101
|
Abstract
Obesity is closely associated with the development of type 2 diabetes. Many strategies have been used in the past to combat these two conditions, but very few provide for stable and durable glycemic control. Bariatric surgery has emerged as a powerful tool for treating obesity and in over 70 % of cases provides a short-term cure for diabetes. While the acute metabolic effects of surgery are striking, it remains important for us to also consider the long-term effects. This review aims to summarize the chronic or long-term metabolic and physiological effects of Roux-en-Y gastric bypass (RYGB) surgery on pancreatic function, skeletal muscle and hepatic insulin sensitivity, and gastrointestinal remodeling. An increased understanding of the current state of research in these areas can provide the basis for stimulating further research that would contribute to new treatment and management strategies for obesity and diabetes.
Collapse
Affiliation(s)
- J David Mosinski
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - John P Kirwan
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
- Metabolic Translational Research Center, Cleveland Clinic, Cleveland, OH, 44195, USA.
| |
Collapse
|
102
|
Lee CJ, Craig Wood G, Lazo M, Brown TT, Clark JM, Still C, Benotti P. Risk of post-gastric bypass surgery hypoglycemia in nondiabetic individuals: A single center experience. Obesity (Silver Spring) 2016; 24:1342-8. [PMID: 27225597 PMCID: PMC4919116 DOI: 10.1002/oby.21479] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/28/2015] [Accepted: 01/07/2016] [Indexed: 01/06/2023]
Abstract
OBJECTIVE The epidemiology of post-gastric bypass surgery hypoglycemia (PGBH) is incompletely understood. This study aimed to evaluate the risk of PGBH among nondiabetic patients and associated factors. METHODS A cohort study of nondiabetic patients who underwent Roux-en-Y gastric bypass (RYGB) was conducted. PGBH was defined by any postoperative record of glucose < 60 mg/dL, diagnosis of hypoglycemia, or any medication use for treatment of PGBH. Kaplan-Meier analysis was used to describe PGBH occurrence, log-rank tests, and Cox regression to examine associated factors. RESULTS Of the 1,206 eligible patients, 86% were female with mean age of 43.7 years, mean preoperative BMI of 48.7 kg/m(2) , and a mean follow-up of 4.8 years. The cumulative incidence of hypoglycemia at 1 and 5 years post-RYGB was 2.7% and 13.3%, respectively. Incidence of PGBH was identified in 158 patients and was associated with lower preoperative BMI (P = 0.048), lower preoperative HbA1c (P = 0.012), and higher 6-month percent of excess body weight loss (%EWL) (P = 0.001). A lower preoperative HbA1c (HR = 1.73, P = 0.0034) and higher 6-month %EWL (HR = 1.96, P = 0.0074) remained independently correlated with increased risk for PGBH in multi-regression analysis. CONCLUSIONS The 5-year incidence of PGBH among nondiabetic individuals was 13.3% and was associated with a lower preoperative HbA1c and greater weight loss at 6 months following surgery.
Collapse
Affiliation(s)
- Clare J. Lee
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, the Johns Hopkins University, Baltimore, Maryland, USA
| | - G. Craig Wood
- Geisinger Obesity Research Institute, Geisinger Clinic, Danville, Pennsylvania, USA
| | - Mariana Lazo
- Division of General Internal Medicine, Department of Medicine, the Johns Hopkins University, Baltimore, Maryland, USA
| | - Todd T. Brown
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, the Johns Hopkins University, Baltimore, Maryland, USA
| | - Jeanne M. Clark
- Division of General Internal Medicine, Department of Medicine, the Johns Hopkins University, Baltimore, Maryland, USA
| | - Christopher Still
- Geisinger Obesity Research Institute, Geisinger Clinic, Danville, Pennsylvania, USA
| | - Peter Benotti
- Geisinger Obesity Research Institute, Geisinger Clinic, Danville, Pennsylvania, USA
| |
Collapse
|
103
|
Stefanidis A, Forrest N, Brown WA, Dixon JB, O’Brien PB, Juliane Kampe, Oldfield BJ. An investigation of the neural mechanisms underlying the efficacy of the adjustable gastric band. Surg Obes Relat Dis 2016; 12:828-838. [DOI: 10.1016/j.soard.2015.11.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/19/2015] [Accepted: 11/19/2015] [Indexed: 02/08/2023]
|
104
|
El-Salhy M, Mazzawi T, Hausken T, Hatlebakk JG. Interaction between diet and gastrointestinal endocrine cells. Biomed Rep 2016; 4:651-656. [PMID: 27284402 PMCID: PMC4887949 DOI: 10.3892/br.2016.649] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/01/2016] [Indexed: 02/07/2023] Open
Abstract
The gastrointestinal endocrine cells are essential for life. They regulate the gastrointestinal motility, secretion, visceral sensitivity, absorption, local immune defense, cell proliferation and appetite. These cells act as sensory cells with specialized microvilli that project into the lumen that sense the gut contents (mostly nutrients and/or bacteria byproducts), and respond to luminal stimuli by releasing hormones into the lamina propria. These released hormones exert their actions by entering the circulating blood and reaching distant targets (endocrine mode), nearby structures (paracrine mode) or via afferent and efferent synaptic transmission. The mature intestinal endocrine cells are capable of expressing several hormones. A change in diet not only affects the release of gastrointestinal hormones, but also alters the densities of the gut endocrine cells. The interaction between ingested foodstuffs and the gastrointestinal endocrine cells can be utilized for the clinical management of gastrointestinal and metabolic diseases, such as irritable bowel syndrome, obesity and diabetes.
Collapse
Affiliation(s)
- Magdy El-Salhy
- Section for Gastroenterology, Department of Medicine, Stord Helse-Fonna Hospital, 5409 Stord, Norway; Section for Gastroenterology, Department of Clinical Medicine, University of Bergen, 5020 Bergen, Norway; Department of Medicine, National Centre for Functional Gastrointestinal Disorders, Haukeland University Hospital, 5021 Bergen, Norway
| | - Tarek Mazzawi
- Section for Gastroenterology, Department of Clinical Medicine, University of Bergen, 5020 Bergen, Norway; Department of Medicine, National Centre for Functional Gastrointestinal Disorders, Haukeland University Hospital, 5021 Bergen, Norway
| | - Trygve Hausken
- Section for Gastroenterology, Department of Clinical Medicine, University of Bergen, 5020 Bergen, Norway; Department of Medicine, National Centre for Functional Gastrointestinal Disorders, Haukeland University Hospital, 5021 Bergen, Norway
| | - Jan Gunnar Hatlebakk
- Section for Gastroenterology, Department of Clinical Medicine, University of Bergen, 5020 Bergen, Norway; Department of Medicine, National Centre for Functional Gastrointestinal Disorders, Haukeland University Hospital, 5021 Bergen, Norway
| |
Collapse
|
105
|
Svane MS, Bojsen-Møller KN, Nielsen S, Jørgensen NB, Dirksen C, Bendtsen F, Kristiansen VB, Hartmann B, Holst JJ, Madsbad S. Effects of endogenous GLP-1 and GIP on glucose tolerance after Roux-en-Y gastric bypass surgery. Am J Physiol Endocrinol Metab 2016; 310:E505-14. [PMID: 26786780 DOI: 10.1152/ajpendo.00471.2015] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/11/2016] [Indexed: 02/07/2023]
Abstract
Exaggerated secretion of glucagon-like peptide 1 (GLP-1) is important for postprandial glucose tolerance after Roux-en-Y gastric bypass (RYGB), whereas the role of glucose-dependent insulinotropic polypeptide (GIP) remains to be resolved. We aimed to explore the relative importance of endogenously secreted GLP-1 and GIP on glucose tolerance and β-cell function after RYGB. We used DPP-4 inhibition to enhance concentrations of intact GIP and GLP-1 and the GLP-1 receptor antagonist exendin-(9-39) (Ex-9) for specific blockage of GLP-1 actions. Twelve glucose-tolerant patients were studied after RYGB in a randomized, placebo-controlled, 4-day crossover study with standard mixed-meal tests and concurrent administration of placebo, oral sitagliptin, Ex-9 infusion, or combined Ex-9-sitagliptin. GLP-1 receptor antagonism increased glucose excursions, clearly attenuated β-cell function, and aggravated postprandial hyperglucagonemia compared with placebo, whereas sitagliptin had no effect despite two- to threefold increased concentrations of intact GLP-1 and GIP. Similarly, sitagliptin did not affect glucose tolerance or β-cell function during GLP-1R blockage. This study confirms the importance of GLP-1 for glucose tolerance after RYGB via increased insulin and attenuated glucagon secretion in the postprandial state, whereas amplification of the GIP signal (or other DPP-4-sensitive glucose-lowering mechanisms) did not appear to contribute to the improved glucose tolerance seen after RYGB.
Collapse
Affiliation(s)
- Maria S Svane
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| | - Kirstine N Bojsen-Møller
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Signe Nielsen
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Nils B Jørgensen
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Carsten Dirksen
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Flemming Bendtsen
- Department of Surgical and Medical Gastroenterology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; and
| | - Viggo B Kristiansen
- Department of Surgical and Medical Gastroenterology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; and
| | - Bolette Hartmann
- NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
106
|
Holst JJ, Madsbad S. Mechanisms of surgical control of type 2 diabetes: GLP-1 is key factor. Surg Obes Relat Dis 2016; 12:1236-42. [PMID: 27313194 DOI: 10.1016/j.soard.2016.02.033] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 02/23/2016] [Indexed: 01/07/2023]
Abstract
GLP-1 secretion in response to meals is dramatically increased after gastric bypass operations. GLP-1 is a powerful insulinotropic and anorectic hormone, and analogs of GLP-1 are widely used for the treatment of diabetes and recently approved also for obesity treatment. It is, therefore, reasonable to assume that the exaggerated GLP-1 secretion contributes to the antidiabetic and anorectic effects of gastric bypass. Indeed, human experiments with the GLP-1 receptor antagonist, Exendin 9-39, have shown that the improved insulin secretion, which is responsible for part of the antidiabetic effect of the operation, is reduced and or abolished after GLP-1 receptor blockade. Also the postoperative improvement of glucose tolerance is eliminated and or reduced by the antagonist, pointing to a key role for the exaggerated GLP-1 secretion. Indeed, there is evidence that the exaggerated GLP-1 secretion is also responsible for postprandial hypoglycemia sometimes observed after bypass. Other operations (biliopancreatic-diversion and or sleeve gastrectomy) appear to involve different and/or additional mechanisms, and so does experimental bariatric surgery in rodents. However, unlike bypass surgery in humans, the rodent operations are generally associated with increased energy metabolism pointing to an entirely different mechanism of action in the animals.
