Imperial College London

ProfessorCarelLe Roux

Faculty of MedicineDepartment of Metabolism, Digestion and Reproduction

Visiting Professor
 
 
 
//

Contact

 

+44 (0)7970 719 453c.leroux

 
 
//

Location

 

08, east wingCharing Cross HospitalCharing Cross Campus

//

Summary

 

Publications

Publication Type
Year
to

345 results found

Qvigstad E, Gulseth HL, Risstad H, le Roux CW, Berg TJ, Mala T, Kristinsson JAet al., 2016, A novel technique of Roux-en-Y gastric bypass reversal for postprandial hyperinsulinemic hypoglycaemia: a case report, International Journal of Surgery Case Reports, Vol: 21, Pages: 91-94, ISSN: 2210-2612

BACKGROUND: We describe an evaluation of the effects of partial Roux-en-Y gastric bypass (RYGB) reversal on postprandial hyperinsulinaemic hypoglycaemia, insulin and GLP-1 levels. CASE SUMMARY: A 37 year old man was admitted with neuroglycopenia (plasma-glucose 1.6mmol/l) 18 months after RYGB, with normal 72h fasting test and abdominal CT. Despite dietary modifications and medical treatment, the hypoglycaemic episodes escalated in frequency. Feeding by a gastrostomy tube positioned in the gastric remnant did not prevent severe episodes of hypoglycaemia. A modified reversal of the RYGB was performed. Mixed meal tests were done perorally (PO), through the gastrostomy tube 1 (GT1), 4 weeks (GT2) after placement and 4 weeks after reversal (POr), with assessment of glucose, insulin and GLP-1 levels. RESULTS: Plasma-glucose increased to a maximum of 9.6, 5.4, 6.5 and 5.8mmol/l at the PO, GT1, GT2 and POr tests respectively. The corresponding insulin levels were 2939, 731, 725 and 463pmol/l. A decrease of plasma-glucose followed: 2.2, 3.0, 3.9 and 2.9mmol/l respectively and insulin levels were suppressed at 150min: 45, 22, 21 and 14pmol/l, respectively. GLP-1 levels increased in the PO test (60min: 122pmol/l, 21 fold of basal), but was attenuated in the two latter tests (12-23pmol/l at 60min). CONCLUSIONS: Reduction of plasma-glucose, insulin and GLP-1 excursions and symptoms were seen after gastric tube placement and partial RYGB reversal. This attenuation of GLP-1 response to feeding could reflect an adaptation to nutrients.

Journal article

van Avesaat M, Troost FJ, Westerterp-Plantenga MS, Helyes Z, Le Roux CW, Dekker J, Masclee AAM, Keszthelyi Det al., 2016, Capsaicin-induced satiety is associated with gastrointestinal distress but not with the release of satiety hormones, AMERICAN JOURNAL OF CLINICAL NUTRITION, Vol: 103, Pages: 305-313, ISSN: 0002-9165

Journal article

Coluzzi I, Raparelli L, Guarnacci L, Paone E, Del Genio G, le Roux CW, Silecchia Get al., 2016, Food Intake and Changes in Eating Behavior After Laparoscopic Sleeve Gastrectomy, Obesity Surgery, Vol: 26, Pages: 2059-2067, ISSN: 1708-0428

