1
|
Zevallos A, Sanches EE, Parmar C, Ribeiro R, Pouwels S. Remission of hypertension after laparoscopic sleeve gastrectomy versus Roux-en-Y-gastric bypass: a systematic review of randomized control trials. Surg Obes Relat Dis 2025; 21:271-278. [PMID: 39477737 DOI: 10.1016/j.soard.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/29/2024] [Accepted: 10/05/2024] [Indexed: 02/09/2025]
Abstract
BACKGROUND Besides its benefits for weight loss, current findings suggest that bariatric surgery can induce remission of hypertension. Limited data report the effect of bariatric surgery on this obesity-associated comorbidity. OBJECTIVE Compare the short-term, mid-term, and long-term remission of hypertension after sleeve gastrectomy versus Roux-en-Y gastric bypass. SETTING Meta-analysis of randomized controlled trials (RCTs). METHODS Four databases (Embase, PubMed, Scopus, and Science Direct) were searched for RCTs that compared the effects of sleeve gastrectomy (SG) versus Roux-en-Y gastric bypass (RYGB) on hypertension remission at <1 year, 2-4 years, and ≥5 years. Patients with a history of hypertension and who had primary bariatric surgery were included. RESULTS After reviewing 11,814 studies, only 11 RCTs were included. In total, the analysis included 2323 patients, with 1158 in the SG group (49.85%) and 1165 in the RYGB group (50.15%). It was found that SG and RYGB procedures had comparable hypertension remission at ≤1 year (Relative risk: 1.11, 95% CI .83-1.48, P = .49), and between 2 and 4 years (Relative risk: 1.11, 95% CI .90-1.37, P = .34). However, there was a significant difference in hypertension remission at ≥ 5 years, favoring RYGB (relative risk: 1.39, 95% CI 1.06-1.82, P = .02). CONCLUSION This systematic review and meta-analysis of RCTs demonstrates that RYGB is superior to SG in resolving hypertension beyond 5 years postoperatively. These findings highlight the long-term benefits of RYGB over SG in managing hypertension, providing valuable insights for surgical decision-making and patient counseling.
Collapse
Affiliation(s)
- Alba Zevallos
- Department of Medicine, Universidad Cientifica del Sur, Lima, Peru
| | - Elijah E Sanches
- Netherlands School of Public and Occupational Health, Utrecht, the Netherlands
| | - Chetan Parmar
- Department of Surgery, The Whittington Hospital NHS Trust, London, United Kingdom; Apollo Hospitals Education and Research Foundation, New Delhi, India; University College London, London, United Kingdom
| | - Rui Ribeiro
- Centro Multidisciplinar do Tratamento da Obesidade, Hospital Lusíadas Amadora, Lisbon, Portugal
| | - Sjaak Pouwels
- Department of Intensive Care Medicine, Elisabeth-Tweesteden Hospital, Tilburg, the Netherlands; Department of Surgery, Marien Hospital Herne, University Hospital of Ruhr University Bochum, Herne, NRW, Germany.
| |
Collapse
|
2
|
Pino-Zuñiga J, Lillo-Urzua P, Olivares-Galvez M, Palacio-Aguero A, Duque JC, Luengas R, Cancino-Lopez J. Adherence to an Early Exercise Plan Promotes Visceral Fat Loss in the First Month Following Bariatric Surgery. Obes Surg 2025; 35:746-754. [PMID: 39953328 DOI: 10.1007/s11695-025-07685-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/06/2025] [Accepted: 01/11/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND The evidence supporting the benefits of early exercise in post-bariatric patients is growing. This study analyzed the effects of early exercise (1-week post-bariatric surgery) on body composition in patients with overweight and obesity 1 month after surgery. METHODS Thirty patients (age 36.5 ± 12.3 [range, 18-65] years; body mass index [BMI], 36.2 ± 12.3 kg/m2, range, 29-48) who underwent laparoscopic sleeve gastrectomy for bariatric surgery were instructed to participate in an exercise training program initiated on day 3 post-surgery and to follow a recommended protein intake of 60 g/day. After 1-month post-surgery, patients were stratified into those who adhered to exercise recommendations and those who did not. Pre- and post-differences in total weight loss (TWL), skeletal muscle mass (SMM), fat mass (FM), and visceral fat mass (VFM) were compared. RESULTS TWL, SMM, and FM loss were similar between non-adherent and adherent subjects (10.2 ± 3.5 kg and 11.9 ± 3.6 kg; p = 0.2; 2.9 ± 1.0 kg and 3.2 ± 1.2; p = 0.2; 6.2 ± 2.1 kg and 7.5 ± 3.6 kg; p = 0.2, respectively), whereas VFM was markedly reduced in the adherent group (29.9 ± 18.2 cm2 vs 14.6 ± 9.4 cm2; p = 0.01) compared to the non-adherent group. When the group was divided according to adherence to exercise and protein intake or non-adherence to both conditions, there was a significant difference in TWL, FM, and VFM losses (p < 0.05). In contrast, no differences in SMM were found. CONCLUSIONS Early exercise training accelerated visceral fat mass loss during the initial recovery period in patients after bariatric surgery. Additionally, adherence to daily protein intake recommendations can increase total body weight and fat mass loss.
Collapse
Affiliation(s)
| | - Paloma Lillo-Urzua
- School of Kinesiology, Faculty of Health Science, Católica Silva Henríquez, University, Santiago, Chile.
| | | | - Ana Palacio-Aguero
- Bariatric Center BIO, Santiago, Chile
- University of Desarrollo, Faculty of Health Science, Santiago, Chile
| | | | | | - Jorge Cancino-Lopez
- Exercise Physiology and Metabolism Laboratory, School of Kinesiology, Faculty of Medicine, Finis Terrae University, Santiago, Chile.
- NFT Center, Santiago, Chile.
| |
Collapse
|
3
|
Li H, He J, Hou J, He C, Dai X, Song Z, Liu Q, Wang Z, Huang H, Ding Y, Qi T, Zhang H, Wu L. Intestinal rearrangement of biliopancreatic limbs, alimentary limbs, and common limbs in obese type 2 diabetic mice after duodenal jejunal bypass surgery. Front Endocrinol (Lausanne) 2025; 15:1456885. [PMID: 39845886 PMCID: PMC11750664 DOI: 10.3389/fendo.2024.1456885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Bariatric surgery is an effective treatment for type 2 Diabetes Mellitus (T2DM), yet the precise mechanisms underlying its effectiveness remain incompletely understood. While previous research has emphasized the role of rearrangement of the gastrointestinal anatomy, gaps persist regarding the specific impact on the gut microbiota and barriers within the biliopancreatic, alimentary, and common limbs. This study aimed to investigate the effects of duodenal-jejunal bypass (DJB) surgery on obese T2DM mice. We performed DJB and SHAM surgery in obese T2DM mice to investigate changes in the gut microbiota and barrier across different intestinal limbs. The effects on serum metabolism and potential associations with T2DM improvement were also investigated. Following DJB surgery, there was an increased abundance of commensals across various limbs. Additionally, the surgery improved intestinal permeability and inflammation in the alimentary and common limbs, while reducing inflammation in the biliopancreatic limbs. Furthermore, DJB surgery also improved T2DM by increasing L-glutamine, short-chain fatty acids, and bile acids and decreasing branched-chain amino acids. This study underscores the role of intestinal rearrangement in reshaping gut microbiota composition and enhancing gut barrier function, thereby contributing to the amelioration of T2DM following bariatric surgery, and providing new insights for further research on bariatric surgery.