Collapse
Affiliation(s)
- Jens Juul Holst
- NNF Center for Basic Metabolic Research and Department of Biomedical Research, the Panum Institute, Copenhagen, Denmark.
| | - Sten Madsbad
- Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
107
|
Meek CL, Lewis HB, Reimann F, Gribble FM, Park AJ. The effect of bariatric surgery on gastrointestinal and pancreatic peptide hormones. Peptides 2016; 77:28-37. [PMID: 26344355 DOI: 10.1016/j.peptides.2015.08.013] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 08/18/2015] [Accepted: 08/19/2015] [Indexed: 12/17/2022]
Abstract
Bariatric surgery for obesity has proved to be an extremely effective method of promoting long-term weight reduction with additional beneficial metabolic effects, such as improved glucose tolerance and remission of type 2 diabetes. A range of bariatric procedures are in common use, including gastric banding, sleeve gastrectomy and the Roux-en-Y gastric bypass. Although the mechanisms underlying the efficacy of bariatric surgery are unclear, gastrointestinal and pancreatic peptides are thought to play an important role. The aim of this review is to summarise the effects of different bariatric surgery procedures upon gastrointestinal and pancreatic peptides, including ghrelin, gastrin, cholecystokinin (CCK), glucose-dependent insulinotropic hormone (GIP), glucagon-like peptide 1 (GLP-1), peptide YY (PYY), oxyntomodulin, insulin, glucagon and somatostatin.
Collapse
Affiliation(s)
- Claire L Meek
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes's Hospital, Box 289, Hills Road, Cambridge CB2 0QQ, United Kingdom; Department of Clinical Biochemistry, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Hannah B Lewis
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes's Hospital, Box 289, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Frank Reimann
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes's Hospital, Box 289, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Fiona M Gribble
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes's Hospital, Box 289, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Adrian J Park
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes's Hospital, Box 289, Hills Road, Cambridge CB2 0QQ, United Kingdom; Department of Clinical Biochemistry, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
108
|
Chaudhry S, Bernardes M, Harris PE, Maffei A. Gastrointestinal dopamine as an anti-incretin and its possible role in bypass surgery as therapy for type 2 diabetes with associated obesity. MINERVA ENDOCRINOL 2016; 41:43-56. [PMID: 26505694 PMCID: PMC5079753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The objective of this review was to summarize and integrate specific clinical observations from the field of gastric bypass surgery and recent findings in beta cell biology. When considered together, these data sets suggest a previously unrecognized physiological mechanism which may explain how Roux-en-Y gastric bypass (RYGB) surgery mediates the early rapid reversal of hyperglycemia, observed before weight loss, in certain type 2 diabetes mellitus (T2DM) patients. The novel mechanism is based on a recently recognized inhibitory circuit of glucose stimulated insulin secretion driven by DA stored in β-cell vesicles and the gut. We propose that DA and glucagon-like peptide 1 (GLP-1) represent two opposing arms of a glucose stimulated insulin secretion (GSIS) regulatory system and hypothesize that dopamine represents the "anti-incretin" hypothesized to explain the beneficial effects of bariatric surgery on T2DM. These new hypotheses and the research driven by them may directly impact our understanding of: 1) the mechanisms underlying improved glucose homeostasis seen before weight loss following bariatric surgery; and 2) the regulation of glucose stimulated insulin secretion within islets. On a practical level, these studies may result in the development of novels drugs to modulate insulin secretion and/or methods to quantitatively asses in real time beta cell function and mass.
Collapse
Affiliation(s)
- Suleman Chaudhry
- Department of Surgery, Columbia University Medical Center, New York, NY, USA -
| | | | | | | |
Collapse
|
109
|
Hao Z, Mumphrey MB, Townsend RL, Morrison CD, Münzberg H, Ye J, Berthoud HR. Reprogramming of defended body weight after Roux-En-Y gastric bypass surgery in diet-induced obese mice. Obesity (Silver Spring) 2016; 24:654-60. [PMID: 26847390 PMCID: PMC4769678 DOI: 10.1002/oby.21400] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 10/26/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Roux-en-Y gastric bypass surgery (RYGB) results in sustained lowering of body weight in most patients, but the mechanisms involved are poorly understood. The aim of this study was to obtain support for the notion that reprogramming of defended body weight, rather than passive restriction of energy intake, is a fundamental mechanism of RYGB. METHODS Male C57BL6J mice reaching different degrees of obesity on a high-fat diet either with ad libitum access or with caloric restriction (weight-reduced) were subjected to RYGB. RESULTS RYGB-induced weight loss and fat mass loss were proportional to pre-surgical levels, with moderately obese mice losing less body weight and fat compared with very obese mice. Remarkably, mice that were weight-reduced to the level of chow controls before surgery immediately gained weight after surgery, exclusively accounted for by lean mass gain. CONCLUSIONS The results provide additional evidence for reprogramming of a new defended body weight as an important principle by which RYGB lastingly suppresses body weight. RYGB appears to selectively abolish defense of a higher fat mass level, while remaining sensitive to the defense of lean mass. The molecular and physiological mechanisms underlying this reprogramming remain to be elucidated.
Collapse
Affiliation(s)
- Zheng Hao
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Michael B Mumphrey
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - R Leigh Townsend
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Christopher D Morrison
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Heike Münzberg
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Jianping Ye
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Hans-Rudolf Berthoud
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| |
Collapse
|
110
|
Abstract
BACKGROUND Bariatric surgery is a recommended treatment for diabetes in severely obese patients. Their immediate post-operative anti-hyperglycemic requirements differ from other hospitalized diabetics, yet no standardized protocols addressing glycemic control for this group exist. OBJECTIVE We aimed to create a safe, easily implemented protocol for immediate post-operative glycemic control, which we defined as the first 30 days. METHODS The protocol was designed by an interdisciplinary workgroup using review of available literature, approved institutional glycemic guidelines, and team members' experience with caring for bariatric surgery patients. RESULTS Patients are offered post-discharge recommendations using the inpatient glycemic protocol. CONCLUSION We designed a protocol with low risk of hypoglycemia that addresses the unique glycemic needs of diabetic bariatric population in the immediate post-operative period.
Collapse
|
111
|
Xu B, Yan X, Shao Y, Shen Q, Hua R, Ding R, Yao Q. A Comparative Study of the Effect of Gastric Bypass, Sleeve Gastrectomy, and Duodenal-Jejunal Bypass on Type-2 Diabetes in non-Obese Rats. Obes Surg 2016; 25:1966-75. [PMID: 26254879 DOI: 10.1007/s11695-015-1835-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND We compared the therapeutic effects of Roux-en-Y gastric bypass (RYGB), sleeve gastrectomy (SG), and duodenal-jejunal bypass (DJB) on type-2 diabetes mellitus (T2DM) in non-obese rats using clamp testing. METHODS Goto-Kakizaki rats (non-obese rats with T2DM) underwent surgery: RYGB, SG, or DJB. Rats were observed for 8 weeks after surgery to evaluate weight changes. Levels of glucose, insulin, and glucagon-like peptide (GLP)-1 were determined 2, 4, 6, and 8 weeks after surgery. An oral glucose tolerance test (OGTT) and clamp test was used to evaluate glucose tolerance and insulin resistance. RESULTS Rats in RYGB, SG, and DJB groups weighed significantly less than sham-group rats 6 and 8 weeks after surgery. Fasting blood glucose levels of RYGB, SG, and DJB rats were significantly lower than preoperative levels. One month after surgery, the area under the curve of the OGTT (in mmol•h/L) for RYGB, SG, DJB, and sham surgery groups was 38.9 ± 5.9, 50.9 ± 2.9, 46.8 ± 3.3, and 67.4 ± 6.0, respectively; there was no significant difference in glucose levels of SG and DJB groups. Glucose infusion rates (in mg/(kg•min)) were 18.3 ± 2.7, 17.2 ± 2.1, and 16.8 ± 1.9 in hyperinsulinemic-euglycemic-clamped RYGB, DJB, and SG rats, respectively, 8 weeks after surgery. The rate in the sham surgery group was 6.3 ± 0.9. Area under plasma insulin curves 8 weeks after surgery in hyperglycemic-clamped RYGB, DJB, SG, and sham surgery rats (in mU•h/L) were 98.8 ± 7.0, 84.4 ± 6.1, 89.0 ± 7.1, and 22.6 ± 2.6, respectively. CONCLUSIONS The three surgical methods described alleviated T2DM and reduced insulin resistance in non-obese rats with T2DM.
Collapse
Affiliation(s)
- Bo Xu
- Department of General Surgery, Huashan Hospital, Fudan University, No 12, Middle Wulumuqi Road, Shanghai, 200040, China,
| | | | | | | | | | | | | |
Collapse
|
112
|
Abstract
The incretin hormones glucose-dependent insulinotropic polypeptide (GIP) and glucagon like peptide-1 (GLP-1) are secreted from enteroendocrine cells in the gut and regulate physiological and homeostatic functions related to glucose control, metabolism and food intake. This review provides a systematic summary of the molecular mechanisms underlying secretion from incretin cells, and an understanding of how they sense and interact with lumen and vascular factors and the enteric nervous system through transporters and G-protein coupled receptors (GPCRs) present on their surface to ultimately culminate in hormone release. Some of the molecules described below such as sodium coupled glucose transporter 1 (SGLT1), G-protein coupled receptor (GPR) 119 and GPR40 are targets of novel therapeutics designed to enhance endogenous gut hormone release. Synthetic ligands at these receptors aimed at treating obesity and type 2 diabetes are currently under investigation.
Collapse
Affiliation(s)
- Ramona Pais
- The Wellcome Trust–MRC Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Fiona M. Gribble
- The Wellcome Trust–MRC Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes’s Hospital, Box 289, Hills Road, Cambridge, CB2 0QQ, UK
| | | |
Collapse
|
113
|
Lee CJ, Brown TT, Cheskin LJ, Choi P, Moran TH, Peterson L, Matuk R, Steele KE. Effects of meal composition on postprandial incretin, glucose and insulin responses after surgical and medical weight loss. Obes Sci Pract 2015; 1:104-109. [PMID: 27774253 PMCID: PMC5064622 DOI: 10.1002/osp4.17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/26/2015] [Accepted: 11/03/2015] [Indexed: 12/13/2022] Open
Abstract
Background Meal tolerance tests are frequently used to study dynamic incretin and insulin responses in the postprandial state; however, the optimal meal that is best tolerated and suited for hormonal response following surgical and medical weight loss has yet to be determined. Objective To evaluate the tolerability and effectiveness of different test meals in inducing detectable changes in markers of glucose metabolism in individuals who have undergone a weight loss intervention. Methods Six individuals who underwent surgical or medical weight loss (two Roux‐en‐Y gastric bypass, two sleeve gastrectomy and two medical weight loss) each completed three meal tolerance tests using liquid‐mixed, solid‐mixed and high‐fat test meals. The tolerability of each test meal, as determined by the total amount consumed and palatability, as well as fasting and meal‐stimulated glucagon‐like peptide, glucose‐dependent insulinotropic polypeptide, insulin and glucose were measured. Results Among the six individuals, the liquid‐mixed meal was better and more uniformly tolerated with a median meal completion rate of 99%. Among the four bariatric surgical patients, liquid‐mixed meal stimulated on average a higher glucagon‐like peptide (percent difference: 83.7, 89), insulin secretion (percent difference: 155.1, 158.7) and glucose‐dependent insulinotropic polypeptide (percent difference: 113.5, 34.3) compared with solid‐mixed and high‐fat meals. Conclusions The liquid‐mixed meal was better tolerated with higher incretin and insulin response compared with the high‐fat and solid‐mixed meals and is best suited for the evaluation of stimulated glucose homeostasis.
