1
|
van der Kolk BW, Pirinen E, Nicoll R, Pietiläinen KH, Heinonen S. Subcutaneous adipose tissue and skeletal muscle mitochondria following weight loss. Trends Endocrinol Metab 2025; 36:339-363. [PMID: 39289110 DOI: 10.1016/j.tem.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/19/2024]
Abstract
Obesity is a major global health issue with various metabolic complications. Both bariatric surgery and dieting achieve weight loss and improve whole-body metabolism, but vary in their ability to maintain these improvements over time. Adipose tissue and skeletal muscle metabolism are crucial in weight regulation, and obesity is linked to mitochondrial dysfunction in both tissues. The impact of bariatric surgery versus dieting on adipose tissue and skeletal muscle mitochondrial metabolism remains to be elucidated. Understanding the molecular pathways that modulate tissue metabolism following weight loss holds potential for identifying novel therapeutic targets in obesity management. This narrative review summarizes current knowledge on mitochondrial metabolism following bariatric surgery and diet-induced weight loss in adipose tissue and skeletal muscle, and sheds light on their respective effects.
Collapse
Affiliation(s)
- Birgitta W van der Kolk
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Eija Pirinen
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Finland; Faculty of Medicine, Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Rachel Nicoll
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; HealthyWeightHub, Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Internal Medicine, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
2
|
Meiring S, Aydin Ö, van Baar ACG, van der Vossen EWJ, Rampanelli E, van Grieken NCT, Holleman F, Nieuwdorp M, Bergman JJGHM. From Endoscopic Inspection to Gene-Expression: A Thorough Assessment of the Duodenal Mucosa After Resurfacing-A Prospective Study. Dig Dis Sci 2025; 70:1052-1063. [PMID: 39779586 PMCID: PMC11920325 DOI: 10.1007/s10620-024-08710-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 10/22/2024] [Indexed: 01/11/2025]
Abstract
AIMS Duodenal Mucosal Resurfacing (DMR) is an endoscopic ablation technique aimed at improving glycemia in patients with type 2 diabetes mellitus (T2DM). Although the exact underlying mechanism is still unclear, it is postulated that the DMR-induced improvements are the result of changes in the duodenal mucosa. For this reason, we assessed macroscopic and microscopic changes in the duodenal mucosa induced by DMR + GLP-1RA. METHODS We included 16 patients with T2DM using basal insulin that received a combination treatment of a single DMR and GLP-1RA. Endoscopic evaluation was performed before the DMR procedure and 3 month after, and duodenal biopsies were obtained. Histological evaluation was performed and L and K cell density was calculated. In addition, gene-expression analysis and Western blotting was performed. RESULTS Endoscopic evaluation at 3 month showed duodenal mucosa with a normal appearance. In line, microscopic histological evaluation showed no signs of villous atrophy or inflammation and unchanged L and K cell density. Unbiased transcriptome profiling and western blotting revealed that PDZK1 expression was higher in responders at baseline and after DMR. GATA6 expression was significantly increased in responders after DMR compared to non-responders. CONCLUSION The absence of macroscopic and microscopic changes after 3 month suggest that improvements in glycemic parameters after DMR do not result from significant histological changes in duodenal mucosa. It is more likely that these improvements result from more subtle changes in enteroendocrine signaling. PDZK1 and GATA6 expression might play a role in DMR; this needs to be confirmed in pre-clinical studies.
Collapse
Affiliation(s)
- S Meiring
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centres, Location AMC, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Ö Aydin
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centres, Location AMC, Amsterdam, The Netherlands
- Department of Bariatric Surgery, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - A C G van Baar
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centres, Location AMC, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.
| | - E W J van der Vossen
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centres, Location AMC, Amsterdam, The Netherlands
| | - E Rampanelli
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centres, Location AMC, Amsterdam, The Netherlands
| | - N C T van Grieken
- Department of Pathology, Amsterdam University Medical Centres, Location VUmc, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - F Holleman
- Department of Internal Medicine, Amsterdam University Medical Centres, Location AMC, Amsterdam, The Netherlands
| | - M Nieuwdorp
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centres, Location AMC, Amsterdam, The Netherlands
| | - J J G H M Bergman
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centres, Location AMC, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Çalık Başaran N, Dotan I, Dicker D. Post metabolic bariatric surgery weight regain: the importance of GLP-1 levels. Int J Obes (Lond) 2025; 49:412-417. [PMID: 38225284 PMCID: PMC11971041 DOI: 10.1038/s41366-024-01461-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/17/2024]
Abstract
Weight regain and insufficient weight loss are essential problems after metabolic bariatric surgery (MBS) in people living with obesity. Changes in the level of glucagon-like peptide-1 (GLP-1) secreted from the gut after bariatric surgery are one of the underlying mechanisms for successful initial weight loss. Studies and meta-analyses have revealed that postprandial GLP-1 levels increase after the Roux-en-Y gastric bypass and sleeve gastrectomy, but fasting GLP-1 levels do not increase significantly. Some observational studies have shown the relationship between higher postprandial GLP-1 levels and successful weight loss after bariatric surgery. There is growing evidence that GLP-1-receptor agonist (GLP-1-RA) use in patients who regained weight after bariatric surgery has resulted in significant weight loss. In this review, we aimed to summarize the changes in endogenous GLP-1 levels and their association with weight loss after MBS, describe the effects of GLP-1-RA use on weight loss after MBS, and emphasize metabolic adaptations in light of the recent literature. We hypothesized that maintaining higher basal-bolus GLP-1-RA levels may be a promising treatment choice in people with obesity who failed to lose weight after bariatric surgery.
Collapse
Affiliation(s)
- Nursel Çalık Başaran
- Hacettepe University, Faculty of Medicine, Department of Internal Medicine, General Internal Medicine, Ankara, Türkiye.
| | - Idit Dotan
- Rabin Medical Center, Beilinson Hospital, Department of Endocrinology and Obesity Clinic, Petah Tikva, Israel
- Tel Aviv University, Faculty of Medicine, Tel Aviv, Israel
| | - Dror Dicker
- Tel Aviv University, Faculty of Medicine, Tel Aviv, Israel
- Rabin Medical Center, Hasharon Hospital, Department of Internal Medicine and Obesity Clinic, Petah Tikva, Israel
| |
Collapse
|
4
|
Karlsson C, Johnson LK, Greasley PJ, Retterstøl K, Hedberg J, Hall M, Hawker N, Robertsen I, Havsol J, Hertel JK, Sandbu R, Skovlund E, Olsen T, Christensen H, Jansson-Löfmark R, Andersson S, Åsberg A, Hjelmesæth J. Gastric Bypass vs Diet and Cardiovascular Risk Factors: A Nonrandomized Controlled Trial. JAMA Surg 2024; 159:971-980. [PMID: 38959017 PMCID: PMC11223056 DOI: 10.1001/jamasurg.2024.2162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/13/2024] [Indexed: 07/04/2024]
Abstract
Importance Roux-en-Y gastric bypass (RYGB) is associated with reduced cardiovascular (CV) risk factors, morbidity, and mortality. Whether these effects are specifically induced by the surgical procedure or the weight loss is unclear. Objective To compare 6-week changes in CV risk factors in patients with obesity undergoing matching caloric restriction and weight loss by RYGB or a very low-energy diet (VLED). Design, Setting, and Participants This nonrandomized controlled study (Impact of Body Weight, Low Calorie Diet, and Gastric Bypass on Drug Bioavailability, Cardiovascular Risk Factors, and Metabolic Biomarkers [COCKTAIL]) was conducted at a tertiary care obesity center in Norway. Participants were individuals with severe obesity preparing for RYGB or a VLED. Recruitment began February 26, 2015; the first patient visit was on March 18, 2015, and the last patient visit (9-week follow-up) was on August 9, 2017. Data were analyzed from April 30, 2021, through June 29, 2023. Interventions VLED alone for 6 weeks or VLED for 6 weeks after RYGB; both interventions were preceded by 3-week LED. Main Outcomes and Measures Between-group comparisons of 6-week changes in CV risk factors. Results Among 78 patients included in the analyses, the mean (SD) age was 47.5 (9.7) years; 51 (65%) were women, and 27 (35%) were men. Except for a slightly higher mean (SD) body mass index of 44.5 (6.2) in the RYGB group (n = 41) vs 41.9 (5.4) in the VLED group (n = 37), baseline demographic and clinical characteristics were similar between groups. Major atherogenic blood lipids (low-density lipoprotein cholesterol, non-high-density lipoprotein cholesterol, apolipoprotein B, lipoprotein[a]) were reduced after RYGB in comparison with VLED despite a similar fat mass loss. Mean between-group differences were -17.7 mg/dL (95% CI, -27.9 to -7.5), -17.4 mg/dL (95% CI, -29.8 to -5.0) mg/dL, -9.94 mg/dL (95% CI, -15.75 to -4.14), and geometric mean ratio was 0.55 U/L (95% CI, 0.42 to 0.72), respectively. Changes in glycemic control and blood pressure were similar between groups. Conclusions and Relevance This study found that clinically meaningful reductions in major atherogenic blood lipids were demonstrated after RYGB, indicating that RYGB may reduce CV risk independent of weight loss. Trial Registration ClinicalTrials.gov Identifier: NCT02386917.
Collapse
Affiliation(s)
- Cecilia Karlsson
- Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Line Kristin Johnson
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, Tønsberg, Norway
| | - Peter J. Greasley
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Kjetil Retterstøl
- The Lipid Clinic, Oslo University Hospital, Oslo, Norway
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jonatan Hedberg
- Medical Evidence and Observational Research, Global Medical BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Martin Hall
- Early Biometrics & Statistical Innovation, Data Science & Artificial Intelligence, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Noele Hawker
- Early Biometrics & Statistical Innovation, Data Science & Artificial Intelligence, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ida Robertsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Jesper Havsol
- Data Science and Artificial Intelligence, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jens Kristoffer Hertel
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, Tønsberg, Norway
| | - Rune Sandbu
- Department of Surgery, Vestfold Hospital Trust, Tønsberg, Norway
| | - Eva Skovlund
- Department of Public Health and Nursing, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Hege Christensen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Rasmus Jansson-Löfmark
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Shalini Andersson
- Research and Early Development, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anders Åsberg
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
- Department of Transplantation Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Jøran Hjelmesæth
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, Tønsberg, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
5
|
Busch CBE, Meiring S, van Baar ACG, Gastaldelli A, DeFronzo R, Mingrone G, Hagen M, White K, Rajagopalan H, Nieuwdorp M, Bergman JJGHM. Insulin sensitivity and beta cell function after duodenal mucosal resurfacing: an open-label, mechanistic, pilot study. Gastrointest Endosc 2024; 100:473-480.e1. [PMID: 38280531 DOI: 10.1016/j.gie.2024.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 01/15/2024] [Accepted: 01/21/2024] [Indexed: 01/29/2024]
Abstract
BACKGROUND AND AIMS The duodenum has been shown to play a key role in glucose homeostasis. Duodenal mucosal resurfacing (DMR) is an endoscopic procedure for patients with type 2 diabetes (T2D) in which the duodenal mucosa is hydrothermally ablated. DMR improves glycemic control, but the underlying mechanisms remain unclear. Here, we report changes in glucoregulatory hormones and indices of insulin sensitivity and beta cell function after DMR. METHODS We included 28 patients on noninsulin glucose-lowering medications who underwent open-label DMR and a mixed meal test (MMT) in Revita-1 or Revita-2 studies. Inclusion criteria were a hemoglobin A1c from 7.6% to 10.4% and a body mass index of 24 to 40 kg/m2. Baseline and 3-month MMT data included plasma glucose, insulin, C-peptide, glucagon-like peptide-1 (GLP-1), and gastric inhibitory polypeptide (GIP) concentrations. Glucoregulatory hormones, insulin sensitivity indices (Homeostatic Model Assessment for Insulin Resistance [HOMA-IR], Matsuda index [MI], and hepatic insulin resistance) and beta cell function (insulinogenic index, disposition index [DI], and insulin secretion rate [ISR]) were assessed. RESULTS Fasting insulin, glucagon, and C-peptide decreased significantly. Insulin sensitivity (HOMA-IR, MI, and hepatic insulin resistance) and beta cell function (DI and ISR) all improved significantly. Declines in postprandial glucose, mainly driven by a decrease in fasting levels, and in postprandial glucagon were observed, whereas GLP-1 and GIP did not change. CONCLUSIONS Insulin sensitivity and insulin secretion improved 3 months after DMR. It is unlikely that incretin changes are responsible for improved glucose control after DMR. These data add to the growing evidence validating the duodenum as a therapeutic target for patients with T2D. (Clinical trial registration numbers: NCT02413567 and NCT03653091.).
Collapse
Affiliation(s)
- Celine B E Busch
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Suzanne Meiring
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Annieke C G van Baar
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Amalia Gastaldelli
- Cardiometabolic Risk Laboratory, CNR Institute of Clinical Physiology, Pisa, Italy
| | - Ralph DeFronzo
- Diabetes Division, University of Texas Health Science Center, Texas Diabetes Institute, San Antonio, Texas, USA
| | - Geltrude Mingrone
- Division of Obesity and Metabolic Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Diabetes, School of Life Course Sciences, King's College London, London, UK
| | - Moira Hagen
- Fractyl Health Inc., Lexington, Massachusetts, USA
| | - Kelly White
- Fractyl Health Inc., Lexington, Massachusetts, USA
| | | | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Jacques J G H M Bergman
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| |
Collapse
|
6
|
Hur KY. Enteroendocrine Reprogramming by Altered Epithelial-Mesenchymal Crosstalk in Metabolic Surgery. JOURNAL OF METABOLIC AND BARIATRIC SURGERY 2024; 13:1-7. [PMID: 38974890 PMCID: PMC11224006 DOI: 10.17476/jmbs.2024.13.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 05/25/2024] [Accepted: 05/30/2024] [Indexed: 07/09/2024]
Abstract
Metabolic surgery is an effective treatment option for type 2 diabetes. However, the therapeutic scope has been limited by unexpected inconsistent outcomes. This study aims to overcome these obstacles by determining fundamental mechanisms from a novel perspective by analyzing and comparing the surgical anatomy, clinical characteristics, and outcomes of metabolic surgery, including duodenal-jejunal bypass, Roux-en-Y gastric bypass, biliopancreatic diversion, one anastomosis gastric bypass, and their modified procedures, predominantly focusing on nonobese patients to mitigate confounding effects from overweighted type 2 diabetes. Regional epithelial cell growth and unique villus formation along the anterior-posterior axis of the small intestine depend on crosstalk between the epithelium and the underlying mesenchyme. Due to altered crosstalk between the epithelium and the opposite mesenchyme at the anastomotic site, the enteroendocrine lineage of the distal intestine is replaced by the proximal epithelium after the bypass procedure. Subsequent intestinal compensatory proliferation accelerates the expansion of the replaced epithelium, including enteroendocrine cells. The primary reasons for unsatisfactory results are incomplete duodenal exclusion and insufficient biliopancreatic limb length. We anticipate that this novel mechanism will have a significant impact on metabolic surgery outcomes and provide valuable insight into optimizing its effectiveness in type 2 diabetes.
