1
|
Sandforth L, Raverdy V, Sandforth A, Bauvin P, Chatelain E, Verkindt H, Mingrone G, Guidone C, Verrastro O, Zhou K, Archid R, Mihaljevic A, Caiazzo R, Baud G, Marciniak C, Chetboun M, Ganslmeier M, Minelli Faiao V, Heni M, Fritsche L, Moller A, Kantartzis K, Peter A, Lehmann R, Wagner R, Prystupa K, Fritsche A, Stefan N, Preissl H, Birkenfeld AL, Jumpertz von Schwartzenberg R, Pattou F. Subphenotype-Dependent Benefits of Bariatric Surgery for Individuals at Risk for Type 2 Diabetes. Diabetes Care 2025; 48:996-1006. [PMID: 40214701 DOI: 10.2337/dc25-0160] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/23/2025] [Indexed: 05/22/2025]
Abstract
OBJECTIVE Bariatric surgery is an effective treatment option for individuals with obesity and type 2 diabetes (T2D). However, whether outcomes in subtypes of individuals at risk for T2D and/or comorbidities (Tübingen Clusters) differ, is unknown. Of these, cluster 5 (C5) and cluster 6 (C6) are high-risk clusters for developing T2D and/or comorbidities, while cluster 4 (C4) is a low-risk cluster. We investigated bariatric surgery outcomes, hypothesizing that high-risk clusters benefit most due to great potential for metabolic improvement. RESEARCH DESIGN AND METHODS We allocated participants without T2D but at risk for T2D, defined by elevated BMI, to the Tübingen Clusters. Participants had normal glucose regulation or prediabetes according to American Diabetes Association criteria. Two cohorts underwent bariatric surgery: a discovery (Lille, France) and a replication cohort (Rome, Italy). A control cohort (Tübingen, Germany) received behavioral modification counseling. Main outcomes included alteration of glucose regulation parameters and prediabetes remission. RESULTS In the discovery cohort, 15.0% of participants (n = 121) were allocated to C4, 22.3% (n = 180) to C5, and 62.4% (n = 503) to C6. Relative body weight loss was similar among all clusters; however, reduction of insulin resistance and improvement of β-cell function were strongest in C5. Prediabetes remission rate was lowest in low-risk C4 and highest in high-risk C5. Individuals from high-risk clusters changed to low-risk clusters in both bariatric surgery cohorts but not in the control cohort. CONCLUSIONS Participants in C5 had the highest benefit from bariatric surgery in terms of improvement in insulin resistance, β-cell function, and prediabetes remission. This novel classification might help identify individuals who will benefit specifically from bariatric surgery.
Collapse
Affiliation(s)
- Leontine Sandforth
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Violeta Raverdy
- INSERM, CHU Lille, Institut Pasteur de Lille, UMR 1190 Translational Research for Diabetes, European Genomic Institute for Diabetes, University Lille, Lille, France
- Integrated Center for Obesity, General and Endocrine Surgery, CHU Lille, Lille, France
| | - Arvid Sandforth
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Pierre Bauvin
- INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID, University Lille, Lille, France
| | - Estelle Chatelain
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, US 41-UAR 2014-PLBS, University Lille, Lille, France
| | - Helene Verkindt
- INSERM, CHU Lille, Institut Pasteur de Lille, UMR 1190 Translational Research for Diabetes, European Genomic Institute for Diabetes, University Lille, Lille, France
- Integrated Center for Obesity, General and Endocrine Surgery, CHU Lille, Lille, France
| | - Geltrude Mingrone
- Department of Internal Medicine, Catholic University of the Sacred Heart, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Division of Diabetes and Nutritional Sciences, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, U.K
| | - Caterina Guidone
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Ornella Verrastro
- Department of Internal Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Karin Zhou
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Rami Archid
- Department of General, Visceral, and Transplant Surgery, University Hospital of Tübingen, Tübingen, Germany
| | - André Mihaljevic
- Department of General, Visceral, and Transplant Surgery, University Hospital of Tübingen, Tübingen, Germany
| | - Robert Caiazzo
- INSERM, CHU Lille, Institut Pasteur de Lille, UMR 1190 Translational Research for Diabetes, European Genomic Institute for Diabetes, University Lille, Lille, France
- Integrated Center for Obesity, General and Endocrine Surgery, CHU Lille, Lille, France
| | - Gregory Baud
- INSERM, CHU Lille, Institut Pasteur de Lille, UMR 1190 Translational Research for Diabetes, European Genomic Institute for Diabetes, University Lille, Lille, France
- Integrated Center for Obesity, General and Endocrine Surgery, CHU Lille, Lille, France
| | - Camille Marciniak
- INSERM, CHU Lille, Institut Pasteur de Lille, UMR 1190 Translational Research for Diabetes, European Genomic Institute for Diabetes, University Lille, Lille, France
- Integrated Center for Obesity, General and Endocrine Surgery, CHU Lille, Lille, France
| | - Mikael Chetboun
- INSERM, CHU Lille, Institut Pasteur de Lille, UMR 1190 Translational Research for Diabetes, European Genomic Institute for Diabetes, University Lille, Lille, France
- Integrated Center for Obesity, General and Endocrine Surgery, CHU Lille, Lille, France
| | - Marlene Ganslmeier
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Vitória Minelli Faiao
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Martin Heni
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital of Tübingen, Tübingen, Germany
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Louise Fritsche
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Anja Moller
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Konstantinos Kantartzis
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Andreas Peter
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Rainer Lehmann
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Robert Wagner
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf (DDZ), Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - Katsiaryna Prystupa
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf (DDZ), Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - Andreas Fritsche
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Norbert Stefan
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Hubert Preissl
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Pharmacy and Biochemistry, Institute of Pharmaceutical Sciences, and Interfaculty Centre for Pharmacogenomics and Pharma Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, U.K
| | - Reiner Jumpertz von Schwartzenberg
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections" (CMFI), University of Tübingen, Tübingen, Germany
- M3 Research Center, Tübingen, Germany
| | - François Pattou
- INSERM, CHU Lille, Institut Pasteur de Lille, UMR 1190 Translational Research for Diabetes, European Genomic Institute for Diabetes, University Lille, Lille, France
- Integrated Center for Obesity, General and Endocrine Surgery, CHU Lille, Lille, France
| |
Collapse
|
2
|
Chen B, Lu Q, Hu B, Sun D, Ying T. Protocol of quantitative ultrasound techniques for noninvasive assessing of hepatic steatosis after bariatric surgery. Front Surg 2024; 10:1244199. [PMID: 38239667 PMCID: PMC10794322 DOI: 10.3389/fsurg.2023.1244199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 11/27/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction Roux-en-Y gastric bypass surgery can effectively improve steatosis, necroinflammatory activity, and hepatic fibrosis in individuals diagnosed with morbid obesity or nonalcoholic steatohepatitis (NASH). Common methods such as body mass index (BMI) to evaluate the postoperative effect of clinical bariatric surgery cannot differentiate subcutaneous fats from visceral fats and muscles. Several Quantitative ultrasound (QUS)-based approaches have been developed to quantify hepatic steatosis. QUS techniques (tissue attenuation imaging (TAI), tissue scatter distribution imaging (TSI)) from radio frequency (RF) data analysis as a means for the detection and grading of hepatic steatosis has been posited as an objective and noninvasive approach. The implementation and standardization of QUS techniques (TAI, TSI) in assessing hepatic steatosis quantitatively after bariatric surgery is of high-priority. Our study is aimed to assess hepatic steatosis with QUS techniques (TAI, TSI) in morbidly obese individuals before and after bariatric surgery, and to compare with anthropometric measurements, laboratory assessments and other imaging methods. Methods and analysis The present investigation, a self-discipline examination of navigational capacity devoid of visual cues, is designed as a single-site, forward-looking evaluation of efficacy with the imprimatur of the institutional review board. The duration of the study has been provisionally determined to span from 1 January 2023 through 31 December 2025. Our cohort shall encompass one hundred participants, who was scheduled to undergo Roux-en-Y gastric bypass (RYGB) at Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine. All patients will undergo anthropometric measurements, blood-based biochemical analyses, ultrasonic examination and magnetic resonance imaging proton density fat fraction (MRI-PDFF). The primary endpoint is the analysis of evaluating the efficacy of QUS techniques assessing hepatic steatosis compared to other methods before and after bariatric surgery. Results Prior to the fomal study, we recruited 21 obese Chinese participants who received ultrasonic examination (TAI, TSI) and MRI-PDFF. AC-TAI showed moderate correlations with MRI-PDFF (adjusted r = 0.632; P < 0.05). For MRI-PDFF ≥10%, SC-TSI showed moderate correlations with MRI-PDFF (adjusted r = 0.677; P < 0.05). Conclusion Our pre-experiment results signified that using QUS techniques for postoperative evaluation of bariatric surgery is promising. QUS techniques will be signed a widespread availability, real-time functionality, and low-cost approach for assessing hepatic steatosis before and after bariatric surgery in obese individuals, thus is capable for subsequent scale-up liver fat quantification. Ethics and dissemination The present research endeavor has been bestowed with the imprimatur of the Ethics Committee of the Hospital, as indicated by its Approval Number: 2023-KY-015. In due course, upon completion of the study, we intend to disseminate our findings by publishing them in a suitable academic journal, thereby facilitating their widespread utilization. Registration The trial is duly registered with the Chinese Clinical Trial Registry, and with a unique Trial Registration Number, ChiCTR2300069892, approved on March 28, 2023.