Collapse
Affiliation(s)
- C J Lee
- Division of Endocrinology, Diabetes and Metabolism The Johns Hopkins University Baltimore Maryland USA
| | - T T Brown
- Division of Endocrinology, Diabetes and Metabolism The Johns Hopkins University Baltimore Maryland USA
| | - L J Cheskin
- Department of Health, Behavior and Society The Johns Hopkins Bloomberg School of Public Health Baltimore Maryland USA
| | - P Choi
- Department of Psychiatry The Johns Hopkins University Baltimore Maryland USA
| | - T H Moran
- Department of Psychiatry The Johns Hopkins University Baltimore Maryland USA
| | - L Peterson
- Department of Surgery The Johns Hopkins University Baltimore Maryland USA
| | - R Matuk
- Department of Surgery The Johns Hopkins University Baltimore Maryland USA
| | - K E Steele
- Department of Surgery The Johns Hopkins University Baltimore Maryland USA
| |
Collapse
|
114
|
Distal small bowel bypass for weight regain after gastric bypass: safety and efficacy threshold occurs at<70% bypass. Surg Obes Relat Dis 2015; 11:1248-55. [DOI: 10.1016/j.soard.2015.08.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 07/26/2015] [Accepted: 08/03/2015] [Indexed: 11/21/2022]
|
115
|
Malik S, Mitchell JE, Steffen K, Engel S, Wiisanen R, Garcia L, Malik SA. Recognition and management of hyperinsulinemic hypoglycemia after bariatric surgery. Obes Res Clin Pract 2015; 10:1-14. [PMID: 26522879 DOI: 10.1016/j.orcp.2015.07.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 06/17/2015] [Accepted: 07/06/2015] [Indexed: 02/06/2023]
Abstract
Hyperinsulinemic hypoglycemia with neuroglycopenia is an increasingly recognized complication of Roux-en-Y gastric bypass (RYGB) due to the changes in gut hormonal milieu. Physicians should be aware of this complication to ensure timely and effective treatment of post-RYGB patients, who present to them with hypoglycemic symptoms. Possible causes of hypoglycemia in these patients include late dumping syndrome, nesidioblastosis and rarely insulinoma. Systematic evaluation including history, biochemical analysis, and diagnostic testing might help in distinguishing among these diagnoses. Continuous glucose monitoring is also a valuable tool, revealing the episodes in the natural environment and can also be used to monitor treatment success. Treatment should begin with strict low carbohydrate diet, followed by medication therapy. Therapy with diazoxide, acarbose, calcium channel blockers and octreotide have been proven to be beneficial, but the response apparently is highly variable. When other treatment options fail, surgical options can be considered.
Collapse
Affiliation(s)
- Sarah Malik
- Neuropsychiatric Research Institute, Fargo, ND, United States; University of North Dakota School of Medicine and Health Sciences, Fargo, ND, United States
| | - James E Mitchell
- Neuropsychiatric Research Institute, Fargo, ND, United States; University of North Dakota School of Medicine and Health Sciences, Fargo, ND, United States.
| | - Kristine Steffen
- Neuropsychiatric Research Institute, Fargo, ND, United States; North Dakota State University, United States
| | - Scott Engel
- Neuropsychiatric Research Institute, Fargo, ND, United States; University of North Dakota School of Medicine and Health Sciences, Fargo, ND, United States
| | | | | | - Shahbaz Ali Malik
- University of North Dakota School of Medicine and Health Sciences, Fargo, ND, United States
| |
Collapse
|
116
|
Williams KE, Washington MC, Johnson-Rouse T, Johnson RE, Freeman C, Reed C, Heath J, Sayegh AI. Exogenous glucagon-like peptide-1 acts in sites supplied by the cranial mesenteric artery to reduce meal size and prolong the intermeal interval in rats. Appetite 2015; 96:254-259. [PMID: 26431682 DOI: 10.1016/j.appet.2015.09.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 09/24/2015] [Accepted: 09/25/2015] [Indexed: 10/23/2022]
Abstract
Three experiments were done to better assess the gastrointestinal (GI) site(s) of action of GLP-1 on food intake in rats. First, near-spontaneous nocturnal chow meal size (MS), intermeal intervals (IMI) length and satiety ratios (SR = MS/IMI) were measured after infusion of saline, 0.025 or 0.5 nmol/kg GLP-1 into the celiac artery (CA, supplying the stomach and upper duodenum), cranial mesenteric artery (CMA, supplying small and all of the large intestine except the rectum), femoral artery (FA, control) or portal vein (PV, control). Second, infusion of 0.5 nmol/kg GLP-1 was tested after pretreatment with the GLP-1 receptor (GLP-1R) antagonist exendin-4(3-39) via the same routes. Third, the regional distribution of GLP-1R in the rat GI tract was determined using rtPCR. CA, CMA and FA GLP-1 reduced first MS relative to saline, with the CMA route more effective than the others. Only CMA GLP-1 prolonged the IMI. None of the infusions affected second MS or later eating. CA and CMA GLP-1 increased the SR, with the CMA route more effective than the CA route. CMA exendin-4 (3-39) infusion reduced the effect of CMA GLP-1. Finally GLP-1R expression was found throughout the GI tract. The results suggest that exogenous GLP-1 acts in multiple GI sites to reduce feeding under our conditions and that GLP-1R in the area supplied by the CMA, i.e., the small and part of the large intestine, plays the leading role.
Collapse
Affiliation(s)
- Kasey E Williams
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA
| | - Martha C Washington
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA
| | - Tanisha Johnson-Rouse
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA
| | - Ruth E Johnson
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA
| | - Corren Freeman
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA
| | - Chris Reed
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA
| | - John Heath
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA
| | - Ayman I Sayegh
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA.
| |
Collapse
|
117
|
Christensen LW, Kuhre RE, Janus C, Svendsen B, Holst JJ. Vascular, but not luminal, activation of FFAR1 (GPR40) stimulates GLP-1 secretion from isolated perfused rat small intestine. Physiol Rep 2015; 3:e12551. [PMID: 26381015 PMCID: PMC4600392 DOI: 10.14814/phy2.12551] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 08/25/2015] [Accepted: 08/29/2015] [Indexed: 12/23/2022] Open
Abstract
Glucagon-like peptide 1 (GLP-1) plays a central role in modern treatment of type 2 diabetes (T2DM) in the form of GLP-1 enhancers and GLP-1 mimetics. An alternative treatment strategy is to stimulate endogenous GLP-1 secretion from enteroendocrine L cells using a targeted approach. The G-protein-coupled receptor, FFAR1 (previously GPR40), expressed on L cells and activated by long-chain fatty acids (LCFAs) is a potential target. A link between FFAR1 activation and GLP-1 secretion has been demonstrated in cellular models and small-molecule FFAR1 agonists have been developed. In this study, we examined the effect of FFAR1 activation on GLP-1 secretion using isolated, perfused small intestines from rats, a physiologically relevant model allowing distinction between direct and indirect effects of FFAR1 activation. The endogenous FFAR1 ligand, linoleic acid (LA), and four synthetic FFAR1 agonists (TAK-875, AMG 837, AM-1638, and AM-5262) were administered through intraluminal and intra-arterial routes, respectively, and dynamic changes in GLP-1 secretion were evaluated. Vascular administration of 10 μmol/L TAK-875, 10 μmol/L AMG 837, 1 μmol/L and 0.1 μmol/L AM-1638, 1 μmol/L AM-6252, and 1 mmol/L LA, all significantly increased GLP-1 secretion compared to basal levels (P < 0.05), whereas luminal administration of LA and FFAR1 agonists was ineffective. Thus, both natural and small-molecule agonists of the FFAR1 receptor appear to require absorption prior to stimulating GLP-1 secretion, indicating that therapies based on activation of nutrient sensing may be more complex than hitherto expected.
Collapse
Affiliation(s)
- Louise W Christensen
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Panum Institute University of Copenhagen, Copenhagen, Denmark
| | - Rune E Kuhre
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Panum Institute University of Copenhagen, Copenhagen, Denmark
| | - Charlotte Janus
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Panum Institute University of Copenhagen, Copenhagen, Denmark
| | - Berit Svendsen
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Panum Institute University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Panum Institute University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
118
|
Nguyen KT, Billington CJ, Vella A, Wang Q, Ahmed L, Bantle JP, Bessler M, Connett JE, Inabnet WB, Thomas A, Ikramuddin S, Korner J. Preserved Insulin Secretory Capacity and Weight Loss Are the Predominant Predictors of Glycemic Control in Patients With Type 2 Diabetes Randomized to Roux-en-Y Gastric Bypass. Diabetes 2015; 64:3104-10. [PMID: 25901097 PMCID: PMC4542441 DOI: 10.2337/db14-1870] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/16/2015] [Indexed: 12/12/2022]
Abstract
Improvement in type 2 diabetes after Roux-en-Y gastric bypass (RYGB) has been attributed partly to weight loss, but mechanisms beyond weight loss remain unclear. We performed an ancillary study to the Diabetes Surgery Study to assess changes in incretins, insulin sensitivity, and secretion 1 year after randomization to lifestyle modification and intensive medical management (LS/IMM) alone (n = 34) or in conjunction with RYGB (n = 34). The RYGB group lost more weight and had greater improvement in HbA1c. Fasting glucose was lower after RYGB than after LS/IMM, although the glucose area under the curve decreased comparably for both groups. Insulin sensitivity increased in both groups. Insulin secretion was unchanged after LS/IMM but decreased after RYGB, except for a rapid increase during the first 30 min after meal ingestion. Glucagon-like peptide 1 (GLP-1) was substantially increased after RYGB, while gastric inhibitory polypeptide and glucagon decreased. Lower HbA1c was most strongly correlated with the percentage of weight loss for both groups. At baseline, a greater C-peptide index and 90-min postprandial C-peptide level were predictive of lower HbA1c at 1 year after RYGB. β-Cell glucose sensitivity, which improved only after RYGB, and improved disposition index were associated with lower HbA1c in both groups, independent of weight loss. Weight loss and preserved β-cell function both predominantly determine the greatest glycemic benefit after RYGB.