Collapse
Affiliation(s)
- Kyung Yul Hur
- Department of Surgery, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, Seoul, Korea
| |
Collapse
|
7
|
Lannoo M, Simoens C, Vangoitsenhoven R, Gillard P, D'Hoore A, De Vadder M, Mertens A, Deleus E, Steenackers N, Mathieu C, Van der Schueren B. Comparative impact of Roux-en-Y gastric bypass, sleeve gastrectomy or diet alone on beta-cell function in insulin-treated type 2 diabetes patients. Sci Rep 2024; 14:8211. [PMID: 38589596 PMCID: PMC11001928 DOI: 10.1038/s41598-024-59048-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 04/05/2024] [Indexed: 04/10/2024] Open
Abstract
Although bariatric surgery is an effective treatment for type 2 diabetes by inducing weight loss and augmenting gut hormone secretion, the immediate effect on beta-cell function itself remains to be elucidated in type 2 diabetes. Therefore, a prospective, randomized trial was performed in 30 patients with insulin-treated type 2 diabetes and a body mass index ≥ 35 kg/m2. Patients were randomly assigned (1:1:1) to Roux-en-Y gastric bypass (RYGB) or sleeve gastrectomy (SG) in combination with protein-sparing modified fast (PSMF), or to PSMF alone. Eu- and hyperglycemic clamps were performed before and 3 weeks after surgery and/or PSMF initiation. The primary outcome was the evolution of insulin sensitivity and beta-cell function after surgery, calculated using the composite measures of glucose disposal rate, insulin secretion rate, and disposition index (DI). Results revealed that markers of insulin sensitivity increased similarly in all arms (p = 0.43). A higher marker for maximal beta-cell function was observed when comparing SG to PSMF (p = 0.007). The DI showed a clear positive evolution after RYGB and SG, but not after PSMF alone. Altogether, these findings indicate that bariatric surgery results in an immediate beta-cell function recovery in insulin-treated type 2 diabetes.
Collapse
Affiliation(s)
- Matthias Lannoo
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Caroline Simoens
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Roman Vangoitsenhoven
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Pieter Gillard
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - André D'Hoore
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Mieke De Vadder
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Ann Mertens
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Ellen Deleus
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Nele Steenackers
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Bart Van der Schueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium.
- Department of Endocrinology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
8
|
Polkinghorne MD, West HW, Antoniades C. Adipose Tissue in Cardiovascular Disease: From Basic Science to Clinical Translation. Annu Rev Physiol 2024; 86:175-198. [PMID: 37931169 DOI: 10.1146/annurev-physiol-042222-021346] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
The perception of adipose tissue as a metabolically quiescent tissue, primarily responsible for lipid storage and energy balance (with some endocrine, thermogenic, and insulation functions), has changed. It is now accepted that adipose tissue is a crucial regulator of metabolic health, maintaining bidirectional communication with other organs including the cardiovascular system. Additionally, adipose tissue depots are functionally and morphologically heterogeneous, acting not only as sources of bioactive molecules that regulate the physiological functioning of the vasculature and myocardium but also as biosensors of the paracrine and endocrine signals arising from these tissues. In this way, adipose tissue undergoes phenotypic switching in response to vascular and/or myocardial signals (proinflammatory, profibrotic, prolipolytic), a process that novel imaging technologies are able to visualize and quantify with implications for clinical prognosis. Furthermore, a range of therapeutic modalities have emerged targeting adipose tissue metabolism and altering its secretome, potentially benefiting those at risk of cardiovascular disease.
Collapse
Affiliation(s)
- Murray D Polkinghorne
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom;
- Acute Multidisciplinary Imaging and Interventional Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Henry W West
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom;
- Acute Multidisciplinary Imaging and Interventional Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- Central Clinical School, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom;
- Acute Multidisciplinary Imaging and Interventional Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
9
|
Younes YR, Cron N, Field BC, Nayyar V, Clark J, Zachariah S, Lakshmipathy K, Isuga JO, Maghsoodi N, Emmanuel J. Proposed treatment strategy for reactive hypoglycaemia. Front Endocrinol (Lausanne) 2024; 15:1332702. [PMID: 38370356 PMCID: PMC10869498 DOI: 10.3389/fendo.2024.1332702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/15/2024] [Indexed: 02/20/2024] Open
Abstract
Background/aim Managing reactive hypoglycaemia (RH) poses challenges due to limited and often ineffective treatment options. We report a case series and draw on this to propose a stepwise treatment approach consisting of lifestyle modifications, metformin, GLP-1 analogues, and the use of flash glucose monitoring technology. Method A retrospective review was conducted to analyse the management of 11 cases presenting with recurrent RH symptoms. Result Two patients experienced successful resolution of symptoms through lifestyle modifications. Metformin alone was effective in treating seven out of nine patients who received pharmacological treatment. Two patients with previous upper gastrointestinal surgery showed a partial response to metformin and benefited further from additional long-acting GLP-1 analogue. Pharmacological intervention led to significant reductions in insulin and C-peptide levels in repeat mixed meal tolerance tests (P-values 0.043 for insulin and 0.006 for C-peptide). Finally, flash glucose monitoring technology was useful in early detection and preventing episodes of hypoglycaemia in one of these patients with persistent symptoms. Conclusion These findings highlight the potential efficacy of escalated treatment strategies for RH, including the use of metformin, GLP-1 analogues, and flash glucose monitoring technology.
Collapse
Affiliation(s)
- Younes R. Younes
- Department of Diabetes & Endocrinology, East Surrey Hospital, Surrey & Sussex Healthcare NHS Trust, Redhill, United Kingdom
| | - Nicholas Cron
- Department of Statistics, London School of Economics, London, United Kingdom
| | - Benjamin C.T. Field
- Department of Diabetes & Endocrinology, East Surrey Hospital, Surrey & Sussex Healthcare NHS Trust, Redhill, United Kingdom
- Section of Clinical Medicine, Faculty of Health & Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Vidhu Nayyar
- Department of Diabetes & Endocrinology, East Surrey Hospital, Surrey & Sussex Healthcare NHS Trust, Redhill, United Kingdom
| | - James Clark
- Department of Diabetes & Endocrinology, East Surrey Hospital, Surrey & Sussex Healthcare NHS Trust, Redhill, United Kingdom
| | - Sunil Zachariah
- Department of Diabetes & Endocrinology, East Surrey Hospital, Surrey & Sussex Healthcare NHS Trust, Redhill, United Kingdom
| | - Kavitha Lakshmipathy
- Department of Diabetes & Endocrinology, East Surrey Hospital, Surrey & Sussex Healthcare NHS Trust, Redhill, United Kingdom
| | - Jimboy O. Isuga
- Department of Diabetes & Endocrinology, East Surrey Hospital, Surrey & Sussex Healthcare NHS Trust, Redhill, United Kingdom
| | - Negar Maghsoodi
- Chemical Pathology Department, University Hospitals Sussex NHS Foundation Trust, Royal Sussex County Hospital, Brighton, United Kingdom
| | - Julian Emmanuel
- Department of Diabetes & Endocrinology, East Surrey Hospital, Surrey & Sussex Healthcare NHS Trust, Redhill, United Kingdom
| |
Collapse
|
10
|
Thaker VV, Kwee LC, Chen H, Bahson J, Ilkayeva O, Muehlbauer MJ, Wolfe B, Purnell JQ, Pi-Sunyer X, Newgard CB, Shah SH, Laferrère B, Look AHEAD Research Group. Metabolite signature of diabetes remission in individuals with obesity undergoing weight loss interventions. Obesity (Silver Spring) 2024; 32:304-314. [PMID: 37962326 PMCID: PMC11201087 DOI: 10.1002/oby.23943] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/25/2023] [Accepted: 09/19/2023] [Indexed: 11/15/2023]
Abstract
OBJECTIVE This observational study investigated metabolomic changes in individuals with type 2 diabetes (T2D) after weight loss. We hypothesized that metabolite changes associated with T2D-relevant phenotypes are signatures of improved health. METHODS Fasting plasma samples from individuals undergoing bariatric surgery (n = 71 Roux-en-Y gastric bypass [RYGB], n = 22 gastric banding), lifestyle intervention (n = 66), or usual care (n = 14) were profiled for 139 metabolites before and 2 years after weight loss. Principal component analysis grouped correlated metabolites into factors. Association of preintervention metabolites was tested with preintervention clinical features and changes in T2D markers. Association between change in metabolites/metabolite factors and change in T2D remission markers, homeostasis model assessment of β-cell function, homeostasis model assessment of insulin resistance, and glycated hemoglobin (HbA1c) was assessed. RESULTS Branched-chain amino acids (BCAAs) were associated with preintervention adiposity. Changes in BCAAs (valine, leucine/isoleucine) and branched-chain ketoacids were positively associated with change in HbA1c (false discovery rate q value ≤ 0.001) that persisted after adjustment for percentage weight change and RYGB (p ≤ 0.02). In analyses stratified by RYGB or other weight loss method, some metabolites showed association with non-RYGB weight loss. CONCLUSIONS This study confirmed known metabolite associations with obesity/T2D and showed an association of BCAAs with HbA1c change after weight loss, independent of the method or magnitude of weight loss.
Collapse
Affiliation(s)
- Vidhu V. Thaker
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | | | - Haiying Chen
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Salem, NC
| | - Judy Bahson
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Salem, NC
| | - Olga Ilkayeva
- Duke Molecular Physiology Institute
- Sarah W. Stedman Nutrition and Metabolism Center
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University School of Medicine, Durham, NC
| | | | - Bruce Wolfe
- Departments of Surgery and Medicine, Oregon Health & Science University, Portland, OR
| | - Jonathan Q Purnell
- Departments of Surgery and Medicine, Oregon Health & Science University, Portland, OR
| | - Xavier Pi-Sunyer
- New York Obesity Research Center, Division of Endocrinology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Christopher B. Newgard
- Duke Molecular Physiology Institute
- Sarah W. Stedman Nutrition and Metabolism Center
- Department of Pharmacology & Cancer Biology and Division of Endocrinology, Department of Medicine, Duke University, Durham, NC
| | - Svati H. Shah
- Duke Molecular Physiology Institute
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Blandine Laferrère
- New York Obesity Research Center, Division of Endocrinology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | | |
Collapse
|
11
|
Heinonen S, Saarinen T, Meriläinen S, Sammalkorpi H, Penttilä AK, Koivikko M, Siira P, Karppinen J, Säiläkivi U, Rosengård-Bärlund M, Koivukangas V, Pietiläinen KH, Juuti A. Roux-en-Y versus one-anastomosis gastric bypass (RYSA study): weight loss, metabolic improvements, and nutrition at 1 year after surgery, a multicenter randomized controlled trial. Obesity (Silver Spring) 2023; 31:2909-2923. [PMID: 37987183 DOI: 10.1002/oby.23852] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 11/22/2023]
Abstract
OBJECTIVE Although it has been suggested that one-anastomosis gastric bypass (OAGB) is metabolically superior to the "gold standard," i.e., Roux-en-Y gastric bypass (RYGB), there is little robust evidence to prove it. Because this result may arise from the typically longer length of bypassed intestine in OAGB, here, the authors standardized the bypass length in RYGB and OAGB and compared weight loss and metabolic outcomes in a randomized controlled trial. METHODS The authors randomized 121 bariatric patients to RYGB (n = 61) or OAGB (n = 60) in two Finnish University Hospitals and measured weight; body composition; metabolic features (insulin sensitivity, lipids, inflammation, nutrition); and comorbidities before and 6 and 12 months after the operation. RESULTS Total weight loss was similar in RYGB and OAGB at 6 months (mean: 21.2% [95% CI: 19.4-23.0] vs. 22.8% [95% CI: 21.5-24.1], p = 0.136) and 12 months (25.4% [95% CI: 23.4-27.5] vs. 26.1% [95% CI: 24.2-28.9], p = 0.635). Insulin sensitivity, lipids, and inflammation improved similarly between the groups (p > 0.05). Remission of type 2 diabetes and hypercholesterolemia was marked and similar (p > 0.05) but the use of antihypertensive medications was lower (p = 0.037) and hypertension tended to improve more (p = 0.053) with RYGB versus OAGB at 12 months. Higher rates of vitamin D-25 deficiency (p < 0.05) and lower D-25 levels were observed with OAGB versus RYGB throughout the follow-up (p < 0.001). No differences in adverse effects were observed. CONCLUSIONS RYGB and OAGB were comparable in weight loss, metabolic improvement, remission of diabetes and hypercholesterolemia, and nutrition at 1-year follow-up. Vitamin D-25 deficiency was more prevalent with OAGB, whereas reduction in antihypertensive medications and hypertension was greater with RYGB. There is no need to change the current practices of RYGB in favor of OAGB.
Collapse
Affiliation(s)
- Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Departement of Internal Medicine, Helsinki University Hospital, Helsinki, Finland
| | - Tuure Saarinen
- Department of Gastrointestinal Surgery, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Sanna Meriläinen
- Abdominal Center, Oulu University Hospital, Medical Research Center Oulu, University of Oulu, Oulu, Finland
| | - Henna Sammalkorpi
- Department of Gastrointestinal Surgery, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Anne K Penttilä
- Department of Gastrointestinal Surgery, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Minna Koivikko
- Center of Internal Medicine, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Pertti Siira
- Department of Physical Medicine and Rehabilitation, Oulu University Hospital, Oulu, Finland
| | - Jaro Karppinen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Ulla Säiläkivi
- Department of Gastrointestinal Surgery, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Milla Rosengård-Bärlund
- HealthyWeightHub, Endocrinology, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Vesa Koivukangas
- Abdominal Center, Oulu University Hospital, Medical Research Center Oulu, University of Oulu, Oulu, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- HealthyWeightHub, Endocrinology, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Anne Juuti
- Department of Gastrointestinal Surgery, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
12
|
Al‐Alsheikh AS, Alabdulkader S, Miras AD, Goldstone AP. Effects of bariatric surgery and dietary interventions for obesity on brain neurotransmitter systems and metabolism: A systematic review of positron emission tomography (PET) and single-photon emission computed tomography (SPECT) studies. Obes Rev 2023; 24:e13620. [PMID: 37699864 PMCID: PMC10909448 DOI: 10.1111/obr.13620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 04/05/2023] [Accepted: 07/10/2023] [Indexed: 09/14/2023]
Abstract
This systematic review collates studies of dietary or bariatric surgery interventions for obesity using positron emission tomography and single-photon emission computed tomography. Of 604 publications identified, 22 met inclusion criteria. Twelve studies assessed bariatric surgery (seven gastric bypass, five gastric bypass/sleeve gastrectomy), and ten dietary interventions (six low-calorie diet, three very low-calorie diet, one prolonged fasting). Thirteen studies examined neurotransmitter systems (six used tracers for dopamine DRD2/3 receptors: two each for 11 C-raclopride, 18 F-fallypride, 123 I-IBZM; one for dopamine transporter, 123 I-FP-CIT; one used tracer for serotonin 5-HT2A receptor, 18 F-altanserin; two used tracers for serotonin transporter, 11 C-DASB or 123 I-FP-CIT; two used tracer for μ-opioid receptor, 11 C-carfentanil; one used tracer for noradrenaline transporter, 11 C-MRB); seven studies assessed glucose uptake using 18 F-fluorodeoxyglucose; four studies assessed regional cerebral blood flow using 15 O-H2 O (one study also used arterial spin labeling); and two studies measured fatty acid uptake using 18 F-FTHA and one using 11 C-palmitate. The review summarizes findings and correlations with clinical outcomes, eating behavior, and mechanistic mediators. The small number of studies using each tracer and intervention, lack of dietary intervention control groups in any surgical studies, heterogeneity in time since intervention and degree of weight loss, and small sample sizes hindered the drawing of robust conclusions across studies.