Collapse
Affiliation(s)
- Heng Li
- Department of Metabolic Surgery, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jipei He
- Department of Basic Medical Research, General Hospital of Southern Theater Command of People's Liberation Army (PLA), Guangzhou, China
| | - Jie Hou
- Department of Metabolic Surgery, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chengjun He
- Department of Metabolic Surgery, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaojiang Dai
- Department of Metabolic Surgery, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhigao Song
- Department of Cardiovascular Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Qing Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Guangzhou, China
| | - Zixin Wang
- Department of Metabolic Surgery, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongyan Huang
- Department of Metabolic Surgery, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yunfa Ding
- Department of Metabolic Surgery, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tengfei Qi
- Department of Metabolic Surgery, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongbin Zhang
- Department of Basic Medical Research, General Hospital of Southern Theater Command of People's Liberation Army (PLA), Guangzhou, China
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Liangping Wu
- Department of Metabolic Surgery, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangzhou Hualiang Qingying Biotechnology Co. Ltd, Guangzhou, China
| |
Collapse
|
4
|
Müller L, Hoffmann A, Bernhart SH, Ghosh A, Zhong J, Hagemann T, Sun W, Dong H, Noé F, Wolfrum C, Dietrich A, Stumvoll M, Massier L, Blüher M, Kovacs P, Chakaroun R, Keller M. Blood methylation pattern reflects epigenetic remodelling in adipose tissue after bariatric surgery. EBioMedicine 2024; 106:105242. [PMID: 39002385 PMCID: PMC11284569 DOI: 10.1016/j.ebiom.2024.105242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND Studies on DNA methylation following bariatric surgery have primarily focused on blood cells, while it is unclear to which extend it may reflect DNA methylation profiles in specific metabolically relevant organs such as adipose tissue. Here, we investigated whether adipose tissue depots specific methylation changes after bariatric surgery are mirrored in blood. METHODS Using Illumina 850K EPIC technology, we analysed genome-wide DNA methylation in paired blood, subcutaneous and omental visceral AT (SAT/OVAT) samples from nine individuals (N = 6 female) with severe obesity pre- and post-surgery. FINDINGS The numbers and effect sizes of differentially methylated regions (DMRs) post-bariatric surgery were more pronounced in AT (SAT: 12,865 DMRs from -11.5 to 10.8%; OVAT: 14,632 DMRs from -13.7 to 12.8%) than in blood (9267 DMRs from -8.8 to 7.7%). Cross-tissue DMRs implicated immune-related genes. Among them, 49 regions could be validated with similar methylation changes in blood from independent individuals. Fourteen DMRs correlated with differentially expressed genes in AT post bariatric surgery, including downregulation of PIK3AP1 in both SAT and OVAT. DNA methylation age acceleration was significantly higher in AT compared to blood, but remained unaffected after surgery. INTERPRETATION Concurrent methylation pattern changes in blood and AT, particularly in immune-related genes, suggest blood DNA methylation mirrors AT's inflammatory state post-bariatric surgery. FUNDING The funding sources are listed in the Acknowledgments section.
Collapse
Affiliation(s)
- Luise Müller
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, 04103, Germany
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, 04103, Germany
| | - Stephan H Bernhart
- Interdisciplinary Center for Bioinformatics, University of Leipzig, 04107, Leipzig, Germany; Bioinformatics Group, Department of Computer, University of Leipzig, 04107, Leipzig, Germany; Transcriptome Bioinformatics, LIFE Research Center for Civilization Diseases, University of Leipzig, 04107, Leipzig, Germany
| | - Adhideb Ghosh
- Institute of Food, Nutrition and Health, ETH Zurich, 8092, Schwerzenbach, Switzerland
| | - Jiawei Zhong
- Department of Medicine Huddinge (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, 141 83, Huddinge, Sweden
| | - Tobias Hagemann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, 04103, Germany
| | - Wenfei Sun
- Institute of Food, Nutrition and Health, ETH Zurich, 8092, Schwerzenbach, Switzerland
| | - Hua Dong
- Institute of Food, Nutrition and Health, ETH Zurich, 8092, Schwerzenbach, Switzerland
| | - Falko Noé
- Institute of Food, Nutrition and Health, ETH Zurich, 8092, Schwerzenbach, Switzerland
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zurich, 8092, Schwerzenbach, Switzerland
| | - Arne Dietrich
- Leipzig University Hospital, Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Section of Bariatric Surgery, 04103, Leipzig, Germany
| | - Michael Stumvoll
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, 04103, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, 04103, Germany; Deutsches Zentrum für Diabetesforschung e.V., 85764, Neuherberg, Germany
| | - Lucas Massier
- Department of Medicine Huddinge (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, 141 83, Huddinge, Sweden
| | - Matthias Blüher
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, 04103, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, 04103, Germany; Deutsches Zentrum für Diabetesforschung e.V., 85764, Neuherberg, Germany
| | - Peter Kovacs
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, 04103, Germany; Deutsches Zentrum für Diabetesforschung e.V., 85764, Neuherberg, Germany
| | - Rima Chakaroun
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, 04103, Germany; The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 41345, Gothenburg, Sweden
| | - Maria Keller
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, 04103, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, 04103, Germany.
| |
Collapse
|
5
|
Retnakaran R, Kashyap SR, Gerstein HC, Aroda VR. Contemporary Clinical Perspectives on Targeting Remission of Type 2 Diabetes. J Clin Endocrinol Metab 2024; 109:1179-1188. [PMID: 38108415 DOI: 10.1210/clinem/dgad746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/06/2023] [Accepted: 12/15/2023] [Indexed: 12/19/2023]
Abstract
It has long been known that some patients with type 2 diabetes (T2DM) can experience sustained metabolic improvement to near-normal levels of glycemia either spontaneously or after medical intervention. Now recognized as remission of diabetes, this intriguing state is currently more feasible than ever before due to profound advances in metabolic surgery, pharmacologic therapy, and regimens of lifestyle modification. This enhanced capacity to induce remission has revealed new pathophysiologic insights, including the presence of a reversible component of the pancreatic beta-cell dysfunction that otherwise drives the chronic progressive nature of T2DM. In doing so, it has changed the therapeutic landscape by offering new potential management objectives and considerations for patients and providers. However, the excitement around these developments must also be tempered by the sobering realities of our current understanding of remission, including the recognition that this condition may not be permanent (resulting in glycemic relapse over time) and that beta-cell function may not be normalized in the setting of remission. These limitations highlight both the many gaps in our current understanding of remission and the caution with which clinical discussions must be handled for clear patient-directed communication of the pros and cons of targeting this outcome in practice. In this mini-review, we consider this rapidly growing literature, including its implications and its limitations, and thereby seek to provide objective balanced perspectives on targeting remission of T2DM in current clinical care.
Collapse
Affiliation(s)
- Ravi Retnakaran
- Leadership Sinai Centre for Diabetes, Mount Sinai Hospital, Toronto, ON M5T 3L9, Canada
- Division of Endocrinology, University of Toronto, Toronto, ON M5S 3H2, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Sangeeta R Kashyap
- Division of Endocrinology, Diabetes, and Metabolism, New York Presbyterian-Weill Cornell Medicine, New York, NY 10021, USA
| | - Hertzel C Gerstein
- Division of Endocrinology, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Vanita R Aroda
- Division of Endocrinology, Diabetes & Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
van der Meer R, Mohamed SA, Monpellier VM, Liem RSL, Hazebroek EJ, Franks PW, Frayling TM, Janssen IMC, Serlie MJ. Genetic variants associated with weight loss and metabolic outcomes after bariatric surgery: A systematic review. Obes Rev 2023; 24:e13626. [PMID: 37632325 DOI: 10.1111/obr.13626] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 08/27/2023]
Abstract
The extent to which genetic variations contribute to interindividual differences in weight loss and metabolic outcomes after bariatric surgery is unknown. Identifying genetic variants that impact surgery outcomes may contribute to clinical decision making. This review evaluates current evidence addressing the association of genetic variants with weight loss and changes in metabolic parameters after bariatric surgery. A search was conducted using Medline, Embase, Scopus, Web of Science, and Cochrane Library. Fifty-two eligible studies were identified. Single nucleotide polymorphisms (SNPs) at ADIPOQ (rs226729, rs1501299, rs3774261, and rs17300539) showed a positive association with postoperative change in measures of glucose homeostasis and lipid profiles (n = 4), but not with weight loss after surgery (n = 6). SNPs at FTO (rs11075986, rs16952482, rs8050136, rs9939609, rs9930506, and rs16945088) (n = 10) and MC4R (rs11152213, rs476828, rs2229616, rs9947255, rs17773430, rs5282087, and rs17782313) (n = 9) were inconsistently associated with weight loss and metabolic improvement. Four studies examining the UCP2 SNP rs660339 reported associations with postsurgical weight loss. In summary, there is limited evidence supporting a role for specific genetic variants in surgical outcomes after bariatric surgery. Most studies have adopted a candidate gene approach, limiting the scope for discovery, suggesting that the absence of compelling evidence is not evidence of absence.