BACKGROUND: Laparoscopic sleeve gastrectomy (LSG) results in reduced calorie intake and weight loss. Whether patients consume the same types of food before and after surgery or whether they reduce the volume and calorie density of the foods they consume remains unknown. OBJECTIVES: The aim of this prospective study was to evaluate the changes in daily caloric and macronutrient intake after LSG and the relation between changes of taste and food tolerance over 2 years. METHODS: Thirty morbidly obese patients with median body mass index (BMI) of 43.9 kg/m(2) (39.5-57.3) were prospectively enrolled prior to LSG. Weight, BMI, %EWL, weight loss percentage (%WL), and daily intake were evaluated preoperatively at 1, 3, 6, 12, and 24 months after surgery along with a questionnaire evaluating food choices, quality of eating, tolerance of certain types of food, frequency of vomiting, and changes in taste. RESULTS: The median %EWL and %WL at 12 and 24 months was 65 % (33.9-93.6 %), 27.3 % (14.2-45.5 %) and 71.5 % (39.6-101.1 %), 31 % (19.1-50.3 %) respectively. Six months after surgery, the daily caloric intake reduced by 68 % and the reduction was maintained until 24 months. The median score of the eating questionnaire was 18 (10-27) at 6 months, 22 (16-26) at 12 months, and 23 (10-27) at 24 months, suggesting that the quality of nutrition improved over time. At 6, 12, and 24 months, 75 % of the patients reported changes in taste with reduced interest in sweets, high fat food, and alcoholic drinks. However, at 24 months, 20 % of patients reported a heightened interest in sweets compared to 12 months previously. CONCLUSIONS: LSG reduced calorie intake both through volume of food and the calorie density of the food consumed. The mechanisms for the changes in food preferences may involve both unconditioned and conditioned effects. The influence of dietary counseling on le

Journal article

le Roux CW, Engström M, Björnfot N, Fändriks L, Docherty NGet al., 2015, Equivalent Increases in Circulating GLP-1 Following Jejunal Delivery of Intact and Hydrolysed Casein: Relevance to Satiety Induction Following Bariatric Surgery., Obesity Surgery, Vol: 26, Pages: 1851-1858, ISSN: 1708-0428

BACKGROUND: Both Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG) decrease the latency of food delivery to the proximal small intestine. This is implicated in exaggerated post-prandial release of glucagon-like peptide 1 (GLP-1), which provokes early satiety and reductions in food intake. Altered stomach anatomy also creates a deficit in enzymatic pre-processing. The impact of this state effect as a modulator of gut hormone responses remains underexplored. METHODS: A double-blind cross-over trial study was conducted in 13 healthy subjects assigned to receive in the fasted state and in random order at 1 week apart, a direct jejunal infusion of either intact casein or a casein hydrolysate. Downstream effects on GLP-1 release, ratings of hunger and fullness and food and water intake on each study day were recorded when an ad libitum meal was provided 30 min after the infusion. RESULTS: Circulating GLP-1 was increased 25 min after infusions and peaked to a similar degree at 15 min post-meal initiation. The hormone surge had no impact on ratings of hunger and fullness ahead of the ad libitum meal. The kinetic and magnitude of satiation following each infusion was not significantly different. Food and water intake were likewise not differentially impacted by the two infusion types. CONCLUSIONS: Protein macronutrient state upon arrival in the small intestine does not in isolation impact upon GLP-1 responses and subsequent onset of satiety. This potentially points to rate of delivery being the dominant factor in exaggerated post-prandial GLP-1 responses in patients post-RYGB and VSG.

Journal article

del Genio G, Gagner M, Limongelli P, Tolone S, Pournaras D, le Roux CW, Brusciano L, Mozzillo AL, del Genio F, Docimo Let al., 2015, Remission of type 2 diabetes in patients undergoing biliointestinal bypass for morbid obesity: a new surgical treatment, Surgery for Obesity and Related Diseases, Vol: 12, Pages: 815-821, ISSN: 1878-7533

