after laparoscopic antecolic antegastric Roux-en-Y gastric bypass for morbid obesity. J Am Coll Surg. 2006;202:262–8.PubMedCrossRef Cho M, Carrodeguas L, Pinto D, et al. Diagnosis and management of partial small bowel obstruction after laparoscopic antecolic antegastric Roux-en-Y gastric bypass for morbid obesity. J Am Coll Surg. 2006;202:262–8.PubMedCrossRef
176.
Zurück zum Zitat Filip JE, Mattar SG, Bowers SP, Smith CD. Internal hernia formation after laparoscopic Roux-en-Y gastric bypass for morbid obesity. Am Surg. 2002;68:640–3.PubMed Filip JE, Mattar SG, Bowers SP, Smith CD. Internal hernia formation after laparoscopic Roux-en-Y gastric bypass for morbid obesity. Am Surg. 2002;68:640–3.PubMed
177.
Zurück zum Zitat Martin LC, Merkle EM, Thompson WM. Review of internal hernias: radiographic and clinical findings. Am J Roentgenol. 2006;186:703–17.CrossRef Martin LC, Merkle EM, Thompson WM. Review of internal hernias: radiographic and clinical findings. Am J Roentgenol. 2006;186:703–17.CrossRef
178.
Zurück zum Zitat Reddy SA, Yang C, McGinnis LA, Seggerman RE, Garza E, Ford III KL. Diagnosis of transmesocolic internal hernia as a complication of retrocolic gastric bypass: CT imaging criteria. Am J Roentgenol. 2007;189:52–5.CrossRef Reddy SA, Yang C, McGinnis LA, Seggerman RE, Garza E, Ford III KL. Diagnosis of transmesocolic internal hernia as a complication of retrocolic gastric bypass: CT imaging criteria. Am J Roentgenol. 2007;189:52–5.CrossRef
179.
Zurück zum Zitat Swartz D.E., Gonzalez V., Felix E.L. Anastomotic stenosis after Roux-en-Y gastric bypass: a rational approach to treatment. Surgery for Obesity and Related Diseases 2:632–636 Swartz D.E., Gonzalez V., Felix E.L. Anastomotic stenosis after Roux-en-Y gastric bypass: a rational approach to treatment. Surgery for Obesity and Related Diseases 2:632–636
180.
Zurück zum Zitat Halverson JD. Micronutrient deficiencies after gastric bypass for morbid obesity. Am Surg. 1986;52(11):594–8.PubMed Halverson JD. Micronutrient deficiencies after gastric bypass for morbid obesity. Am Surg. 1986;52(11):594–8.PubMed
181.
Zurück zum Zitat Smith CD, Herkes SB, Behrns KE, et al. Gastric acid secretion and vitamin B12 absorption after vertical Roux-gastric-Y bypass for morbid obesity. Ann Surg. 1993;218:91–6.PubMedPubMedCentralCrossRef Smith CD, Herkes SB, Behrns KE, et al. Gastric acid secretion and vitamin B12 absorption after vertical Roux-gastric-Y bypass for morbid obesity. Ann Surg. 1993;218:91–6.PubMedPubMedCentralCrossRef
182.
Zurück zum Zitat Decker GA, Swain JM, Crowell MD, Scolapio JS. Gastrointestinal and nutritional complications after bariatric surgery. Am J Gastroenterol. 2007;102:2571–80.PubMedCrossRef Decker GA, Swain JM, Crowell MD, Scolapio JS. Gastrointestinal and nutritional complications after bariatric surgery. Am J Gastroenterol. 2007;102:2571–80.PubMedCrossRef
183.
Zurück zum Zitat Balsiger, Bruno M., Kennedy, Frank P., Abu-Lebdeh, Haitham S., Collazo-Clavell, Maria, Jensen, Michael D., O'Brien, Timothy, Hensrud, Donald D., Dinneen, Sean F., Thompson, Geoffrey B., and Que, Florencia G. Prospective evaluation of Roux-en-Y gastric bypass as primary operation for medically complicated obesity. Mayo Clinic Proceedings 75[7], 673–680. 2000. Elsevier. Ref Type: Conference Proceeding Balsiger, Bruno M., Kennedy, Frank P., Abu-Lebdeh, Haitham S., Collazo-Clavell, Maria, Jensen, Michael D., O'Brien, Timothy, Hensrud, Donald D., Dinneen, Sean F., Thompson, Geoffrey B., and Que, Florencia G. Prospective evaluation of Roux-en-Y gastric bypass as primary operation for medically complicated obesity. Mayo Clinic Proceedings 75[7], 673–680. 2000. Elsevier. Ref Type: Conference Proceeding
184.
Zurück zum Zitat Avinoah E, Ovnat A, Charuzi I. Nutritional status seven years after Roux-en-Y gastric bypass surgery. Surgery. 1992;111:137–42.PubMed Avinoah E, Ovnat A, Charuzi I. Nutritional status seven years after Roux-en-Y gastric bypass surgery. Surgery. 1992;111:137–42.PubMed
185.