Collapse
Affiliation(s)
- Alhanouf S. Al‐Alsheikh
- Department of Metabolism, Digestion and Reproduction, Imperial College LondonHammersmith HospitalLondonUK
- Department of Community Health Sciences, College of Applied Medical SciencesKing Saud UniversityRiyadhSaudi Arabia
| | - Shahd Alabdulkader
- Department of Metabolism, Digestion and Reproduction, Imperial College LondonHammersmith HospitalLondonUK
- Department of Health Sciences, College of Health and Rehabilitation SciencesPrincess Nourah Bint Abdulrahman UniversityRiyadhSaudi Arabia
| | - Alexander D. Miras
- Department of Metabolism, Digestion and Reproduction, Imperial College LondonHammersmith HospitalLondonUK
- School of Medicine, Faculty of Life and Health SciencesUlster UniversityLondonderryUK
| | - Anthony P. Goldstone
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College LondonHammersmith HospitalLondonUK
| |
Collapse
|
13
|
Salehi M, Peterson R, Tripathy D, Pezzica S, DeFronzo R, Gastaldelli A. Differential effect of gastric bypass versus sleeve gastrectomy on insulinotropic action of endogenous incretins. Obesity (Silver Spring) 2023; 31:2774-2785. [PMID: 37853989 PMCID: PMC10593483 DOI: 10.1002/oby.23872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/09/2023] [Accepted: 06/26/2023] [Indexed: 10/20/2023]
Abstract
OBJECTIVE Prandial hyperinsulinemia after Roux-en-Y gastric bypass surgery (GB), and to lesser degree after sleeve gastrectomy (SG), has been attributed to rapid glucose flux from the gut and increased insulinotropic gut hormones. However, β-cell sensitivity to exogenous incretin is reduced after GB. This study examines the effect of GB versus SG on prandial glycemia and β-cell response to increasing concentrations of endogenous incretins. METHODS Glucose kinetics, insulin secretion rate (ISR), and incretin responses to 50-g oral glucose ingestion were compared between ten nondiabetic participants with GB versus nine matched individuals with SG and seven nonoperated normal glucose tolerant control individuals (CN) with and without administration of 200 mg of sitagliptin. RESULTS Fasting glucose and hormonal levels were similar among three groups. Increasing plasma concentrations of endogenous incretins by two- to three-fold diminished prandial glycemia and increased β-cell secretion in all three groups (p < 0.05), but insulin secretion per insulin sensitivity (i.e., disposition index) was increased only in GB (p < 0.05 for interaction). However, plot of the slope of ISR (from premeal to peak values) versus plasma glucagon-like peptide-1 concentration was smaller after GB compared with SG and CN. CONCLUSIONS After GB, increasing incretin activity augments prandial β-cell response whereas the β-cell sensitivity to increasing plasma concentrations of endogenous incretin is diminished.
Collapse
Affiliation(s)
- Marzieh Salehi
- Division of Diabetes, University of Texas at San Antonio, San Antonio, TX, United States
- STVHCS, Audie Murphy Hospital, San Antonio, TX, United States
| | - Richard Peterson
- Department of Surgery, University of Texas at San Antonio, San Antonio, TX, United States
| | - Devjit Tripathy
- Division of Diabetes, University of Texas at San Antonio, San Antonio, TX, United States
| | - Samantha Pezzica
- Cardiometabolic Risk Unit, CNR Institute of Clinical Physiology, Pisa, Italy
| | - Ralph DeFronzo
- Division of Diabetes, University of Texas at San Antonio, San Antonio, TX, United States
| | - Amalia Gastaldelli
- Division of Diabetes, University of Texas at San Antonio, San Antonio, TX, United States
- Cardiometabolic Risk Unit, CNR Institute of Clinical Physiology, Pisa, Italy
| |
Collapse
|
14
|
Chang YC, Hsu CN, Chong K, Yang PJ, Ser KH, Lee PC, Chen SC, Hsuan CF, Lee YC, Hsu CC, Lee HL, Liao KCW, Hsieh ML, Chuang GT, Yang WS, Chu SL, Li WY, Chuang LM, Lee WJ. Roux-en-Y and One-Anastomosis Gastric Bypass Surgery Are Superior to Sleeve Gastrectomy in Lowering Glucose and Cholesterol Levels Independent of Weight Loss: a Propensity-Score Weighting Analysis. Obes Surg 2023; 33:3035-3050. [PMID: 37612578 DOI: 10.1007/s11695-023-06656-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND The superior effects of gastric bypass surgery in preventing cardiovascular diseases compared with sleeve gastrectomy are well-established. However, whether these effects are independent of weight loss is not known. METHODS In this retrospective cohort study, we compared the change in cardiometabolic risks of 1073 diabetic patients undergoing Roux-en-Y gastric bypass (RYGB) (n = 265), one-anastomosis gastric bypass (OAGB) (n = 619), and sleeve gastrectomy (SG) (n = 189) with equivalent weight loss from the Min-Shen General Hospital. Propensity score-weighting, multivariate regression, and matching were performed to adjust for baseline differences. RESULTS After 12 months, OAGB and, to a lesser extent, RYGB exhibited superior effects on glycemic control compared with SG in patients with equivalent weight loss. The effect was significant in patients with mild-to-modest BMI reduction but diminished in patients with severe BMI reduction. RYGB and OAGB had significantly greater effects in lowering total and low-density lipoprotein cholesterol than SG, regardless of weight loss. The results of matching patients with equivalent weight loss yielded similar results. The longer length of bypassed biliopancreatic (BP) limbs was correlated with a greater decrease in glycemic levels, insulin resistance index, lipids, C-reactive protein (CRP) levels, and creatinine levels in patients receiving RYBG. It was correlated with greater decreases in BMI, fasting insulin, insulin resistance index, and C-reactive protein levels in patients receiving OAGB. CONCLUSION Diabetic patients receiving OAGB and RYGB had lower glucose and cholesterol levels compared with SG independent of weight loss. Our results suggest diabetic patients with cardiovascular risk factors such as hypercholesterolemia to receive bypass surgery.
Collapse
Affiliation(s)
- Yi-Cheng Chang
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, 100, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Center for Obesity, Lifestyle, and Metabolic Surgery, National Taiwan University Hospital, Taipei, 100, Taiwan
- Graduate Institute of Molecular Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Chih-Neng Hsu
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, 640, Taiwan
| | - Keong Chong
- Department of Medicine, Min-Sheng General Hospital, Taoyuan, 330, Taiwan
| | - Po-Jen Yang
- Center for Obesity, Lifestyle, and Metabolic Surgery, National Taiwan University Hospital, Taipei, 100, Taiwan
- Department of Surgery, National Taiwan University Hospital, Taipei, 100, Taiwan
- Department of Surgery, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Kong-Han Ser
- Department of Surgery, Ten-Chen General Hospital, Taoyuan, 326, Taiwan
| | - Po-Chu Lee
- Department of Surgery, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Shu-Chun Chen
- Department of Nursing, Ming-Sheng General Hospital, Taoyuan, 330, Taiwan
| | - Chin-Feng Hsuan
- Department of Internal Medicine, Division of Cardiology, E-Da Hospital, Kaohsiung, 824, Taiwan
- Department of Internal Medicine, Division of Cardiology, E-Da Dachang Hospital, Kaohsiung, 824, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, 840, Taiwan
| | - Yi-Chih Lee
- Department of International Business, Chien Hsin University of Science and Technology, Taoyuan, 320, Taiwan
| | - Chih-Cheng Hsu
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, 350, Taiwan
| | - Hsiao-Lin Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Karen Chia-Wen Liao
- Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Meng-Lun Hsieh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Gwo-Tsann Chuang
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, 100, Taiwan
- Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Wei-Shun Yang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, 302, Taiwan
| | - Shao-Lun Chu
- Department of Medicine, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Wen-Yi Li
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, 640, Taiwan
| | - Lee-Ming Chuang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan.
- Graduate Institute of Molecular Medicine, National Taiwan University, Taipei, 100, Taiwan.
- Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, 100, Taiwan.
| | - Wei-Jei Lee
- Department of Medicine, Min-Sheng General Hospital, Taoyuan, 330, Taiwan.
- Department of Surgery, Min-Sheng General Hospital, Taoyuan, 330, Taiwan.
| |
Collapse
|
15
|
Yin M, Wang Y, Han M, Liang R, Li S, Wang G, Gang X. Mechanisms of bariatric surgery for weight loss and diabetes remission. J Diabetes 2023; 15:736-752. [PMID: 37442561 PMCID: PMC10509523 DOI: 10.1111/1753-0407.13443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/12/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Obesity and type 2 diabetes(T2D) lead to defects in intestinal hormones secretion, abnormalities in the composition of bile acids (BAs), increased systemic and adipose tissue inflammation, defects of branched-chain amino acids (BCAAs) catabolism, and dysbiosis of gut microbiota. Bariatric surgery (BS) has been shown to be highly effective in the treatment of obesity and T2D, which allows us to view BS not simply as weight-loss surgery but as a means of alleviating obesity and its comorbidities, especially T2D. In recent years, accumulating studies have focused on the mechanisms of BS to find out which metabolic parameters are affected by BS through which pathways, such as which hormones and inflammatory processes are altered. The literatures are saturated with the role of intestinal hormones and the gut-brain axis formed by their interaction with neural networks in the remission of obesity and T2D following BS. In addition, BAs, gut microbiota and other factors are also involved in these benefits after BS. The interaction of these factors makes the mechanisms of metabolic improvement induced by BS more complicated. To date, we do not fully understand the exact mechanisms of the metabolic alterations induced by BS and its impact on the disease process of T2D itself. This review summarizes the changes of intestinal hormones, BAs, BCAAs, gut microbiota, signaling proteins, growth differentiation factor 15, exosomes, adipose tissue, brain function, and food preferences after BS, so as to fully understand the actual working mechanisms of BS and provide nonsurgical therapeutic strategies for obesity and T2D.
Collapse
Affiliation(s)
- Mengsha Yin
- Department of Endocrinology and MetabolismThe First Hospital of Jilin UniversityChangchunChina
| | - Yao Wang
- Department of OrthopedicsThe Second Hospital Jilin UniversityChangchunChina
| | - Mingyue Han
- Department of Endocrinology and MetabolismThe First Hospital of Jilin UniversityChangchunChina
| | - Ruishuang Liang
- Department of Endocrinology and MetabolismThe First Hospital of Jilin UniversityChangchunChina
| | - Shanshan Li
- Department of Endocrinology and MetabolismThe First Hospital of Jilin UniversityChangchunChina
| | - Guixia Wang
- Department of Endocrinology and MetabolismThe First Hospital of Jilin UniversityChangchunChina
| | - Xiaokun Gang
- Department of Endocrinology and MetabolismThe First Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
16
|
Kim ER, Yun JH, Kim HJ, Park HY, Heo Y, Park YS, Park DJ, Koo SK. Evaluation of hormonal and circulating inflammatory biomarker profiles in the year following bariatric surgery. Front Endocrinol (Lausanne) 2023; 14:1171675. [PMID: 37564975 PMCID: PMC10411526 DOI: 10.3389/fendo.2023.1171675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/06/2023] [Indexed: 08/12/2023] Open
Abstract
Background Bariatric surgery (BS) has a superior effect on reducing body weight and fat in patients with morbid obesity. As a result, BS mitigates obesity-related complications such as type 2 diabetes (T2D). However, few studies have shown the mechanism underlying diabetes remission after surgery. This study aimed to investigate the differences in serum hormone and inflammatory cytokine levels related to diabetes before surgery and during 12 months of follow-up in Korean patients with obesity. Methods The study participants were patients with morbid obesity (n=63) who underwent sleeve gastrectomy (SG) or Roux-en-Y gastric bypass (RYGB) between 2016 - 2017 at seven tertiary hospitals in Korea. The patients were followed for 1 year after surgery. Results Sixty-three patients had significant weight loss after surgery and showed improvements in clinical parameters and hormonal and inflammatory profiles. Among them, 23 patients who were diabetic preoperatively showed different remission after surgery. The levels of inflammation-related clinical parameters changed significantly in the remission group, and serum inflammatory cytokine and hormones significantly decreased at certain points and showed an overall decreasing trend. Conclusions Our study found postoperative changes of factors in blood samples, and the changes in hormones secreted from the three major metabolic tissue (pancreas, adipose, and gut) along with the differences in multi-origin inflammatory cytokines between remission and non-remission groups provide a path for understanding how the effect of BS in improving glucose metabolism is mediated.
Collapse
Affiliation(s)
- Eun Ran Kim
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence, Korea National Institute of Health, Cheongju-si, Chungbuk, Republic of Korea
| | - Ji Ho Yun
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence, Korea National Institute of Health, Cheongju-si, Chungbuk, Republic of Korea
| | - Hyo-Jin Kim
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence, Korea National Institute of Health, Cheongju-si, Chungbuk, Republic of Korea
| | - Hyeon Young Park
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence, Korea National Institute of Health, Cheongju-si, Chungbuk, Republic of Korea
| | - Yoonseok Heo
- Department of Surgery, School of Medicine, Inha University, Incheon, Republic of Korea
| | - Young Suk Park
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Do Joong Park
- Department of Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Soo Kyung Koo
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence, Korea National Institute of Health, Cheongju-si, Chungbuk, Republic of Korea
| |
Collapse
|
17
|
Abstract
Many people with obesity and type 2 diabetes achieve remission of their diabetes after Roux-en-Y Gastric Bypass Surgery but the mechanisms remain disputed. We provide our perspective on competing data sets that either point towards this effect being entirely due to the loss of weight, or due to weight loss-independent effects.