Collapse
Affiliation(s)
- Rieneke van der Meer
- Nederlandse Obesitas Kliniek, Huis ter Heide, The Netherlands
- Department of Endocrinology & Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Siham A Mohamed
- Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | | | - Ronald S L Liem
- Department of Surgery, Groene Hart Hospital, Gouda, The Netherlands
- Nederlandse Obesitas Kliniek, The Hague and Gouda, The Netherlands
| | - Eric J Hazebroek
- Department of Surgery, Rijnstate Hospital/Vitalys Clinics, Arnhem, The Netherlands
| | - Paul W Franks
- Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | | | - Mireille J Serlie
- Department of Endocrinology & Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Department of Endocrinology & Metabolism, Yale University, New Haven, CT, USA
| |
Collapse
|
7
|
Custers E, Franco A, Kiliaan AJ. Bariatric Surgery and Gut-Brain-Axis Driven Alterations in Cognition and Inflammation. J Inflamm Res 2023; 16:5495-5514. [PMID: 38026245 PMCID: PMC10676679 DOI: 10.2147/jir.s437156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Obesity is associated with systemic inflammation, comorbidities like diabetes, cardiovascular disease and several cancers, cognitive decline and structural and functional brain changes. To treat, or potentially prevent these related comorbidities, individuals with obesity must achieve long-term sustainable weight loss. Often life style interventions, such as dieting and increased physical activity are not successful in achieving long-term weight loss. Meanwhile bariatric surgery has emerged as a safe and effective procedure to treat obesity. Bariatric surgery causes changes in physiological processes, but it is still not fully understood which exact mechanisms are involved. The successful weight loss after bariatric surgery might depend on changes in various energy regulating hormones, such as ghrelin, glucagon-like peptide-1 and peptide YY. Moreover, changes in microbiota composition and white adipose tissue functionality might play a role. Here, we review the effect of obesity on neuroendocrine effects, microbiota composition and adipose tissue and how these may affect inflammation, brain structure and cognition. Finally, we will discuss how these obesity-related changes may improve after bariatric surgery.
Collapse
Affiliation(s)
- Emma Custers
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, the Netherlands
| | - Ayla Franco
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, the Netherlands
| | - Amanda Johanne Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, the Netherlands
| |
Collapse
|
8
|
Lee JS, Sohn M, Kim K, Yoon YS, Lim S. Glucose Regulation after Partial Pancreatectomy: A Comparison of Pancreaticoduodenectomy and Distal Pancreatectomy in the Short and Long Term. Diabetes Metab J 2023; 47:703-714. [PMID: 37349082 PMCID: PMC10555545 DOI: 10.4093/dmj.2022.0205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/07/2022] [Indexed: 06/24/2023] Open
Abstract
BACKGRUOUND Long term quality of life is becoming increasingly crucial as survival following partial pancreatectomy rises. The purpose of this study was to investigate the difference in glucose dysregulation after pancreaticoduodenectomy (PD) or distal pancreatectomy (DP). METHODS In this prospective observational study from 2015 to 2018, 224 patients who underwent partial pancreatectomy were selected: 152 (67.9%) received PD and 72 (32.1%) received DP. Comprehensive assessment for glucose regulation, including a 75 g oral glucose tolerance test was conducted preoperatively, and 1, 12, and 52 weeks after surgery. Patients were further monitored up to 3 years to investigate development of new-onset diabetes mellitus (NODM) in patients without diabetes mellitus (DM) at baseline or worsening of glucose regulation (≥1% increase in glycosylated hemoglobin [HbA1c]) in those with preexisting DM. RESULTS The disposition index, an integrated measure of β-cell function, decreased 1 week after surgery in both groups, but it increased more than baseline level in the PD group while its decreased level was maintained in the DP group, resulting in a between-group difference at the 1-year examination (P<0.001). During follow-up, the DP group showed higher incidence of NODM and worsening of glucose regulation than the PD group with hazard ratio (HR) 4.29 (95% confidence interval [CI], 1.49 to 12.3) and HR 2.15 (95% CI, 1.09 to 4.24), respectively, in the multivariate analysis including dynamic glycemic excursion profile. In the DP procedure, distal DP and spleen preservation were associated with better glucose regulation. DP had a stronger association with glucose dysregulation than PD. CONCLUSION Proactive surveillance of glucose dysregulation is advised, particularly for patients who receive DP.
Collapse
Affiliation(s)
- Jun Suh Lee
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Minji Sohn
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Kyuho Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Yoo-Seok Yoon
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Soo Lim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
9
|
Tang H, Ling J, Meng H, Wu L, Zhu L, Zhu S. Temporal Relationship Between Insulin Resistance and Lipid Accumulation After Bariatric Surgery: a Multicenter Cohort Study. Obes Surg 2023:10.1007/s11695-023-06508-3. [PMID: 37060490 DOI: 10.1007/s11695-023-06508-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 06/12/2022] [Accepted: 06/15/2022] [Indexed: 04/16/2023]
Abstract
PURPOSE Insulin resistance (IR) is closely associated with lipid accumulation. Here, we investigated the temporal relationship between the two conditions after bariatric surgery. MATERIALS AND METHODS In total, 409 participants were enrolled from three bariatric centers in China from 2009 to 2018. We evaluated whether baseline IR (proxied by homeostasis model assessment of insulin resistance (HOMA-IR)) and lipid accumulation (proxied by visceral adiposity index (VAI) and lipid accumulation product (LAP)) were associated with follow-up IR and lipid accumulation (3 months postoperatively) using linear regression models. We then conducted a cross-lagged panel analysis model to simultaneously examine the bidirectional relationship between IR and lipid accumulation. RESULTS Multivariable linear regression analyses showed that baseline HOMA-IR was associated with follow-up VAI (β = 0.430, 95% CI: 0.082-0.778, p = 0.016) and LAP (β = 0.070, 95% CI: 0.010-0.130, p = 0.022). There was no relationship between baseline lipid accumulation and follow-up IR. Further cross-lagged panel analyses indicated that the path coefficient from baseline HOMA-IR to follow-up VAI (β2 = 0.145, p = 0.003) was significantly greater than the coefficient from baseline VAI to follow-up HOMA-IR (β1 = - 0.013, p = 0.777). Similarly, the path coefficient from baseline HOMA-IR to follow-up LAP (β2 = 0.141, p = 0.003) was significantly greater than the coefficient from baseline LAP to follow-up HOMA-IR (β1 = 0.041, p = 0.391). CONCLUSION Our study demonstrated a unidirectional relationship from HOMA-IR to VAI and LAP, suggesting that the change in IR may precede lipid accumulation after surgery.
Collapse
Affiliation(s)
- Haibo Tang
- Department of Metabolic and Bariatric Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jiapu Ling
- Department of Metabolic and Bariatric Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Hua Meng
- Department of General Surgery, The China-Japan Friendship Hospital, Beijing, China
| | - Liangping Wu
- Department of Metabolic Surgery, The Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liyong Zhu
- Department of Metabolic and Bariatric Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shaihong Zhu
- Department of Metabolic and Bariatric Surgery, The Third Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
10
|
Albaugh VL, He Y, Münzberg H, Morrison CD, Yu S, Berthoud HR. Regulation of body weight: Lessons learned from bariatric surgery. Mol Metab 2023; 68:101517. [PMID: 35644477 PMCID: PMC9938317 DOI: 10.1016/j.molmet.2022.101517] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 05/04/2022] [Accepted: 05/21/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bariatric or weight loss surgery is currently the most effective treatment for obesity and metabolic disease. Unlike dieting and pharmacology, its beneficial effects are sustained over decades in most patients, and mortality is among the lowest for major surgery. Because there are not nearly enough surgeons to implement bariatric surgery on a global scale, intensive research efforts have begun to identify its mechanisms of action on a molecular level in order to replace surgery with targeted behavioral or pharmacological treatments. To date, however, there is no consensus as to the critical mechanisms involved. SCOPE OF REVIEW The purpose of this non-systematic review is to evaluate the existing evidence for specific molecular and inter-organ signaling pathways that play major roles in bariatric surgery-induced weight loss and metabolic benefits, with a focus on Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG), in both humans and rodents. MAJOR CONCLUSIONS Gut-brain communication and its brain targets of food intake control and energy balance regulation are complex and redundant. Although the relatively young science of bariatric surgery has generated a number of hypotheses, no clear and unique mechanism has yet emerged. It seems increasingly likely that the broad physiological and behavioral effects produced by bariatric surgery do not involve a single mechanism, but rather multiple signaling pathways. Besides a need to improve and better validate surgeries in animals, advanced techniques, including inducible, tissue-specific knockout models, and the use of humanized physiological traits will be necessary. State-of-the-art genetically-guided neural identification techniques should be used to more selectively manipulate function-specific pathways.