Zurück zum Zitat Rhode BM, Arseneau P, Cooper BA, Katz M, Gilfix BM, MacLean LD. Vitamin B-12 deficiency after gastric surgery for obesity. Am J Clin Nutr. 1996;63:103–9.PubMed Rhode BM, Arseneau P, Cooper BA, Katz M, Gilfix BM, MacLean LD. Vitamin B-12 deficiency after gastric surgery for obesity. Am J Clin Nutr. 1996;63:103–9.PubMed
186.
Zurück zum Zitat Brolin RE, Gorman JH, Petschenik AJ, Bradley LJ, Kenler HA, Cody RP. Are vitamin B12 and folate deficiency clinically important after roux-en-Y gastric bypass? J Gastrointest Surg. 1998;2:436–42.PubMedCrossRef Brolin RE, Gorman JH, Petschenik AJ, Bradley LJ, Kenler HA, Cody RP. Are vitamin B12 and folate deficiency clinically important after roux-en-Y gastric bypass? J Gastrointest Surg. 1998;2:436–42.PubMedCrossRef
187.
Zurück zum Zitat Updegraff TA, Neufeld NJ. Protein, iron, and folate status of patients prior to and following surgery for morbid obesity. J Am Diet Assoc. 1981;78:135.PubMed Updegraff TA, Neufeld NJ. Protein, iron, and folate status of patients prior to and following surgery for morbid obesity. J Am Diet Assoc. 1981;78:135.PubMed
188.
Zurück zum Zitat Brolin RE, LaMarca LB, Kenler HA, Cody RP. Malabsorptive gastric bypass in patients with superobesity. J Gastrointest Surg. 2002;6:195–205.PubMedCrossRef Brolin RE, LaMarca LB, Kenler HA, Cody RP. Malabsorptive gastric bypass in patients with superobesity. J Gastrointest Surg. 2002;6:195–205.PubMedCrossRef
189.
Zurück zum Zitat Wortsman J, Matsuoka LY, Chen TC, Lu Z, Holick MF. Decreased bioavailability of vitamin D in obesity. Am J Clin Nutr. 2000;72:690–3.PubMed Wortsman J, Matsuoka LY, Chen TC, Lu Z, Holick MF. Decreased bioavailability of vitamin D in obesity. Am J Clin Nutr. 2000;72:690–3.PubMed
190.
Zurück zum Zitat Coates PS, Fernstrom JD, Fernstrom MH, Schauer PR, Greenspan SL. Gastric bypass surgery for morbid obesity leads to an increase in bone turnover and a decrease in bone mass. J Clin Endocrinol Metabol. 2004;89:1061–5.CrossRef Coates PS, Fernstrom JD, Fernstrom MH, Schauer PR, Greenspan SL. Gastric bypass surgery for morbid obesity leads to an increase in bone turnover and a decrease in bone mass. J Clin Endocrinol Metabol. 2004;89:1061–5.CrossRef
191.
Zurück zum Zitat von Mach MA, Stoeckli R, Bilz S, Kraenzlin M, Langer I, Keller U. Changes in bone mineral content after surgical treatment of morbid obesity. Metabolism. 2004;53:918–21.CrossRef von Mach MA, Stoeckli R, Bilz S, Kraenzlin M, Langer I, Keller U. Changes in bone mineral content after surgical treatment of morbid obesity. Metabolism. 2004;53:918–21.CrossRef
192.
Zurück zum Zitat Shah M, Simha V, Garg A. Long-term impact of bariatric surgery on body weight, comorbidities, and nutritional status. J Clin Endocrinol Metabol. 2006;91:4223–31.CrossRef Shah M, Simha V, Garg A. Long-term impact of bariatric surgery on body weight, comorbidities, and nutritional status. J Clin Endocrinol Metabol. 2006;91:4223–31.CrossRef
193.
Zurück zum Zitat Abegg K, Gehring N, Wagner CA, et al. Roux-en-Y gastric bypass surgery reduces bone mineral density and induces metabolic acidosis in rats. Am J Physiol Regul Integr Comp Physiol. 2013;305:R999–R1009.PubMedCrossRef Abegg K, Gehring N, Wagner CA, et al. Roux-en-Y gastric bypass surgery reduces bone mineral density and induces metabolic acidosis in rats. Am J Physiol Regul Integr Comp Physiol. 2013;305:R999–R1009.PubMedCrossRef
194.
Zurück zum Zitat Lalmohamed A, de Vries F, Bazelier MT, et al. Risk of fracture after bariatric surgery in the United Kingdom: population based, retrospective cohort study. BMJ: Br Med J . 2012;345. Lalmohamed A, de Vries F, Bazelier MT, et al. Risk of fracture after bariatric surgery in the United Kingdom: population based, retrospective cohort study. BMJ: Br Med J . 2012;345.
195.
Zurück zum Zitat Langlois JA, Mussolino ME, Visser M, Looker AC, Harris T, Madans J. Weight loss from maximum body weight among middle-age and older white women and the risk of hip fracture: the NHANES I epidemiologic follow-up study. Osteoporos Int. 2011;12:763–8.CrossRef Langlois JA, Mussolino ME, Visser M, Looker AC, Harris T, Madans J. Weight loss from maximum body weight among middle-age and older white women and the risk of hip fracture: the NHANES I epidemiologic follow-up study. Osteoporos Int. 2011;12:763–8.CrossRef