Collapse
Affiliation(s)
- Samuel Klein
- Center for Human Nutrition, Washington University, St. Louis, Missouri, USA
- Sansum Diabetes Research Institute, Santa Barbara, CA, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Shah H, Kramer A, Mullins CA, Mattern M, Gannaban RB, Townsend RL, Campagna SR, Morrison CD, Berthoud HR, Shin AC. Reduction of Plasma BCAAs following Roux-en-Y Gastric Bypass Surgery Is Primarily Mediated by FGF21. Nutrients 2023; 15:1713. [PMID: 37049555 PMCID: PMC10096671 DOI: 10.3390/nu15071713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/26/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023] Open
Abstract
Type 2 diabetes (T2D) is a challenging health concern worldwide. A lifestyle intervention to treat T2D is difficult to adhere, and the effectiveness of approved medications such as metformin, thiazolidinediones (TZDs), and sulfonylureas are suboptimal. On the other hand, bariatric procedures such as Roux-en-Y gastric bypass (RYGB) are being recognized for their remarkable ability to achieve diabetes remission, although the underlying mechanism is not clear. Recent evidence points to branched-chain amino acids (BCAAs) as a potential contributor to glucose impairment and insulin resistance. RYGB has been shown to effectively lower plasma BCAAs in insulin-resistant or T2D patients that may help improve glycemic control, but the underlying mechanism for BCAA reduction is not understood. Hence, we attempted to explore the mechanism by which RYGB reduces BCAAs. To this end, we randomized diet-induced obese (DIO) mice into three groups that underwent either sham or RYGB surgery or food restriction to match the weight of RYGB mice. We also included regular chow-diet-fed healthy mice as an additional control group. Here, we show that compared to sham surgery, RYGB in DIO mice markedly lowered serum BCAAs most likely by rescuing BCAA breakdown in both liver and white adipose tissues. Importantly, the restored BCAA metabolism following RYGB was independent of caloric intake. Fasting insulin and HOMA-IR were decreased as expected, and serum valine was strongly associated with insulin resistance. While gut hormones such as glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) are postulated to mediate various surgery-induced metabolic benefits, mice lacking these hormonal signals (GLP-1R/Y2R double KO) were still able to effectively lower plasma BCAAs and improve glucose tolerance, similar to mice with intact GLP-1 and PYY signaling. On the other hand, mice deficient in fibroblast growth factor 21 (FGF21), another candidate hormone implicated in enhanced glucoregulatory action following RYGB, failed to decrease plasma BCAAs and normalize hepatic BCAA degradation following surgery. This is the first study using an animal model to successfully recapitulate the RYGB-led reduction of circulating BCAAs observed in humans. Our findings unmasked a critical role of FGF21 in mediating the rescue of BCAA metabolism following surgery. It would be interesting to explore the possibility of whether RYGB-induced improvement in glucose homeostasis is partly through decreased BCAAs.
Collapse
Affiliation(s)
- Harsh Shah
- Neurobiology of Nutrition Laboratory, Department of Nutritional Sciences, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Alyssa Kramer
- Neurobiology of Nutrition Laboratory, Department of Nutritional Sciences, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Caitlyn A. Mullins
- Neurobiology of Nutrition Laboratory, Department of Nutritional Sciences, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Marie Mattern
- Neurobiology of Nutrition Laboratory, Department of Nutritional Sciences, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Ritchel B. Gannaban
- Neurobiology of Nutrition Laboratory, Department of Nutritional Sciences, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - R. Leigh Townsend
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Shawn R. Campagna
- Department of Chemistry, University of Tennessee, Knoxville, TN 37996, USA
| | - Christopher D. Morrison
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Andrew C. Shin
- Neurobiology of Nutrition Laboratory, Department of Nutritional Sciences, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
19
|
Salehi M, Peterson R, Tripathy D, Pezzica S, DeFronzo R, Gastaldelli A. Insulinotropic effect of endogenous incretins is greater after gastric bypass than sleeve gastrectomy despite diminished beta-cell sensitivity to plasma incretins. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.28.23287755. [PMID: 37034666 PMCID: PMC10081422 DOI: 10.1101/2023.03.28.23287755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
BACKGROUND/AIMS Prandial hyperinsulinemia after Roux-en Y gastric bypass surgery (GB), and to lesser degree after sleeve gastrectomy (SG), has been attributed to rapid glucose flux from the gut and increased insulinotropic gut hormones. However, β-cell sensitivity to exogenous incretin is markedly reduced after GB. This study examines the effect of GB versus SG on prandial glycemia and β-cell response to increasing concentrations of endogenous incretins. METHODS Glucose kinetics, insulin secretion rate (ISR), and incretin responses to 50-gram oral glucose ingestion were compared between 10 non-diabetic subjects with GB versus 9 matched individuals with SG and 7 non-operated normal glucose tolerant controls (CN) on two days with and without administration of 200 mg sitagliptin. RESULTS Fasting glucose and hormonal levels were similar among 3 groups. Increasing plasma concentrations of endogenous incretins by 2-3-fold diminished post-OGTT glycemia and increased β-cell secretion in all 3 groups (p<0.05), but insulin secretion per insulin sensitivity (i.e., disposition index) was increased only in GB (p<0.05 for interaction). As a result, sitagliptin administration led to hypoglycemia in 3 of 10 GB. Yet, plot of the slope of ISR versus the increase in endogenous incretin concentration was smaller after GB compared to both SG and CN. CONCLUSION Augmented glycemic-induced β-cell response caused by enhanced incretin activity is unique to GB and not shared with SG. However, the β-cell sensitivity to increasing concentrations of endogenous incretin is smaller after bariatric surgery, particularly after GB, compared to non-operated controls, indicating a long-term adaptation of gut-pancreas axis after these procedures. HIGHLIGHTS What is known?: Glycemic effects of gastric bypass (GB) and sleeve gastrectomy (SG) is attributed to rapid nutrient flux and enhanced insulinotropic effects of gut hormones but β-cell sensitivity to exogenous GLP-1 or GIP is diminished after GB. What the present findings add?: Post-OGTT β-cell sensitivity to enhanced endogenous incretins by DPP4i is markedly reduced in bariatric subjects versus non-operated controls, and yet insulin secretory response (disposition index) is increased leading to hypoglycemia in GB and not SG. Significance?: Blunted sensitivity to GLP-1 may represent β-cell adaptation to massive elevation in GLP-1 secretion following bariatric surgery to protect against hypoglycemia.The differential effect of enhanced concentrations of incretins on post-OGTT insulin response (disposition index) among GB versus SG highlights a distinct adaptive process among the two procedures.Augmented insulinotropic effects of gut hormones on postprandial insulin secretory response after GB despite a reduced beta-cell sensitivity to plasma concentrations of GLP-1 makes a case for non-hormonal mechanisms of GLP-1 action after GB.Better understanding of long-term effects of bariatric surgery on gut-pancreas axis activity is critical in development of GLP-1-based strategies to address glucose abnormalities (both hyperglycemia and hypoglycemia) in these settings.
Collapse
|
20
|
Albaugh VL, Axelrod C, Belmont KP, Kirwan JP. Physiology Reconfigured: How Does Bariatric Surgery Lead to Diabetes Remission? Endocrinol Metab Clin North Am 2023; 52:49-64. [PMID: 36754497 DOI: 10.1016/j.ecl.2022.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Bariatric surgery improves glucose homeostasis and glycemic control in patients with type 2 diabetes. Over the past 20 years, a breadth of studies has been conducted in humans and rodents aimed to identify the regulatory nodes responsible for surgical remission of type 2 diabetes. The review herein discusses central mechanisms of type 2 diabetes remission associated with weight loss and surgical modification of the gastrointestinal tract.
Collapse
Affiliation(s)
- Vance L Albaugh
- Metamor Institute, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA; Integrative Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Louisiana State University, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | - Christopher Axelrod
- Integrative Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Louisiana State University, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | - Kathryn P Belmont
- Integrative Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Louisiana State University, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | - John P Kirwan
- Integrative Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Louisiana State University, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| |
Collapse
|
21
|
Liu T, Zou X, Ruze R, Xu Q. Bariatric Surgery: Targeting pancreatic β cells to treat type II diabetes. Front Endocrinol (Lausanne) 2023; 14:1031610. [PMID: 36875493 PMCID: PMC9975540 DOI: 10.3389/fendo.2023.1031610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 01/19/2023] [Indexed: 02/17/2023] Open
Abstract
Pancreatic β-cell function impairment and insulin resistance are central to the development of obesity-related type 2 diabetes mellitus (T2DM). Bariatric surgery (BS) is a practical treatment approach to treat morbid obesity and achieve lasting T2DM remission. Traditionally, sustained postoperative glycemic control was considered a direct result of decreased nutrient intake and weight loss. However, mounting evidence in recent years implicated a weight-independent mechanism that involves pancreatic islet reconstruction and improved β-cell function. In this article, we summarize the role of β-cell in the pathogenesis of T2DM, review recent research progress focusing on the impact of Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG) on pancreatic β-cell pathophysiology, and finally discuss therapeutics that have the potential to assist in the treatment effect of surgery and prevent T2D relapse.
Collapse
Affiliation(s)
- Tiantong Liu
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
- School of Medicine, Tsinghua University, Beijing, China
| | - Xi Zou
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rexiati Ruze
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiang Xu
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
22
|
Feris F, McRae A, Kellogg TA, McKenzie T, Ghanem O, Acosta A. Mucosal and hormonal adaptations after Roux-en-Y gastric bypass. Surg Obes Relat Dis 2023; 19:37-49. [PMID: 36243547 PMCID: PMC9797451 DOI: 10.1016/j.soard.2022.08.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 01/12/2023]
Abstract
The aim of this study was to perform a comprehensive literature review regarding the relevant hormonal and histologic changes observed after Roux-en-Y gastric bypass (RYGB). We aimed to describe the relevant hormonal (glucagon-like peptides 1 and 2 [GLP-1 and GLP-2], peptide YY [PYY], oxyntomodulin [OXM], bile acids [BA], cholecystokinin [CCK], ghrelin, glucagon, gastric inhibitory polypeptide [GIP], and amylin) profiles, as well as the histologic (mucosal cellular) adaptations happening after patients undergo RYGB. Our review compiles the current evidence and furthers the understanding of the rationale behind the food intake regulatory adaptations occurring after RYGB surgery. We identify gaps in the literature where the potential for future investigations and therapeutics may lie. We performed a comprehensive database search without language restrictions looking for RYGB bariatric surgery outcomes in patients with pre- and postoperative blood work hormonal profiling and/or gut mucosal biopsies. We gathered the relevant study results and describe them in this review. Where human findings were lacking, we included animal model studies. The amalgamation of physiologic, metabolic, and cellular adaptations following RYGB is yet to be fully characterized. This constitutes a fundamental aspiration for enhancing and individualizing obesity therapy.
Collapse
Affiliation(s)
- Fauzi Feris
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Alison McRae
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Todd A Kellogg
- Division of Endocrine and Metabolic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Travis McKenzie
- Division of Endocrine and Metabolic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Omar Ghanem
- Division of Endocrine and Metabolic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
23
|
Valadez LZM, Frigolet ME, Dominguez RM, Pescarus R, Zerrweck C, Boudreau V, Doumouras A, Cookson T, Anvari M. Metabolic and Bariatric Surgery in Diabetes Management. THE DIABETES TEXTBOOK 2023:673-690. [DOI: 10.1007/978-3-031-25519-9_42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
24
|
Caloric restriction improves glycaemic control without reducing plasma branched-chain amino acids or keto-acids in obese men. Sci Rep 2022; 12:19273. [PMID: 36369511 PMCID: PMC9652417 DOI: 10.1038/s41598-022-21814-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 10/04/2022] [Indexed: 11/13/2022] Open
Abstract
Higher plasma leucine, isoleucine and valine (BCAA) concentrations are associated with diabetes, obesity and insulin resistance (IR). Here, we evaluated the effects of 6-weeks very-low calorie diet (VLCD) upon fasting BCAA in overweight (OW) non-diabetic men, to explore associations between circulating BCAA and IR, before and after a weight loss intervention. Fasting plasma BCAAs were quantified in an OW (n = 26; BMI 32.4 ± 3 kg/m2; mean age 44 ± 9 y) and a normal-weight (NW) group (n = 26; BMI 24 ± 3.1 kg/m2; mean age 32 ± 12.3 y). Ten of the OW group (BMI 32.2 ± 4 kg/m2; 46 ± 8 y) then underwent 6-weeks of VLCD (600-800 kcal/day). Fasting plasma BCAA (gas chromatography-mass spectrometry), insulin sensitivity (HOMA-IR) and body-composition (DXA) were assessed before and after VLCD. Total BCAA were higher in OW individuals (sum leucine/isoleucine/valine: 457 ± 85 µM) compared to NW control individuals (365 ± 78 µM, p < 0.001). Despite significant weight loss (baseline 103.9 ± 12.3 to 93 ± 9.6 kg and BMI 32.2 ± 4 to 28.9 ± 3.6 kg/m2), no changes were observed in BCAAs after 6-weeks of VLCD. Moreover, although VLCD resulted in a significant reduction in HOMA-IR (baseline 1.19 ± 0.62 to 0.51 ± 0.21 post-VLCD; p < 0.001), Pearson's r revealed no relationships between BCAA and HOMA-IR, either before (leucine R2: 2.49e-005, p = 0.98; isoleucine R2: 1.211-e006, p = 0.9; valine R2: 0.004, p = 0.85) or after VLCD (leucine R2: 0.003, p = 0.86; isoleucine R2: 0.006, p = 0.82; valine R2: 0.002, p = 0.65). Plasma BCAA are higher in OW compared to NW individuals. However, while 6-weeks VLCD reduced body weight and IR in OW individuals, this was not associated with reductions in BCAA. This suggests that studies demonstrating links between BCAA and insulin resistance in OW individuals, are complex and are not normalised by simply losing weight.
Collapse
|
25
|
Barrea L, Salzano C, Pugliese G, Laudisio D, Frias-Toral E, Savastano S, Colao A, Muscogiuri G. The challenge of weight loss maintenance in obesity: a review of the evidence on the best strategies available. Int J Food Sci Nutr 2022; 73:1030-1046. [PMID: 36245260 DOI: 10.1080/09637486.2022.2130186] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Long-term weight loss maintenance represents a big challenge for the management of obesity. This narrative review aims to provide an overview of the main endocrine mechanisms involved in weight regain in subjects with obesity and to review the current evidence on the best lifestyle approaches, including diet and physical activity. Weight regain after weight loss occurs in about 50% of subjects with obesity in the absence of lifestyle changes. The primary endocrine mechanism responsible for weight regain involves the brain-gut axis, which encourages food intake and thus weight regain through the secretion and action of several gastrointestinal hormones, such as ghrelin, leptin and cholecystokinin. Several evidence reported changes of secretion of these hormones during weight loss and weight loss maintenance programs. Endurance training is the most effective physical activity to lose and keep weight loss; the association of endurance with resistance training is recommended for remodelling body shape.