Collapse
Affiliation(s)
- Vance L Albaugh
- Translational and Integrative Gastrointestinal and Endocrine Research Laboratory, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Yanlin He
- Brain Glycemic and Metabolism Control Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Heike Münzberg
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Christopher D Morrison
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Sangho Yu
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
11
|
Regulation of Pancreatic TXNIP-Insulin Expression Levels after Bariatric Surgery Using Diabetic Rodent Model. BIOMED RESEARCH INTERNATIONAL 2023; 2023:9563359. [PMID: 36733403 PMCID: PMC9889143 DOI: 10.1155/2023/9563359] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/27/2022] [Accepted: 12/31/2022] [Indexed: 01/26/2023]
Abstract
Purpose The purpose of this study was to investigate the effect of bariatric surgery on pancreatic thioredoxin-interacting protein (TXNIP) and insulin expression levels. The research question is does bariatric surgery induce changes in the pancreatic TXNIP level, given that TXNIP has been proposed as a key glucose control factor? Methods Using nondiabetic and diabetic rats, we investigated whether our streptozotocin-induced diabetic rat models exhibited changes in pancreatic TXNIP regulation. Following this confirmation, we randomly divided the diabetic rats into the following three groups: the gastric bypass group (n = 16), pair-fed group (n = 10), and sham group (n = 10). Preoperatively and 3 weeks postoperatively, all the rats underwent an oral glucose tolerance test, insulin tolerance test, and blood sampling procedures for hormonal analysis. Results The TXNIP messenger ribonucleic acid (mRNA) and protein expression levels were significantly lower in the gastric bypass group than in the other groups. Regarding the gastric bypass group, the pancreatic mRNA expression levels of microRNA-204 (miR-204) and MafA were significantly lower and higher, respectively, than in the other groups. Furthermore, the levels of pancreatic insulin expression at the mRNA and protein levels were also significantly higher in the gastric bypass group than in the other groups. Conclusion Bariatric surgery significantly improved glucose control and regulated the pancreatic insulin production pathways of TXNIP, miR-204, and MafA. The regulation of TXNIP, miR-204, and MafA might play an important role in the mechanism of diabetes remission following bariatric surgery.
Collapse
|
12
|
Bhandari M, Kosta S, Bhandari M, Reddy M, Mathur W, Gupta M. Effects of Bariatric Surgery on People with Obesity and Polycystic Ovary Syndrome: a Large Single Center Study from India. Obes Surg 2022; 32:3305-3312. [PMID: 35882755 DOI: 10.1007/s11695-022-06209-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 07/09/2022] [Accepted: 07/14/2022] [Indexed: 11/24/2022]
Abstract
PURPOSE Bariatric surgery has been proven to be a successful management strategy for morbid obesity, but limited studies exist on its effect on polycystic ovary syndrome (PCOS) in terms of clinical, hormonal, and comorbidities. MATERIALS AND METHODS This is a prospective observational study of 1013 PCOS patients who underwent bariatric surgery from a single high-volume center. Assessment of demographic data and menstrual irregularity as well as hirsutism and comorbidities was done preoperatively 6-month and yearly follow-up, whereas data regarding %TWL and %EWL was taken at follow-up visits conducted at regular intervals of 6 months, and 1, 2, 3, 4, and 5 years following surgery. RESULTS Out of 1013 PCOS patients, 993 patients had hirsutism before surgery, and 741 (74.6%; p < 0.001) had complete resolution of hirsutism at end of 6 months' follow-up. A total of 202 (20.3%) had moderate resolution at follow-up of 1 year, 5 patients had minimal resolution at end of 2 years, and 45 (4.5%) patients reported no change in their hirsutism at 4 and 5 years of follow-up. Among 1007 women with PCOS who had menstrual dysfunction, 936 (93% p < 0.0001) women restored their normal menstrual cycle at 6 months post-surgery with 55.4% EWL while remaining other 71 (7%) women reported regular menses at 2 years post-surgery at 74.2% EWL and continued to have normal menstrual pattern during the entire follow-up period. Similarly, all the associated comorbidities T2DM (79.7%), HTN (78.7%), DLP (93.2%), and OSA (98.5%) and symptoms of PCOS were statistically (p < 0.0001) and completely resolved at end years of follow-up. CONCLUSION Bariatric surgery is a good option for women with obesity and PCOS. It is effectively reducing weight along with PCOS and its disorder including hirsutism and menstrual irregularity in women with obesity and PCOS.
Collapse
Affiliation(s)
- Mohit Bhandari
- Mohak Bariatrics and Robotics Center, Sri Aurobindo University, Indore, Madhya Pradesh, India.
| | - Susmit Kosta
- Mohak Bariatrics and Robotics Center, SAIMS Campus, Indore- Ujjain Highway, Indore, Madhya Pradesh, India
| | - Mahak Bhandari
- Mohak Bariatrics and Robotics Center, Sri Aurobindo University, Indore, Madhya Pradesh, India
| | - Manoj Reddy
- Mohak Bariatrics and Robotics Center, Sri Aurobindo University, Indore, Madhya Pradesh, India
| | - Winni Mathur
- Mohak Bariatrics and Robotics Center, Sri Aurobindo University, Indore, Madhya Pradesh, India
| | - Mahendra Gupta
- Sri Aurobindo Medical College and PG Institute, Sri Aurobindo University, Indore, Madhya Pradesh, India
| |
Collapse
|
13
|
Ning C, Jiao Y, Wang J, Li W, Zhou J, Lee YC, Ma DL, Leung CH, Zhu R, David Wang HM. Recent advances in the managements of type 2 diabetes mellitus and natural hypoglycemic substances. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
14
|
Fanni G, Katsogiannos P, Nandi Jui B, Sundbom M, Hetty S, Pereira MJ, Eriksson JW. Response of multiple hormones to glucose and arginine challenge in T2DM after gastric bypass. Endocr Connect 2022; 11:e220172. [PMID: 35904227 PMCID: PMC9346340 DOI: 10.1530/ec-22-0172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/27/2022] [Indexed: 11/08/2022]
Abstract
Purpose In patients with type 2 diabetes mellitus (T2DM), Roux-en-Y gastric bypass (RYGB) leads to beneficial metabolic adaptations, including enhanced incretin secretion, beta-cell function, and systemic insulin sensitivity. We explored the impact of RYGB on pituitary, pancreatic, gut hormones, and cortisol responses to parenteral and enteral nutrient stimulation in patients with obesity and T2DM with repeated sampling up to 2 years after intervention. Methods We performed exploratory post hoc analyses in a previously reported randomized trial. Levels of adrenocorticotropic hormone (ACTH), cortisol, growth hormone (GH), glucagon-like peptide 1 (GLP-1), glucose-dependent insulinotropic peptide (GIP), peptide YY (PYY), ACTH, insulin, and glucagon were measured in 13 patients with T2DM and obesity at four different visits: before and 4, 24, and 104 weeks after RYGB; and in three sequential conditions on the same day: fasting, intravenous arginine challenge, and OGTT. Results RYGB surprisingly induced a rise in ACTH, cortisol, and GH levels upon an oral glucose load, together with enhanced GLP-1 and PYY responses. Fasting and post-arginine GH levels were higher after RYGB, whereas insulin, glucagon, GLP-1, GIP, and cortisol were lower. These endocrine adaptations were seen as early as 4 weeks after surgery and were maintained for up to 2 years. Conclusion These findings indicate adaptations of glucose sensing mechanisms and responses in multiple endocrine organs after RYGB, involving the gut, pancreatic islets, the pituitary gland, the adrenals, and the brain.