Collapse
Affiliation(s)
- Kim T Nguyen
- Division of Endocrinology, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Charles J Billington
- Division of Endocrinology and Diabetes, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Adrian Vella
- Division of Endocrinology and Diabetes, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Qi Wang
- Division of Biostatistics, University of Minnesota, Minneapolis, MN
| | - Leaque Ahmed
- Department of Surgery, Columbia University Medical Center, New York, NY
| | - John P Bantle
- Division of Endocrinology and Diabetes, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Marc Bessler
- Department of Surgery, Columbia University Medical Center, New York, NY
| | - John E Connett
- Division of Biostatistics, University of Minnesota, Minneapolis, MN
| | | | - Avis Thomas
- Division of Biostatistics, University of Minnesota, Minneapolis, MN
| | | | - Judith Korner
- Division of Endocrinology, Department of Medicine, Columbia University Medical Center, New York, NY
| |
Collapse
|
119
|
Zhou J, Hao Z, Irwin N, Berthoud HR, Ye J. Gastric inhibitory polypeptide (GIP) is selectively decreased in the roux-limb of dietary obese mice after RYGB surgery. PLoS One 2015; 10:e0134728. [PMID: 26266950 PMCID: PMC4534413 DOI: 10.1371/journal.pone.0134728] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 07/13/2015] [Indexed: 01/27/2023] Open
Abstract
Gastric inhibitory polypeptide (GIP, glucose-dependent insulinotropic polypeptide) is expressed by intestinal K cells to regulate glucose-induced insulin secretion. The impact of Roux-en Y bypass (RYGB) surgery on blood GIP is highly contraversial. This study was conducted to address the mechanism of controversy. GIP mRNA was examined in the intestine, and serum GIP was determined using Luminex and ELISA in diet-induced obese (DIO) mice. The assays were conducted in RYGB mice in fasting and fed conditions. Food preference, weight loss and insulin sensitivity were monitored in RYGB mice. In DIO mice, GIP mRNA was increased by 80% in all sections of the small intestine over the lean control. The increase was observed in both fasting and fed conditions. After RYGB surgery, the food-induced GIP expression was selectively reduced in the Roux-limb, but not in the biliopancreatic and common limbs of intestine in fed condition. Lack of stimulation by glucose or cholesterol contributed to the reduction. Jejunal mucosa of Roux-limb exhibited hypertrophy, but villous surface was decreased by the undigested food. Serum GIP (total) was significantly higher in the fasting condition, but not in the fed condition due to attenuated GIP response to food intake in RYGB mice. The GIP alteration was associated with chow diet preference, sustained weight loss and insulin sensitization in RYGB mice. RYGB increased serum GIP in the fasting, but not in the fed conditions. The loss of food-induced GIP response in Roux-limb of intestine likely contributes to the attenuated serum GIP response to feeding.
Collapse
Affiliation(s)
- Jiaqiang Zhou
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Zheng Hao
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Nigel Irwin
- School of Biomedical Sciences, University of Ulster, Coleraine, United Kingdom
| | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Jianping Ye
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| |
Collapse
|
120
|
Gögebakan Ö, Osterhoff MA, Schüler R, Pivovarova O, Kruse M, Seltmann AC, Mosig AS, Rudovich N, Nauck M, Pfeiffer AFH. GIP increases adipose tissue expression and blood levels of MCP-1 in humans and links high energy diets to inflammation: a randomised trial. Diabetologia 2015; 58:1759-68. [PMID: 25994074 DOI: 10.1007/s00125-015-3618-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 04/15/2015] [Indexed: 12/25/2022]
Abstract
AIMS/HYPOTHESIS Obesity is associated with elevated monocyte chemoattractant protein-1 (MCP-1), a proinflammatory chemokine related to diabetes and cardiovascular disease. Since obesity is triggered by energy dense diets, we hypothesised that nutrient induced intestinal hormones such as glucose-dependent insulinotropic peptide (GIP) may directly stimulate the release of chemokines from adipose tissue and induce low-grade inflammation. METHODS GIP effects on gene expression and secretion of inflammatory markers were studied by microarray analysis and PCR from human subcutaneous fat biopsies of slightly obese but healthy volunteers in the metabolic ward of German Institute of Human Nutrition, Department of Clinical Nutrition, Potsdam-Rehbrücke. To allocate the participants to the study arms they were numbered in order of their recruitment and then assigned to the groups by a random number generator. In a randomised, single-blind (participants) crossover design, the participants received GIP infusions in postprandial concentrations (2 pmol kg(-1) min(-1)) or saline (154 mmol/l NaCl) infusions for 240 min either alone, in combination with hyperinsulinaemic-euglycaemic (EU) or hyperinsulinaemic-hyperglycaemic (HC) clamps. Possible mechanisms of GIP effects were investigated in single and co-cultures of macrophage and adipocyte cell lines and in primary human monocytes, macrophages and adipocytes. RESULTS A total of 17 participants were randomised to the following groups: EU with GIP infusion (n = 9); EU with NaCl infusion (n = 9); HC with GIP infusion (n = 8); HC with NaCl infusion (n = 8); sole GIP infusion (n = 11) and sole placebo infusion (n = 11). All 17 individuals were analysed. The study is completed. In human subcutaneous adipose tissue (hSCAT), infusions of GIP significantly increased inflammatory chemokine and cytokine gene networks in transcriptomic microarray analyses. Particularly MCP-1 (180 ± 26%), MCP-2 (246 ± 58%) and IL-6 (234 ± 40%) mRNA levels in adipose tissue as well as circulating plasma concentrations of MCP-1 (165 ± 12 vs 135 ± 13 pg/ml; GIP vs saline after 240 min; p < 0.05 for all variables) in humans increased independently of circulating insulin or glucose plasma concentrations. GIP stimulation increased Mcp-1 mRNA-expression in co-cultures of differentiated 3T3L1-adipocytes and RAW 264.7 macrophages but not in the isolated cell lines. Similarly, GIP increased MCP-1 transcripts in co-cultures of primary human macrophages with human adipocytes. GIP receptor (GIPR) transcripts were present in primary monocytes and the different cell lines and induced activation of extracellular related kinase (ERK) as well as increases in cAMP, indicating functional receptors. CONCLUSIONS/INTERPRETATION Our findings suggest that the nutrient induced gut hormone GIP may initiate adipose tissue inflammation by triggering a crosstalk of adipocytes and macrophages involving MCP-1. TRIAL REGISTRATION ClinicalTrials.gov NCT00774488. FUNDING This work was supported by the German Research Foundation (DFG): grant No. Pf164/021002.
Collapse
Affiliation(s)
- Özlem Gögebakan
- Department of Clinical Nutrition, German Institute of Human Nutrition, Potsdam-Rehbruecke, Arthur-Scheunert-Allee 155, 14558, Nuthetal, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Sandoval DA, D'Alessio DA. Physiology of proglucagon peptides: role of glucagon and GLP-1 in health and disease. Physiol Rev 2015; 95:513-48. [PMID: 25834231 DOI: 10.1152/physrev.00013.2014] [Citation(s) in RCA: 310] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The preproglucagon gene (Gcg) is expressed by specific enteroendocrine cells (L-cells) of the intestinal mucosa, pancreatic islet α-cells, and a discrete set of neurons within the nucleus of the solitary tract. Gcg encodes multiple peptides including glucagon, glucagon-like peptide-1, glucagon-like peptide-2, oxyntomodulin, and glicentin. Of these, glucagon and GLP-1 have received the most attention because of important roles in glucose metabolism, involvement in diabetes and other disorders, and application to therapeutics. The generally accepted model is that GLP-1 improves glucose homeostasis indirectly via stimulation of nutrient-induced insulin release and by reducing glucagon secretion. Yet the body of literature surrounding GLP-1 physiology reveals an incompletely understood and complex system that includes peripheral and central GLP-1 actions to regulate energy and glucose homeostasis. On the other hand, glucagon is established principally as a counterregulatory hormone, increasing in response to physiological challenges that threaten adequate blood glucose levels and driving glucose production to restore euglycemia. However, there also exists a potential role for glucagon in regulating energy expenditure that has recently been suggested in pharmacological studies. It is also becoming apparent that there is cross-talk between the proglucagon derived-peptides, e.g., GLP-1 inhibits glucagon secretion, and some additive or synergistic pharmacological interaction between GLP-1 and glucagon, e.g., dual glucagon/GLP-1 agonists cause more weight loss than single agonists. In this review, we discuss the physiological functions of both glucagon and GLP-1 by comparing and contrasting how these peptides function, variably in concert and opposition, to regulate glucose and energy homeostasis.
Collapse
Affiliation(s)
- Darleen A Sandoval
- Division of Endocrinology and Metabolism, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David A D'Alessio
- Division of Endocrinology and Metabolism, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
122
|
Abstract
Bariatric surgery is arguably the most effective therapy for weight loss, and Rouen-Y gastric bypass (RYGB) is considered the "gold-standard" procedure. However, sleeve gastrectomy (SG) surgery has become more prevalent in recent years and it is unclear if weight loss differences occur between these procedures. Herein, we discuss evidence from randomized clinical trials comparing the effectiveness of RYGB and SG on weight loss. Moreover, we highlight gut hormones (e.g., GLP-1, ghrelin, bile acids, etc.) as potentially important mechanisms that contribute to the durability of decreased appetite and opposed fat storage following RYGB and SG. Collectively, although a subtle (∼ 3-5 kg) weight loss difference may exist in favor of RYGB up to 3 years post-operation, it appears that RYGB and SG induce comparable weight loss and changes in gut physiology that parallel reduced disease risk. These findings are clinically relevant for optimizing treatment strategies that combat obesity-related diabetes and cardiovascular disease.
Collapse
Affiliation(s)
- Steven K Malin
- Department of Kinesiology, University of Virginia, 210 Memorial Gymnasium, Charlottesville, VA, USA.
- Division of Endocrinology and Metabolism, University of Virginia, 210 Memorial Gymnasium, Charlottesville, VA, USA.
| | - Sangeeta R Kashyap
- Department of Endocrinology, Diabetes, and Metabolism, Cleveland Clinic, 9500 Euclid Ave (NE40), Cleveland, OH, 44195, USA.
| |
Collapse
|
123
|
Grams J, Garvey WT. Weight Loss and the Prevention and Treatment of Type 2 Diabetes Using Lifestyle Therapy, Pharmacotherapy, and Bariatric Surgery: Mechanisms of Action. Curr Obes Rep 2015; 4:287-302. [PMID: 26627223 DOI: 10.1007/s13679-015-0155-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Weight loss, whether achieved by lifestyle intervention, pharmacotherapy, or bariatric surgery, is highly effective as a primary interventional strategy in both the prevention and treatment of type 2 diabetes. In high-risk patients with prediabetes and/or metabolic syndrome, weight loss effectively prevents progression to type 2 diabetes mellitus (T2DM) and improves cardiovascular risk factors. These benefits are the result of improvements in insulin resistance, which is central to the pathophysiology of cardiometabolic disease. In patients with T2DM, weight loss improves glycemia, while reducing the need for conventional glucose-lowering medicines, by affecting all three processes that produce and sustain the hyperglycemic state, namely via increments in peripheral insulin sensitivity with improvements in insulin signal transduction at the cellular level, more robust insulin secretory responses, and reduced rates of hepatic glucose production. In both nondiabetic and diabetic subjects, hypocaloric feeding (e.g., treatment with very low-calorie diet or bariatric surgery) produces a rapid improvement in insulin sensitivity due to mobilization of fat from the intramyocellular, intrahepatocellular, and intra-abdominal compartments, and via a more long-term mechanism that correlates with the loss of total body fat. In diabetes, by improving glycemia, weight loss also enhances glucose homeostasis by reversing the defects in insulin action and secretion attributable to glucose toxicity. Regardless of the therapeutic approach, weight loss of ∼ 10 % maximally prevents future diabetes in patients with prediabetes or metabolic syndrome. In T2DM, greater degrees of weight loss lead to progressive improvements in glucose homeostasis. Therefore, when accompanied by greater weight loss, the metabolic benefits following bariatric surgery are generally more pronounced than those achieved following lifestyle and medical treatment. In addition, the mechanisms by which bariatric operations improve diabetes may include both weight-dependent and weight-independent mechanisms, and the latter may involve changes in gut hormones, bile acids, or gut microflora.