Collapse
Affiliation(s)
- Luigi Barrea
- Dipartimento di Scienze Umanistiche, Università Telematica Pegaso, Napoli, Italy.,Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy
| | - Ciro Salzano
- Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy
| | - Gabriella Pugliese
- Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy.,Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Federico II University Medical School of Naples, Naples, Italy
| | - Daniela Laudisio
- Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy.,Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Federico II University Medical School of Naples, Naples, Italy
| | - Evelyn Frias-Toral
- Clinical Research Associate Professor for Palliative Care Residency from Universidad Católica Santiago de Guayaquil, Guayaquil, Ecuador
| | - Silvia Savastano
- Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy.,Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Federico II University Medical School of Naples, Naples, Italy
| | - Annamaria Colao
- Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy.,Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Federico II University Medical School of Naples, Naples, Italy.,Cattedra Unesco "Educazione alla salute e allo sviluppo sostenibile", University Federico II, Naples, Italy
| | - Giovanna Muscogiuri
- Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy.,Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Federico II University Medical School of Naples, Naples, Italy.,Cattedra Unesco "Educazione alla salute e allo sviluppo sostenibile", University Federico II, Naples, Italy
| |
Collapse
|
26
|
Sridhar A, Khan D, Abdelaal M, Elliott JA, Naughton V, Flatt PR, Le Roux CW, Docherty NG, Moffett CR. Differential effects of RYGB surgery and best medical treatment for obesity-diabetes on intestinal and islet adaptations in obese-diabetic ZDSD rats. PLoS One 2022; 17:e0274788. [PMID: 36137097 PMCID: PMC9499270 DOI: 10.1371/journal.pone.0274788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022] Open
Abstract
Modification of gut-islet secretions after Roux-En-Y gastric bypass (RYBG) surgery contributes to its metabolic and anti-diabetic benefits. However, there is limited knowledge on tissue-specific hormone distribution post-RYGB surgery and how this compares with best medical treatment (BMT). In the present study, pancreatic and ileal tissues were excised from male Zucker-Diabetic Sprague Dawley (ZDSD) rats 8-weeks after RYGB, BMT (daily oral dosing with metformin 300mg/kg, fenofibrate 100mg/kg, ramipril 1mg/kg, rosuvastatin 10mg/kg and subcutaneous liraglutide 0.2mg/kg) or sham operation (laparotomy). Insulin, glucagon, somatostatin, PYY, GLP-1 and GIP expression patterns were assessed using immunocytochemistry and analyzed using ImageJ. After RYGB and BMT, body weight and plasma glucose were decreased. Intestinal morphometry was unaltered by RYGB, but crypt depth was decreased by BMT. Intestinal PYY cells were increased by both interventions. GLP-1- and GIP-cell counts were unchanged by RYGB but BMT increased ileal GLP-1-cells and decreased those expressing GIP. The intestinal contents of PYY and GLP-1 were significantly enhanced by RYGB, whereas BMT decreased ileal GLP-1. No changes of islet and beta-cell area or proliferation were observed, but the extent of beta-cell apoptosis and islet integrity calculated using circularity index were improved by both treatments. Significantly decreased islet alpha-cell areas were observed in both groups, while beta- and PYY-cell areas were unchanged. RYGB also induced a decrease in islet delta-cell area. PYY and GLP-1 colocalization with glucagon in islets was significantly decreased in both groups, while co-staining of PYY with glucagon was decreased and that with somatostatin increased. These data characterize significant cellular islet and intestinal adaptations following RYGB and BMT associated with amelioration of obesity-diabetes in ZDSD rats. The differential responses observed and particularly those within islets, may provide important clues to the unique ability of RYGB to cause diabetes remission.
Collapse
Affiliation(s)
- Ananyaa Sridhar
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Dawood Khan
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
- * E-mail:
| | - Mahmoud Abdelaal
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Jessie A. Elliott
- Department of Surgery, Trinity Centre for Health Sciences and St. James’s Hospital, Dublin, Ireland
| | - Violetta Naughton
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Peter R. Flatt
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Carel W. Le Roux
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Neil G. Docherty
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Charlotte R. Moffett
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| |
Collapse
|
27
|
Salehi M, DeFronzo R, Gastaldelli A. Altered Insulin Clearance after Gastric Bypass and Sleeve Gastrectomy in the Fasting and Prandial Conditions. Int J Mol Sci 2022; 23:ijms23147667. [PMID: 35887007 PMCID: PMC9324232 DOI: 10.3390/ijms23147667] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/01/2022] [Accepted: 07/03/2022] [Indexed: 11/17/2022] Open
Abstract
Background: The liver has the capacity to regulate glucose metabolism by altering the insulin clearance rate (ICR). The decreased fasting insulin concentrations and enhanced prandial hyperinsulinemia after Roux-en-Y gastric-bypass (GB) surgery and sleeve gastrectomy (SG) are well documented. Here, we investigated the effect of GB or SG on insulin kinetics in the fasting and fed states. Method: ICR was measured (i) during a mixed-meal test (MMT) in obese non-diabetic GB (n = 9) and SG (n = 7) subjects and (ii) during a MMT combined with a hyperinsulinemic hypoglycemic clamp in the same GB and SG subjects. Five BMI-matched and non-diabetic subjects served as age-matched non-operated controls (CN). Results: The enhanced ICR during the fasting state after GB and SC compared with CN (p < 0.05) was mainly attributed to augmented hepatic insulin clearance rather than non-liver organs. The dose-response slope of the total insulin extraction rate (InsExt) of exogenous insulin per circulatory insulin value was greater in the GB and SG subjects than in the CN subjects, despite the similar peripheral insulin sensitivity among the three groups. Compared to the SG or the CN subjects, the GB subjects had greater prandial insulin secretion (ISR), independent of glycemic levels. The larger post-meal ISR following GB compared with SG was associated with a greater InsExt until it reached a plateau, leading to a similar reduction in meal-induced ICR among the GB and SG subjects. Conclusions: GB and SG alter ICR in the presence or absence of meal stimulus. Further, altered ICR after bariatric surgery results from changes in hepatic insulin clearance and not from a change in peripheral insulin sensitivity.
Collapse
Affiliation(s)
- Marzieh Salehi
- Division of Diabetes, University of Texas Health at San Antonio, San Antonio, TX 78229, USA;
- South Texas Veteran Health Care System, Audie Murphy Hospital, San Antonio, TX 78229, USA
- Correspondence: (M.S.); (A.G.); Tel.: +1-(210)-450-8560 (M.S.)
| | - Ralph DeFronzo
- Division of Diabetes, University of Texas Health at San Antonio, San Antonio, TX 78229, USA;
| | - Amalia Gastaldelli
- Division of Diabetes, University of Texas Health at San Antonio, San Antonio, TX 78229, USA;
- Cardiometabolic Risk Unit, CNR Institute of Clinical Physiology, 56124 Pisa, Italy
- Correspondence: (M.S.); (A.G.); Tel.: +1-(210)-450-8560 (M.S.)
| |
Collapse
|
28
|
Grespan E, Guolo A, Muscelli E, Ferrannini E, Mari A. Loss of the Incretin Effect in Type 2 Diabetes: A Systematic Review and Meta-analysis. J Clin Endocrinol Metab 2022; 107:2092-2100. [PMID: 35397169 DOI: 10.1210/clinem/dgac213] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Loss of the incretin effect (IE) in type 2 diabetes (T2D) contributes to hyperglycemia and the mechanisms underlying this impairment are unclear. OBJECTIVE To quantify the IE impairment in T2D and to investigate the factors associated with it using a meta-analytic approach. METHODS PubMed, Scopus, and Web-of-Science were searched. Studies measuring IE by the gold-standard protocol employing an oral glucose tolerance test (OGTT) and an intravenous glucose infusion at matched glucose levels were selected. We extracted IE, sex, age, body mass index (BMI), and hemoglobin A1c, fasting values, and area under curve (AUC) of glucose, insulin, C-peptide, glucose-dependent insulinotropic peptide (GIP) and glucagon-like peptide 1 (GLP-1). In subjects with T2D, we also recorded T2D duration, age at diagnosis, and the percentage of subjects taking antidiabetic medications. RESULTS The IE weighted mean difference between subjects with T2D and those with normal glucose tolerance (NGT) was -27.3% (CI -36.5% to -18.1%; P < .001; I2 = 86.6%) and was affected by age (P < .005). By meta-regression of combined NGT and T2D data, IE was inversely associated with glucose tolerance (lower IE in T2D), BMI, and fasting GIP (P < .05). By meta-regression of T2D studies only, IE was associated with the OGTT glucose dose (P < .0001). IE from insulin was larger than IE from C-peptide (weighted mean difference 11.2%, CI 9.2-13.2%; P < .0001; I2 = 28.1%); the IE difference was inversely associated with glucose tolerance and fasting glucose. CONCLUSION The IE impairment in T2D vs NGT is consistent though considerably variable, age being a possible factor affecting the IE difference. Glucose tolerance, BMI, and fasting GIP are independently associated with IE; in subjects with T2D only, the OGTT dose is a significant covariate.
Collapse
Affiliation(s)
| | - Annamaria Guolo
- Department of Statistical Sciences, University of Padua, Padua, Italy
| | - Elza Muscelli
- Department of Internal Medicine, School of Medical Sciences, State University of Campinas, Campinas, Brazil
| | | | - Andrea Mari
- C.N.R. Institute of Neuroscience, Padua, Italy
| |
Collapse
|
29
|
Cheng A, Yeoh E, Moh A, Low S, Tan CH, Lam B, Sum CF, Subramaniam T, Lim SC. Roux-en-Y gastric bypass versus best medical treatment for type 2 diabetes mellitus in adults with body mass index between 27 and 32 kg/m 2: A 5-year randomized controlled trial. Diabetes Res Clin Pract 2022; 188:109900. [PMID: 35513159 DOI: 10.1016/j.diabres.2022.109900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 11/26/2022]
Abstract
AIMS To evaluate the effects of Roux-en-Y gastric bypass (RYGB) versus best medical treatment in Asians with type 2 diabetes mellitus (T2DM) and class I obesity. METHODS In this 5-year single-centre, open-label randomized controlled trial, participants were randomized to RYGB or medical treatment including newer classes of diabetes medications (ClinicalTrials.gov:NCT02041234). The primary endpoint was diabetes remission defined as HbA1c ≤ 6% (≤42 mmol/mol) and discontinuation of glucose-lowering medication at 12 months post-intervention and beyond. Glycaemia and weight changes were assessed. Continuous glucose monitoring was performed. RESULTS Of 28 subjects randomized, 26 were analyzed in the final cohort (14 medical, 12 RYGB; age:44 ± 10 years, 34.6% males, BMI:29.4 ± 1.6 kg/m2). At 12 months, 50% of RYGB subjects achieved diabetes remission; 83% stopped all glucose-lowering medications. By year 5, 42% were in remission. None attained diabetes remission in the medical group. Percentage declines in fasting plasma glucose, HbA1c and BMI were significantly greater in the RYGB arm (all P < 0.05). Early improvements in glycaemic variability and time in range were similar in both treatment arms. Hypoglycaemia and surgical complications were observed in some RYGB subjects. CONCLUSIONS Over 5 years, RYGB outperforms best medical treatment in glycemia and weight improvements for Asians with T2DM and class I obesity.
Collapse
Affiliation(s)
- Anton Cheng
- Department of General Surgery, Khoo Teck Puat Hospital, Singapore; Clinical Research Unit, Khoo Teck Puat Hospital, Singapore.
| | - Ester Yeoh
- Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore
| | - Angela Moh
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | - Serena Low
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore; Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore
| | - Chun Hai Tan
- Department of General Surgery, Khoo Teck Puat Hospital, Singapore; Integrated Centre for Obesity and Diabetes, Khoo Teck Puat Hospital, Singapore
| | - Benjamin Lam
- Integrated Centre for Obesity and Diabetes, Khoo Teck Puat Hospital, Singapore
| | - Chee Fang Sum
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore; Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore
| | - Tavintharan Subramaniam
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore; Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore
| | - Su Chi Lim
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore; Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore; Saw Swee Hock School of Public Health, National University Hospital, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
30
|
Gao Z, Yang J, Liang Y, Yang S, Zhang T, Gong Z, Li M. Changes in Gastric Inhibitory Polypeptide (GIP) After Roux-en-Y Gastric Bypass in Obese Patients: a Meta-analysis. Obes Surg 2022; 32:2706-2716. [PMID: 35597875 DOI: 10.1007/s11695-022-05959-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/25/2022] [Accepted: 02/03/2022] [Indexed: 01/19/2023]
Abstract
This meta-analysis aimed to evaluate changes in GIP after RYGB in obese patients. We searched PubMed, EMBASE, and CENTRAL for relevant studies from database inception through July 2021. Articles were eligible for inclusion if they reported pre-operative and post-operative fasting GIP levels. We found fasting GIP levels had a decreasing tendency. The decrease in fasting glucose and postprandial GIP levels was also observed. Subgroup analysis indicated diabetic subjects tended to have a more obvious fasting GIP reduction compared to non-diabetic individuals. Meta-regression showed that the amount of weight loss (% total body weight), gastric pouch volume, alimentary limb length, and biliopancreatic limb length were not related to fasting GIP decrease. Fasting GIP levels decreased significantly after RYGB in obese people, especially in diabetic patients.
Collapse
Affiliation(s)
- Zhiguang Gao
- Department of Gastrointestinal Surgery, The affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, 523320, China.
| | - Jingge Yang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yuzhi Liang
- Department of Medical Imaging, The affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, 523320, China
| | - Sen Yang
- Guangdong Medical University, Zhanjiang, Guangdong Province, China
| | - Tao Zhang
- Department of Gastrointestinal Surgery, The affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, 523320, China
| | - Zuyuan Gong
- Department of Gastrointestinal Surgery, The affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, 523320, China
| | - Min Li
- Department of Gastrointestinal Surgery, The affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, 523320, China
| |
Collapse
|
31
|
L’intestin un organe endocrine : de la physiologie aux implications thérapeutiques en nutrition. NUTR CLIN METAB 2022. [DOI: 10.1016/j.nupar.2021.12.179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
32
|
Jecht M. Änderungen der Betazellfunktion nach einer Magenbypassoperation. DIABETOLOGE 2022. [DOI: 10.1007/s11428-022-00877-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
33
|
Laparoscopic sleeve gastrectomy for morbid obesity improves gut microbiota balance, increases colonic mucosal-associated invariant T cells and decreases circulating regulatory T cells. Surg Endosc 2022; 36:7312-7324. [PMID: 35182212 DOI: 10.1007/s00464-022-09122-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 02/07/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Laparoscopic sleeve gastrectomy (LSG) for morbid obesity may improve gut microbiota balance and decrease chronic inflammation. This study examines the changes in gut microbiota and immune environment, including mucosal-associated invariant T cells (MAIT cells) and regulatory T cells (Treg cells) caused by LSG. METHODS Ten morbidly obese patients underwent LSG at our institution between December 2018 and March 2020. Flow cytometry for Th1/Th2/Th17 cells, Treg cells and MAIT cells in peripheral blood and colonic mucosa and 16S rRNA analysis of gut microbiota were performed preoperatively and then 12 months postoperatively. RESULTS Twelve months after LSG, the median percent total weight loss was 30.3% and the median percent excess weight loss was 66.9%. According to laboratory data, adiponectin increased, leptin decreased, and chronic inflammation improved after LSG. In the gut microbiota, Bacteroidetes and Fusobacteria increased after LSG, and indices of alpha diversity increased after LSG. In colonic mucosa, the frequency of MAIT cells increased after LSG. In peripheral blood, the frequency of Th1 cells and effector Treg cells decreased after LSG. CONCLUSIONS After LSG for morbid obesity, improvement in chronic inflammation in obesity is suggested by change in the constituent bacterial species, increase in the diversity of gut microbiota, increase in MAIT cells in the colonic mucosa, and decrease in effector Treg cells in the peripheral blood.