BackgroundNutrient interaction with the mid-gut may play a role in improving type 2 diabetes mellitus (T2D) after bariatric surgery. However, Roux-en-Y gastric bypass, biliopancreatic diversion, and sleeve gastrectomy include diversion of food from the duodenum and/or partial gastrectomy. Biliointestinal bypass (BIBP) was introduced to eliminate the major side effects of jejunoileal bypass. It does not involve any change to the anatomy of the stomach or the duodenum. A prospective evaluation of the role of BIBP in glycemic control has not been reported.ObjectivesLongitudinal evaluation of T2D after BIBP.SettingUniversity hospitals in Europe and Canada.MethodThe effects of BIBP on metabolism and glycemia in 28 consecutive patients with T2D were evaluated over 2 years.ResultsDecreases (P<.001) in fasting glycemia, insulinemia, and homeostasis model assessment were observed 3 months after surgery, were improved after 1 year, and remained stable after 2 years. Glycosylated hemoglobin levels decreased at 3, 12, and 24 months after surgery (from 9.2±2.1 to 6.3±1.1 (P<.0001), 4.9±1.7 (P<.0001), and 4.8±1.1 (P<.0001), after 3, 12, and 24 months, respectively). Medical therapy was discontinued in 83% (20 of 24) of the patients; for the remaining 17% (4 of 24), therapy was reduced to oral hypoglycemic agents.ConclusionBIBP had a favorable risk–benefit relationship and positive metabolic effects in the short term. How BIBP achieves optimal glycemic control and whether it improves β-cell function and/or insulin sensitivity require further study.

Journal article

Boxwell AJ, Chen Z, Mathes CM, Spector AC, Le Roux CW, Travers SP, Travers JBet al., 2015, Effects of high-fat diet and gastric bypass on neurons in the caudal solitary nucleus, PHYSIOLOGY & BEHAVIOR, Vol: 152, Pages: 329-339, ISSN: 0031-9384

Journal article

Docherty NG, Le Roux CW, 2015, Physiological adaptations following Roux-en-Y gastric bypass and the identification of targets for bariatric mimetic pharmacotherapy, CURRENT OPINION IN PHARMACOLOGY, Vol: 25, Pages: 23-29, ISSN: 1471-4892

Journal article

Goldstone AP, Miras AD, Scholtz S, Jackson S, Neff KJ, Penicaud L, Geoghegan J, Chhina N, Durighel G, Bell JD, Meillon S, le Roux CWet al., 2015, Link between increased satiety gut hormones and reduced food reward following gastric bypass surgery for obesity, Journal of Clinical Endocrinology & Metabolism, Vol: 101, Pages: 599-609, ISSN: 1945-7197

Context: Roux-en-Y gastric bypass (RYGB) surgery is an effective long-term intervention for weightloss maintenance, reducing appetite, and also food reward, via unclear mechanisms.Objective: To investigate the role of elevated satiety gut hormones after RYGB, we examined foodhedonic-reward responses following their acute post-prandial suppression.Design: Randomised placebo-controlled double-blind cross-over experimental medicine studies.Patients: Two groups, over 5 months after RYGB for obesity (n7–11), compared with non-obesecontrols (n10), or patients after gastric banding (BAND) surgery (n9).Intervention: Studies were performed after acute administration of the somatostatin analogueOctreotide or saline. In one study, patients after RYGB, and non-obese controls, performed abehavioral progressive ratio task (PRT) for chocolate sweets. In another study, patients after RYGB,and controls after BAND surgery, performed a functional magnetic resonance imaging (fMRI) foodpicture evaluation task.Main outcome measures: Octreotide increased both appetitive food reward (breakpoint) in thePRT (n9), and food appeal (n9) and reward system blood oxygen level dependent (BOLD) signal(n7) in the fMRI task, in the RYGB group, but not in control groups.ISSN

Journal article

Abdeen G, le Roux CW, 2015, Mechanism Underlying the Weight Loss and Complications of Roux-en-Y Gastric Bypass. Review, Obesity Surgery, Vol: 26, Pages: 410-421, ISSN: 1708-0428

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.