196.
197.
Zurück zum Zitat Reid IR. Relationships among body mass, its components, and bone. IBMS BoneKEy. 2002. Reid IR. Relationships among body mass, its components, and bone. IBMS BoneKEy. 2002.
198.
Zurück zum Zitat Asplin JR, Coe FL. Hyperoxaluria in kidney stone formers treated with modern bariatric surgery. J Urol. 2007;177:565–9.PubMedCrossRef Asplin JR, Coe FL. Hyperoxaluria in kidney stone formers treated with modern bariatric surgery. J Urol. 2007;177:565–9.PubMedCrossRef
199.
Zurück zum Zitat Duffey BG, Pedro RN, Makhlouf A, et al. Roux-en-Y gastric bypass is associated with early increased risk factors for development of calcium oxalate nephrolithiasis. J Am Coll Surg. 2008;206:1145–53.PubMedCrossRef Duffey BG, Pedro RN, Makhlouf A, et al. Roux-en-Y gastric bypass is associated with early increased risk factors for development of calcium oxalate nephrolithiasis. J Am Coll Surg. 2008;206:1145–53.PubMedCrossRef
200.
Zurück zum Zitat Sinha MK, Collazo-Clavell ML, Rule A, et al. Hyperoxaluric nephrolithiasis is a complication of Roux-en-Y gastric bypass surgery. Kidney Int. 2007;72:100–7.PubMedCrossRef Sinha MK, Collazo-Clavell ML, Rule A, et al. Hyperoxaluric nephrolithiasis is a complication of Roux-en-Y gastric bypass surgery. Kidney Int. 2007;72:100–7.PubMedCrossRef
201.
Zurück zum Zitat Mole DR, Tomson CRV, Mortensen N, Winearls CG. Renal complications of jejuno-ileal bypass for obesity. QJM. 2001;94:69–77.PubMedCrossRef Mole DR, Tomson CRV, Mortensen N, Winearls CG. Renal complications of jejuno-ileal bypass for obesity. QJM. 2001;94:69–77.PubMedCrossRef
202.
Zurück zum Zitat Matlaga BR, Shore AD, Magnuson T, Clark JM, Johns R, Makary MA. Effect of gastric bypass surgery on kidney stone disease. J Urol. 2009;181:2573–7.PubMedCrossRef Matlaga BR, Shore AD, Magnuson T, Clark JM, Johns R, Makary MA. Effect of gastric bypass surgery on kidney stone disease. J Urol. 2009;181:2573–7.PubMedCrossRef
203.
Zurück zum Zitat Pinkney J. Consensus at last? The International Diabetes Federation statement on bariatric surgery in the treatment of obese type 2 diabetes. Diabet Med. 2011;28:884–5.PubMedCrossRef Pinkney J. Consensus at last? The International Diabetes Federation statement on bariatric surgery in the treatment of obese type 2 diabetes. Diabet Med. 2011;28:884–5.PubMedCrossRef
204.
Zurück zum Zitat Cummings D.E. Endocrine mechanisms mediating remission of diabetes after gastric bypass surgery. Int J Obes 0 AD; 33:S33-S40. Cummings D.E. Endocrine mechanisms mediating remission of diabetes after gastric bypass surgery. Int J Obes 0 AD; 33:S33-S40.
205.
Zurück zum Zitat Fruhbeck G. Bariatric and metabolic surgery: a shift in eligibility and success criteria. Nat Rev Endocrinol. 2015. Fruhbeck G. Bariatric and metabolic surgery: a shift in eligibility and success criteria. Nat Rev Endocrinol. 2015.
206.
Zurück zum Zitat Shin AC, Zheng H, Berthoud HR. 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.PubMedPubMedCentralCrossRef Shin AC, Zheng H, Berthoud HR. 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.PubMedPubMedCentralCrossRef
Metadaten
Titel
Mechanism Underlying the Weight Loss and Complications of Roux-en-Y Gastric Bypass. Review
verfasst von
G Abdeen
CW le Roux
Publikationsdatum
01.02.2016
Verlag
Springer US
Erschienen in
Obesity Surgery / Ausgabe 2/2016
Print ISSN: 0960-8923
Elektronische ISSN: 1708-0428
DOI
https://doi.org/10.1007/s11695-015-1945-7

Weitere Artikel der Ausgabe 2/2016

Obesity Surgery 2/2016 Zur Ausgabe

Original Contributions

Duodenal Electric Stimulation

Update Chirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

CME: 2 Punkte

Prof. Dr. med. Gregor Antoniadis Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

CME: 2 Punkte

Dr. med. Benjamin Meyknecht, PD Dr. med. Oliver Pieske Das Webinar S2e-Leitlinie „Distale Radiusfraktur“ beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

CME: 2 Punkte

Dr. med. Mihailo Andric
Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.