Collapse
Affiliation(s)
- Giovanni Fanni
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Petros Katsogiannos
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Bipasha Nandi Jui
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Magnus Sundbom
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Susanne Hetty
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Maria J Pereira
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| |
Collapse
|
15
|
Effect of Bariatric Surgery on Long-Term Cardiovascular Outcomes: A Systematic Review and Meta-analysis of Population-Based Cohort Studies. Surg Obes Relat Dis 2022; 18:1074-1086. [DOI: 10.1016/j.soard.2022.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/13/2022] [Accepted: 05/04/2022] [Indexed: 02/07/2023]
|
16
|
Jiang B, Wang H, Li N, Yan Q, Wang W, Wang Y, Xue H, Ma S, Li X, Diao W, Pan R, Gao Z, Qu MH. Role of Proximal Intestinal Glucose Sensing and Metabolism in the Blood Glucose Control in Type 2 Diabetic Rats After Duodenal Jejunal Bypass Surgery. Obes Surg 2022; 32:1119-1129. [PMID: 35080701 DOI: 10.1007/s11695-021-05871-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Although gastric surgery can significantly improve blood glucose homeostasis in type 2 diabetes mellitus (T2DM), its mechanism remains unclear. This study evaluated the role of intestinal glucose sensing, glucose transport, and metabolism in the alimentary limb (A limb) of T2DM rats after duodenal jejunal bypass (DJB) surgery. METHODS A T2DM rat model was induced via a high-glucose high-fat diet and low-dose streptozotocin injection. The diabetic rats were divided into two groups: the DJB surgery (T2DM-DJB) group and the sham surgery (T2DM-Sham) group. Wistar rats were used as wild-type control (Control). Small animal PET was used to assess the change in glucose metabolic status in the intestine. The intestinal villi height and the number of EECs after DJB were evaluated. The expressions of sweet taste receptors (T1R2/T1R3), glucose transporters (SGLT1/GLUT2), and key enzymes involved in glucose metabolism (HK2, PFK2, PKM2, G6Pase, and PCK1) in the A limb after DJB was detected by Western blot and qRT-PCR. RESULTS Small animal PET analysis showed the intestinal glucose metabolism increased significantly 6 weeks after DJB surgery. The intestinal villi height and the number of EECs in the A limb 6 weeks after surgery increased significantly in T2DM-DJB rats comparing to T2DM-Sham rats. The mRNA and protein expression of T1R1/T1R3 and SGLT1/GLUT2 were downregulated in DJB-T2DM rats, while enzymes involved in glucose metabolism was upregulated in the A limb in T2DM-DJB rats. CONCLUSION Proximal intestinal glucose sensing and metabolism play an important role in blood glucose homeostasis by DJB.
Collapse
Affiliation(s)
- Bin Jiang
- Translational Medical Center, Weifang Second People's Hospital, Weifang, 261041, China
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, China
| | - Huaijie Wang
- Translational Medical Center, Weifang Second People's Hospital, Weifang, 261041, China
| | - Na Li
- Translational Medical Center, Weifang Second People's Hospital, Weifang, 261041, China
| | - Qingtao Yan
- Department of Pediatric Surgery, Weifang People's Hospital, The First Affiliated Hospital of Weifang Medical University, Weifang, 261041, China
| | - Weiyu Wang
- Translational Medical Center, Weifang Second People's Hospital, Weifang, 261041, China
| | - Yubing Wang
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, China
| | - Hantao Xue
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, China
| | - Shengyao Ma
- Translational Medical Center, Weifang Second People's Hospital, Weifang, 261041, China
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China
| | - Xiaocheng Li
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, China
| | - Wenbin Diao
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, China
| | - Ruiyan Pan
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China.
| | - Zhiqin Gao
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, China.
| | - Mei-Hua Qu
- Translational Medical Center, Weifang Second People's Hospital, Weifang, 261041, China.
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, China.
| |
Collapse
|
17
|
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
|
18
|
An Z, Wang H, Mokadem M. Role of the Autonomic Nervous System in Mechanism of Energy and Glucose Regulation Post Bariatric Surgery. Front Neurosci 2021; 15:770690. [PMID: 34887725 PMCID: PMC8649921 DOI: 10.3389/fnins.2021.770690] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/15/2021] [Indexed: 01/06/2023] Open
Abstract
Even though lifestyle changes are the mainstay approach to address obesity, Sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB) are the most effective and durable treatments facing this pandemic and its associated metabolic conditions. The traditional classifications of bariatric surgeries labeled them as “restrictive,” “malabsorptive,” or “mixed” types of procedures depending on the anatomical rearrangement of each one of them. This conventional categorization of bariatric surgeries assumed that the “restrictive” procedures induce their weight loss and metabolic effects by reducing gastric content and therefore having a smaller reservoir. Similarly, the “malabsorptive” procedures were thought to induce their main energy homeostatic effects from fecal calorie loss due to intestinal malabsorption. Observational data from human subjects and several studies from rodent models of bariatric surgery showed that neither of those concepts is completely true, at least in explaining the multiple metabolic changes and the alteration in energy balance that those two surgeries induce. Rather, neuro-hormonal mechanisms have been postulated to underly the physiologic effects of those two most performed bariatric procedures. In this review, we go over the role the autonomic nervous system plays- through its parasympathetic and sympathetic branches- in regulating weight balance and glucose homeostasis after SG and RYGB.
Collapse
Affiliation(s)
- Zhibo An
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States
| | - Haiying Wang
- Department of Physiology, Basic Medical School of Jining Medical University, Jining, China
| | - Mohamad Mokadem
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States.,Fraternal Order of Eagles Diabetes Research Center, The University of Iowa, Iowa City, IA, United States.,Obesity Research and Education Initiative, The University of Iowa, Iowa City, IA, United States.,Iowa City Veterans Affairs Health Care System, Iowa City, IA, United States
| |
Collapse
|
19
|
Luo P, Cao Y, Li P, Wang G, Song Z, Li W, Su Z, Zhou H, Yi X, Fu Z, Sun X, Tang H, Cui B, Yu Q, Zhu L, Zhu S. Insulin Resistance Remission Following Laparoscopic Roux-en-Y Gastric Bypass and Laparoscopic Sleeve Gastrectomy in Chinese Type 2 Diabetes Mellitus Patients With a Body Mass Index of 27.5-32.5 kg/m 2. Front Physiol 2021; 12:772577. [PMID: 34819878 PMCID: PMC8606571 DOI: 10.3389/fphys.2021.772577] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Insulin resistance (IR) is closely associated with the pathogenesis of type 2 diabetes mellitus (T2DM). However, remission of insulin sensitivity after bariatric surgery in patients with T2DM and a body mass index (BMI) of 27.5–32.5 kg/m2 has not been fully elucidated. Methods: Thirty-six T2DM patients with a BMI of 27.5–32.5 kg/m2 were prospectively consecutively recruited for laparoscopic Roux-en-Y gastric bypass (LRYGB) or laparoscopic sleeve gastrectomy (LSG). Hyperinsulinemic euglycemic clamp, oral glucose tolerance test (OGTT), and other indicators were tested at baseline and 6 months postoperative. Glucose disposal rate (GDR), time to reach euglycemia, homeostatic model assessment of IR, quantitative insulin sensitivity check index (QUICKI), triglyceride glucose (TyG) index, 30-min insulinogenic index (IGI30), and disposition index (DI) were calculated at baseline and 6 months after surgery. The criterion for remission in T2DM patients was the achievement of the triple composite endpoint. Results: Anthropometric and glucolipid metabolism parameters significantly improved following surgery. The GDR increased significantly from baseline to 6 months after LRYGB (from 4.28 ± 1.70 mg/kg/min to 8.47 ± 1.89 mg/kg/min, p < 0.0001) and LSG (from 3.18 ± 1.36 mg/kg/min to 7.09 ± 1.69 mg/kg/min, p < 0.001). The TyG index decreased after surgery (RYGB group, from 9.93 ± 1.03 to 8.60 ± 0.43, p < 0.0001; LSG group, from 10.04 ± 0.79 to 8.72 ± 0.65, p = 0.0002). There was a significant reduction in the IGI30 (RYGB group, from 2.04 ± 2.12 to 0.83 ± 0.47, p = 0.005; LSG group, from 2.12 ± 1.73 to 0.92 ± 0.66, p = 0.001). The mean DI significantly increased from 1.14 ± 1.35 to 7.11 ± 4.93 in the RYGB group (p = 0.0001) and from 1.25 ± 1.78 to 5.60 ± 4.58 in the LSG group (p = 0.003). Compared with baseline, HOMR-IR, QUICKI, area under the curve-C-peptide release test (AUC-CRT), and AUC-OGTT were significantly changed at 6 months postoperative. Overall, 52.63% of patients in the LRYGB group versus 29.41% of patients in the LSG group achieved the triple composite endpoint. Conclusion: Both LRYGB and LSG effectively induced remission of IR in patients with T2DM and a BMI of 27.5–32.5 kg/m2.