Journal article

Elliott JA, Jackson S, King S, McHugh R, Docherty NG, Reynolds JV, le Roux CWet al., 2015, Gut Hormone Suppression Increases Food Intake After Esophagectomy With Gastric Conduit Reconstruction, ANNALS OF SURGERY, Vol: 262, Pages: 824-830, ISSN: 0003-4932

Journal article

Lho Y, le Roux CW, Park HS, Kim GS, Jung J, Hwang G-S, Seo YK, Ha TK, Ha Eet al., 2015, Changes in Glucose Metabolism in Vertical Sleeve Gastrectomy, OBESITY SURGERY, Vol: 25, Pages: 2002-2010, ISSN: 0960-8923

Journal article

Pournaras DJ, le Roux CW, 2015, Bariatric Surgery and Decreasing Vascular Risk, Angiology, Vol: 67, Pages: 610-611, ISSN: 1940-1574

Journal article

Mathes CM, Bohnenkamp RA, le Roux CW, Spector ACet al., 2015, Reduced sweet and fatty fluid intake after Roux-en-Y gastric bypass in rats is dependent on experience without change in stimulus motivational potency, AMERICAN JOURNAL OF PHYSIOLOGY-REGULATORY INTEGRATIVE AND COMPARATIVE PHYSIOLOGY, Vol: 309, Pages: R864-R874, ISSN: 0363-6119

Journal article

Maguire PJ, Power KA, O'Higgins AC, Jackson S, Harley R, le Roux CW, Turner MJet al., 2015, Maternal C-reactive protein in early pregnancy, EUROPEAN JOURNAL OF OBSTETRICS & GYNECOLOGY AND REPRODUCTIVE BIOLOGY, Vol: 193, Pages: 79-82, ISSN: 0301-2115

Journal article

Goldstone AP, 2015, Changes in Reward after Gastric Bypass: the Advantages and Disadvantages, Current Atherosclerosis Reports, Vol: 17, ISSN: 1534-6242

Gastric bypass surgery is an effective long-term weight loss intervention. Key to its success appears a putative shift in food preference away from high-energy-density foods associated with a reduced appetitive drive and loss of neural reactivity in the reward system of the brain towards food. Post-prandial exaggerated satiety gut hormone responses have been implicated as mediators. Whilst the positive impact of bariatric surgery on both physical and psychological outcomes for many patients is clearly evident, a subset of patients appear to be detrimentally affected by this loss of reward from food and by a lack of alternative strategies for regulating affect after surgery. Mindfulness training has emerged as a potential tool in reducing the need for immediate reward that underpins much of eating behaviour. Further research is needed to help identify patients who may be more vulnerable after gastric bypass and which forms of support may be most beneficial.

Journal article

Pournaras DJ, le Roux CW, 2015, Type 2 diabetes: multimodal treatment of a complex disease, LANCET, Vol: 386, Pages: 936-937, ISSN: 0140-6736

Journal article

Belgaumkar AP, Vincent RP, Carswell KA, Hughes RD, Alaghband-Zadeh J, Mitry RR, le Roux CW, Patel AGet al., 2015, Changes in Bile Acid Profile After Laparoscopic Sleeve Gastrectomy are Associated with Improvements in Metabolic Profile and Fatty Liver Disease, Obesity Surgery, Vol: 26, Pages: 1195-1202, ISSN: 1708-0428

BACKGROUND: Bile acids (BA) modulate lipid and glucose metabolism in a feedback loop through production of fibroblast growth factor (FGF) 19 in the terminal ileum. Changes in BA after bariatric surgery may lead to improvements in the metabolic syndrome, including fatty liver disease. This study investigated the relationship between BA and metabolic and inflammatory profiles after laparoscopic sleeve gastrectomy (LSG). METHODS: Patients undergoing LSG had fasting blood samples taken pre-operatively and 6 months post-surgery. Liver injury was measured using cytokeratin (CK) 18 fragments. BA were measured using liquid chromatography tandem-mass spectrometry. FGF-19 was measured using enzyme-linked immunosorbent assay. RESULTS: The study included 18 patients (12 females), with mean age 46.3 years (SEM ± 2.9) and BMI 60.1 kg/m(2) (±2.6). After 6 months, patients lost 39.8 kg (±3.1; p < 0.001). Fourteen patients (78 %) had steatosis. FGF-19 increased from median 128.1 (IQR 89.4-210.1) to 177.1 (121.8-288.9, p = 0.045) at 6 months. Although total BA did not change, primary glycine- and taurine-conjugated BA, cholic acid decreased, and secondary BA, glycine-conjugated urodeoxycholic acid increased over the study period. These changes are associated with reduction in insulin resistance, pro-inflammatory cytokines and CK-18 levels. CONCLUSIONS: The profile of individual BA is altered after LSG. These changes occur in the presence of reductions in inflammatory cytokines and markers of liver injury. This study supports evidence from recent animal models that LSG may have an effect on fatty liver through changes in BA metabolism.