Collapse
Affiliation(s)
- Bin Chen
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qijie Lu
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Hu
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Sun
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Ying
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Hazelwood E, Sanderson E, Tan VY, Ruth KS, Frayling TM, Dimou N, Gunter MJ, Dossus L, Newton C, Ryan N, Pournaras DJ, O'Mara TA, Davey Smith G, Martin RM, Yarmolinsky J. Identifying molecular mediators of the relationship between body mass index and endometrial cancer risk: a Mendelian randomization analysis. BMC Med 2022; 20:125. [PMID: 35436960 PMCID: PMC9017004 DOI: 10.1186/s12916-022-02322-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/03/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Endometrial cancer is the most common gynaecological cancer in high-income countries. Elevated body mass index (BMI) is an established modifiable risk factor for this condition and is estimated to confer a larger effect on endometrial cancer risk than any other cancer site. However, the molecular mechanisms underpinning this association remain unclear. We used Mendelian randomization (MR) to evaluate the causal role of 14 molecular risk factors (hormonal, metabolic and inflammatory markers) in endometrial cancer risk. We then evaluated and quantified the potential mediating role of these molecular traits in the relationship between BMI and endometrial cancer using multivariable MR. METHODS Genetic instruments to proxy 14 molecular risk factors and BMI were constructed by identifying single-nucleotide polymorphisms (SNPs) reliably associated (P < 5.0 × 10-8) with each respective risk factor in previous genome-wide association studies (GWAS). Summary statistics for the association of these SNPs with overall and subtype-specific endometrial cancer risk (12,906 cases and 108,979 controls) were obtained from a GWAS meta-analysis of the Endometrial Cancer Association Consortium (ECAC), Epidemiology of Endometrial Cancer Consortium (E2C2) and UK Biobank. SNPs were combined into multi-allelic models and odds ratios (ORs) and 95% confidence intervals (95% CIs) were generated using inverse-variance weighted random-effects models. The mediating roles of the molecular risk factors in the relationship between BMI and endometrial cancer were then estimated using multivariable MR. RESULTS In MR analyses, there was strong evidence that BMI (OR per standard deviation (SD) increase 1.88, 95% CI 1.69 to 2.09, P = 3.87 × 10-31), total testosterone (OR per inverse-normal transformed nmol/L increase 1.64, 95% CI 1.43 to 1.88, P = 1.71 × 10-12), bioavailable testosterone (OR per natural log transformed nmol/L increase: 1.46, 95% CI 1.29 to 1.65, P = 3.48 × 10-9), fasting insulin (OR per natural log transformed pmol/L increase: 3.93, 95% CI 2.29 to 6.74, P = 7.18 × 10-7) and sex hormone-binding globulin (SHBG, OR per inverse-normal transformed nmol/L increase 0.71, 95% CI 0.59 to 0.85, P = 2.07 × 10-4) had a causal effect on endometrial cancer risk. Additionally, there was suggestive evidence that total serum cholesterol (OR per mg/dL increase 0.90, 95% CI 0.81 to 1.00, P = 4.01 × 10-2) had an effect on endometrial cancer risk. In mediation analysis, we found evidence for a mediating role of fasting insulin (19% total effect mediated, 95% CI 5 to 34%, P = 9.17 × 10-3), bioavailable testosterone (15% mediated, 95% CI 10 to 20%, P = 1.43 × 10-8) and SHBG (7% mediated, 95% CI 1 to 12%, P = 1.81 × 10-2) in the relationship between BMI and endometrial cancer risk. CONCLUSIONS Our comprehensive MR analysis provides insight into potential causal mechanisms linking BMI with endometrial cancer risk and suggests targeting of insulinemic and hormonal traits as a potential strategy for the prevention of endometrial cancer.
Collapse
Affiliation(s)
- Emma Hazelwood
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Bristol Medical School, University of Bristol, Bristol, UK
| | - Eleanor Sanderson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Bristol Medical School, University of Bristol, Bristol, UK
| | - Vanessa Y Tan
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Bristol Medical School, University of Bristol, Bristol, UK
| | - Katherine S Ruth
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Timothy M Frayling
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Niki Dimou
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Marc J Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Laure Dossus
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Claire Newton
- Department of Gynecology, St Michaels Hospital University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Neil Ryan
- Department of Gynecology, St Michaels Hospital University Hospitals Bristol NHS Foundation Trust, Bristol, UK
- The Academic Women's Health Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Dimitri J Pournaras
- Department of Upper GI and Bariatric/Metabolic Surgery, North Bristol NHS Trust, Southmead Hospital, Bristol, UK
| | - Tracy A O'Mara
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Bristol Medical School, University of Bristol, Bristol, UK
| | - Richard M Martin
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Bristol Medical School, University of Bristol, Bristol, UK
- National Institute for Health Research Bristol Biomedical Research Centre, University of Bristol, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - James Yarmolinsky
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
- Bristol Medical School, University of Bristol, Bristol, UK.
| |
Collapse
|
4
|
Martinou E, Stefanova I, Iosif E, Angelidi AM. Neurohormonal Changes in the Gut-Brain Axis and Underlying Neuroendocrine Mechanisms following Bariatric Surgery. Int J Mol Sci 2022; 23:3339. [PMID: 35328759 PMCID: PMC8954280 DOI: 10.3390/ijms23063339] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 02/05/2023] Open
Abstract
Obesity is a complex, multifactorial disease that is a major public health issue worldwide. Currently approved anti-obesity medications and lifestyle interventions lack the efficacy and durability needed to combat obesity, especially in individuals with more severe forms or coexisting metabolic disorders, such as poorly controlled type 2 diabetes. Bariatric surgery is considered an effective therapeutic modality with sustained weight loss and metabolic benefits. Numerous genetic and environmental factors have been associated with the pathogenesis of obesity, while cumulative evidence has highlighted the gut-brain axis as a complex bidirectional communication axis that plays a crucial role in energy homeostasis. This has led to increased research on the roles of neuroendocrine signaling pathways and various gastrointestinal peptides as key mediators of the beneficial effects following weight-loss surgery. The accumulate evidence suggests that the development of gut-peptide-based agents can mimic the effects of bariatric surgery and thus is a highly promising treatment strategy that could be explored in future research. This article aims to elucidate the potential underlying neuroendocrine mechanisms of the gut-brain axis and comprehensively review the observed changes of gut hormones associated with bariatric surgery. Moreover, the emerging role of post-bariatric gut microbiota modulation is briefly discussed.
Collapse
Affiliation(s)
- Eirini Martinou
- Department of Upper Gastrointestinal Surgery, Frimley Health NHS Foundation Trust, Camberley GU16 7UJ, UK;
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Irena Stefanova
- Department of General Surgery, Frimley Health NHS Foundation Trust, Camberley GU16 7UJ, UK;
| | - Evangelia Iosif
- Department of General Surgery, Royal Surrey County Hospital, Guildford GU2 7XX, UK;
| | - Angeliki M. Angelidi
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
5
|
Preservation of Fat-free Mass in the first year after Bariatric Surgery: A systematic review and meta-analysis of 122 studies and 10758 participants. Surg Obes Relat Dis 2022; 18:964-982. [DOI: 10.1016/j.soard.2022.02.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 02/07/2022] [Accepted: 02/21/2022] [Indexed: 02/07/2023]
|
6
|
Fatima F, Hjelmesæth J, Birkeland KI, Gulseth HL, Hertel JK, Svanevik M, Sandbu R, Småstuen MC, Hartmann B, Holst JJ, Hofsø D. Gastrointestinal Hormones and β-Cell Function After Gastric Bypass and Sleeve Gastrectomy: A Randomized Controlled Trial (Oseberg). J Clin Endocrinol Metab 2022; 107:e756-e766. [PMID: 34463768 DOI: 10.1210/clinem/dgab643] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Indexed: 02/04/2023]
Abstract
CONTEXT Whether Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG) differentially affect postprandial gastrointestinal hormones and β-cell function in type 2 diabetes remains unclear. OBJECTIVE We aimed to compare gastrointestinal hormones and β-cell function, assessed by an oral glucose tolerance test (OGTT) 5 weeks and 1 year after surgery, hypothesizing higher glucagon-like peptide-1 (GLP-1) levels and greater β-cell response to glucose after RYGB than after SG. METHODS This study was a randomized, triple-blind, single-center trial at a tertiary care center in Norway. The primary outcomes were diabetes remission and IVGTT-derived β-cell function. Participants with obesity and type 2 diabetes were allocated (1:1) to RYGB or SG. We measured gastrointestinal hormone profiles and insulin secretion as β-cell glucose sensitivity (β-GS) derived from 180-minute OGTTs. RESULTS Participants were 106 patients (67% women), mean (SD) age 48 (10) years. Diabetes remission rates at 1 year were higher after RYGB than after SG (77% vs 48%; P = 0.002). Incremental area under the curve (iAUC0-180) GLP-1 and β-GS increased more after RYGB than after SG, with 1-year between-group difference 1173 pmol/L*min (95% CI, 569-1776; P = 0.0010) and 0.45 pmol/kg/min/mmol (95% CI, 0.15-0.75; P = 0.0032), respectively. After surgery, fasting and postprandial ghrelin levels were higher and decremental AUC0-180 ghrelin, iAUC0-180 glucose-dependent insulinotropic polypeptide, and iAUC0-60 glucagon were greater after RYGB than after SG. Diabetes remission at 1 year was associated with higher β-GS and higher GLP-1 secretion. CONCLUSION RYGB was associated with greater improvement in β-cell function and higher postprandial GLP-1 levels than SG.