Collapse
|
34
|
Prasad M, Mark V, Ligon C, Dutia R, Nair N, Shah A, Laferrère B. Role of the Gut in the Temporal Changes of β-Cell Function After Gastric Bypass in Individuals With and Without Diabetes Remission. Diabetes Care 2022; 45:469-476. [PMID: 34857533 PMCID: PMC8914419 DOI: 10.2337/dc21-1270] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 11/10/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE The role of the gut in diabetes remission after Roux-en-Y gastric bypass (RYGB) is incompletely understood. We assessed the temporal change in insulin secretory capacity after RYGB, using oral and intravenous (IV) glucose, in individuals with type 2 diabetes. RESEARCH DESIGN AND METHODS Longitudinal, prospective measures of β-cell function were assessed after oral glucose intake and graded glucose infusion in individuals with severe obesity and diabetes studied at 0, 3 (n = 29), 12 (n = 24), and 24 (n = 20) months after RYGB. Data were collected between 2015 and 2019 in an academic clinical research center. RESULTS The decreases in body weight, fat mass, waist circumference, and insulin resistance after surgery (all P < 0.001 at 12 and 24 months) did not differ according to diabetes remission status. In contrast, both the magnitude and temporal changes in β-cell glucose sensitivity after oral glucose intake differed by remission status (P = 0.04): greater (6.5-fold; P < 0.01) and sustained in those in full remission, moderate and not sustained past 12 months in those with partial remission (3.3-fold; P < 0.001), and minimal in those not experiencing remission (2.7-fold; P = not significant). The improvement in β-cell function after IV glucose administration was not apparent until 12 months, significant only in those in full remission, and only ∼33% of that observed after oral glucose intake. Preintervention β-cell function and its change after surgery predicted remission; weight loss and insulin sensitivity did not. CONCLUSIONS Our data show the time course of changes in β-cell function after RYGB. The improvement in β-cell function after RYGB, but not changes in weight loss or insulin sensitivity, drives diabetes remission.
Collapse
Affiliation(s)
- Malini Prasad
- New York Nutrition Obesity Research Center, Columbia University Irving Medical Center, New York, NY
| | - Victoria Mark
- New York Nutrition Obesity Research Center, Columbia University Irving Medical Center, New York, NY
| | - Chanel Ligon
- Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Roxanne Dutia
- New York Nutrition Obesity Research Center, Columbia University Irving Medical Center, New York, NY
| | - Nandini Nair
- Division of Endocrinology, Columbia University Irving Medical Center, New York, NY
| | - Ankit Shah
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ
| | - Blandine Laferrère
- New York Nutrition Obesity Research Center, Columbia University Irving Medical Center, New York, NY.,Department of Medicine, Columbia University Irving Medical Center, New York, NY.,Division of Endocrinology, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
35
|
Sakran N, Graham Y, Pintar T, Yang W, Kassir R, Willigendael EM, Singhal R, Kooreman ZE, Ramnarain D, Mahawar K, Parmar C, Madhok B, Pouwels S. The many faces of diabetes. Is there a need for re-classification? A narrative review. BMC Endocr Disord 2022; 22:9. [PMID: 34991585 PMCID: PMC8740476 DOI: 10.1186/s12902-021-00927-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 12/22/2021] [Indexed: 12/13/2022] Open
Abstract
The alarming rise in the worldwide prevalence of obesity and associated type 2 diabetes mellitus (T2DM) have reached epidemic portions. Diabetes in its many forms and T2DM have different physiological backgrounds and are difficult to classify. Bariatric surgery (BS) is considered the most effective treatment for obesity in terms of weight loss and comorbidity resolution, improves diabetes, and has been proven superior to medical management for the treatment of diabetes. The term metabolic surgery (MS) describes bariatric surgical procedures used primarily to treat T2DM and related metabolic conditions. MS is the most effective means of obtaining substantial and durable weight loss in individuals with obesity. Originally, BS was used as an alternative weight-loss therapy for patients with severe obesity, but clinical data revealed its metabolic benefits in patients with T2DM. MS is more effective than lifestyle or medical management in achieving glycaemic control, sustained weight loss, and reducing diabetes comorbidities. New guidelines for T2DM expand the use of MS to patients with a lower body mass index.Evidence has shown that endocrine changes resulting from BS translate into metabolic benefits that improve the comorbid conditions associated with obesity, such as hypertension, dyslipidemia, and T2DM. Other changes include bacterial flora rearrangement, bile acids secretion, and adipose tissue effect.This review aims to examine the physiological mechanisms in diabetes, risks for complications, the effects of bariatric and metabolic surgery and will shed light on whether diabetes should be reclassified.
Collapse
Affiliation(s)
- Nasser Sakran
- Department of Surgery, Holy Family Hospital, Nazareth, Israel
- the Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Yitka Graham
- Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, UK
- Facultad de Psycologia, Universidad Anahuac Mexico, Mexico City, Mexico
| | - Tadeja Pintar
- Department of Abdominal Surgery, University Medical Center Ljubljana, Zaloška cesta, Ljubljana, Slovenia
| | - Wah Yang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, 613 Huangpu Avenue West, Guangzhou, Guangdong Province, China
| | - Radwan Kassir
- CHU Félix Guyon, Allée des Topazes, Saint-Denis, France
| | - Edith M Willigendael
- Department of Vascular Surgery, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Rishi Singhal
- Bariatric and Upper GI Unit, Birmingham Heartlands Hospital, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | - Zoë E Kooreman
- Department of Dermatology, Amphia Hospital, Breda, The Netherlands
| | - Dharmanand Ramnarain
- Department of Intensive Care Medicine, Elisabeth-Tweesteden Hospital, Tilburg, The Netherlands
| | - Kamal Mahawar
- Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, UK
- Bariatric Unit, South Tyneside and Sunderland NHS Foundation Trust, Sunderland, UK
| | - Chetan Parmar
- Department of Surgery, Whittington Health NHS Trust, London, UK
| | - Brijesh Madhok
- East Midlands Bariatric and Metabolic Institute, University Hospital of Derby and Burton NHS Foundation Trust, Burton, UK
| | - Sjaak Pouwels
- Department of Intensive Care Medicine, Elisabeth-Tweesteden Hospital, Tilburg, The Netherlands.
- Department of Intensive Care Medicine, ETZ Elisabeth, Hilvarenbeekseweg 60, P.O. Box 90151, 5000 LC, Tilburg, The Netherlands.
| |
Collapse
|
36
|
Bakbak E, Terenzi DC, Trac JZ, Teoh H, Quan A, Glazer SA, Rotstein OD, Al-Omran M, Verma S, Hess DA. Lessons from bariatric surgery: Can increased GLP-1 enhance vascular repair during cardiometabolic-based chronic disease? Rev Endocr Metab Disord 2021; 22:1171-1188. [PMID: 34228302 DOI: 10.1007/s11154-021-09669-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/23/2021] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes (T2D) and obesity represent entangled pandemics that accelerate the development of cardiovascular disease (CVD). Given the immense burden of CVD in society, non-invasive prevention and treatment strategies to promote cardiovascular health are desperately needed. During T2D and obesity, chronic dysglycemia and abnormal adiposity result in systemic oxidative stress and inflammation that deplete the vascular regenerative cell reservoir in the bone marrow that impairs blood vessel repair and exacerbates the penetrance of CVD co-morbidities. This novel translational paradigm, termed 'regenerative cell exhaustion' (RCE), can be detected as the depletion and dysfunction of hematopoietic and endothelial progenitor cell lineages in the peripheral blood of individuals with established T2D and/or obesity. The reversal of vascular RCE has been observed after administration of the sodium-glucose cotransporter-2 inhibitor (SGLT2i), empagliflozin, or after bariatric surgery for severe obesity. In this review, we explore emerging evidence that links improved dysglycemia to a reduction in systemic oxidative stress and recovery of circulating pro-vascular progenitor cell content required for blood vessel repair. Given that bariatric surgery consistently increases systemic glucagon-like-peptide 1 (GLP-1) release, we also focus on evidence that the use of GLP-1 receptor agonists (GLP-1RA) during obesity may act to inhibit the progression of systemic dysglycemia and adiposity, and indirectly reduce inflammation and oxidative stress, thereby limiting the impact of RCE. Therefore, therapeutic intervention with currently-available GLP-1RA may provide a less-invasive modality to reverse RCE, bolster vascular repair mechanisms, and improve cardiometabolic risk in individuals living with T2D and obesity.
Collapse
Affiliation(s)
- Ehab Bakbak
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Daniella C Terenzi
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Justin Z Trac
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Hwee Teoh
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, ON, Canada
- Division of Endocrinology and Metabolism, St. Michael's Hospital, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
| | - Adrian Quan
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
| | - Stephen A Glazer
- Department of Internal Medicine, Humber River Hospital, Toronto, ON, Canada
- Division of Endocrinology and Metabolism, Queen's University, Kingston, ON, Canada
| | - Ori D Rotstein
- Division of Endocrinology and Metabolism, St. Michael's Hospital, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Mohammed Al-Omran
- Division of Vascular Surgery, St. Michael's Hospital, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Subodh Verma
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - David A Hess
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, ON, Canada.
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.
- Molecular Medicine Research Laboratories, Robarts Research Institute, London, ON, Canada.
- Department of Physiology and Pharmacology, Western University, London, ON, Canada.
| |
Collapse
|
37
|
Oppenländer L, Palit S, Stemmer K, Greisle T, Sterr M, Salinno C, Bastidas-Ponce A, Feuchtinger A, Böttcher A, Ansarullah, Theis FJ, Lickert H. Vertical sleeve gastrectomy triggers fast β-cell recovery upon overt diabetes. Mol Metab 2021; 54:101330. [PMID: 34500108 PMCID: PMC8487975 DOI: 10.1016/j.molmet.2021.101330] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/17/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE The effectiveness of bariatric surgery in restoring β-cell function has been described in type-2 diabetes (T2D) patients and animal models for years, whereas the mechanistic underpinnings are largely unknown. The possibility of vertical sleeve gastrectomy (VSG) to rescue far-progressed, clinically-relevant T2D and to promote β-cell recovery has not been investigated on a single-cell level. Nevertheless, characterization of the heterogeneity and functional states of β-cells after VSG is a fundamental step to understand mechanisms of glycaemic recovery and to ultimately develop alternative, less-invasive therapies. METHODS We performed VSG in late-stage diabetic db/db mice and analyzed the islet transcriptome using single-cell RNA sequencing (scRNA-seq). Immunohistochemical analyses and quantification of β-cell area and proliferation complement our findings from scRNA-seq. RESULTS We report that VSG was superior to calorie restriction in late-stage T2D and rapidly restored normoglycaemia in morbidly obese and overt diabetic db/db mice. Single-cell profiling of islets of Langerhans showed that VSG induced distinct, intrinsic changes in the β-cell transcriptome, but not in that of α-, δ-, and PP-cells. VSG triggered fast β-cell redifferentiation and functional improvement within only two weeks of intervention, which is not seen upon calorie restriction. Furthermore, VSG expanded β-cell area by means of redifferentiation and by creating a proliferation competent β-cell state. CONCLUSION Collectively, our study reveals the superiority of VSG in the remission of far-progressed T2D and presents paths of β-cell regeneration and molecular pathways underlying the glycaemic benefits of VSG.
Collapse
Affiliation(s)
- Lena Oppenländer
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, 81675, Munich, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Subarna Palit
- Institute of Computational Biology, Helmholtz Center Munich, 85764, Neuherberg, Germany; Technical University of Munich, TUM School of Life Sciences Weihenstephan, 85354, Freising, Germany
| | - Kerstin Stemmer
- Institute of Diabetes and Obesity, Helmholtz Center Munich, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany; Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392, Giessen, Germany
| | - Tobias Greisle
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, 81675, Munich, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Michael Sterr
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Ciro Salinno
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, 81675, Munich, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, 81675, Munich, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Annette Feuchtinger
- Core Facility Pathology and Tissue Analytics, Helmholtz Center Munich, 85764, Neuherberg, Germany
| | - Anika Böttcher
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Ansarullah
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany.
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Center Munich, 85764, Neuherberg, Germany; Department of Mathematics, Technical University of Munich, 85748, Garching, Germany; Technical University of Munich, TUM School of Life Sciences Weihenstephan, 85354, Freising, Germany.
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, 81675, Munich, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany; Department of Medicine, Technical University of Munich, 81675, Munich, Germany.
| |
Collapse
|
38
|
Otten J, Stomby A, Waling M, Chorell E, Ryberg M, Svensson M, Holst JJ, Olsson T. The liver-alpha-cell axis after a mixed meal and during weight loss in type 2 diabetes. Endocr Connect 2021; 10:1101-1110. [PMID: 34382579 PMCID: PMC8494406 DOI: 10.1530/ec-21-0171] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 08/11/2021] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Glucagon and amino acids may be regulated in a feedback loop called the liver-alpha-cell axis with alanine or glutamine as suggested signal molecules. We assessed this concept in individuals with type 2 diabetes in the fasting state, after ingestion of a protein-rich meal, and during weight loss. Moreover, we investigated if postprandial glucagon secretion and hepatic insulin sensitivity were related. METHODS This is a secondary analysis of a 12-week weight-loss trial (Paleolithic diet ± exercise) in 29 individuals with type 2 diabetes. Before and after the intervention, plasma glucagon and amino acids were measured in the fasting state and during 180 min after a protein-rich mixed meal. Hepatic insulin sensitivity was measured using the hyperinsulinemic-euglycemic clamp with [6,6-2H2]glucose as a tracer. RESULTS The postprandial increase of plasma glucagon was associated with the postprandial increase of alanine and several other amino acids but not glutamine. In the fasted state and after the meal, glucagon levels were negatively correlated with hepatic insulin sensitivity (rS = -0.51/r = -0.58, respectively; both P < 0.05). Improved hepatic insulin sensitivity with weight loss was correlated with decreased postprandial glucagon response (r = -0.78; P < 0.001). CONCLUSIONS Several amino acids, notably alanine, but not glutamine could be key signals to the alpha cell to increase glucagon secretion. Amino acids may be part of a feedback mechanism as glucagon increases endogenous glucose production and ureagenesis in the liver. Moreover, postprandial glucagon secretion seems to be tightly related to hepatic insulin sensitivity.