Journal article

Cohen R, Galvao Neto MP, Le Roux CW, 2015, Another look at the (endoscopic duodenal liner) ENDO trial, or how to avoid the burial of a valuable antidiabetic tool, SURGERY FOR OBESITY AND RELATED DISEASES, Vol: 12, Pages: 702-704, ISSN: 1550-7289

Journal article

Tremaroli V, Karlsson F, Werling M, Ståhlman M, Kovatcheva-Datchary P, Olbers T, Fändriks L, le Roux CW, Nielsen J, Bäckhed Fet al., 2015, Roux-en-Y Gastric Bypass and Vertical Banded Gastroplasty Induce Long-Term Changes on the Human Gut Microbiome Contributing to Fat Mass Regulation, Cell Metabolism, Vol: 22, Pages: 228-238, ISSN: 1932-7420

Bariatric surgery is currently the most effective procedure for the treatment of obesity. Given the role of the gut microbiota in regulating host metabolism and adiposity, we investigated the long-term effects of bariatric surgery on the microbiome of patients randomized to Roux-en-Y gastric bypass or vertical banded gastroplasty and matched for weight and fat mass loss. The two surgical procedures induced similar and durable changes on the gut microbiome that were not dependent on body mass index and resulted in altered levels of fecal and circulating metabolites compared with obese controls. By colonizing germ-free mice with stools from the patients, we demonstrated that the surgically altered microbiota promoted reduced fat deposition in recipient mice. These mice also had a lower respiratory quotient, indicating decreased utilization of carbohydrates as fuel. Our results suggest that the gut microbiota may play a direct role in the reduction of adiposity observed after bariatric surgery.

Journal article

del Genio G, Ferreri C, Marfella R, Pournaras D, le Roux CW, del Genio F, Paolo L, Tolone S, Docimo L, Puca AAet al., 2015, Morbid Obesity is Associated to Altered Fatty Acid Profile of Erythrocyte Membranes, JOURNAL OF DIABETES & METABOLISM, Vol: 6, ISSN: 2155-6156

Journal article

Docherty NG, Canney AL, le Roux CW, 2015, Weight Loss Interventions and Progression of Diabetic Kidney Disease, CURRENT DIABETES REPORTS, Vol: 15, ISSN: 1534-4827

Journal article

Pi-Sunyer X, Astrup A, Fujioka K, Greenway F, Halpern A, Krempf M, Lau DCW, le Roux CW, Violante Ortiz R, Jensen CB, Wilding JPHet al., 2015, A Randomized, Controlled Trial of 3.0 mg of Liraglutide in Weight Management, NEW ENGLAND JOURNAL OF MEDICINE, Vol: 373, Pages: 11-22, ISSN: 0028-4793

Journal article

Miras AD, Chuah LL, Khalil N, Nicotra A, Vusirikala A, Baqai N, Graham C, Ravindra S, Lascaratos G, Oliver N, le Roux CWet al., 2015, Type 2 diabetes mellitus and microvascular complications 1 year after Roux-en-Y gastric bypass: a case-control study, DIABETOLOGIA, Vol: 58, Pages: 1443-1447, ISSN: 0012-186X