Collapse
Affiliation(s)
- Farhat Fatima
- Morbid Obesity Centre, Vestfold Hospital Trust, 3103 Tønsberg, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway
| | - Jøran Hjelmesæth
- Morbid Obesity Centre, Vestfold Hospital Trust, 3103 Tønsberg, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
| | - Kåre Inge Birkeland
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway
- Department of Transplantation, Oslo University Hospital, 0424 Oslo, Norway
| | - Hanne Løvdal Gulseth
- Department of Chronic Diseases and Ageing, Norwegian Institute of Public Health, 0213 Oslo, Norway
| | | | - Marius Svanevik
- Morbid Obesity Centre, Vestfold Hospital Trust, 3103 Tønsberg, Norway
- Department of Surgery, Vestfold Hospital Trust, 3103 Tønsberg, Norway
| | - Rune Sandbu
- Morbid Obesity Centre, Vestfold Hospital Trust, 3103 Tønsberg, Norway
- Department of Surgery, Vestfold Hospital Trust, 3103 Tønsberg, Norway
| | - Milada Cvancarova Småstuen
- Morbid Obesity Centre, Vestfold Hospital Trust, 3103 Tønsberg, Norway
- Department of Nutrition and Management, Oslo Metropolitan University, 0130 Oslo, Norway
| | - Bolette Hartmann
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jens Juul Holst
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Dag Hofsø
- Morbid Obesity Centre, Vestfold Hospital Trust, 3103 Tønsberg, Norway
| |
Collapse
|
7
|
Kim KH, Kim YH, Seo KW, Yoon KY, Shin YM, Choi YS, Kim BK. Immediate Changes of Glucose Metabolism After Gastretomy for Early Gastric Cancer in Patients with Type 2 Diabetes. KOSIN MEDICAL JOURNAL 2021. [DOI: 10.7180/kmj.2021.36.1.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Objectives It is well known that type 2 diabetes (T2DM) is dramatically improved after bariatric surgery, although the mechanisms have not been clearly identified. The skill required for gastric surgery for gastric cancer is very similar to that needed in bariatric surgery. In this study, we evaluated the immediate improvement of T2DM after gastrectomy for gastric cancer. Methods A total of nine patients who were diagnosed with early gastric cancer (EGC) and already had T2DM underwent a 75 g oral glucose tolerance test (OGTT) before surgery and within two weeks after gastrectomy. Glucose, insulin, and c-peptide were measured before, and 30 and 60 minutes after ingesting 75 g of glucose. From these trials, we calculated the HOMA-IR, insulinogenic index, Matsuda index, and area under the curve (AUC). Results The mean age of participants was 57.23 ± 11.08 years and eight of them were men. HOMA-IR (4.2 vs. 2.3, P = 0.012) levels were decreased after surgery. There were no significant differences of insulinogenic index, fasting blood sugar before and after surgery. The Matsuda index (3.3 vs. 8.3, P = 0.002) was significantly increased and AUC (512.9 vs. 388.7 mg-hr/dL, P > 0.001) upon 75 g OGTT was significantly decreased after surgery. Conclusions Insulin sensitivity was immediately improved after gastrectomy for early gastric cancer in patients with T2DM.
Collapse
|
8
|
Haghighat N, Ashtari-Larky D, Aghakhani L, Asbaghi O, Hoseinpour H, Hosseini B, Shahabinezhad A, Pourmohammad A, Hosseini SV, Amini M, Clark CCT, Bananzadeh A. How Does Fat Mass Change in the First Year After Bariatric Surgery? A Systemic Review and Meta-Analysis. Obes Surg 2021; 31:3799-3821. [PMID: 34089442 DOI: 10.1007/s11695-021-05512-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 12/29/2022]
Abstract
This systematic review and meta-analysis investigated the time-course effect of different type of bariatric surgeries (BS) up to 1 year post-surgery on fat mass (FM) and body fat percentage (BFP) in patients with morbid obesity. We searched PubMed, Scopus, EMBASE, ISI web of science, and Cochrane databases from October 2002 until May 2020 with no restriction in the English language, to find studies examining the effect of BS on FM (kg) and BFP (%) in morbid obese patients. Meta-analysis of 103 studies carried out on data of 18,166 and 14,575 morbid obese patients following BS, showed that BS was associated with a substantial decrease in FM and BFP, respectively, in 1 month (- 8.17 kg [95% CI - 9.07, - 7.27] and - 1.51% [95% CI - 2.56, - 0.46]), 3 months (- 15.75 [95% CI - 17.49, - 14.0] and - 4.90 [95% CI - 5.97, - 3.83]), 6 months (- 22.51 [95% CI - 23.93, - 21.09] and - 8.56% [95% CI - 9.63, - 7.49]), and 12 months (- 29.69 [95% CI - 31.3, - 28.09] and - 13.49% [95% CI - 14.52, - 12.40]) after the surgery. In conclusion, BS was associated with sustained declines in FM and BFP, from 1 to 12 months, with no indication of plateau phase post-surgery post-operatively. The present study emphasizes that post-bariatric care should have more focus on FM loss during 1-year post-surgery to identify the patients at risk for fat loss plateau.
Collapse
Affiliation(s)
- Neda Haghighat
- Laparoscopy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Damoon Ashtari-Larky
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ladan Aghakhani
- Laparoscopy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omid Asbaghi
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran, Tehran, Iran
| | - Hamidreza Hoseinpour
- Laparoscopy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Babak Hosseini
- Laparoscopy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Shahabinezhad
- Laparoscopy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Arash Pourmohammad
- Laparoscopy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Vahid Hosseini
- Laparoscopy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Colorectal Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoud Amini
- Laparoscopy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Cain C T Clark
- Center for Intelligent Healthcare, Coventry University, Coventry, CV1 5FB, UK
| | - Alimohammad Bananzadeh
- Laparoscopy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran. .,Colorectal Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
9
|
Conversion from Prediabetes to Diabetes in Individuals with Obesity, 5-Years Post-Band, Sleeve, and Gastric Bypass Surgeries. Obes Surg 2019; 29:3901-3906. [PMID: 31313239 DOI: 10.1007/s11695-019-04090-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND Identifying risk factors for conversion to diabetes among individuals with obesity and prediabetes is important for preventing diabetes. PURPOSE We assessed conversion rates to diabetes 5 years after three types of metabolic surgery and examined predictors of diabetes development. METHODS We accessed data of individuals with prediabetes, defined as fasting glucose (FG) 100-125 mg/dL (5.6-6.9 mmol/L) or HbA1c 5.7-6.4% at baseline (preoperatively), who underwent metabolic surgeries in Clalit Health Services during 2002-2011. RESULTS Of 1,756 individuals with prediabetes, 819 underwent gastric banding (GB), 845 sleeve gastrectomy (SG), and 92 Roux-en-Y gastric bypass (RYGB). Mean age was 41.6 years and 73.5% were women. Five years post-surgery, 177 (10.1%) had developed diabetes. Conversion rates by type of surgery were 14.4%, 6.3%, and 6.5% for GB, SG, and RYGB, respectively (p < 0.001). Conversion was more rapid following GB than SG or RYGB (χ2(2) = 29.67, p < 0.005). In a multiple-logistic-regression model, predictors of diabetes development 5 years postoperatively were (1) weight loss during the first postoperative year and (2) preoperative levels of both FG and HbA1c within the prediabetes range. Baseline weight, age, and sex, were not associated with conversion to diabetes. Conversion rates were lower (4.7%) five years postoperatively for patients who lost > 25% of their baseline weight, compared to those who lost less than 15% of their weight during the first postoperative year: (14.0% < 0.001). CONCLUSIONS Our findings emphasize the importance of preoperative glycemic control and weight loss during the first year postoperatively, for the long-term prevention of diabetes in patients with prediabetes undergoing metabolic surgery.
Collapse
|
10
|
Jirapinyo P, Jin DX, Qazi T, Mishra N, Thompson CC. A Meta-Analysis of GLP-1 After Roux-En-Y Gastric Bypass: Impact of Surgical Technique and Measurement Strategy. Obes Surg 2018; 28:615-626. [PMID: 28871519 DOI: 10.1007/s11695-017-2913-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Roux-en-Y gastric bypass (RYGB) is an effective treatment for diabetes. Glucagon-like peptide-1 (GLP-1) is a gut hormone that is important to glucose homeostasis. OBJECTIVE This study aimed to assess GLP-1 level and its predictors after RYGB. METHODS The study design was a meta-analysis. The data sources were MEDLINE, EMBASE, Web of Science, and the Cochrane Databases. The study selection composed of studies with pre- and post-RYGB levels. The main outcomes were as follows: Primary outcome was the change in postprandial GLP-1 levels after RYGB. Secondary outcomes included the changes in fasting glucose, fasting insulin, and fasting GLP-1 levels after RYGB. Meta-regression to determine predictors of changes in GLP-1 levels was performed. Outcomes were reported using Hedge's g. RESULTS Twenty-four studies with 368 patients were included. Postprandial GLP-1 levels increased after RYGB (Hedge's g = 1.29, p < 0.0001), while fasting GLP-1 did not change (p = 0.23). Peak postprandial GLP-1 levels gave the most consistent results (I 2 = 9.11). Fasting glucose and insulin levels decreased after RYGB (p < 0.0001). Roux limb length was a significant predictor for amount of GLP-1 increase (β = - 0.01, p = 0.02). Diabetes status, amount of weight loss, length of biliopancreatic limb, and time of measurement were not significant predictors (p > 0.05). CONCLUSION Postprandial GLP-1 levels increase after RYGB, while fasting levels remain unchanged. Shorter Roux limb length is associated with greater increase in postprandial GLP-1, which may lead to better glycemic control in this population.