Collapse
Affiliation(s)
- Ping Luo
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yaoquan Cao
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Pengzhou Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Guohui Wang
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Song
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Weizheng Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhihong Su
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Hui Zhou
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xianhao Yi
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhibing Fu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xulong Sun
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Haibo Tang
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Beibei Cui
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Qianqian Yu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Liyong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Shaihong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
20
|
Guerreiro V, Maia I, Neves JS, Salazar D, Ferreira MJ, Mendonça F, Silva MM, Viana S, Costa C, Pedro J, Varela A, Lau E, Freitas P, Carvalho D. Adequate magnesium level as an associated factor of pre-diabetes and diabetes mellitus remission in patients with obesity submitted to bariatric surgery. Sci Rep 2021; 11:21223. [PMID: 34707130 PMCID: PMC8551299 DOI: 10.1038/s41598-021-00584-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 09/14/2021] [Indexed: 01/19/2023] Open
Abstract
Bariatric surgery (BS) can lead to remission of type 2 diabetes mellitus (T2DM), however, the evidence on the influence of preoperative serum magnesium levels on this reversal is scarce. To study the influence of preoperative serum magnesium levels on the pre-T2DM and T2DM remission one year after BS. Retrospective study carried out among 1656 patients with obesity who underwent BS in the Centro Hospitalar Universitário São João. T2DM and pre-T2DM remission were defined as being normal glycaemic measures of at least one year's after BS and without pharmacological therapy. To assess the association between preoperative serum magnesium levels and pre- and T2DM remission, logistic regression models, crude and adjusted for sex, age and body mass index were computed. Patients with normoglycaemia presented hypomagnesaemia less often than those patients with pre-T2DM and T2DM (17.0% vs. 21.3% vs. 39.9%) (p < 0.001). One year after BS, 62.9% of patients with pre-T2DM or T2DM before BS showed remission. Adequate magnesium levels were positively associated with T2DM and pre-T2DM remission, one year after BS (OR 1.79; 95% CI 1.34-2.38), independently of sex, age, and body mass index. Adequate preoperative serum magnesium levels showed to be an important clinical parameter for pre-T2DM and T2DM remission.
Collapse
Affiliation(s)
- Vanessa Guerreiro
- Departamento de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal. .,Faculdade de Medicina da Universidade do Porto, Porto, Portugal. .,Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal. .,Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal.
| | - Isabel Maia
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal.,Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
| | - João Sérgio Neves
- Departamento de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
| | - Daniela Salazar
- Departamento de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
| | - Maria João Ferreira
- Departamento de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
| | - Fernando Mendonça
- Departamento de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
| | - Maria Manuel Silva
- Departamento de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
| | - Sara Viana
- Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal.,Serviço de Medicina Interna, Unidade de Saúde Local do Norte Alentejo EPE, Alentejo, Portugal
| | - Cláudia Costa
- Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal.,Serviço de Endocrinologia, Instituto Portugês de Oncologia, Porto, Portugal
| | - Jorge Pedro
- Departamento de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
| | - Ana Varela
- Departamento de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal.,Serviço de Endocrinologia, Instituto Portugês de Oncologia, Porto, Portugal
| | - Eva Lau
- Departamento de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
| | - Paula Freitas
- Departamento de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
| | - Davide Carvalho
- Departamento de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
| |
Collapse
|
21
|
Jin ZL, Liu W. Progress in treatment of type 2 diabetes by bariatric surgery. World J Diabetes 2021; 12:1187-1199. [PMID: 34512886 PMCID: PMC8394224 DOI: 10.4239/wjd.v12.i8.1187] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/29/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023] Open
Abstract
The incidence of type 2 diabetes (T2D) is increasing at an alarming rate worldwide. Bariatric surgical procedures, such as the vertical sleeve gastrectomy and Roux-en-Y gastric bypass, are the most efficient approaches to obtain substantial and durable remission of T2D. The benefits of bariatric surgery are realized through the consequent increased satiety and alterations in gastrointestinal hormones, bile acids, and the intestinal microbiota. A comprehensive understanding of the mechanisms by which various bariatric surgical procedures exert their benefits on T2D could contribute to the design of better non-surgical treatments for T2D. In this review, we describe the classification and evolution of bariatric surgery and explore the multiple mechanisms underlying the effect of bariatric surgery on insulin resistance. Based upon our summarization of the current knowledge on the underlying mechanisms, we speculate that the gut might act as a new target for improving T2D. Our ultimate goal with this review is to provide a better understanding of T2D pathophysiology in order to support development of T2D treatments that are less invasive and more scalable.
Collapse
Affiliation(s)
- Zhang-Liu Jin
- Department of General Surgery & Department of Biliopancreatic and Metabolic Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Wei Liu
- Department of General Surgery & Department of Biliopancreatic and Metabolic Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| |
Collapse
|
22
|
The Effect of Laparoscopic Gastric Bypass Surgery on Insulin Resistance and Glycosylated Hemoglobin A1c: a 2-Year Follow-up Study. Obes Surg 2021; 30:3489-3495. [PMID: 32314253 PMCID: PMC7378103 DOI: 10.1007/s11695-020-04599-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Bariatric surgery improves insulin sensitivity and secretion in patients with type 2 diabetes, but the effect on patients with prediabetes or even normal glucose tolerance deserves further consideration. METHODS Cohort study including patients operated with laparoscopic Roux-en-Y gastric bypass surgery (LRYGB) between November 2012 and June 2017 at the Örebro University Hospital (n = 813) with follow-up of 742 patients 2 years after surgery. Fasting insulin, glucose, glycosylated hemoglobin (HbA1c), and homeostatic model assessment of insulin resistance (HOMA-IR) were analyzed at baseline and 2 years after surgery for patients with overt type 2 diabetes, prediabetes, or non-diabetes. RESULTS Fasting insulin levels improved for all groups (diabetics baseline 25.5 mIU/L, IQR 17.5-38.0, 2 years 7.6 mIU/L, IQR 5.4-11.1, p < 0.001; prediabetics baseline 25.0 mIU/L, IQR 17.5-35.0, 2 years 6.7mIU/L, IQR 5.3-8.8, p < 0.001; non-diabetics baseline 20.0 mIU/L, IQR 14.0-30.0, 2 years 6.4 mIU/L, IQR 5.0-8.5, p < 0.001). HbA1c improved in all groups (diabetics baseline 56 mmol/mol, IQR 49-74 [7.3%, IQP 6.6-8.9], 2 years 38 mmol/mol, IQR 36-47 [5.6%, IQR 5.4-6.4], p < 0.001; prediabetics baseline 40 mmol/mol, IQR 39-42 [5.8%, IQR5.7-6.0], 2 years 36 mmol/mol, IQR 34-38 [5.5%, IQR 5.3-5.6], p < 0.001; non-diabetics baseline 35 mmol/mol, IQR 33-37 [5.4%, IQR 5.2-5.5]; 2 years 34 mmol/mol, IQR 31-36 [5.3%, IQR 5.0-5.4], p < 0.001). HOMA-IR improved in all groups (diabetics baseline 9.3 mmol/mol, IQR 5.4-12.9, 2 years 1.9 mmol/mol, IQR 1.4-2.7, p < 0.001; prediabetics baseline 7.0 mmol/mol, IQR 4.3-9.9, 2 years 1.6 mmol/mol, IQR 1.2-2.1, p < 0.001; non-diabetics 4.9 mmol/mol, IQR 3.4-7.3, 2 years 1.4 mmol/mol, IQR 1.1-1.9, p < 0.001). CONCLUSION Insulin homeostasis and glucometabolic control improve in all patients after LRYGB, not only in diabetics but also in prediabetics and non-diabetic obese patients, and this improvement is sustained 2 years after surgery.