Journal article

Alsters SIM, Goldstone AP, Buxton JL, Zekavati A, sosinsky A, Yiorkas AM, Holder S, Klaber RE, Bridges N, Haelst MM, le roux CW, Walley AJ, Walters RG, Mueller M, Blakemore AIFet al., 2015, Truncating homozygous mutation of carboxypeptidase E (CPE) in a morbidly obese female with type 2 diabetes mellitus, intellectual disability and hypogonadotrophic hypogonadism, PLOS One, Vol: 10, ISSN: 1932-6203

Carboxypeptidase E is a peptide processing enzyme, involved in cleaving numerous peptide precursors, including neuropeptides and hormones involved in appetite control and glucose metabolism. Exome sequencing of a morbidly obese female from a consanguineous family revealed homozygosity for a truncating mutation of the CPE gene (c.76_98del; p.E26RfsX68). Analysis detected no CPE expression in whole blood-derived RNA from the proband, consistent with nonsense-mediated decay. The morbid obesity, intellectual disability, abnormal glucose homeostasis and hypogonadotrophic hypogonadism seen in this individual recapitulates phenotypes in the previously described fat/fat and Cpe knockout mouse models, evidencing the importance of this peptide/hormone-processing enzyme in regulating body weight, metabolism, and brain and reproductive function in humans.

Journal article

Werling M, Fandriks L, Olbers T, Bueter M, Sjostrom L, Lonroth H, Wallenius V, Stenlof K, le Roux CWet al., 2015, Roux-en-Y Gastric Bypass Surgery Increases Respiratory Quotient and Energy Expenditure during Food Intake, PLOS ONE, Vol: 10, ISSN: 1932-6203

Journal article

Arora T, Velagapudi V, Pournaras DJ, Welbourn R, le Roux CW, Oresic M, Backhed Fet al., 2015, Roux-en-Y Gastric Bypass Surgery Induces Early Plasma Metabolomic and Lipidomic Alterations in Humans Associated with Diabetes Remission, PLOS ONE, Vol: 10, ISSN: 1932-6203

Journal article

Elliott JA, Le Roux CW, 2015, How long should we make the biliopancreatic limb during Roux-en-Y gastric bypass?, SURGERY FOR OBESITY AND RELATED DISEASES, Vol: 11, Pages: 1246-1247, ISSN: 1550-7289

Journal article

Dutia R, Embrey M, O'Brien S, Haeusler RA, Agenor KK, Homel P, McGinty J, Vincent RP, Alaghband-Zadeh J, Staels B, le Roux CW, Yu J, Laferrere Bet al., 2015, Temporal changes in bile acid levels and 12 alpha-hydroxylation after Roux-en-Y gastric bypass surgery in type 2 diabetes, INTERNATIONAL JOURNAL OF OBESITY, Vol: 39, Pages: 806-813, ISSN: 0307-0565

Journal article

Tham JC, le Roux CW, Docherty NG, 2015, Cardiovascular, Renal and Overall Health Outcomes After Bariatric Surgery, CURRENT CARDIOLOGY REPORTS, Vol: 17, ISSN: 1523-3782

Journal article

Mathes CM, Bohnenkamp RA, Blonde GD, Letourneau C, Corteville C, Bueter M, Lutz TA, le Roux CW, Spector ACet al., 2015, Gastric bypass in rats does not decrease appetitive behavior towards sweet or fatty fluids despite blunting preferential intake of sugar and fat, PHYSIOLOGY & BEHAVIOR, Vol: 142, Pages: 179-188, ISSN: 0031-9384

Journal article

This data is extracted from the Web of Science and reproduced under a licence from Thomson Reuters. You may not copy or re-distribute this data in whole or in part without the written consent of the Science business of Thomson Reuters.

Request URL: http://wlsprd.imperial.ac.uk:80/respub/WEB-INF/jsp/search-html.jsp Request URI: /respub/WEB-INF/jsp/search-html.jsp Query String: id=00303866&limit=30&person=true&page=6&respub-action=search.html