Collapse
Affiliation(s)
- Pichamol Jirapinyo
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - David X Jin
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Taha Qazi
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Nitin Mishra
- Department of Surgery, Mayo Clinic Hospital, Phoenix, AZ, USA
| | - Christopher C Thompson
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|
11
|
Abstract
The prevalence of obesity in combination with sarcopenia (the age-related loss of muscle mass and strength or physical function) is increasing in adults aged 65 years and older. A major subset of adults over the age of 65 is now classified as having sarcopenic obesity, a high-risk geriatric syndrome predominantly observed in an ageing population that is at risk of synergistic complications from both sarcopenia and obesity. This Review discusses pathways and mechanisms leading to muscle impairment in older adults with obesity. We explore sex-specific hormonal changes, inflammatory pathways and myocellular mechanisms leading to the development of sarcopenic obesity. We discuss the evolution, controversies and challenges in defining sarcopenic obesity and present current body composition modalities used to assess this condition. Epidemiological surveys form the basis of defining its prevalence and consequences beyond comorbidity and mortality. Current treatment strategies, and the evidence supporting them, are outlined, with a focus on calorie restriction, protein supplementation and aerobic and resistance exercises. We also describe weight loss-induced complications in patients with sarcopenic obesity that are relevant to clinical management. Finally, we review novel and potential future therapies including testosterone, selective androgen receptor modulators, myostatin inhibitors, ghrelin analogues, vitamin K and mesenchymal stem cell therapy.
Collapse
Affiliation(s)
- John A Batsis
- Sections of General Internal Medicine and Weight and Wellness, and the Dartmouth Centers for Health and Aging, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
- Geisel School of Medicine at Dartmouth, The Dartmouth Institute for Health Policy and Clinical Practice, The Health Promotion Research Center and the Norris Cotton Cancer Center, Dartmouth College, Hanover, NH, USA.
| | - Dennis T Villareal
- Division of Endocrinology, Diabetes and Metabolism, Baylor College of Medicine, Houston, TX, USA
- Center for Translational Research on Inflammatory Diseases, Michael E DeBakey VA Medical Center, Houston, TX, USA
| |
Collapse
|
12
|
Zhai H, Li Z, Peng M, Huang Z, Qin T, Chen L, Li H, Zhang H, Zhang W, Xu G. Takeda G Protein-Coupled Receptor 5-Mechanistic Target of Rapamycin Complex 1 Signaling Contributes to the Increment of Glucagon-Like Peptide-1 Production after Roux-en-Y Gastric Bypass. EBioMedicine 2018; 32:201-214. [PMID: 29859856 PMCID: PMC6020750 DOI: 10.1016/j.ebiom.2018.05.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/20/2018] [Accepted: 05/21/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The mechanism by which Roux-en-Y Gastric Bypass (RYGB) increases the secretion of glucagon-like peptide-1 (GLP-1) remains incompletely defined. Here we investigated whether TGR5-mTORC1 signaling mediates the RYGB-induced alteration in GLP-1 production in mice and human beings. METHODS Circulating bile acids, TGR5-mTORC1 signaling, GLP-1 synthesis and secretion were determined in lean or obese male C57BL/6 mice with or without RYGB operation, as well as in normal glycemic subjects, obese patients with type 2 diabetes before and after RYGB. RESULTS Positive relationships were observed among circulating bile acids, ileal mechanistic target of rapamycin complex 1 (mTORC1) signaling and GLP-1 during changes in energy status in the present study. RYGB increased circulating bile acids, ileal Takeda G protein-coupled receptor 5 (TGR5) and mTORC1 signaling activity, as well as GLP-1 production in both mice and human subjects. Inhibition of ileal mTORC1 signaling by rapamycin significantly attenuated the stimulation of bile acid secretion, TGR5 expression and GLP-1 synthesis induced by RYGB in lean and diet-induced obese mice. GLP-1 production and ileal TGR5-mTORC1 signaling were positively correlated with plasma deoxycholic acid (DCA) in mice. Treatment of STC-1 cells with DCA stimulated the production of GLP-1. This effect was associated with a significant enhancement of TGR5-mTORC1 signaling. siRNA knockdown of mTORC1 or TGR5 abolished the enhancement of GLP-1 synthesis induced by DCA. DCA increased interaction between mTOR-regulatory-associated protein of mechanistic target of rapamycin (Raptor) and TGR5 in STC-1 cells. INTERPRETATION Deoxycholic acid-TGR5-mTORC1 signaling contributes to the up-regulation of GLP-1 production after RYGB.
Collapse
Affiliation(s)
- Hening Zhai
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China; Endoscopy Center, The First Affiliated Hospital of Jinan University, 613 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510630, China
| | - Zhi Li
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Miao Peng
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Zhaoqi Huang
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Tingfeng Qin
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Linxi Chen
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Hanbing Li
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Heng Zhang
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Weizhen Zhang
- Shenzhen University Diabetes Center, Shenzhen University Health Science Center, Shenzhen, Guangdong 518060, China; Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI 48109-0346, USA.
| | - Geyang Xu
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China.
| |
Collapse
|
13
|
Göbl CS, Bozkurt L, Tura A, Leutner M, Andrei L, Fahr L, Husslein P, Eppel W, Kautzky-Willer A. Assessment of glucose regulation in pregnancy after gastric bypass surgery. Diabetologia 2017; 60:2504-2513. [PMID: 28918470 PMCID: PMC6448941 DOI: 10.1007/s00125-017-4437-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/19/2017] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS Roux-en-Y gastric bypass (RYGB) surgery is characterised by glycaemic variability. Prospective studies of glucose metabolism in pregnancy after RYGB are not available, therefore this study aimed to evaluate physiological alterations in glucose metabolism in pregnancy following RYGB. METHODS Sixty-three pregnant women (25 who underwent RYGB, 19 non-operated obese control women and 19 normal weight control women) were included. Frequently sampled 3 h OGTTs and 1 h IVGTTs were performed between 24 and 28 weeks of gestation and, in a subgroup, were repeated at 3-6 months after delivery. RESULTS We observed major alterations in glucose kinetics during the OGTT, including an early increase in plasma glucose followed by hypoglycaemia in 90% of women who had previously undergone RYGB. The higher degree of glycaemic variability in this group was accompanied by increased insulin, C-peptide and glucagon concentrations after oral glucose load, whereas no differences in insulin response were observed after parenteral glucose administration (RYGB vs normal weight). IVGTT data suggested improved insulin sensitivity (mean difference 0.226 × 10-4 min-1 [pmol/l]-1 [95% CI 0.104, 0.348]; p < 0.001) and disposition index in pregnancies after RYGB when compared with obese control women. However, subtle alterations in insulin action and beta cell function were still observed when comparing women who had undergone RYGB with the normal-weight control group. Moreover, we observed that fetal growth was associated with maternal glucose nadir levels and insulin secretion in offspring of those who had previously undergone RYGB. CONCLUSIONS/INTERPRETATION Pregnancies after RYGB are affected by altered postprandial glucose, insulin and C-peptide dynamics. Insulin sensitivity is improved by RYGB, although subtle alterations in beta cell function are observed. Longitudinal studies are needed to assess potential consequences for fetal development and pregnancy outcomes.
Collapse
Affiliation(s)
- Christian S Göbl
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-maternal Medicine, Medical University of Vienna, Vienna, Austria
| | - Latife Bozkurt
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Gender Medicine Unit, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Andrea Tura
- Metabolic Unit, Institute of Neuroscience, National Research Council, Padova, Italy
| | - Michael Leutner
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Gender Medicine Unit, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Laura Andrei
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-maternal Medicine, Medical University of Vienna, Vienna, Austria
| | - Lukas Fahr
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-maternal Medicine, Medical University of Vienna, Vienna, Austria
| | - Peter Husslein
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-maternal Medicine, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Eppel
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-maternal Medicine, Medical University of Vienna, Vienna, Austria
| | - Alexandra Kautzky-Willer
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Gender Medicine Unit, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria.