Collapse
Affiliation(s)
- Julia Otten
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
- Correspondence should be addressed to J Otten:
| | - Andreas Stomby
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
- Region Jönköping County, Jönköping, Sweden
| | - Maria Waling
- Department of Food, Nutrition and Culinary Science, Umeå University, Umeå, Sweden
| | - Elin Chorell
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Mats Ryberg
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Michael Svensson
- Department of Community Medicine and Rehabilitation, Section for Sports Medicine, Umeå University, Umeå Sweden
| | - Jens Juul Holst
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tommy Olsson
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
39
|
Choong YS, Lim YY, Soong JX, Savoo N, Guida C, Rhyman L, Ramracheya R, Ramasami P. Theoretical study of the interactions between peptide tyrosine tyrosine [PYY (1-36)], a newly identified modulator in type 2 diabetes pathophysiology, with receptors NPY1R and NPY4R. Hormones (Athens) 2021; 20:557-569. [PMID: 33782920 DOI: 10.1007/s42000-021-00278-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/10/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE Diabetes mellitus is a common condition in the clinically obese. Bariatric surgery is one of the ways to put type 2 diabetes in remission. Recent findings propose the appetite-regulator peptide tyrosine tyrosine (PYY) as a therapeutic option for patients with type 2 diabetes. This novel gut hormone restores impaired insulin and glucagon secretion in pancreatic islets and is implicated in type 2 diabetes reversal after bariatric surgery. The current study elucidates the interactions between PYY and the NPY1R and NPY4R receptors using computational methods. METHODS Protein structure prediction, molecular docking simulation, and molecular dynamics (MD) simulation were performed to elucidate the interactions of PYY with NPY1R and NPY4R. RESULTS The predicted binding models of PYY-NPY receptors are in agreement with those described in the literature, although different interaction partners are presented for the C-terminal tail of PYY. Non-polar interactions are predicted to drive the formation of the protein complex. The calculated binding energies show that PYY has higher affinity for NPY4R (ΔGGBSA = -65.08 and ΔGPBSA = -87.62 kcal/mol) than for NPY1R (ΔGGBSA = -23.11 and ΔGPBSA = -50.56 kcal/mol). CONCLUSIONS Based on the constructed models, the binding conformations obtained from docking and MD simulation for both the PYY-NPY1R and PYY-NPY4R complexes provide a detailed map of possible interactions. The calculated binding energies show a higher affinity of PYY for NPY4R. These findings may help to understand the mechanisms behind the improvement of diabetes following bariatric surgery.
Collapse
Affiliation(s)
- Yee Siew Choong
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Penang, Malaysia.
| | - Yee Ying Lim
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Jia Xin Soong
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Nandini Savoo
- Computational Chemistry Group, Department of Chemistry, Faculty of Science, University of Mauritius, Réduit 80837, Mauritius
| | - Claudia Guida
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom
| | - Lydia Rhyman
- Computational Chemistry Group, Department of Chemistry, Faculty of Science, University of Mauritius, Réduit 80837, Mauritius
- Department of Chemical Sciences, University of Johannesburg, Doornfontein, Johannesburg 2028, South Africa
| | - Reshma Ramracheya
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom.
- Pharmaceutical Operations, Centre International de Dévelopment Pharmaceutique, BioPark Mauritius, Socota Phoenicia, Phoenix 73408, Mauritius.
| | - Ponnadurai Ramasami
- Computational Chemistry Group, Department of Chemistry, Faculty of Science, University of Mauritius, Réduit 80837, Mauritius.
- Department of Chemical Sciences, University of Johannesburg, Doornfontein, Johannesburg 2028, South Africa.
| |
Collapse
|
40
|
Schnabl K, Li Y, U-Din M, Klingenspor M. Secretin as a Satiation Whisperer With the Potential to Turn into an Obesity-curbing Knight. Endocrinology 2021; 162:6294014. [PMID: 34089599 DOI: 10.1210/endocr/bqab113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Indexed: 01/01/2023]
Abstract
The obesity pandemic requires effective preventative and therapeutic intervention strategies. Successful and sustained obesity treatment is currently limited to bariatric surgery. Modulating the release of gut hormones is considered promising to mimic bariatric surgery with its beneficial effects on food intake, body weight, and blood glucose levels. The gut peptide secretin was the first molecule to be termed a hormone; nevertheless, only recently has it been established as a legitimate anorexigenic peptide. In contrast to gut hormones that crosstalk with the brain either directly or by afferent neuronal projections, secretin mediates meal-associated brown fat thermogenesis to induce meal termination, thereby qualifying this physiological mechanism as an attractive, peripheral target for the treatment of obesity. In this perspective, it is of pivotal interest to deepen our as yet superficial knowledge on the physiological roles of secretin as well as meal-associated thermogenesis in energy balance and body weight regulation. Of note, the emerging differences between meal-associated thermogenesis and cold-induced thermogenesis must be taken into account. In fact, there is no correlation between these 2 entities. In addition, the investigation of potential effects of secretin in hedonic-driven food intake, bariatric surgery and chronic treatment using suitable application strategies to overcome pharmacokinetic limitations will provide further insight into its potential to influence energy balance. The aim of this article is to review the facts on secretin's metabolic effects, address prevailing gaps in our knowledge, and provide an overview on the opportunities and challenges of the therapeutic potential of secretin in body weight control.
Collapse
Affiliation(s)
- Katharina Schnabl
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ-Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Yongguo Li
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ-Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
| | - Mueez U-Din
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ-Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany
| |
Collapse
|
41
|
Akkus G, Tetiker T. Which predictors could effect on remission of type 2 diabetes mellitus after the metabolic surgery: A general perspective of current studies? World J Diabetes 2021; 12:1312-1324. [PMID: 34512896 PMCID: PMC8394232 DOI: 10.4239/wjd.v12.i8.1312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/18/2021] [Accepted: 04/25/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The alarming rise in the worldwide prevalence of obesity is paralleled by an increasing burden of type 2 diabetes mellitus (T2DM). Metabolic surgery is the most effective means of obtaining substantial and durable weight loss in individual obese patients with T2DM. There are randomized trials that justify the inclusion of metabolic surgery into the treatment algorithm for patients with T2DM, but remission rates of T2DM after metabolic surgery can display great variability. AIM To discuss the most commonly used surgical options including vertical sleeve gastrectomy, adjustable gastric banding, Roux-en-Y gastric bypass, and biliopancreatic diversion with duodenal switch. METHODS We also report from observational and randomized controlled studies on rate of remission of T2DM after the surgical procedures. RESULTS In light of the recent findings, metabolic surgery is a safe and effective treatment option for obese patient with T2DM, but further studies are needed to clarify better the rate of diabetes remission. CONCLUSION In light of the recent findings, metabolic surgery is a safe and effective treatment option for obese patients with T2DM, but further studies are needed to clarify better the rate of diabetes remission.
Collapse
Affiliation(s)
- Gamze Akkus
- Department of Internal Medicine, Cukurova University Medical Faculty, Adana 33170, Turkey
- Department of Endocrinology, Cukurova University, Adana 33170, Turkey
| | - Tamer Tetiker
- Department of Internal Medicine, Cukurova University Medical Faculty, Adana 33170, Turkey
- Department of Endocrinology, Cukurova University, Adana 33170, Turkey
| |
Collapse
|
42
|
Ligon C, Shah A, Prasad M, Laferrère B. Preintervention Clinical Determinants and Measured β-Cell Function As Predictors of Type 2 Diabetes Remission After Roux-en-Y Gastric Bypass Surgery. Diabetes Care 2021; 44:dc210395. [PMID: 34400479 PMCID: PMC8929185 DOI: 10.2337/dc21-0395] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/20/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Bariatric surgery results in improved glycemic control in individuals with type 2 diabetes. Single and clusters of clinical determinants have been identified as presurgery predictors of postsurgery diabetes remission. Our goal was to assess whether the addition of measured preoperative β-cell function would improve established clinical models of prediction of diabetes remission. RESEARCH DESIGN AND METHODS Presurgery clinical characteristics, metabolic markers, and β-cell function after oral and intravenous (IV) glucose challenges were assessed in 73 individuals with severe obesity and type 2 diabetes and again 1 year after gastric bypass surgery. Single and multivariate analyses were conducted with preoperative variables to determine the best predictive models of remission. RESULTS Presurgery β-cell glucose sensitivity, a surrogate of β-cell function, was negatively correlated with known diabetes duration, HbA1c, insulin use, and the diabetes remission scores DiaRem and advanced (Ad)-DiaRem (all P < 0.001). Measured β-cell function after oral glucose was 1.6-fold greater than after the IV glucose challenge and more strongly correlated with preoperative clinical and metabolic characteristics. The addition of preoperative β-cell function to clinical models containing well-defined diabetes remission scores did not improve the model's ability to predict diabetes remission after Roux-en-Y gastric bypass. CONCLUSIONS The addition of measured β-cell function does not add predictive value to defined clinical models of diabetes remission 1 year after surgical weight loss.
Collapse
Affiliation(s)
- Chanel Ligon
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Ankit Shah
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ
| | - Malini Prasad
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
- New York Obesity Research Center, Division of Endocrinology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Blandine Laferrère
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
- New York Obesity Research Center, Division of Endocrinology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| |
Collapse
|
43
|
Angelini G, Salinari S, Castagneto-Gissey L, Bertuzzi A, Casella-Mariolo J, Ahlin S, Boskoski I, Gaggini M, Raffaelli M, Costamagna G, Casella G, Marini PL, Gastaldelli A, Bornstein S, Mingrone G. Small intestinal metabolism is central to whole-body insulin resistance. Gut 2021; 70:1098-1109. [PMID: 32994312 DOI: 10.1136/gutjnl-2020-322073] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/14/2020] [Accepted: 07/24/2020] [Indexed: 12/27/2022]
Abstract
OBJECTIVE To assess the role of jejunum in insulin resistance in humans and in experimental animals. DESIGN Twenty-four subjects undergoing biliopancreatic diversion (BPD) or Roux-en-Y gastric bypass (RYGB) were enrolled. Insulin sensitivity was measured at baseline and at 1 week after surgery using oral glucose minimal model.We excluded the jejunum from intestinal continuity in pigs and created a jejunal loop with its vascular and nerve supply intact accessible from two cutaneous stomas, and reconnected the bowel with an end-to-end anastomosis. Glucose stable isotopes were given in the stomach or in the jejunal loop.In vitro studies using primary porcine and human hepatocytes or myoblasts tested the effects of plasma on gluconeogenesis or glucose uptake and insulin signalling. RESULTS Whole-body insulin sensitivity (SI∙104: 0.54±0.12 before vs 0.82±0.11 after BPD, p=0.024 and 0.41±0.09 before vs 0.65±0.09/pM/min after RYGB, p=not significant) and Glucose Disposition Index increased only after BPD. In pigs, insulin sensitivity was significantly lower when glucose was administered in the jejunal loop than in the stomach (glucose rate of disappearance (Rd) area under the curve (AUC)/insulin AUC∙10: 1.82±0.31 vs 2.96±0.33 mmol/pM/min, p=0.0017).Metabolomics showed a similar pattern before surgery and during jejunal-loop stimulation, pointing to a higher expression of gluconeogenetic substrates, a metabolic signature of impaired insulin sensitivity.A greater hepatocyte phosphoenolpyruvate-carboxykinase and glucose-6-phosphatase gene expression was elicited with plasma from porcine jejunal loop or before surgery compared with plasma from jejunectomy in pigs or jejunal bypass in humans.Stimulation of myoblasts with plasma from porcine jejunal loop or before surgery reduced glucose uptake, Ser473-Akt phosphorylation and GLUT4 expression compared with plasma obtained during gastric glucose administration after jejunectomy in pigs or after jejunal bypass in humans. CONCLUSION Proximal gut plays a crucial role in controlling insulin sensitivity through a distinctive metabolic signature involving hepatic gluconeogenesis and muscle insulin resistance. Bypassing the jejunum is beneficial in terms of insulin-mediated glucose disposal in obesity. TRIAL REGISTRATION NUMBER NCT03111953.
Collapse
Affiliation(s)
- Giulia Angelini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Serenella Salinari
- Department of Computer, Control, and Management Engineering "Antonio Ruberti", Universityof Rome "Sapienza", Rome, Italy
| | | | | | | | - Sofie Ahlin
- Department of Molecular and Clinical Medicine, Institute of Medicine, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Ivo Boskoski
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Melania Gaggini
- Cardiometabolic Risk Laboratory, Institute of Clinical Physiology, CNR, Pisa, Italy
| | - Marco Raffaelli
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Guido Costamagna
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Casella
- Department of Surgical Sciences, Sapienza University of Rome, Rome, Italy
| | - Pier Luigi Marini
- Department of Surgery, Azienda Ospedaliera S. Camillo Forlanini, Rome, Italy
| | - Amalia Gastaldelli
- Cardiometabolic Risk Laboratory, Institute of Clinical Physiology, CNR, Pisa, Italy
| | - Stefan Bornstein
- Division of Diabetes & Nutritional Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK.,Department of Medicine III, Universitätsklinikum Carl Gustav Carus an der Technischen, Universität Dresden, Dresden, Germany
| | - Geltrude Mingrone
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy .,Università Cattolica del Sacro Cuore, Rome, Italy.,Division of Diabetes & Nutritional Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| |
Collapse
|
44
|
Ding B, Hu Y, Yuan L, Yan RN, Ma JH. Effectiveness of beinaglutide in a patient with late dumping syndrome after gastrectomy: A case report. Medicine (Baltimore) 2021; 100:e26086. [PMID: 34032745 PMCID: PMC8154494 DOI: 10.1097/md.0000000000026086] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/23/2021] [Accepted: 05/06/2021] [Indexed: 01/04/2023] Open
Abstract
RATIONALE Dumping syndrome is a frequent and potentially severe complication after gastric surgery. Beinaglutide, a recombinant human glucagon-like peptide-1 (GLP-1) which shares 100% homology with human GLP-1(7-36), has never been reported in the treatment of dumping syndrome before. PATIENT CONCERNS The patient had undergone distal gastrectomy for gastric signet ring cell carcinoma 16 months ago. He presented with symptoms of paroxysmal palpitation, sweating, and dizziness for 4 months. DIAGNOSIS He was diagnosed with late dumping syndrome. INTERVENTIONS AND OUTCOMES The patient was treated with dietary changes and acarbose for 4 months before admitted to our hospital. The treatment with dietary changes and acarbose did not prevent postprandial hyperinsulinemia and hypoglycemia according to the 75 g oral glucose tolerance test (OGTT) and continuous glucose monitoring (CGM) on admission.Therefore, the patient was treated with beinaglutide 0.1 mg before breakfast and lunch instead of acarbose. After the treatment of beinaglutide for 1 month, OGTT showed a reduction in postprandial hyperinsulinemia compared with before starting treatment, and the time in the range of 3.9 to 10 mmol/L became 100% in CGM. No side effect was observed in this patient during beinaglutide treatment. LESSONS These findings suggest that beinaglutide may be effective for treating post-gastrectomy late dumping syndrome.