Collapse
Affiliation(s)
- J Grams
- Department of Surgery, University of Alabama at Birmingham and the Birmingham VA Medical Center, KB401, 1720 2nd Ave S, Birmingham, AL, 35294-0016, USA.
| | - W Timothy Garvey
- Department of Nutrition Sciences, University of Alabama at Birmingham and the Birmingham VA Medical Center, 1675 University Boulevard, Birmingham, AL, 35294-3360, USA.
| |
Collapse
|
124
|
Kang CM, Lee JH. Pathophysiology after pancreaticoduodenectomy. World J Gastroenterol 2015; 21:5794-5804. [PMID: 26019443 PMCID: PMC4438013 DOI: 10.3748/wjg.v21.i19.5794] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 02/25/2015] [Accepted: 04/17/2015] [Indexed: 02/06/2023] Open
Abstract
Pancreaticoduodenectomy (PD) will result in removal of important multiorgans in upper intestinal tract and subsequently secondary physiologic change. In the past, surgeons just focused on the safety of surgical procedure; however, PD is regarded as safe and widely applied to treatment of periampullary lesions. Practical issues after PD, such as, effect of duodenectomy, metabolic surgery-like effect, alignment effect of gastrointestinal continuity, and non-alcoholic fatty liver disease were summarized and discussed.
Collapse
|
125
|
Lee CJ, Clark JM, Schweitzer M, Magnuson T, Steele K, Koerner O, Brown TT. Prevalence of and risk factors for hypoglycemic symptoms after gastric bypass and sleeve gastrectomy. Obesity (Silver Spring) 2015; 23:1079-84. [PMID: 25866150 PMCID: PMC4414701 DOI: 10.1002/oby.21042] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/06/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To determine the prevalence of and risk factors for postprandial hypoglycemic symptoms among bariatric surgery patients. METHODS A questionnaire including the Edinburgh hypoglycemia scale was mailed to patients who underwent either Roux-en-Y gastric bypass (RYGB) or vertical sleeve gastrectomy (VSG) at a single center. Based on the questionnaire, the patients were categorized as having high or low suspicion for post surgical, postprandial hypoglycemic symptoms. RESULTS Of the 1119 patients with valid addresses, 40.2% (N = 450) responded. Among the respondents, 34.2% had a high suspicion for symptoms of post bariatric surgery hypoglycemia. In multivariate analyses, in addition to female sex (P = 0.001), RYGB (P = 0.004), longer time since surgery (P = 0.013), and lack of diabetes (P = 0.040), the high suspicion group was more likely to report pre-operative symptoms of hypoglycemia (P < 0.001), compared to the low suspicion group. Similar results were observed when the high suspicion group was restricted to those requiring assistance from others, syncope, seizure with severe symptoms, or medically confirmed hypoglycemia (N = 52). CONCLUSIONS One third of patients who underwent RYGB or VSG reported postprandial symptoms concerning for postsurgical hypoglycemia, which was related to the presence of pre-operative hypoglycemic symptoms. Pre-operative screening for hypoglycemic symptoms may identify a group of patients at increased risk of postbariatric surgery hypoglycemia.
Collapse
Affiliation(s)
- Clare J. Lee
- Division of Endocrinology, Diabetes and Metabolism, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Jeanne M. Clark
- Department of Medicine, Division of General Internal Medicine, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Michael Schweitzer
- Department of Surgery, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Thomas Magnuson
- Department of Surgery, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Kimberley Steele
- Department of Surgery, The Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Todd T. Brown
- Division of Endocrinology, Diabetes and Metabolism, The Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
126
|
Camilleri M. Peripheral mechanisms in appetite regulation. Gastroenterology 2015; 148:1219-33. [PMID: 25241326 PMCID: PMC4369188 DOI: 10.1053/j.gastro.2014.09.016] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/09/2014] [Accepted: 09/15/2014] [Indexed: 12/13/2022]
Abstract
Peripheral mechanisms in appetite regulation include the motor functions of the stomach, such as the rate of emptying and accommodation, which convey symptoms of satiation to the brain. The rich repertoire of peripherally released peptides and hormones provides feedback from the arrival of nutrients in different regions of the gut from where they are released to exert effects on satiation, or regulate metabolism through their incretin effects. Ultimately, these peripheral factors provide input to the highly organized hypothalamic circuitry and vagal complex of nuclei to determine cessation of energy intake during meal ingestion, and the return of appetite and hunger after fasting. Understanding these mechanisms is key to the physiological control of feeding and the derangements that occur in obesity and their restoration with treatment (as shown by the effects of bariatric surgery).
Collapse
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research, Mayo Clinic College of Medicine, Rochester, Minnesota.
| |
Collapse
|
127
|
Abstract
The gastrointestinal (GI) tract comprises a large endocrine organ that regulates not only nutrient sensing and metabolising but also satiety and energy homeostasis. More than 20 hormones secreted from the stomach, intestine, and pancreas as well as signaling mediators of the gut microbiome are involved in this process. A better understanding of how related pathways affect body weight and food intake will help us to find new strategies and drugs to treat obesity. For example, weight loss secondary to lifestyle intervention is often accompanied by unfavorable changes in multiple GI hormones, which may cause difficulties in maintaining a lower body weight status. Conversely, bariatric surgery favorably changes the hormone profile to support improved satiety and metabolic function. This partially explains stronger sustained body weight reduction resulting in better long-term results of improved metabolic functions. This review focuses on GI hormones and signaling mediators of the microbiome involved in satiety regulation and energy homeostasis and summarizes their changes following weight loss. Furthermore, the potential role of GI hormones as anti-obesity drugs is discussed.
Collapse
Affiliation(s)
- Thomas Reinehr
- Vestische Hospital for Children and Adolescents Datteln, Institute for Pediatric Endocrinology, Diabetes and Nutrition Medicine, University of Witten/Herdecke, Datteln, Germany,
| | | |
Collapse
|
128
|
Chen MZ, Hudson CA, Vincent EE, de Berker DAR, May MT, Hers I, Dayan CM, Andrews RC, Tavaré JM. Bariatric surgery in morbidly obese insulin resistant humans normalises insulin signalling but not insulin-stimulated glucose disposal. PLoS One 2015; 10:e0120084. [PMID: 25876175 PMCID: PMC4395354 DOI: 10.1371/journal.pone.0120084] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 01/19/2015] [Indexed: 01/14/2023] Open
Abstract
Aims Weight-loss after bariatric surgery improves insulin sensitivity, but the underlying molecular mechanism is not clear. To ascertain the effect of bariatric surgery on insulin signalling, we examined glucose disposal and Akt activation in morbidly obese volunteers before and after Roux-en-Y gastric bypass surgery (RYGB), and compared this to lean volunteers. Materials and Methods The hyperinsulinaemic euglycaemic clamp, at five infusion rates, was used to determine glucose disposal rates (GDR) in eight morbidly obese (body mass index, BMI=47.3±2.2 kg/m2) patients, before and after RYGB, and in eight lean volunteers (BMI=20.7±0.7 kg/m2). Biopsies of brachioradialis muscle, taken at fasting and insulin concentrations that induced half-maximal (GDR50) and maximal (GDR100) GDR in each subject, were used to examine the phosphorylation of Akt-Thr308, Akt-473, and pras40, in vivo biomarkers for Akt activity. Results Pre-operatively, insulin-stimulated GDR was lower in the obese compared to the lean individuals (P<0.001). Weight-loss of 29.9±4 kg after surgery significantly improved GDR50 (P=0.004) but not GDR100 (P=0.3). These subjects still remained significantly more insulin resistant than the lean individuals (p<0.001). Weight loss increased insulin-stimulated skeletal muscle Akt-Thr308 and Akt-Ser473 phosphorylation, P=0.02 and P=0.03 respectively (MANCOVA), and Akt activity towards the substrate PRAS40 (P=0.003, MANCOVA), and in contrast to GDR, were fully normalised after the surgery (obese vs lean, P=0.6, P=0.35, P=0.46, respectively). Conclusions Our data show that although Akt activity substantially improved after surgery, it did not lead to a full restoration of insulin-stimulated glucose disposal. This suggests that a major defect downstream of, or parallel to, Akt signalling remains after significant weight-loss.
Collapse
Affiliation(s)
- Mimi Z. Chen
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Claire A. Hudson
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Emma E. Vincent
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | | | - Margaret T. May
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Ingeborg Hers
- School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom
| | - Colin M. Dayan
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Robert C. Andrews
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
- * E-mail:
| | - Jeremy M. Tavaré
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
129
|
Scavini M, Dugnani E, Pasquale V, Liberati D, Aleotti F, Di Terlizzi G, Petrella G, Balzano G, Piemonti L. Diabetes after pancreatic surgery: novel issues. Curr Diab Rep 2015; 15:16. [PMID: 25702096 DOI: 10.1007/s11892-015-0589-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In the developed world, pancreatic surgery is becoming more common, with an increasing number of patients developing diabetes because of either partial or total pancreatectomy, with a significant impact on quality of life and survival. Although these patients are expected to consume increasing health care resources in the near future, many aspects of diabetes after pancreatectomy are still not well defined. The treatment of diabetes in these patients takes advantage of the therapies used in type 1 and 2 diabetes; however, no specific guidelines for its management, both immediately after pancreatic surgery or in the long term, have been developed. In this article, on the basis of both the literature and our clinical experience, we address the open issues and discuss the most appropriate therapeutic options for patients with diabetes after pancreatectomy.
Collapse
Affiliation(s)
- Marina Scavini
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Yu YH, Vasselli JR, Zhang Y, Mechanick JI, Korner J, Peterli R. Metabolic vs. hedonic obesity: a conceptual distinction and its clinical implications. Obes Rev 2015; 16:234-47. [PMID: 25588316 PMCID: PMC5053237 DOI: 10.1111/obr.12246] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 11/19/2014] [Accepted: 11/19/2014] [Indexed: 01/01/2023]
Abstract
Body weight is determined via both metabolic and hedonic mechanisms. Metabolic regulation of body weight centres around the 'body weight set point', which is programmed by energy balance circuitry in the hypothalamus and other specific brain regions. The metabolic body weight set point has a genetic basis, but exposure to an obesogenic environment may elicit allostatic responses and upward drift of the set point, leading to a higher maintained body weight. However, an elevated steady-state body weight may also be achieved without an alteration of the metabolic set point, via sustained hedonic over-eating, which is governed by the reward system of the brain and can override homeostatic metabolic signals. While hedonic signals are potent influences in determining food intake, metabolic regulation involves the active control of both food intake and energy expenditure. When overweight is due to elevation of the metabolic set point ('metabolic obesity'), energy expenditure theoretically falls onto the standard energy-mass regression line. In contrast, when a steady-state weight is above the metabolic set point due to hedonic over-eating ('hedonic obesity'), a persistent compensatory increase in energy expenditure per unit metabolic mass may be demonstrable. Recognition of the two types of obesity may lead to more effective treatment and prevention of obesity.