| |
Collapse
|
14
|
Contribution of Malabsorption to Weight Loss After Roux-en-Y Gastric Bypass: a Systematic Review. Obes Surg 2017; 27:2194-2206. [PMID: 28585108 DOI: 10.1007/s11695-017-2762-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
15
|
Kalra J, Dhar A. Double-stranded RNA-dependent protein kinase signalling and paradigms of cardiometabolic syndrome. Fundam Clin Pharmacol 2017; 31:265-279. [PMID: 27992964 DOI: 10.1111/fcp.12261] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 11/30/2016] [Accepted: 12/16/2016] [Indexed: 12/28/2022]
Affiliation(s)
- Jaspreet Kalra
- Department of Pharmacy; Birla Institute of Technology and Sciences Pilani, Hyderabad Campus; Jawahar Nagar Shameerpet, Hyderabad Andhra Pradesh 500078 India
| | - Arti Dhar
- Department of Pharmacy; Birla Institute of Technology and Sciences Pilani, Hyderabad Campus; Jawahar Nagar Shameerpet, Hyderabad Andhra Pradesh 500078 India
| |
Collapse
|
16
|
Campos J, Ramos A, Szego T, Zilberstein B, Feitosa H, Cohen R. THE ROLE OF METABOLIC SURGERY FOR PATIENTS WITH OBESITY GRADE I AND TYPE 2 DIABETES NOT CONTROLLED CLINICALLY. ACTA ACUST UNITED AC 2017; 29 Suppl 1:102-106. [PMID: 27409057 PMCID: PMC5064276 DOI: 10.1590/0102-6720201600s10025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/19/2016] [Indexed: 12/13/2022]
Abstract
Introduction Even considering the advance of the medical treatment in the last 20 years with new and more effective drugs, the outcomes are still disappointing as the control of obesity and type 2 Diabetes Mellitus (T2DM) with a large number of patients under the medical treatment still not reaching the desired outcomes. Objective: To present a Metabolic Risk Score to better guide the surgical indication for T2DM patients with body mass index (BMI) where surgery for obesity is still controversial. Method: Research was conducted in Pubmed, Medline, Pubmed Central, Scielo and Lilacs between 2003-2015 correlating headings: metabolic surgery, obesity and type 2 diabetes mellitus. In addition, representatives of the societies involved, as an expert panel, issued opinions. Results: Forty-five related articles were analyzed by evidence-based medicine criteria. Grouped opinions sought to answer the following questions: Why metabolic and not bariatric surgery?; Mechanisms involved in glycemic control; BMI as a single criterion for surgical indication for uncontrolled T2DM; Results of metabolic surgery studies in BMI<35 kg/m2; Safety of metabolic surgery in patients with BMI<35 kg/m2; Long-term effects of surgery in patients with baseline BMI<35 kg/m2 and Proposal for a Metabolic Risk Score. Conclusion: Metabolic surgery has well-defined mechanisms of action both in experimental and human studies. Gastrointestinal interventions in T2DM patients with IMC≤35 kg/m2 has similar safety and efficacy when compared to groups with greater BMIs, leading to the improvement of diabetes in a superior manner than clinical treatment and lifestyle changes, in part through weight loss independent mechanisms . There is no correlation between baseline BMI and weight loss in the long term with the success rate after any surgical treatment. Gastrointestinal surgery treatment may be an option for patients with T2DM without adequate clinical control, with a BMI between 30 and 35, after thorough evaluation following the parameters detailed in Metabolic Risk Score defined by the surgical societies. Roux-en-Y gastric bypass (RYGB), because of its well known safety and efficacy and longer follow-up studies, is the main surgical technique indicated for patients eligible for surgery through the Metabolic Risk Score. The vertical sleeve gastrectomy may be considered if there is an absolute contraindication for the RYGB. T2DM patients should be evaluated by the multiprofessional team that will assess surgical eligibility, preoperative work up, follow up and long term monitoring for micro and macrovascular complications.
Collapse
Affiliation(s)
- Josemberg Campos
- Inter Societary guideline bythe Brazilian Society for Bariatric and Metabolic Surgery (SBCBM), Brazilian College of Surgeons (CBC) and Brazilian College of Digestive Surgery (CBCD), São Paulo, SP, Brazil
| | - Almino Ramos
- Inter Societary guideline bythe Brazilian Society for Bariatric and Metabolic Surgery (SBCBM), Brazilian College of Surgeons (CBC) and Brazilian College of Digestive Surgery (CBCD), São Paulo, SP, Brazil
| | - Thomaz Szego
- Inter Societary guideline bythe Brazilian Society for Bariatric and Metabolic Surgery (SBCBM), Brazilian College of Surgeons (CBC) and Brazilian College of Digestive Surgery (CBCD), São Paulo, SP, Brazil
| | - Bruno Zilberstein
- Inter Societary guideline bythe Brazilian Society for Bariatric and Metabolic Surgery (SBCBM), Brazilian College of Surgeons (CBC) and Brazilian College of Digestive Surgery (CBCD), São Paulo, SP, Brazil
| | - Heládio Feitosa
- Inter Societary guideline bythe Brazilian Society for Bariatric and Metabolic Surgery (SBCBM), Brazilian College of Surgeons (CBC) and Brazilian College of Digestive Surgery (CBCD), São Paulo, SP, Brazil
| | - Ricardo Cohen
- Inter Societary guideline bythe Brazilian Society for Bariatric and Metabolic Surgery (SBCBM), Brazilian College of Surgeons (CBC) and Brazilian College of Digestive Surgery (CBCD), São Paulo, SP, Brazil
| |
Collapse
|
17
|
Changes of insulin sensitivity and secretion after bariatric/metabolic surgery. Surg Obes Relat Dis 2016; 12:1199-205. [PMID: 27568471 DOI: 10.1016/j.soard.2016.05.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 05/11/2016] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes (T2D) is classically characterized by failure of pancreatic β-cell function and insulin secretion to compensate for a prevailing level of insulin resistance, typically associated with visceral obesity. Although this is usually a chronic, progressive disease in which delay of end-organ complications is the primary therapeutic goal for medical and behavioral approaches, several types of bariatric surgery, especially those that include intestinal bypass components, exert powerful antidiabetes effects to yield remission of T2D in most cases. It has become increasingly clear that in addition to the known benefits of acute caloric restriction and chronic weight loss to ameliorate T2D, bariatric/metabolic operations also engage a variety of weight-independent mechanisms to improve glucose homeostasis, enhancing insulin sensitivity and secretion to varying degrees depending on the specific operation. In this paper, we review the effects of Roux-en-Y gastric bypass, biliopancreatic diversion, and vertical sleeve gastrectomy on the primary determinants of glucose homeostasis: insulin sensitivity, insulin secretion, and, to the lesser extent that it is known, insulin-independent glucose disposal. A full understanding of these effects should help optimize surgical and device-based designs to provide maximal antidiabetes impact, and it holds the promise to identify targets for possible novel diabetes pharmacotherapeutics. These insights also contribute to the conceptual rationale for use of bariatric operations as "metabolic surgery," employed primarily to treat T2D, including among patients not obese enough to qualify for surgery based on traditional criteria related to high body mass index.
Collapse
|
18
|
Andrade HFDA, Pedrosa W, Diniz MDFHS, Passos VMA. Adverse effects during the oral glucose tolerance test in post-bariatric surgery patients. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2016; 60:307-13. [PMID: 26910630 PMCID: PMC10118721 DOI: 10.1590/2359-3997000000149] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 11/29/2015] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The oral glucose tolerance test (OGTT) is used in the screening of gestational diabetes, in diagnosis of type 2 diabetes in conjunction with fasting blood glucose and glycated hemoglobin. The aim of this study was to examine the incidence and risk factors of adverse effects of OGTT in patients who underwent bariatric surgery, in addition to proposing standardization for ordering the OGTT in these patients. SUBJECTS AND METHODS This study assessed the incidence of adverse effects in 128 post-bariatric surgery patients who underwent the OGTT. Descriptive and logistic regression analysis were performed, the dependent variables were defined as the presence of signs (tremor, profuse sweating, tachycardia), symptoms (nausea, diarrhea, dizziness, weakness), and hypoglycemia (blood glucose ≤ 50 mg/dL). RESULTS One hundred and seventeen participants (91.4%) were female; 38 (29.7%) participants were pregnant. High incidence (64.8%) of adverse effects was observed: nausea (38.4%), dizziness (30.5%), weakness (25.8%), diarrhea (23.4%), hypoglycemia (14.8%), tachycardia (14.1%), tremor (13.3%), profuse sweating (12.5%) and one case of severe hypoglycemia (24 mg/dL). The presence of signs was associated with hypoglycemia (OR = 8.1, CI 95% 2.6-25.1). The arterial hypertension persisted as a risk factor for the incidence of signs (OR = 3.6, CI 95% 1.2-11.3). Fasting glucose below 75 mg/dL increased the risk of hypoglycemia during the test (OR = 9.5, CI 95% 2.6-35.1). CONCLUSION In this study, high incidence of adverse effects during the OGTT was observed in post-bariatric surgery patients. If these results are confirmed by further studies, the indication and regulation of the OGTT procedure must be reviewed for these patients.
Collapse
Affiliation(s)
- Heliana Fernanda de Albuquerque Andrade
- Departamento de Clínica Médica, Programa de Pós-Graduação em Ciências Aplicadas à Saúde do Adulto, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brasil
| | - William Pedrosa
- Departamento de Clínica Médica, Programa de Pós-Graduação em Ciências Aplicadas à Saúde do Adulto, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brasil
| | - Maria de Fátima Haueisen Sander Diniz
- Departamento de Clínica Médica, Programa de Pós-Graduação em Ciências Aplicadas à Saúde do Adulto, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brasil
| | - Valéria Maria Azeredo Passos
- Departamento de Clínica Médica, Programa de Pós-Graduação em Ciências Aplicadas à Saúde do Adulto, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brasil
| |
Collapse
|
19
|
Manning S, Pucci A, Batterham RL. GLP-1: a mediator of the beneficial metabolic effects of bariatric surgery? Physiology (Bethesda) 2015; 30:50-62. [PMID: 25559155 DOI: 10.1152/physiol.00027.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There has been increasing interest in the role that gut hormones may play in contributing to the physiological changes produced by certain bariatric procedures, such as Roux-en-Y gastric bypass and sleeve gastrectomy. Here, we review the evidence implicating one such gut hormone, glucagon-like peptide-1, as a mediator of the metabolic benefits of these two procedures.