Collapse
|
45
|
Lawler H, Salehi M. Diabetes Remission After Gastric Bypass: To Be or Not to Be Dependent on Weight Loss. J Clin Endocrinol Metab 2021; 106:e2362-e2364. [PMID: 33524103 PMCID: PMC8063235 DOI: 10.1210/clinem/dgab008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Indexed: 11/19/2022]
Affiliation(s)
- Helen Lawler
- University of Colorado, Division of Endocrinology, Metabolism, and Diabetes, Aurora, CO, USA
| | - Marzieh Salehi
- University of Texas Health Science Center, San Antonio, TX, USA
- STVHC, Audie Murphy Hospital, San Antonio, TX, USA
- Correspondence: Marzieh Salehi, MD MS, Associate Professor of Medicine, Medical Director, Bartter Research Unit, Audie Murphy Hospital, 7703 Floyd Curl Drive, University of Texas Health at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
46
|
Faramia J, Hao Z, Mumphrey MB, Townsend RL, Miard S, Carreau AM, Nadeau M, Frisch F, Baraboi ED, Grenier-Larouche T, Noll C, Li M, Biertho L, Marceau S, Hould FS, Lebel S, Morrison CD, Münzberg H, Richard D, Carpentier AC, Tchernof A, Berthoud HR, Picard F. IGFBP-2 partly mediates the early metabolic improvements caused by bariatric surgery. Cell Rep Med 2021; 2:100248. [PMID: 33948578 PMCID: PMC8080239 DOI: 10.1016/j.xcrm.2021.100248] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/21/2020] [Accepted: 03/23/2021] [Indexed: 12/21/2022]
Abstract
Insulin-like growth factor-binding protein (IGFBP)-2 is a circulating biomarker of cardiometabolic health. Here, we report that circulating IGFBP-2 concentrations robustly increase after different bariatric procedures in humans, reaching higher levels after biliopancreatic diversion with duodenal switch (BPD-DS) than after Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG). This increase is closely associated with insulin sensitization. In mice and rats, BPD-DS and RYGB operations also increase circulating IGFBP-2 levels, which are not affected by SG or caloric restriction. In mice, Igfbp2 deficiency significantly impairs surgery-induced loss in adiposity and early improvement in insulin sensitivity but does not affect long-term enhancement in glucose homeostasis. This study demonstrates that the modulation of circulating IGFBP-2 may play a role in the early improvement of insulin sensitivity and loss of adiposity brought about by bariatric surgery.
Collapse
Affiliation(s)
- Justine Faramia
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
- Faculty of Pharmacy, Université Laval, Québec, QC, Canada
| | - Zheng Hao
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Michael B. Mumphrey
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - R. Leigh Townsend
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | | | - Anne-Marie Carreau
- Division of Endocrinology, Department of Medicine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mélanie Nadeau
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
| | - Frédérique Frisch
- Division of Endocrinology, Department of Medicine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Elena-Dana Baraboi
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
| | - Thomas Grenier-Larouche
- Division of Endocrinology, Department of Medicine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Christophe Noll
- Division of Endocrinology, Department of Medicine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Meng Li
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
- Faculty of Pharmacy, Université Laval, Québec, QC, Canada
| | - Laurent Biertho
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
| | - Simon Marceau
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
| | - Frédéric-Simon Hould
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
| | - Stéfane Lebel
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
| | - Christopher D. Morrison
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Heike Münzberg
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Denis Richard
- Faculty of Pharmacy, Université Laval, Québec, QC, Canada
| | - André C. Carpentier
- Division of Endocrinology, Department of Medicine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - André Tchernof
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
| | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Frédéric Picard
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
- Faculty of Pharmacy, Université Laval, Québec, QC, Canada
| |
Collapse
|
47
|
Di Prospero NA, Yee J, Frustaci ME, Samtani MN, Alba M, Fleck P. Efficacy and safety of glucagon-like peptide-1/glucagon receptor co-agonist JNJ-64565111 in individuals with type 2 diabetes mellitus and obesity: A randomized dose-ranging study. Clin Obes 2021; 11:e12433. [PMID: 33475251 DOI: 10.1111/cob.12433] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/17/2020] [Accepted: 11/29/2020] [Indexed: 12/15/2022]
Abstract
Weight loss has been shown to improve metabolic parameters and cardiovascular risk in people with type 2 diabetes mellitus (T2DM). This phase 2 study evaluated the safety and efficacy of JNJ-64565111, a dual agonist of GLP-1 and glucagon receptors, in individuals with T2DM and class II/III obesity. In this randomized, double-blind study, participants with T2DM (HbA1c 6.5%-9.5%), body mass index of 35 to 50 kg/m2 and stable weight were randomly assigned (1:1:1:1) to placebo or JNJ-64565111 (5.0 mg, 7.4 mg or 10.0 mg). The primary endpoint was percent change from baseline in body weight at week 12. Of 195 dosed participants, 144 (73.8%) completed treatment. At week 12, placebo-subtracted body weight changes were -4.6%, -5.9% and -7.2% with JNJ-64565111 5.0 mg, 7.4 mg and 10.0 mg, respectively. All JNJ-64565111 doses were associated with no change in HbA1c and slight numerical elevation of fasting insulin. Numerical increases in fasting plasma glucose were observed with JNJ-64565111 5.0 mg and 7.4 mg. Incidence of treatment-emergent adverse events, especially nausea and vomiting, was higher with JNJ-64565111 vs placebo. Overall, JNJ-64565111 significantly reduced body weight in a dose-dependent manner vs placebo but was associated with greater incidence of treatment-emergent adverse events, no HbA1c reductions, and increased fasting plasma glucose and fasting insulin.
Collapse
Affiliation(s)
| | - Jaqueline Yee
- Janssen Research & Development, LLC, Raritan, New Jersey, USA
| | - Mary E Frustaci
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | | | - Maria Alba
- Janssen Research & Development, LLC, Raritan, New Jersey, USA
| | - Penny Fleck
- Janssen Research & Development, LLC, Raritan, New Jersey, USA
| |
Collapse
|
48
|
Marciniak C, Chávez-Talavera O, Caiazzo R, Hubert T, Zubiaga L, Baud G, Quenon A, Descat A, Vallez E, Goossens JF, Kouach M, Vangelder V, Gobert M, Daoudi M, Derudas B, Pigny P, Klein A, Gmyr V, Raverdy V, Lestavel S, Laferrère B, Staels B, Tailleux A, Pattou F. Characterization of one anastomosis gastric bypass and impact of biliary and common limbs on bile acid and postprandial glucose metabolism in a minipig model. Am J Physiol Endocrinol Metab 2021; 320:E772-E783. [PMID: 33491532 PMCID: PMC8906817 DOI: 10.1152/ajpendo.00356.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The alimentary limb has been proposed to be a key driver of the weight-loss-independent metabolic improvements that occur upon bariatric surgery. However, the one anastomosis gastric bypass (OAGB) procedure, consisting of one long biliary limb and a short common limb, induces similar beneficial metabolic effects compared to Roux-en-Y Gastric Bypass (RYGB) in humans, despite the lack of an alimentary limb. The aim of this study was to assess the role of the length of biliary and common limbs in the weight loss and metabolic effects that occur upon OAGB. OAGB and sham surgery, with or without modifications of the length of either the biliary limb or the common limb, were performed in Gottingen minipigs. Weight loss, metabolic changes, and the effects on plasma and intestinal bile acids (BAs) were assessed 15 days after surgery. OAGB significantly decreased body weight, improved glucose homeostasis, increased postprandial GLP-1 and fasting plasma BAs, and qualitatively changed the intestinal BA species composition. Resection of the biliary limb prevented the body weight loss effects of OAGB and attenuated the postprandial GLP-1 increase. Improvements in glucose homeostasis along with changes in plasma and intestinal BAs occurred after OAGB regardless of the biliary limb length. Resection of only the common limb reproduced the glucose homeostasis effects and the changes in intestinal BAs. Our results suggest that the changes in glucose metabolism and BAs after OAGB are mainly mediated by the length of the common limb, whereas the length of the biliary limb contributes to body weight loss.NEW & NOTEWORTHY Common limb mediates postprandial glucose metabolism change after gastric bypass whereas biliary limb contributes to weight loss.
Collapse
Affiliation(s)
- Camille Marciniak
- U1190, Institut Pasteur de Lille, University of Lille, Inserm, Lille, France
| | | | - Robert Caiazzo
- U1190, Institut Pasteur de Lille, University of Lille, Inserm, Lille, France
| | - Thomas Hubert
- U1190, Institut Pasteur de Lille, University of Lille, Inserm, Lille, France
| | - Lorea Zubiaga
- U1190, Institut Pasteur de Lille, University of Lille, Inserm, Lille, France
| | - Gregory Baud
- U1190, Institut Pasteur de Lille, University of Lille, Inserm, Lille, France
| | - Audrey Quenon
- U1190, Institut Pasteur de Lille, University of Lille, Inserm, Lille, France
| | - Amandine Descat
- Mass Spectrometry Department, Pharmacy Faculty, PSM-GRITA, Lille, France
| | - Emmanuelle Vallez
- U1011, Institut Pasteur de Lille, University of Lille, Inserm Lille, France
| | | | - Mostafa Kouach
- Mass Spectrometry Department, Pharmacy Faculty, PSM-GRITA, Lille, France
| | - Vincent Vangelder
- U1190, Institut Pasteur de Lille, University of Lille, Inserm, Lille, France
| | - Mathilde Gobert
- U1190, Institut Pasteur de Lille, University of Lille, Inserm, Lille, France
| | - Mehdi Daoudi
- U1190, Institut Pasteur de Lille, University of Lille, Inserm, Lille, France
| | - Bruno Derudas
- U1011, Institut Pasteur de Lille, University of Lille, Inserm Lille, France
| | - Pascal Pigny
- Mass Spectrometry Department, Pharmacy Faculty, PSM-GRITA, Lille, France
| | - André Klein
- Metabolism and Glycosylation Diseases, Biology Pathology Center, Lille, France
| | - Valéry Gmyr
- U1190, Institut Pasteur de Lille, University of Lille, Inserm, Lille, France
| | - Violeta Raverdy
- U1190, Institut Pasteur de Lille, University of Lille, Inserm, Lille, France
| | - Sophie Lestavel
- U1011, Institut Pasteur de Lille, University of Lille, Inserm Lille, France
| | - Blandine Laferrère
- Division of Endocrinology, Department of Medicine, New York Obesity Research Center, Columbia University Irving Medical Center, New York, New York
| | - Bart Staels
- U1011, Institut Pasteur de Lille, University of Lille, Inserm Lille, France
| | - Anne Tailleux
- U1011, Institut Pasteur de Lille, University of Lille, Inserm Lille, France
| | - François Pattou
- U1190, Institut Pasteur de Lille, University of Lille, Inserm, Lille, France
| |
Collapse
|
49
|
West JA, Tsakmaki A, Ghosh SS, Parkes DG, Grønlund RV, Pedersen PJ, Maggs D, Rajagopalan H, Bewick GA. Chronic peptide-based GIP receptor inhibition exhibits modest glucose metabolic changes in mice when administered either alone or combined with GLP-1 agonism. PLoS One 2021; 16:e0249239. [PMID: 33788878 PMCID: PMC8011784 DOI: 10.1371/journal.pone.0249239] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/13/2021] [Indexed: 12/04/2022] Open
Abstract
Combinatorial gut hormone therapy is one of the more promising strategies for identifying improved treatments for metabolic disease. Many approaches combine the established benefits of glucagon-like peptide-1 (GLP-1) agonism with one or more additional molecules with the aim of improving metabolic outcomes. Recent attention has been drawn to the glucose-dependent insulinotropic polypeptide (GIP) system due to compelling pre-clinical evidence describing the metabolic benefits of antagonising the GIP receptor (GIPR). We rationalised that benefit might be accrued from combining GIPR antagonism with GLP-1 agonism. Two GIPR peptide antagonists, GIPA-1 (mouse GIP(3–30)NH2) and GIPA-2 (NαAc-K10[γEγE-C16]-Arg18-hGIP(5–42)), were pharmacologically characterised and both exhibited potent antagonist properties. Acute in vivo administration of GIPA-1 during an oral glucose tolerance test (OGTT) had negligible effects on glucose tolerance and insulin in lean mice. In contrast, GIPA-2 impaired glucose tolerance and attenuated circulating insulin levels. A mouse model of diet-induced obesity (DIO) was used to investigate the potential metabolic benefits of chronic dosing of each antagonist, alone or in combination with liraglutide. Chronic administration studies showed expected effects of liraglutide, lowering food intake, body weight, fasting blood glucose and plasma insulin concentrations while improving glucose sensitivity, whereas delivery of either GIPR antagonist alone had negligible effects on these parameters. Interestingly, chronic dual therapy augmented insulin sensitizing effects and lowered plasma triglycerides and free-fatty acids, with more notable effects observed with GIPA-1 compared to GIPA-2. Thus, the co-administration of both a GIPR antagonist with a GLP1 agonist uncovers interesting beneficial effects on measures of insulin sensitivity, circulating lipids and certain adipose stores that seem influenced by the degree or nature of GIP receptor antagonism.
Collapse
Affiliation(s)
- Jason A. West
- Fractyl Laboratories Inc, Lexington, MA, United States of America
| | - Anastasia Tsakmaki
- Diabetes Research Group, School of Life Course Sciences, Faculty of Life Science and Medicine, King’s College London, London, England, United Kingdom
| | | | | | | | | | - David Maggs
- Fractyl Laboratories Inc, Lexington, MA, United States of America
| | | | - Gavin A. Bewick
- Diabetes Research Group, School of Life Course Sciences, Faculty of Life Science and Medicine, King’s College London, London, England, United Kingdom
- * E-mail:
| |
Collapse
|
50
|
Schalla MA, Taché Y, Stengel A. Neuroendocrine Peptides of the Gut and Their Role in the Regulation of Food Intake. Compr Physiol 2021; 11:1679-1730. [PMID: 33792904 DOI: 10.1002/cphy.c200007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The regulation of food intake encompasses complex interplays between the gut and the brain. Among them, the gastrointestinal tract releases different peptides that communicate the metabolic state to specific nuclei in the hindbrain and the hypothalamus. The present overview gives emphasis on seven peptides that are produced by and secreted from specialized enteroendocrine cells along the gastrointestinal tract in relation with the nutritional status. These established modulators of feeding are ghrelin and nesfatin-1 secreted from gastric X/A-like cells, cholecystokinin (CCK) secreted from duodenal I-cells, glucagon-like peptide 1 (GLP-1), oxyntomodulin, and peptide YY (PYY) secreted from intestinal L-cells and uroguanylin (UGN) released from enterochromaffin (EC) cells. © 2021 American Physiological Society. Compr Physiol 11:1679-1730, 2021.
Collapse
Affiliation(s)
- Martha A Schalla
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Yvette Taché
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, David Geffen School of Medicine, UCLA, Los Angeles, California, USA.,VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Andreas Stengel
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|