Collapse
|
23
|
Comparing the Anti-diabetic Effect of Sleeve Gastrectomy with Transit Bipartition Against Sleeve Gastrectomy and Roux-en-Y Gastric Bypass Using a Diabetic Rodent Model. Obes Surg 2021; 31:2203-2210. [PMID: 33507518 DOI: 10.1007/s11695-021-05256-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE Roux-en-Y gastric bypass (RYGB) has superior long-term diabetes remission outcomes to sleeve gastrectomy (SG). However, in regions with a high prevalence of gastric cancer, RYGB may not be the best option. This study aimed to investigate the anti-diabetic effect of SG with transit bipartition (SG-TB) compared with SG and RYGB. MATERIALS AND METHODS A total of 32 diabetic Sprague-Dawley rat models were assigned to one of four groups: SG (n = 8), RYGB (n = 8), SG-TB (n = 8), and SHAM (n = 8). Body weight, food intake, blood glucose, and hormonal changes (glucagon-like peptide-1 (GLP-1), insulin, and glucagon) were measured to investigate the effect of surgery in all groups. Oral glucose tolerance test and insulin tolerance test were performed before and 8 weeks after surgery. RESULTS There were no significant differences in the postoperative changes in body weight and food intake among the SG, RYGB, and SG-TB groups. Postoperatively, the RYGB and SG-TB groups had significantly higher GLP-1 levels and lower insulin levels than the SG group. Further, RYGB and SG-TB had significantly better glucose control improvements than SG. There were no significant differences in GLP-1, insulin, glucagon, and homeostasis model assessment of insulin resistance levels between RYGB and SG-TB. The preoperative and postoperative values of all variables in the SHAM group did not show significant differences. CONCLUSION In this study using a diabetes-induced rodent model, we found that the anti-diabetic effect of SG-TB is superior to that of SG and non-inferior to that of RYGB.
Collapse
|
24
|
The Metabolic Effects of Pre-probiotic Supplementation After Roux-en-Y Gastric Bypass (RYGB) Surgery: a Prospective, Randomized Controlled Study. Obes Surg 2020; 31:215-223. [PMID: 32803709 DOI: 10.1007/s11695-020-04894-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/25/2020] [Accepted: 07/28/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Following Roux-en-Y gastric bypass (RYGB), positive alterations are observed in gut microbiota and intestinal peptides. Previous studies demonstrated similar alterations observed in cases when pre-probiotics are used without surgery. The aim of this trial was to evaluate the effectiveness of early use of pre-probiotics after RYGB. MATERIAL AND METHODS The operation and follow-up of the patients were performed at Istanbul University Medical Faculty. Thirty-two patients who had undergone RYGB were randomized to pre-probiotic group (PreProBG, n = 16; 200 g/day yogurt plus 10 g/day inulin+oligofructose) and probiotic group (ProBG, n = 16; 200 g/day yogurt only) for 6 months. Blood samples (glucose, insulin, A1c, GLP-1, PYY), anthropometric measurements, and appetite ratings have been evaluated at baseline and 3 (m3) and 6 (m6) months after RYGB. RESULTS Initial anthropometric measurements and appetite ratings decreased significantly after surgery and there were no significant differences between the groups. The decrease of area under the curve(insulin) was less and has a positive correlation with the changes in anthropometric measurements in PreProBG. GLP-1 and PYY which increased dramatically after surgery in all patients were higher in PreProBG. But this increase had a negative correlation with the changes in anthropometric measurements during the study. CONCLUSION Increased insulin, GLP-1, and PYY secretion was more enhanced by pre-probiotic use in early postoperative period. But this increase not only in anthropometric measurements but also in appetite ratings affects negatively, contrary to expectations. In summary, it should be investigated with new studies that use of pre-probiotics in the late postoperative period may be more effective in patients with weak insulin and incretin response and therefore insufficient weight loss. Trial Registration clinicaltrials.gov Identifier: NCT03517345.
Collapse
|
25
|
Vizioli C, Jaime-Lara RB, Franks AT, Ortiz R, Joseph PV. Untargeted Metabolomic Approach Shows No Differences in Subcutaneous Adipose Tissue of Diabetic and Non-Diabetic Subjects Undergoing Bariatric Surgery: An Exploratory Study. Biol Res Nurs 2020; 23:109-118. [PMID: 32762338 DOI: 10.1177/1099800420942900] [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: 12/12/2022]
Abstract
BACKGROUND Obesity plays a major role in the development of insulin resistance (IR) and diabetes (T2DM). Increased adipose tissue (AT) is particularly of interest because it activates a chronic inflammatory response in adipocytes and other tissues. AT plays key endocrine and metabolic functions, acting in the regulation of insulin sensitivity and energy homeostasis. Additionally, it can be easily collected during bariatric surgery. The purpose of this pilot study was to explore the potential differences in AT metabolism, through comparing the untargeted metabolomic profiles of diabetic and non-diabetic obese patients undergoing bariatric surgery. METHODS For this exploratory study, samples were collected from 17 subjects. Subcutaneous AT (SAT) samples from obese-diabetic (n = 8) and Obese-non-Diabetic (n = 9) subjects were obtained from the Human Metabolic Tissue Bank. Untargeted metabolomic profiling was performed by Metabolon® Inc. Statistical analysis was performed using the MetaboAnalyst 4.0 platform. RESULTS Among the 421 metabolites identified and analyzed there were no significant differences between the Obese-Diabetics and the Obese-non-Diabetics. Small changes were observed by fold change analysis mainly in lipid (n = 12; e.g. NEFAs) and amino acid (n = 8; e.g. BCAAs) metabolic pathways. Dysregulation of these metabolites has been associated with IR and other T2DM-related pathophysiological processes. CONCLUSION Obesity may influence SAT metabolism masking T2DM-dependent dysregulation. Better understanding the metabolic differences within SAT in diabetic populations may help identify potential biomarkers for diagnosis and monitoring of T2DM in patients undergoing bariatric surgery.