Collapse
Affiliation(s)
- Sean Manning
- Department of Medicine, Centre for Obesity Research, Rayne Institute, University College London, London, United Kingdom; UCLH Centre for Weight Loss, Metabolic and Endocrine Surgery, University College London Hospitals, London, United Kingdom; National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Andrea Pucci
- Department of Medicine, Centre for Obesity Research, Rayne Institute, University College London, London, United Kingdom; UCLH Centre for Weight Loss, Metabolic and Endocrine Surgery, University College London Hospitals, London, United Kingdom
| | - Rachel L Batterham
- Department of Medicine, Centre for Obesity Research, Rayne Institute, University College London, London, United Kingdom; UCLH Centre for Weight Loss, Metabolic and Endocrine Surgery, University College London Hospitals, London, United Kingdom; National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, United Kingdom
| |
Collapse
|
20
|
Abstract
The obesity epidemic, combined with the lack of available and effective treatments for morbid obesity, is a scientific and public health priority. Worldwide, bariatric and metabolic surgeries are increasingly being performed to effectively aid weight loss in patients with severe obesity, as well as because of the favourable metabolic effects of the procedures. The positive effects of bariatric surgery, especially with respect to improvements in type 2 diabetes mellitus, have expanded the eligibility criteria for metabolic surgery to patients with diabetes mellitus and a BMI of 30-35 kg/m(2). However, the limitations of BMI, both in the diagnosis and follow-up of patients, need to be considered, particularly for determining the actual adiposity and fat distribution of the patients following weight loss. Understanding the characteristics shared by bariatric and metabolic surgeries, as well as their differential aspects and outcomes, is required to enhance patient benefits and operative achievements. For a holistic approach that focuses on the multifactorial effects of bariatric and metabolic surgery to be possible, a paradigm shift that goes beyond the pure semantics is needed. Such a shift could lead to profound clinical implications for eligibility criteria and the definition of success of the surgical approach.
Collapse
Affiliation(s)
- Gema Frühbeck
- Department of Endocrinology &Nutrition, CIBEROBN, Clínica Universidad de Navarra, University of Navarra, IdiSNA, Avda. Pío XII 36, 31008 Pamplona, Spain
| |
Collapse
|
21
|
Nguyen NQ, Debreceni TL, Burgstad CM, Wishart JM, Bellon M, Rayner CK, Wittert GA, Horowitz M. Effects of Posture and Meal Volume on Gastric Emptying, Intestinal Transit, Oral Glucose Tolerance, Blood Pressure and Gastrointestinal Symptoms After Roux-en-Y Gastric Bypass. Obes Surg 2015; 25:1392-1400. [PMID: 25502436 DOI: 10.1007/s11695-014-1531-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The purpose of this study is to determine the effects of posture and drink volume on gastric/pouch emptying (G/PE), intestinal transit, hormones, absorption, glycaemia, blood pressure and gastrointestinal (GI) symptoms after gastric bypass (Roux-en-Y gastric bypass (RYGB)). METHODS Ten RYGB subjects were studied on four occasions in randomized order (sitting vs. supine posture; 50 vs. 150 ml of labelled water mixed with 3 g 3-O-methyl-D-glucose (3-OMG) and 50 g glucose). G/PE, caecal arrival time (CAT), blood glucose, plasma insulin, glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), peptide YY (PYY), 3-OMG, blood pressure, heart rate and GI symptoms were assessed over 240 min. Controls were ten volunteers with no medical condition or previous abdominal surgery, who were studied with the 150-ml drink in the sitting position. RESULTS Compared to controls, PE (P < 0.001) and CAT (P < 0.001) were substantially more rapid in RYGB subjects. In RYGB, PE was more rapid in the sitting position (2.5 ± 0.7 vs. 16.6 ± 5.3 min, P = 0.02) and tends to be faster after 150 ml than the 50-ml drinks (9.5 ± 2.9 vs. 14.0 ± 3.5 min, P = 0.16). The sitting position and larger volume drinks were associated with greater releases of insulin, GLP-1 and PYY, as well as more hypotension (P < 0.01), tachycardia (P < 0.01) and postprandial symptoms (P < 0.001). CONCLUSIONS Pouch emptying, blood pressure and GI symptoms after RYGB are dependent on both posture and meal volume.
Collapse
Affiliation(s)
- Nam Q Nguyen
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, North Terrace, Adelaide, SA, 5000, Australia,
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Valezi AC, Marson AC, Merguizo RA, Costa FL. Roux-en-Y gastric bypass: limb length and weight loss. ABCD-ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA 2015; 27 Suppl 1:56-8. [PMID: 25409968 PMCID: PMC4743521 DOI: 10.1590/s0102-6720201400s100014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 07/15/2014] [Indexed: 12/31/2022]
Abstract
Background Roux-en-Y gastric bypass is a surgical technique widely used in the treatment of
obesity. It is unclear, however, if the length of the biliopancreatic and
alimentary limb interferes with the magnitude of weight loss. Aim To evaluate if the length of these limbs is related to the percentage of weight
loss one year after surgery. Method One hundred and twenty obese people underwent surgery between 2009 and 2011.
Patients were inserted into four groups: A) biliopancreatic limb with 50 cm length
and alimentary limb with100 cm length; B) biliopancreatic limb with 50 cm length
and alimentary limb with 150 cm length; C) biliopancreatic limb with 100 cm length
and alimentary limb with100 cm length; D) biliopancreatic limb with 100 cm length
and alimentary limb with150 cm length. Age, gender, body mass index and the
percentage of total weight loss were analyzed. Data were collected preoperatively
and one year after surgery. The groups were compared and weight loss compared
between groups. Results The follow-up occurred in 78.3% of the sample. The composition of the groups was
similar, with no statistical significance. The average age was 43 years in groups
A, C and D and 42 years in group B. The female gender predominated in all groups
(about 60% of the sample). The mean body mass index was 46 kg/m2 for
groups A, C and D and 42 kg/m2 in group B. The percentage of weight
loss was 33% for group A and 34% for groups B, C and D. There was no significant
difference among groups. Conclusion Different lengths of the biliopancreatic and alimentary limbs did not affect the
percentage of total weight loss.
Collapse
Affiliation(s)
| | - Antonio César Marson
- Departamento de Cirurgia, Universidade Estadual de Londrina, Londrina, PR, Brazil
| | | | - Fernando Leão Costa
- Departamento de Cirurgia, Universidade Estadual de Londrina, Londrina, PR, Brazil
| |
Collapse
|
23
|
Heinzmann SS, Schmitt-Kopplin P. Deep Metabotyping of the Murine Gastrointestinal Tract for the Visualization of Digestion and Microbial Metabolism. J Proteome Res 2015; 14:2267-77. [DOI: 10.1021/acs.jproteome.5b00034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Silke S. Heinzmann
- Research
Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Philippe Schmitt-Kopplin
- Research
Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Chair
of Analytical Food Chemistry, Technische Universität München, 85354 Freising, Germany
| |
Collapse
|
24
|
Anderwald CH, Tura A, Gessl A, Smajis S, Bieglmayer C, Marculescu R, Luger A, Pacini G, Krebs M. Whole-body insulin sensitivity rather than body-mass-index determines fasting and post-glucose-load growth hormone concentrations. PLoS One 2014; 9:e115184. [PMID: 25517727 PMCID: PMC4269423 DOI: 10.1371/journal.pone.0115184] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 11/19/2014] [Indexed: 11/25/2022] Open
Abstract
Background Obese, non-acromegalic persons show lower growth hormone (GH) concentrations at fasting and reduced GH nadir during an oral glucose tolerance test (OGTT). However, this finding has never been studied with regard to whole-body insulin-sensitivity as a possible regulator. Methods In this retrospective analysis, non-acromegalic (NonACRO, n = 161) and acromegalic (ACRO, n = 35), non-diabetic subjects were subdivided into insulin-sensitive (IS) and –resistant (IR) groups according to the Clamp-like Index (CLIX)-threshold of 5 mg·kg−1·min−1 from the OGTT. Results Non-acromegalic IS (CLIX: 8.8±0.4 mg·kg−1·min−1) persons with similar age and sex distribution, but lower (p<0.001) body-mass-index (BMI = 25±0 kg/m2, 84% females, 56±1 years) had 59% and 70%, respectively, higher (p<0.03) fasting GH and OGTT GH area under the curve concentrations than IR (CLIX: 3.5±0.1 mg·kg−1·min−1, p<0.001) subjects (BMI = 29±1 kg/m2, 73% females, 58±1 years). When comparing on average overweight non-acromegalic IS and IR with similar anthropometry (IS: BMI: 27±0 kg/m2, 82% females, 58±2 years; IR: BMI: 27±0 kg/m2, 71% females, 60±1 years), but different CLIX (IS: 8.7±0.9 vs. IR: 3.8±0.1 mg·kg−1·min−1, p<0.001), the results remained almost the same. In addition, when adjusted for OGTT-mediated glucose rise, GH fall was less pronounced in IR. In contrast, in acromegalic subjects, no difference was found between IS and IR patients with regard to fasting and post-glucose-load GH concentrations. Conclusions Circulating GH concentrations at fasting and during the OGTT are lower in non-acromegalic insulin-resistant subjects. This study seems the first to demonstrate that insulin sensitivity rather than body-mass modulates fasting and post-glucose-load GH concentrations in non-diabetic non–acromegalic subjects.