Collapse
Affiliation(s)
- Y-H Yu
- Weight Loss and Diabetes Center, Greenwich Hospital, Greenwich, CT, USA; Endocrinology Associates of Greenwich, Northeast Medical Group, Yale New-Haven Health System, Greenwich, CT, USA
| | | | | | | | | | | |
Collapse
|
131
|
Plant-rich mixed meals based on Palaeolithic diet principles have a dramatic impact on incretin, peptide YY and satiety response, but show little effect on glucose and insulin homeostasis: an acute-effects randomised study. Br J Nutr 2015; 113:574-84. [PMID: 25661189 DOI: 10.1017/s0007114514004012] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
There is evidence for health benefits from 'Palaeolithic' diets; however, there are a few data on the acute effects of rationally designed Palaeolithic-type meals. In the present study, we used Palaeolithic diet principles to construct meals comprising readily available ingredients: fish and a variety of plants, selected to be rich in fibre and phyto-nutrients. We investigated the acute effects of two Palaeolithic-type meals (PAL 1 and PAL 2) and a reference meal based on WHO guidelines (REF), on blood glucose control, gut hormone responses and appetite regulation. Using a randomised cross-over trial design, healthy subjects were given three meals on separate occasions. PAL2 and REF were matched for energy, protein, fat and carbohydrates; PAL1 contained more protein and energy. Plasma glucose, insulin, glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic peptide (GIP) and peptide YY (PYY) concentrations were measured over a period of 180 min. Satiation was assessed using electronic visual analogue scale (EVAS) scores. GLP-1 and PYY concentrations were significantly increased across 180 min for both PAL1 (P= 0·001 and P< 0·001) and PAL2 (P= 0·011 and P= 0·003) compared with the REF. Concomitant EVAS scores showed increased satiety. By contrast, GIP concentration was significantly suppressed. Positive incremental AUC over 120 min for glucose and insulin did not differ between the meals. Consumption of meals based on Palaeolithic diet principles resulted in significant increases in incretin and anorectic gut hormones and increased perceived satiety. Surprisingly, this was independent of the energy or protein content of the meal and therefore suggests potential benefits for reduced risk of obesity.
Collapse
|
132
|
Cătoi AF, Pârvu A, Mureşan A, Busetto L. Metabolic Mechanisms in Obesity and Type 2 Diabetes: Insights from Bariatric/Metabolic Surgery. Obes Facts 2015; 8:350-63. [PMID: 26584027 PMCID: PMC5644813 DOI: 10.1159/000441259] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/17/2015] [Indexed: 12/11/2022] Open
Abstract
Obesity and the related diabetes epidemics represent a real concern worldwide. Bariatric/metabolic surgery emerged in last years as a valuable therapeutic option for obesity and related diseases, including type 2 diabetes mellitus (T2DM). The complicated network of mechanisms involved in obesity and T2DM have not completely defined yet. There is still a debate on which would be the first metabolic defect leading to metabolic deterioration: insulin resistance or hyperinsulinemia? Insight into the metabolic effects of bariatric/metabolic surgery has revealed that, beyond weight loss and food restriction, other mechanisms can be activated by the rearrangements of the gastrointestinal tract, such as the incretinic/anti-incretinic system, changes in bile acid composition and flow, and modifications of gut microbiota; all of them possibly involved in the remission of T2DM. The complete elucidation of these mechanisms will lead to a better understanding of the pathogenesis of this disease. Our aim was to review some of the metabolic mechanisms involved in the development of T2DM in obese patients as well as in the remission of this condition in patients submitted to bariatric/metabolic surgery.
Collapse
Affiliation(s)
- Adriana Florinela Cătoi
- Department of Functional Biosciences, Faculty of Medicine, ‘Iuliu Haţieganu’ University of Medicine and Pharmacy, Cluj-Napoca, Romania, Italy
| | - Alina Pârvu
- Department of Functional Biosciences, Faculty of Medicine, ‘Iuliu Haţieganu’ University of Medicine and Pharmacy, Cluj-Napoca, Romania, Italy
| | - Adriana Mureşan
- Department of Functional Biosciences, Faculty of Medicine, ‘Iuliu Haţieganu’ University of Medicine and Pharmacy, Cluj-Napoca, Romania, Italy
| | - Luca Busetto
- Department of Medicine, University of Padova, Padova, Italy
- *Dr. Luca Busetto, Clinica Medica 3, Policlinico Universitario, Via Giustiniani 2, 30100 Padova, Italy
| |
Collapse
|
133
|
Malin SK, Bena J, Abood B, Pothier CE, Bhatt DL, Nissen S, Brethauer SA, Schauer PR, Kirwan JP, Kashyap SR. Attenuated improvements in adiponectin and fat loss characterize type 2 diabetes non-remission status after bariatric surgery. Diabetes Obes Metab 2014; 16:1230-8. [PMID: 25132119 PMCID: PMC4227926 DOI: 10.1111/dom.12376] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 07/02/2014] [Accepted: 08/04/2014] [Indexed: 12/15/2022]
Abstract
AIM To identify the metabolic determinants of type 2 diabetes non-remission status after bariatric surgery at 12 and 24 months. METHODS A total of 40 adults [mean ± sd body mass index 36 ± 3 kg/m(2) , age 48 ± 9 years, glycated haemoglobin (HbA1c) 9.7 ± 2%) undergoing bariatric surgery [Roux-en-Y gastric bypass (RYGB) or sleeve gastrectomy (SG)] were enrolled in the present study, the Surgical Treatment and Medication Potentially Eradicate Diabetes Efficiently (STAMPEDE) trial. Type 2 diabetes remission was defined as HbA1c <6.5% and fasting glucose <126 mg/dl (i.e. <7 mmol/l) without antidiabetic medication. Indices of insulin secretion and sensitivity were calculated from plasma glucose, insulin and C-peptide values during a 120-min mixed-meal tolerance test. Body fat, incretins (glucagon-like polypeptide-1, gastric inhibitory peptide, ghrelin) and adipokines [adiponectin, leptin, tumour necrosis factor-α, high-sensitivity C-reactive protein (hs-CRP)] were also assessed. RESULTS At 24 months, 37 patients had available follow-up data (RYGB, n = 18; SG, n = 19). Bariatric surgery induced type 2 diabetes remission rates of 40 and 27% at 12 and 24 months, respectively. Total fat/abdominal fat loss, insulin secretion, insulin sensitivity and β-cell function (C-peptide0-120 /glucose0-120 × Matsuda index) improved more in those with remission at 12 and 24 months than in those without remission. Incretin levels were unrelated to type 2 diabetes remission, but, compared with those without remission, hs-CRP decreased and adiponectin increased more in those with remission. Only baseline adiponectin level predicted lower HbA1c levels at 12 and 24 months, and elevated adiponectin correlated with enhanced β-cell function, lower triglyceride levels and fat loss. CONCLUSIONS Smaller rises in adiponectin level, a mediator of insulin action and adipose mass, characterize type 2 diabetes non-remission up to 2 years after bariatric surgery. Adjunctive strategies promoting greater fat loss and/or raising adiponectin may be key to achieving higher type 2 diabetes remission rates after bariatric surgery.
Collapse
Affiliation(s)
- Steven K. Malin
- Dept of Pathobiology, Cleveland Clinic, Cleveland OH, USA
- Dept of Nutrition, Case Western Reserve University Cleveland Clinic, Cleveland OH, USA
| | - James Bena
- Dept of Quantitative Health Sciences, Cleveland Clinic, Cleveland OH, USA
| | - Beth Abood
- Dept of Endocrinology, Diabetes and Metabolism, Cleveland Clinic, Cleveland OH, USA
| | - Claire E. Pothier
- Dept of Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland OH, USA
| | - Deepak L Bhatt
- Brigham and Women’s Hospital, and Harvard Medical School, Boston, Massachusetts, USA
| | - Steven Nissen
- Dept of Heart and Vascular, Cleveland Clinic, Cleveland OH, USA
| | - Stacy A. Brethauer
- Dept of Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland OH, USA
- Metabolic Translational Research Center, Endocrine and Metabolism Institute, Cleveland Clinic, Cleveland OH, USA
| | - Philip R. Schauer
- Dept of Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland OH, USA
- Metabolic Translational Research Center, Endocrine and Metabolism Institute, Cleveland Clinic, Cleveland OH, USA
| | - John P. Kirwan
- Dept of Pathobiology, Cleveland Clinic, Cleveland OH, USA
- Metabolic Translational Research Center, Endocrine and Metabolism Institute, Cleveland Clinic, Cleveland OH, USA
- Dept of Nutrition, Case Western Reserve University Cleveland Clinic, Cleveland OH, USA
| | - Sangeeta R. Kashyap
- Dept of Endocrinology, Diabetes and Metabolism, Cleveland Clinic, Cleveland OH, USA
- Metabolic Translational Research Center, Endocrine and Metabolism Institute, Cleveland Clinic, Cleveland OH, USA
| |
Collapse
|
134
|
Comparative analysis of maple syrup to other natural sweeteners and evaluation of their metabolic responses in healthy rats. J Funct Foods 2014. [DOI: 10.1016/j.jff.2014.10.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
135
|
Pabreja K, Mohd MA, Koole C, Wootten D, Furness SGB. Molecular mechanisms underlying physiological and receptor pleiotropic effects mediated by GLP-1R activation. Br J Pharmacol 2014; 171:1114-28. [PMID: 23889512 DOI: 10.1111/bph.12313] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 07/10/2013] [Accepted: 07/19/2013] [Indexed: 12/22/2022] Open
Abstract
The incidence of type 2 diabetes in developed countries is increasing yearly with a significant negative impact on patient quality of life and an enormous burden on the healthcare system. Current biguanide and thiazolidinedione treatments for type 2 diabetes have a number of clinical limitations, the most serious long-term limitation being the eventual need for insulin replacement therapy (Table 1). Since 2007, drugs targeting the glucagon-like peptide-1 (GLP-1) receptor have been marketed for the treatment of type 2 diabetes. These drugs have enjoyed a great deal of success even though our underlying understanding of the mechanisms for their pleiotropic effects remain poorly characterized even while major pharmaceutical companies actively pursue small molecule alternatives. Coupling of the GLP-1 receptor to more than one signalling pathway (pleiotropic signalling) can result in ligand-dependent signalling bias and for a peptide receptor such as the GLP-1 receptor this can be exaggerated with the use of small molecule agonists. Better consideration of receptor signalling pleiotropy will be necessary for future drug development. This is particularly important given the recent failure of taspoglutide, the report of increased risk of pancreatitis associated with GLP-1 mimetics and the observed clinical differences between liraglutide, exenatide and the newly developed long-acting exenatide long acting release, albiglutide and dulaglutide.
Collapse
Affiliation(s)
- K Pabreja
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic., Australia
| | | | | | | | | |
Collapse
|
136
|
Rashti F, Gupta E, Ebrahimi S, Shope TR, Koch TR, Gostout CJ. Development of minimally invasive techniques for management of medically-complicated obesity. World J Gastroenterol 2014; 20:13424-13445. [PMID: 25309074 PMCID: PMC4188895 DOI: 10.3748/wjg.v20.i37.13424] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/15/2014] [Accepted: 07/16/2014] [Indexed: 02/06/2023] Open
Abstract
The field of bariatric surgery has been rapidly growing and evolving over the past several decades. During the period that obesity has become a worldwide epidemic, new interventions have been developed to combat this complex disorder. The development of new laparoscopic and minimally invasive treatments for medically-complicated obesity has made it essential that gastrointestinal physicians obtain a thorough understanding of past developments and possible future directions in bariatrics. New laparoscopic advancements provide patients and practitioners with a variety of options that have an improved safety profile and better efficacy without open, invasive surgery. The mechanisms of weight loss after bariatric surgery are complex and may in part be related to altered release of regulatory peptide hormones from the gut. Endoscopic techniques designed to mimic the effects of bariatric surgery and endolumenal interventions performed entirely through the gastrointestinal tract offer potential advantages. Several of these new techniques have demonstrated promising, preliminary results. We outline herein historical and current trends in the development of bariatric surgery and its transition to safer and more minimally invasive procedures designed to induce weight loss.