Collapse
Affiliation(s)
- Christian-Heinz Anderwald
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council (ISIB-CNR), Padua, Italy
- Mariahilf Community Pharmacy, Arnoldstein, Austria
- Medical Direction, Specialized Hospital Complex Agathenhof, Micheldorf, Austria
- * E-mail:
| | - Andrea Tura
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council (ISIB-CNR), Padua, Italy
| | - Alois Gessl
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Sabina Smajis
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Christian Bieglmayer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Vienna, Vienna, Austria
| | - Rodrig Marculescu
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Vienna, Vienna, Austria
| | - Anton Luger
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Giovanni Pacini
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council (ISIB-CNR), Padua, Italy
| | - Michael Krebs
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
25
|
Itariu BK, Zeyda M, Prager G, Stulnig TM. Insulin-like growth factor 1 predicts post-load hypoglycemia following bariatric surgery: a prospective cohort study. PLoS One 2014; 9:e94613. [PMID: 24736741 PMCID: PMC3988194 DOI: 10.1371/journal.pone.0094613] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 03/17/2014] [Indexed: 12/19/2022] Open
Abstract
Postprandial hypoglycemia is a complication following gastric bypass surgery, which frequently remains undetected. Severe hypoglycemic episodes, however, put patients at risk, e.g., for syncope. A major cause of hypoglycemia following gastric bypass is hyperinsulinemic nesidioblastosis. Since pancreatic islets in nesidioblastosis overexpress insulin-like growth factor 1 (IGF-1) receptor α and administration of recombinant IGF-1 provokes hypoglycemia, our main objective was to investigate the occurrence of post-load hypoglycemia one year after bariatric surgery and its relation to pre- and post-operative IGF-1 serum concentrations. We evaluated metabolic parameters including 2 h 75 g oral glucose tolerance test (OGTT) and measured IGF-1 serum concentration in thirty-six non-diabetic patients (29 f/7 m), aged 41.3±2.0 y with a median (IQR) BMI of 30.9 kg/m2 (27.5-34.3 kg/m2), who underwent elective bariatric surgery (predominantly gastric bypass, 83%) at our hospital. Post-load hypoglycemia as defined by a 2 h glucose concentration <60 mg/dl was detected in 50% of patients. Serum insulin and C-peptide concentration during the OGTT and HOMA-IR (homeostatic model assessment-insulin resistance) were similar in hypoglycemic and euglycemic patients. Strikingly, pre- and post-operative serum IGF-1 concentrations were significantly higher in hypoglycemic patients (p = 0.012 and p = 0.007 respectively). IGF-1 serum concentration before surgery negatively correlated with 2 h glucose concentration during the OGTT (rho = -0.58, p = 0.0003). Finally, IGF-1 serum concentrations before and after surgery significantly predicted post-load hypoglycemia with odds ratios of 1.28 (95%CI:1.03-1.55, p = 0.029) and 1.18 (95%CI:1.03-1.33, p = 0.015), respectively, for each 10 ng/ml increment. IGF-1 serum concentration could be a valuable biomarker to identify patients at risk for hypoglycemia following bariatric surgery independently of a diagnostic OGTT. Thus, IGF-1 testing could help to prevent a significant complication of gastric bypass surgery.
Collapse
Affiliation(s)
- Bianca K. Itariu
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University Vienna, Vienna, Austria
- Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy, Medical University Vienna, Vienna, Austria
| | - Maximilian Zeyda
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University Vienna, Vienna, Austria
- Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy, Medical University Vienna, Vienna, Austria
| | - Gerhard Prager
- Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Thomas M. Stulnig
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University Vienna, Vienna, Austria
- Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy, Medical University Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
26
|
Cohen R, Caravatto PP, Petry T. Metabolic Surgery for Type 2 Diabetes in Patients with a BMI of <35 kg/m(2): A Surgeon's Perspective. Obes Surg 2014; 23:809-18. [PMID: 23564465 DOI: 10.1007/s11695-013-0930-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bariatric surgery was developed with the aim of weight reduction. Success was defined only by excess weight loss. Other indices of resolution of metabolic comorbidities were reported, but were mostly secondary. Several communications have reported that regardless of body mass index (BMI), complete or partial remission of type 2 diabetes mellitus (T2DM) is possible. These results mostly occur before weight loss, positioning metabolic surgery as a good tool for controlling the current T2DM epidemic. Medical treatment is evolving, but is expensive and not risk-free. Surgery aimed mainly at diseases such as diabetes and not weight loss are referred to as "metabolic surgery." Metabolic surgery has been proven to be safe and effective, and although more data are needed, it is unquestionable that a new discipline has been founded. Metabolic surgery can effectively treat T2DM in individuals with any BMI, including that below 35 kg/m(2).
Collapse
Affiliation(s)
- Ricardo Cohen
- The Center of Excellence for Metabolic and Bariatric Surgery, Hospital Oswaldo Cruz, São Paulo, Brazil.
| | | | | |
Collapse
|
27
|
Grueneberger JM, Karcz-Socha I, Sawczyn T, Kosmowski J, Stygar D, Goos M, Küsters S, Zwirska-Korczala K, Marjanovic G, Keck T, Hopt UT, Karcz WK. Systematic ileal transposition in Zucker rats shows advantage for long segment distal transposition. Surgery 2014; 155:165-72. [DOI: 10.1016/j.surg.2013.05.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Accepted: 05/28/2013] [Indexed: 01/05/2023]
|
28
|
Carvalho BM, Oliveira AG, Ueno M, Araújo TG, Guadagnini D, Carvalho-Filho MA, Geloneze B, Lima MMO, Pareja JC, Carvalheira JBC, Saad MJA. Modulation of double-stranded RNA-activated protein kinase in insulin sensitive tissues of obese humans. Obesity (Silver Spring) 2013; 21:2452-2457. [PMID: 23519983 DOI: 10.1002/oby.20410] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 01/23/2013] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The double-stranded RNA-dependent protein kinase (PKR) was recently implicated in regulating molecular integration of nutrient- and pathogen-sensing pathways in obese mice. However, its modulation in human tissues in situations of insulin resistance has not been investigated. The present study was performed to first determine the tissue expression and phosphorylation levels of PKR in the liver, muscle, and adipose tissue in obese humans, and also the modulation of this protein in the adipose tissue of obese patients after bariatric surgery. DESIGN AND METHODS Eleven obese subjects who were scheduled to undergo Roux-en-Y Gastric Bypass Procedure participated in this study. Nine apparently healthy lean subjects as a control group were also included. RESULTS Our data show that PKR is activated in liver, muscle, and adipose tissue of obese humans and, after bariatric surgery, there is a clear reduction in PKR activation accompanied by a decrease in protein kinase-like endoplasmic reticulum kinase, c-Jun N-terminal kinase, inhibitor of kappa β kinase, and insulin receptor substrate-1 serine 312 phosphorylation in subcutaneous adipose tissue from these patients. CONCLUSION Thus, it is proposed that PKR is an important mediator of obesity-induced insulin resistance and a potential target for the therapy.
Collapse
Affiliation(s)
- Bruno M Carvalho
- Department of Internal Medicine, State University of Campinas, Campinas, SP, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Obesity is a rising epidemic, and it is projected that over 700 million people will be obese by 2015. As the number of people with morbid obesity rises, so will the number of bariatric procedures performed. The goal of this article is to review current surgical and endoscopic options for weight loss in morbidly obese patients including their efficacy and complications. RECENT FINDINGS New bariatric surgical techniques have been developed with the goals of maximizing weight loss and metabolic outcomes, while minimizing complications. In addition, there is a role for therapeutic endoscopy in treating obesity as well as managing bariatric surgical complications. As the metabolic effects of bariatric surgery are better elucidated, bariatric surgeries may provide a role in treatment of metabolic syndrome in mildly obese individuals. For those with insufficient weight loss, revisional bariatric surgeries have been performed with varying success. SUMMARY Bariatric surgery is an effective treatment for obesity and its comorbidities. Several bariatric surgeries are available, and a multidisciplinary approach is recommended for choosing the best procedure for the appropriate candidate, along with providing long-term follow-up care to maximize outcome.
Collapse
|
30
|
Anderwald CH, Tura A, Gessl A, Luger A, Pacini G, Krebs M. Adequately adapted insulin secretion and decreased hepatic insulin extraction cause elevated insulin concentrations in insulin resistant non-diabetic adrenal incidentaloma patients. PLoS One 2013; 8:e77326. [PMID: 24146977 PMCID: PMC3797754 DOI: 10.1371/journal.pone.0077326] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 09/08/2013] [Indexed: 11/19/2022] Open
Abstract
Background Insulin-resistance is commonly found in adrenal incidentaloma (AI) patients. However, little is known about beta-cell secretion in AI, because comparisons are difficult, since beta–cell-function varies with altered insulin-sensitivity. Objectives To retrospectively analyze beta–cell function in non-diabetic AI, compared to healthy controls (CON). Methods AI (n=217, 34%males, 57±1years, body-mass-index:27.7±0.3kg/m2) and CON [n=25, 32%males, 56±1years, 26.7±0.8kg/m2] with comparable anthropometry (p≥0.31) underwent oral-glucose-tolerance-tests (OGTTs) with glucose, insulin, and C–peptide measurements. 1mg-dexamethasone-suppression-tests were performed in AI. AI were divided according to post–dexamethasone-suppression–test cortisol-thresholds of 1.8 and 5µg/dL into 3subgroups: pDexa<1.8µg/dL, pDexa1.8-5µg/dL and pDexa>5µg/dL. Using mathematical modeling, whole-body insulin-sensitivity [Clamp-like-Index (CLIX)], insulinogenic Index, Disposition Index, Adaptation Index, and hepatic insulin extraction were calculated. Results CLIX was lower in AI combined (4.9±0.2mg·kg-1·min-1), pDexa<1.8µg/dL (4.9±0.3) and pDexa1.8-5µg/dL (4.7±0.3, p<0.04 vs.CON:6.7±0.4). Insulinogenic and Disposition Indexes were 35%–97% higher in AI and each subgroup (p<0.008 vs.CON), whereas C–peptide–derived Adaptation Index, compensating for insulin-resistance, was comparable between AI, subgroups, and CON. Mathematical estimation of insulin–derived (insulinogenic and Disposition) Indexes from associations to insulin-sensitivity in CON revealed that AI-subgroups had ~19%-32% higher insulin-secretion than expectable. These insulin-secretion-index differences negatively (r=-0.45, p<0.001) correlated with hepatic insulin extraction, which was 13-16% lower in AI and subgroups (p<0.003 vs.CON). Conclusions AI-patients show insulin-resistance, but adequately adapted insulin secretion with higher insulin concentrations during an OGTT, because of decreased hepatic insulin extraction; this finding affects all AI-patients, regardless of dexamethasone-suppression-test outcome.
Collapse
Affiliation(s)
- Christian-Heinz Anderwald
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council (ISIB-CNR), Padua, Italy
- Mariahilf Community Pharmacy, Arnoldstein, Austria
- Medical Direction, Specialized Hospital Complex Agathenhof, Micheldorf, Austria
- * E-mail:
| | - Andrea Tura
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council (ISIB-CNR), Padua, Italy
| | - Alois Gessl
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Anton Luger
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Giovanni Pacini
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council (ISIB-CNR), Padua, Italy
| | - Michael Krebs
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
31
|
Campos GM, Rabl C, Havel PJ, Rao M, Schwarz JM, Schambelan M, Mulligan K. Changes in post-prandial glucose and pancreatic hormones, and steady-state insulin and free fatty acids after gastric bypass surgery. Surg Obes Relat Dis 2013; 10:1-8. [PMID: 24209879 DOI: 10.1016/j.soard.2013.07.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Revised: 06/25/2013] [Accepted: 07/15/2013] [Indexed: 12/25/2022]
Abstract
BACKGROUND Changes in the multiple mechanisms that regulate glucose metabolism after gastric bypass (RYGB) are still being unveiled. The objective of this study was to compare the changes of glucose and pancreatic hormones [C-peptide, glucagon, and pancreatic polypeptide (PP)] during a meal tolerance test (MTT) and steady-state insulin and free fatty acid (FFA) concentrations during euglycemic-hyperinsulinemic clamp 14 days and 6 months after RYGB in morbidly obese nondiabetic patients. METHODS Two groups were studied at baseline and at 14 days: the RYGB followed by caloric restriction group (RYGB, n = 12) and the equivalent caloric restriction alone group (Diet, n = 10), to control for energy intake and weight loss. The RYGB group was studied again at 6 months to assess the changes after substantial weight loss. During MTT, the early and overall changes in glucose and pancreatic hormone concentrations were determined, and during the clamp, steady-state insulin and FFA concentrations were assessed. RESULTS After 14 days, RYGB patients had enhanced postprandial glucose, C-peptide, and glucagon responses, and decreased postprandial PP concentrations. Steady-state insulin concentrations were decreased at 14 days only in RYGB patients, and FFA increased in both groups. Six months after RYGB and substantial weight loss, the decrease in insulin concentrations during clamp persisted, and there were further changes in postprandial glucose and glucagon responses. FFA concentrations during clamp were significantly lower at 6 months, relative to presurgical values. CONCLUSIONS In morbidly obese nondiabetic patients, RYGB produces early changes in postmeal glucose, C-peptide, glucagon, and PP responses, and it appears to enhance insulin clearance early after RYGB and improve insulin sensitivity in adipose tissue at 6 months postsurgery. The early changes cannot be explained by caloric restriction alone.
Collapse
Affiliation(s)
- Guilherme M Campos
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; Department of Surgery, University of California San Francisco, San Francisco, California.
| | - Charlotte Rabl
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; Department of Surgery, University of California San Francisco, San Francisco, California; Department of Surgery, Paracelsus Medical University, Salzburg, Austria
| | - Peter J Havel
- Department of Molecular Biosciences, School of Veterinary Medicine and Department of Nutrition, University of California Davis, Davis, California
| | - Madhu Rao
- Department of Medicine, University of California San Francisco, San Francisco, California
| | | | - Morris Schambelan
- Department of Medicine, University of California San Francisco, San Francisco, California
| | - Kathleen Mulligan
- Department of Medicine, University of California San Francisco, San Francisco, California
| |
Collapse
|
32
|
Campos GM, Ziemelis M, Paparodis R, Ahmed M, Davis DB. Laparoscopic reversal of Roux-en-Y gastric bypass: technique and utility for treatment of endocrine complications. Surg Obes Relat Dis 2013; 10:36-43. [PMID: 24120983 DOI: 10.1016/j.soard.2013.05.012] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 04/24/2013] [Accepted: 05/25/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND The anatomic and physiologic changes with Roux-en-Y gastric bypass (RYGB) may lead to uncommon but occasionally difficult to treat complications such as hyperinsulinemic hypoglycemia with neuroglycopenia and recalcitrant hypocalcemia associated to hypoparathyroidism. Medical management of these complications is challenging. Laparoscopic reversal of RYGB anatomy with restoration of pyloric function and duodenal continuity is a potential treatment. The objective of this study was to present the indications, surgical technique, and clinical outcomes of laparoscopic reversal of RYGB. METHODS Prospective study of consecutive patients offered laparoscopic reversal of RYGB. RESULTS Five patients with remote laparoscopic RYGB underwent laparoscopic reversal of RYGB to normal anatomy (n = 2) or modified sleeve gastrectomy (n = 3). Indications were medically refractory hyperinsulinemic hypoglycemia with neuroglycopenia (n = 3), recalcitrant hypocalcemia with hypoparathyroidism (n = 1), and both conditions simultaneously (n = 1). Before reversal, all patients had a gastrostomy tube placed in the excluded stomach to document improvement of symptoms. Laparoscopic reversal was accomplished successfully in all patients. Three postoperative complications occurred: bleeding that required transfusion, gallstone pancreatitis, and a superficial trocar site infection. Average length of stay was 3 days. At a mean follow-up of 12 months (range 3 to 22), no additional episodes of neuroglycopenia occurred, average number of hypoglycemic episodes per week decreased from 18.5 ± 12.4 to 1.5 ± 1.9 (P = .05), and hypocalcemia became responsive to oral replacement therapy in both patients. CONCLUSIONS Laparoscopic reversal of RYGB to normal anatomy or modified sleeve gastrectomy is feasible and may be a therapeutic option for selected patients with medically refractory hyperinsulinemic hypoglycemia and/or recalcitrant hypocalcemia associated with hypoparathyroidism.
Collapse
Affiliation(s)
- Guilherme M Campos
- Department of Surgery, Division of General Surgery, Section of Foregut and Bariatric Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| | - Martynas Ziemelis
- Department of Surgery, Division of General Surgery, Section of Foregut and Bariatric Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Rodis Paparodis
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Muhammed Ahmed
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Dawn Belt Davis
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| |
Collapse
|
33
|
Improvements in hippocampal-dependent memory and microglial infiltration with calorie restriction and gastric bypass surgery, but not with vertical sleeve gastrectomy. Int J Obes (Lond) 2013; 38:349-56. [PMID: 23736372 DOI: 10.1038/ijo.2013.100] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 04/19/2013] [Accepted: 05/22/2013] [Indexed: 01/06/2023]
Abstract
BACKGROUND Much recent evidence suggest that obesity and related comorbidities contribute to cognitive decline, including the development of non age-related dementia and Alzheimer's disease. Obesity is a serious threat to public health, and few treatments offer proven long-term weight loss. In fact, bariatric surgery remains the most effective long-term therapy to reduce weight and alleviate other aspects of the metabolic syndrome (MetS). Unlike the demonstrated benefits of caloric restriction to prevent weight gain, few if any studies have compared various means of weight loss on central nervous system function and hippocampal-dependent cognitive processes. DESIGN AND RESULTS Our studies comprise the first direct comparisons of caloric restriction to two bariatric surgeries (Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG)) on cognitive function. Weight loss following caloric restriction, RYGB and VSG was associated with generalized improvements in metabolic health and hippocampal-dependent learning, as measured in the radial arm maze and spontaneous alternation tests. However, VSG-treated rats exhibited deficits on spatial learning tasks in the Morris water maze. In addition, whereas VSG animals had elevated hippocampal inflammation, comparable to that of obese controls, RYGB and calorie-restricted (pair-fed, PF) controls exhibited an amelioration of inflammation, as measured by the microglial protein ionized calcium binding adaptor molecule 1 (IBA1). We also assessed whether GHR (ghrelin) replacement would attenuate hippocampal inflammation in VSG, as post-surgical GHR levels are significantly reduced in VSG relative to RYGB and PF rats. However, GHR treatment did not attenuate the hippocampal inflammation. CONCLUSION Although VSG was comparably effective at reducing body weight and improving glucose regulation as RYGB, VSG did not appear to confer an equal benefit on cognitive function and markers of inflammation.
Collapse
|
34
|
Moran GW, Lal S, McLaughlin JT. Commentary: a comparison of glucagon-like peptides 1 and 2. Aliment Pharmacol Ther 2013; 37:279-80. [PMID: 23252781 DOI: 10.1111/apt.12165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 11/05/2012] [Indexed: 12/08/2022]
Affiliation(s)
- G W Moran
- Institute of Inflammation and Repair, and Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | | | | |
Collapse
|