Collapse
|
137
|
Endogenous Glucagon-Like Peptide-1 as a Potential Mediator of the Resolution of Diabetic Kidney Disease following Roux en Y Gastric Bypass: Evidence and Perspectives. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/503846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Diabetic kidney disease in patients with type 2 diabetes strongly correlates with the incidence of major cardiovascular events and all-cause mortality. Pharmacological and lifestyle based management focusing on glycaemic, lipid, and blood pressure control is the mainstay of treatment but efficacy remains limited. Roux en Y gastric bypass is an efficacious intervention in diabetes. Emerging evidence also supports a role for bypass as an intervention for early diabetic kidney disease. This paper firstly presents level 1 evidence of the effects of bypass on hyperglycaemia and hypertension and then summarises emerging data on its effects on diabetic kidney disease. Glucagon-like peptide-1 is implicated as a central mediator of diabetes resolution following bypass through the incretin effect. It has been ascribed vasodilatory, pronatriuretic, and antioxidant properties and its exogenous administration or optimisation of its endogenous levels via dipeptidyl peptidase IV inhibition results in antioxidant and antiproteinuric effects in preclinical models of DKD. Some evidence is emerging of translation of coherent effects in the clinical setting. These findings raise the question of whether pharmacotherapy targeted at optimising circulating hormone levels may be capable of recapitulating some of the effects of bypass surgery on renal injury.
Collapse
|
138
|
Clinical Course of Diabetes After Gastrectomy According to Type of Reconstruction in Patients with Concurrent Gastric Cancer and Type 2 Diabetes. Obes Surg 2014; 25:673-9. [DOI: 10.1007/s11695-014-1426-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
139
|
Abstract
To date, weight loss surgeries are the most effective treatment for obesity and glycemic control in patients with type 2 diabetes. Roux-en-Y gastric bypass surgery (RYGB) and sleeve gastrectomy (SG), two widely used bariatric procedures for the treatment of obesity, induce diabetes remission independent of weight loss while glucose improvement after adjustable gastric banding (AGB) is proportional to the amount of weight loss. The immediate, weight-loss independent glycemic effect of gastric bypass has been attributed to postprandial hyperinsulinemia and an enhanced incretin effect. The rapid passage of nutrients into the intestine likely accounts for significantly enhanced glucagon like-peptide 1 (GLP-1) secretion, and postprandial hyperinsulinemia after GB is typically attributed to the combined effects of elevated glucose and GLP-1. For this review we focus on the beneficial effects of the three most commonly performed bariatric procedures, RYGB, SG, and AGB, on glucose metabolism and diabetes remission. Central to this discussion will be the extent to which the effects of surgery are mediated by GLP-1. Better understanding of these mechanisms could provide insight to development of novel therapeutic strategies for treatment of diabetes as well as refinement of surgical techniques.
Collapse
Affiliation(s)
- Marzieh Salehi
- Department of Internal Medicine, Division of Endocrinology, Diabetes, & Metabolism, University of Cincinnati College of Medicine, 260 Stetson, Suite 4200, Cincinnati, OH 45219-0547 USA
| | - David A. D’Alessio
- Department of Internal Medicine, Division of Endocrinology, Diabetes, & Metabolism, University of Cincinnati College of Medicine, 260 Stetson, Suite 4200, Cincinnati, OH 45219-0547 USA
- Cincinnati VA Medical Center, Cincinnati, OH USA
| |
Collapse
|
140
|
Selectivity of peptide ligands for the human incretin receptors expressed in HEK-293 cells. Eur J Pharmacol 2014; 741:311-5. [PMID: 25179575 DOI: 10.1016/j.ejphar.2014.08.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 08/19/2014] [Accepted: 08/20/2014] [Indexed: 12/20/2022]
Abstract
The increase in insulin response to oral glucose compared with glucose given by intravenous injection is termed the incretin effect and is mediated by two peptide hormones secreted from the gut in response to nutrient intake: glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP). GLP-1 and GIP exert their insulinotropic effects through their respective receptors expressed on pancreatic β-cells. Both the GLP-1 receptor and the GIP receptor are members of the secretin family of G protein-coupled receptors and couple positively with adenylate cyclase, resulting in an increase in intracellular cAMP. In the present study, we investigated the activity of six previously reported peptide ligands at both the GLP-1 and GIP receptors expressed on HEK-293 cells using a highly sensitive reporter gene assay. GLP-1 and GIP demonstrated almost 100,000-fold selectivity for their respective receptors. Exendin 4 (Ex-4), a long-acting GLP-1 receptor agonist, displayed considerable activity at the GIP receptor. Exendin 9-39 (Ex 9-39) was able to block activity at both the GLP-1 and GIP receptors, and Pro3GIP, a previously-reported GIP receptor antagonist, was shown to act as a partial agonist at the GIP receptor. These data highlight the need for more selective antagonists to study these therapeutically important receptors.
Collapse
|
141
|
Abstract
This review summarizes recent evidence related to the safety, efficacy, and metabolic outcomes of bariatric surgery to guide clinical decision making. Several short term randomized controlled trials have demonstrated the effectiveness of bariatric procedures for inducing weight loss and initial remission of type 2 diabetes. Observational studies have linked bariatric procedures with long term improvements in body weight, type 2 diabetes, survival, cardiovascular events, incident cancer, and quality of life. Perioperative mortality for the average patient is low but varies greatly across subgroups. The incidence of major complications after surgery also varies widely, and emerging data show that some procedures are associated with a greater risk of substance misuse disorders, suicide, and nutritional deficiencies. More research is needed to enable long term outcomes to be compared across various procedures and subpopulations, and to identify those most likely to benefit from surgical intervention. Given uncertainties about the balance between the risks and benefits of bariatric surgery in the long term, the decision to undergo surgery should be based on a high quality shared decision making process.
Collapse
Affiliation(s)
- David E Arterburn
- Group Health Research Institute, Group Health Cooperative, Seattle, WA 98101, USA
| | | |
Collapse
|
142
|
Sala PC, Torrinhas RS, Giannella-Neto D, Waitzberg DL. Relationship between gut hormones and glucose homeostasis after bariatric surgery. Diabetol Metab Syndr 2014; 6:87. [PMID: 25152774 PMCID: PMC4141947 DOI: 10.1186/1758-5996-6-87] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 08/06/2014] [Indexed: 12/18/2022] Open
Abstract
Type 2 diabetes mellitus (T2D) is emerging as a worldwide public health problem, and is mainly associated with an increased incidence of obesity. Bariatric surgery is currently considered the most effective treatment for severely obese patients. After bariatric surgery, T2D patients have shown a significant improvement in glycemic control, even before substantial weight loss and often discontinuation of medication for diabetes control. A central role for enteroendocrine cells from the epithelium of the gastrointestinal tract has been speculated in this postoperative phenomenon. These cells produce and secrete polypeptides - gut hormones - that are associated with regulating energy intake and glucose homeostasis through modulation of peripheral target organs, including the endocrine pancreas. This article reviews and discusses the biological actions of the gut hormones ghrelin, cholecystokinin, incretins, enteroglucagon, and Peptide YY, all of which were recently identified as potential candidates for mediators of glycemic control after bariatric surgery. In conclusion, current data reinforce the hypothesis that T2D reversion after bariatric surgery may be related to glycemic homeostasis developed by the intestine.
Collapse
Affiliation(s)
- Priscila Campos Sala
- />Medical School, Department of Gastroenterology, Digestive Surgery Discipline (LIM 35), University of São Paulo, Av. Dr. Arnaldo, 455, Cerqueira César, CEP: 01246-903, São Paulo, Brazil
| | - Raquel Susana Torrinhas
- />Medical School, Department of Gastroenterology, Digestive Surgery Discipline (LIM 35), University of São Paulo, Av. Dr. Arnaldo, 455, Cerqueira César, CEP: 01246-903, São Paulo, Brazil
| | | | - Dan Linetzky Waitzberg
- />Medical School, Department of Gastroenterology, Digestive Surgery Discipline (LIM 35), University of São Paulo, Av. Dr. Arnaldo, 455, Cerqueira César, CEP: 01246-903, São Paulo, Brazil
| |
Collapse
|
143
|
Uchida A, Zechner JF, Mani BK, Park WM, Aguirre V, Zigman JM. Altered ghrelin secretion in mice in response to diet-induced obesity and Roux-en-Y gastric bypass. Mol Metab 2014; 3:717-30. [PMID: 25353000 PMCID: PMC4209356 DOI: 10.1016/j.molmet.2014.07.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 07/22/2014] [Accepted: 07/25/2014] [Indexed: 01/06/2023] Open
Abstract
The current study examined potential mechanisms for altered circulating ghrelin levels observed in diet-induced obesity (DIO) and following weight loss resulting from Roux-en-Y gastric bypass (RYGB). We hypothesized that circulating ghrelin levels were altered in obesity and after weight loss through changes in ghrelin cell responsiveness to physiological cues. We confirmed lower ghrelin levels in DIO mice and demonstrated elevated ghrelin levels in mice 6 weeks post-RYGB. In both DIO and RYGB settings, these changes in ghrelin levels were associated with altered ghrelin cell responsiveness to two key physiological modulators of ghrelin secretion - glucose and norepinephrine. In DIO mice, increases in ghrelin cell density within both the stomach and duodenum and in somatostatin-immunoreactive D cell density in the duodenum were observed. Our findings provide new insights into the regulation of ghrelin secretion and its relation to circulating ghrelin within the contexts of obesity and weight loss.
Collapse
Affiliation(s)
- Aki Uchida
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA ; Division of Endocrinology & Metabolism, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA ; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Juliet F Zechner
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA ; Division of Digestive and Liver Diseases, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bharath K Mani
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA ; Division of Endocrinology & Metabolism, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA ; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Won-Mee Park
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA ; Division of Endocrinology & Metabolism, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA ; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vincent Aguirre
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA ; Division of Digestive and Liver Diseases, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey M Zigman
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA ; Division of Endocrinology & Metabolism, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA ; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
144
|
Sweeney TE, Morton JM. Metabolic surgery: action via hormonal milieu changes, changes in bile acids or gut microbiota? A summary of the literature. Best Pract Res Clin Gastroenterol 2014; 28:727-40. [PMID: 25194186 PMCID: PMC4399638 DOI: 10.1016/j.bpg.2014.07.016] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 07/21/2014] [Accepted: 07/28/2014] [Indexed: 01/31/2023]
Abstract
Obesity and type 2 diabetes remain epidemic problems. Different bariatric surgical techniques causes weight loss and diabetes remission to varying degrees. The underlying mechanisms of the beneficial effects of bariatric surgery are complex, and include changes in diet and behaviour, as well as changes in hormones, bile acid flow, and gut bacteria. We summarized the effects of multiple different bariatric procedures, and their resulting effects on several hormones (leptin, ghrelin, adiponectin, glucagon-like peptide 1 (GLP-1), peptide YY, and glucagon), bile acid changes in the gut and the serum, and resulting changes to the gut microbiome. As much as possible, we have tried to incorporate multiple studies to try to explain underlying mechanistic changes. What emerges from the data is a picture of clear differences between restrictive and metabolic procedures. The latter, in particular the roux-en-Y gastric bypass, induces large and distinctive changes in most measured fat and gut hormones, including early and sustained increase in GLP-1, possible through intestinal bile acid signalling. The changes in bile flow and the gut microbiome are causally inseparable so far, but new studies show that each contributes to the effects of weight loss and diabetes resolution.
Collapse
Affiliation(s)
- Timothy E Sweeney
- Stanford University, Department of General Surgery, Section of Bariatric and Minimally Invasive (BMI) Surgery, 300 Pasteur Drive, H3680, Stanford, CA 94025, USA
| | - John M Morton
- Stanford University, Department of General Surgery, Section of Bariatric and Minimally Invasive (BMI) Surgery, 300 Pasteur Drive, H3680, Stanford, CA 94025, USA.
| |
Collapse
|
145
|
Dirksen C, Jacobsen SH, Bojsen-Møller KN, Jørgensen NB, Naver LS, Hvolris LE, Worm D, Madsbad S, Holst JJ, Hansen DL. Reduction in cardiovascular risk factors and insulin dose, but no beta-cell regeneration 1 year after Roux-en-Y gastric bypass in an obese patient with type 1 diabetes: a case report. Obes Res Clin Pract 2014; 7:e269-74. [PMID: 24306154 DOI: 10.1016/j.orcp.2012.01.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 01/24/2012] [Accepted: 01/24/2012] [Indexed: 01/06/2023]
Abstract
Experience with Roux-en-Y gastric bypass in patients with type 1 diabetes is very limited, despite an increasing prevalence of obesity also in this population. We describe changes in anthropometric measures, insulin dose, HbA1c, blood pressure, lipid status, and metabolic response to a liquid mixed meal throughout the first year after RYGB in an obese patient with type 1 diabetes. No change in HbA1c was observed, but a 48% reduction in weight-adjusted insulin dose and improvements in cardiovascular risk factors was seen 1 year after surgery. Exaggerated secretions of anorexigenic gut hormones were seen during the meals.
Collapse
Affiliation(s)
- Carsten Dirksen
- Novo Nordisk Foundation Centre for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Incretin response to a standard test meal in a rat model of sleeve gastrectomy with diet-induced obesity. Obes Surg 2014; 24:95-101. [PMID: 23934273 DOI: 10.1007/s11695-013-1056-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND Currently, the most effective treatment for obesity is bariatric surgery. Gastroduodenal bypass surgery produces sustained weight loss and improves glycemic control and insulin sensitivity. Previous studies have shown that sleeve gastrectomy (SG) produces similar results and implicate changes in incretin hormone release in these effects. METHODS Male Sprague-Dawley rats were divided into four groups; lean control (lean), diet-induced obesity (DIO), DIO animals that had undergone SG (SG), and DIO animals that had undergone a sham operation (sham). RESULTS After a 2-week recovery period, the incretin response to a standard test meal was measured. Blood sampling was performed in free-moving rats at various time points using chronic vascular access to the right jugular vein. There was a significant increase in the bodyweight of DIO animals fed a high-fat/high-sugar diet compared with the lean animals, which was reversed by SG. DIO caused an impairment of the GLP-1 response to a standard test meal, but not the GIP response. SG resulted in a dramatic increase in the GLP-1 response to a standard test meal but had no effect on the GIP response. CONCLUSIONS A rapid rise in blood sugar was observed in the SG group following a standard test meal that was followed by reactive hypoglycemia. SG dramatically increases the GLP-1 response to a standard test meal but has no effect on GIP in a rat model of DIO.
Collapse
|
147
|
Tsuei J, Chau T, Mills D, Wan YJY. Bile acid dysregulation, gut dysbiosis, and gastrointestinal cancer. Exp Biol Med (Maywood) 2014; 239:1489-504. [PMID: 24951470 DOI: 10.1177/1535370214538743] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Because of increasingly widespread sedentary lifestyles and diets high in fat and sugar, the global diabetes and obesity epidemic continues to grow unabated. A substantial body of evidence has been accumulated which associates diabetes and obesity to dramatically higher risk of cancer development, particularly in the liver and gastrointestinal tract. Additionally, diabetic and obese individuals have been shown to suffer from dysregulation of bile acid (BA) homeostasis and dysbiosis of the intestinal microbiome. Abnormally elevated levels of cytotoxic secondary BAs and a pro-inflammatory shift in gut microbial profile have individually been linked to numerous enterohepatic diseases including cancer. However, recent findings have implicated a detrimental interplay between BA dysregulation and intestinal dysbiosis that promotes carcinogenesis along the gut-liver axis. This review seeks to examine the currently investigated interactions between the regulation of BA metabolism and activity of the intestinal microbiota and how these interactions can drive cancer formation in the context of diabesity. The precarcinogenic effects of BA dysregulation and gut dysbiosis including excessive inflammation, heightened oxidative DNA damage, and increased cell proliferation are discussed. Furthermore, by focusing on the mediatory roles of BA nuclear receptor farnesoid x receptor, ileal transporter apical sodium dependent BA transporter, and G-coupled protein receptor TGR5, this review attempts to connect BA dysregulation, gut dysbiosis, and enterohepatic carcinogenesis at a mechanistic level. A better understanding of the intricate interplay between BA homeostasis and gut microbiome can yield novel avenues to combat the impending rise in diabesity-related cancers.
Collapse
Affiliation(s)
- Jessica Tsuei
- Department of Pathology and Laboratory Medicine, University of California at Davis Medical Center, Sacramento, CA 95831, USA
| | - Thinh Chau
- Department of Pathology and Laboratory Medicine, University of California at Davis Medical Center, Sacramento, CA 95831, USA
| | - David Mills
- Department of Food Science and Technology, Department of Viticulture and Enology, Foods for Health Institute, University of California, Davis, CA 95616, USA
| | - Yu-Jui Yvonne Wan
- Department of Pathology and Laboratory Medicine, University of California at Davis Medical Center, Sacramento, CA 95831, USA
| |
Collapse
|
148
|
Lips MA, de Groot GH, van Klinken JB, Aarts E, Berends FJ, Janssen IM, Van Ramshorst B, Van Wagensveld BA, Swank DJ, Van Dielen F, Willems van Dijk K, Pijl H. Calorie restriction is a major determinant of the short-term metabolic effects of gastric bypass surgery in obese type 2 diabetic patients. Clin Endocrinol (Oxf) 2014; 80:834-42. [PMID: 23711328 DOI: 10.1111/cen.12254] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 05/02/2013] [Accepted: 05/23/2013] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Roux-en-Y gastric bypass (RYGB) and restrictive weight loss interventions, such as gastric banding (GB) and very-low-calorie diets (VLCD) directly impact glucose metabolism, possibly by calorie restriction and/or altered secretion of gut hormones. We aimed to establish the direct endocrine and metabolic effects of RYGB compared to restrictive interventions in obese glucose-tolerant (NGT) subjects and subjects with type 2 diabetes (T2DM). DESIGN Controlled, nonrandomized observational trial. PATIENTS AND MEASUREMENTS Four groups of obese females received a mixed meal at baseline and 3 weeks after intervention; NGT-GB (n = 11), NGT-RYGB (n = 16), T2DM-RYGB (n = 15) and T2DM-VLCD (n = 12). Normal weight controls (n = 12) were studied once. RESULTS At baseline, all obese subjects were hyperinsulinemic. T2DM was associated with hyperglycaemia and decreased GLP-1 levels. RYGB and VLCD reduced glucose levels to a similar extent in T2DM, insulin levels decreased only after VLCD. Comparison of restrictive intervention vs RYGB showed a more pronounced decrease in glucose and insulin AUC after restriction. In NGT and T2DM subjects, RYGB increased GLP-1 and PYY levels and decreased ghrelin levels, whereas VLCD and GB only increased GIP levels. CONCLUSIONS These data indicate that deterioration of glucose metabolism in T2DM is associated with a decline of GLP-1 levels. Calorie restriction facilitates glucose metabolism and blunts hyperinsulinemia in obese (diabetic) humans. Additional duodenal exclusion through RYGB induces gut hormone release and hyperinsulinemia but does not improve postprandial glucose levels any further. Our data thus strongly suggest that calorie restriction underlies the short-term metabolic benefits of RYGB in obese T2DM patients.
Collapse
Affiliation(s)
- Mirjam A Lips
- Department of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Hayes MR, Mietlicki-Baase EG, Kanoski SE, De Jonghe BC. Incretins and amylin: neuroendocrine communication between the gut, pancreas, and brain in control of food intake and blood glucose. Annu Rev Nutr 2014; 34:237-60. [PMID: 24819325 DOI: 10.1146/annurev-nutr-071812-161201] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Arguably the most fundamental physiological systems for all eukaryotic life are those governing energy balance. Without sufficient energy, an individual is unable to survive and reproduce. Thus, an ever-growing appreciation is that mammalian physiology developed a redundant set of neuroendocrine signals that regulate energy intake and expenditure, which maintains sufficient circulating energy, predominantly in the form of glucose, to ensure that energy needs are met throughout the body. This orchestrated control requires cross talk between the gastrointestinal tract, which senses the incoming meal; the pancreas, which produces glycemic counterregulatory hormones; and the brain, which controls autonomic and behavioral processes regulating energy balance. Therefore, this review highlights the physiological, pharmacological, and pathophysiological effects of the incretin hormones glucagon-like peptide-1 and gastric inhibitory polypeptide, as well as the pancreatic hormone amylin, on energy balance and glycemic control.
Collapse
Affiliation(s)
- Matthew R Hayes
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104;
| | | | | | | |
Collapse
|
150
|
Acosta A, Abu Dayyeh BK, Port JD, Camilleri M. Recent advances in clinical practice challenges and opportunities in the management of obesity. Gut 2014; 63:687-95. [PMID: 24402654 PMCID: PMC4170188 DOI: 10.1136/gutjnl-2013-306235] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Despite advances in understanding the roles of adiposity, food intake, GI and adipocyte-related hormones, inflammatory mediators, the gut-brain axis and the hypothalamic nervous system in the pathophysiology of obesity, the effects of different therapeutic interventions on those pathophysiological mechanisms are controversial. There are still no low-cost, safe, effective treatments for obesity and its complications. Currently, bariatric surgical approaches targeting the GI tract are more effective than non-surgical approaches in inducing weight reduction and resolving obesity-related comorbidities. However, current guidelines emphasise non-surgical approaches through lifestyle modification and medications to achieve slow weight loss, which is not usually sustained and may be associated with medication-related side effects. This review analyses current central, peripheral or hormonal targets to treat obesity and addresses challenges and opportunities to develop novel approaches for obesity.
Collapse
Affiliation(s)
- Andres Acosta
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Barham K. Abu Dayyeh
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - John D. Port
- Division of Neuroradiology, Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota
| |
Collapse
|