Collapse
Affiliation(s)
- Carlotta Vizioli
- Sensory Science & Metabolism Unit, Biobehavioral Branch, Division of Intramural Research, National Institute of Nursing Research, 2511National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Rosario B Jaime-Lara
- Sensory Science & Metabolism Unit, Biobehavioral Branch, Division of Intramural Research, National Institute of Nursing Research, 2511National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Alexis T Franks
- Sensory Science & Metabolism Unit, Biobehavioral Branch, Division of Intramural Research, National Institute of Nursing Research, 2511National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Rodrigo Ortiz
- Sensory Science & Metabolism Unit, Biobehavioral Branch, Division of Intramural Research, National Institute of Nursing Research, 2511National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Paule V Joseph
- Sensory Science & Metabolism Unit, Biobehavioral Branch, Division of Intramural Research, National Institute of Nursing Research, 2511National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| |
Collapse
|
26
|
Amouyal C, Castel J, Guay C, Lacombe A, Denom J, Migrenne-Li S, Rouault C, Marquet F, Georgiadou E, Stylianides T, Luquet S, Le Stunff H, Scharfmann R, Clément K, Rutter GA, Taboureau O, Magnan C, Regazzi R, Andreelli F. A surrogate of Roux-en-Y gastric bypass (the enterogastro anastomosis surgery) regulates multiple beta-cell pathways during resolution of diabetes in ob/ob mice. EBioMedicine 2020; 58:102895. [PMID: 32739864 PMCID: PMC7393530 DOI: 10.1016/j.ebiom.2020.102895] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bariatric surgery is an effective treatment for type 2 diabetes. Early post-surgical enhancement of insulin secretion is key for diabetes remission. The full complement of mechanisms responsible for improved pancreatic beta cell functionality after bariatric surgery is still unclear. Our aim was to identify pathways, evident in the islet transcriptome, that characterize the adaptive response to bariatric surgery independently of body weight changes. METHODS We performed entero-gastro-anastomosis (EGA) with pyloric ligature in leptin-deficient ob/ob mice as a surrogate of Roux-en-Y gastric bypass (RYGB) in humans. Multiple approaches such as determination of glucose tolerance, GLP-1 and insulin secretion, whole body insulin sensitivity, ex vivo glucose-stimulated insulin secretion (GSIS) and functional multicellular Ca2+-imaging, profiling of mRNA and of miRNA expression were utilized to identify significant biological processes involved in pancreatic islet recovery. FINDINGS EGA resolved diabetes, increased pancreatic insulin content and GSIS despite a persistent increase in fat mass, systemic and intra-islet inflammation, and lipotoxicity. Surgery differentially regulated 193 genes in the islet, most of which were involved in the regulation of glucose metabolism, insulin secretion, calcium signaling or beta cell viability, and these were normalized alongside changes in glucose metabolism, intracellular Ca2+ dynamics and the threshold for GSIS. Furthermore, 27 islet miRNAs were differentially regulated, four of them hubs in a miRNA-gene interaction network and four others part of a blood signature of diabetes resolution in ob/ob mice and in humans. INTERPRETATION Taken together, our data highlight novel miRNA-gene interactions in the pancreatic islet during the resolution of diabetes after bariatric surgery that form part of a blood signature of diabetes reversal. FUNDING European Union's Horizon 2020 research and innovation programme via the Innovative Medicines Initiative 2 Joint Undertaking (RHAPSODY), INSERM, Société Francophone du Diabète, Institut Benjamin Delessert, Wellcome Trust Investigator Award (212625/Z/18/Z), MRC Programme grants (MR/R022259/1, MR/J0003042/1, MR/L020149/1), Diabetes UK (BDA/11/0004210, BDA/15/0005275, BDA 16/0005485) project grants, National Science Foundation (310030-188447), Fondation de l'Avenir.
Collapse
Affiliation(s)
- Chloé Amouyal
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic approaches (NutriOmics), Paris, France; AP-HP, Pitié-Salpêtrière Hospital, Diabetology department, F-75013 Paris, France
| | - Julien Castel
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | - Claudiane Guay
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, CH-1005, Lausanne, Switzerland
| | - Amélie Lacombe
- PreclinICAN, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Jessica Denom
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | | | - Christine Rouault
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic approaches (NutriOmics), Paris, France
| | - Florian Marquet
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic approaches (NutriOmics), Paris, France
| | - Eleni Georgiadou
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | | | - Serge Luquet
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | - Hervé Le Stunff
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | - Raphael Scharfmann
- Université de Paris, Cochin Institute, Inserm U1016, Paris 75014, France
| | - Karine Clément
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic approaches (NutriOmics), Paris, France; APHP, Pitié-Salpêtrière Hospital, Nutrition department, F-75013 Paris, France
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK; Lee Kong Chian School of Medicine, Nan Yang Technological University, Singapore
| | - Olivier Taboureau
- Université de Paris, BFA, Team CMPLI, Inserm U1133, CNRS UMR 8251, Paris, France
| | | | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, CH-1005, Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, Rue du Bugnon 7, CH-1005 Lausanne, Switzerland
| | - Fabrizio Andreelli
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic approaches (NutriOmics), Paris, France; AP-HP, Pitié-Salpêtrière Hospital, Diabetology department, F-75013 Paris, France.
| |
Collapse
|
27
|
Singh D, Arumalla K, Aggarwal S, Singla V, Ganie A, Malhotra N. Impact of Bariatric Surgery on Clinical, Biochemical, and Hormonal Parameters in Women with Polycystic Ovary Syndrome (PCOS). Obes Surg 2020; 30:2294-2300. [DOI: 10.1007/s11695-020-04487-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
28
|
Katsogiannos P, Kamble PG, Wiklund U, Sundbom M, Espes D, Hammar U, Karlsson FA, Pereira MJ, Eriksson JW. Rapid changes in neuroendocrine regulation may contribute to reversal of type 2 diabetes after gastric bypass surgery. Endocrine 2020; 67:344-353. [PMID: 31983031 PMCID: PMC7026226 DOI: 10.1007/s12020-020-02203-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/13/2020] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To explore the role of hormones and the autonomic nervous system in the rapid remission of diabetes after Roux-en-Y Gastric Bypass (RYGB). RESEARCH DESIGN AND METHODS Nineteen obese patients with type 2 diabetes, 7 M/12 F, were randomized (2:1) to RYGB or standard-of-care medical treatment (control). At baseline and 4 and 24 weeks post surgery, fasting blood sampling, OGTT, intravenous arginine challenge, and heart-rate variability (HRV) assessments were performed. RESULTS At both 4 and 24 weeks post-RYGB the following effects were found: arginine-stimulated insulin secretion was reduced. GLP-1, GIP, and glucagon rise during OGTT was enhanced. IGF-1 and GH levels increased. In addition, total HRV and spectral components PLF (power of low frequency) and PHF (power of high frequency) increased. At 4 weeks, morning cortisol was lower than baseline and 24 weeks. At 24 weeks, NEFA levels during OGTT, and the PLF/PHF ratio decreased. None of these changes were seen in the control group. CONCLUSIONS There were rapid changes within 4 weeks after RYGB: signs of enhanced parasympathetic nerve activity, reduced morning cortisol, and enhanced incretin and glucagon responses to glucose. The findings suggest that neurohormonal mechanisms can contribute to the rapid improvement of insulin resistance and glycemia following RYGB in type 2 diabetes.
Collapse
Affiliation(s)
- Petros Katsogiannos
- Department of Medical Science, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Prasad G Kamble
- Department of Medical Science, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Urban Wiklund
- Radiation Sciences, Biomedical Engineering & Informatics, Umeå University, Umeå, Sweden
| | | | - Daniel Espes
- Department of Medical Science, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Ulf Hammar
- Department of Medical Science, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - F Anders Karlsson
- Department of Medical Science, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Maria J Pereira
- Department of Medical Science, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Science, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
29
|
van Baar AC, Beuers U, Wong K, Haidry R, Costamagna G, Hafedi A, Deviere J, Ghosh SS, Lopez-Talavera JC, Rodriguez L, Galvao Neto MP, Sanyal A, Bergman JJ. Endoscopic duodenal mucosal resurfacing improves glycaemic and hepatic indices in type 2 diabetes: 6-month multicentre results. JHEP Rep 2019; 1:429-437. [PMID: 32039394 PMCID: PMC7005649 DOI: 10.1016/j.jhepr.2019.10.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 09/27/2019] [Accepted: 10/29/2019] [Indexed: 02/06/2023] Open
Abstract
Insulin resistance is a core pathophysiological defect underscoring type 2 diabetes mellitus (T2DM) and non-alcoholic fatty liver disease (NAFLD). Both conditions improve with duodenal exclusion surgery. Duodenal mucosal resurfacing (DMR) is an endoscopic intervention developed to treat metabolic disease which has been shown to improve glycaemia in patients with poorly controlled T2DM. Herein, we aimed to further analyse the effects of DMR on hepatic and metabolic parameters in this patient cohort. METHODS Eighty-five patients with T2DM who received endoscopic DMR treatment were enrolled from 5 centres and followed up for 6 months. We assessed safety in all patients. Efficacy was evaluated in patients who received at least 9 cm of duodenal ablation (n = 67). Endpoints included HbA1c, fasting plasma glucose, weight and aminotransferase levels. Metabolomic analysis was conducted in a subgroup (n = 14). Data were analysed using paired t test or ANOVA for repeated measures with Bonferroni correction and correction for initial weight loss if applicable. RESULTS The DMR procedure was completed with no intraprocedural complications in the entire cohort. HbA1c was lower 6 months after DMR than at baseline (7.9 ± 0.2% vs. 9.0 ± 0.2% [mean ± SE], p ≪0.001). Fasting plasma glucose was also significantly lower 6 months after DMR compared to baseline (161 ± 7 mg/dl vs. 189 ± 6 mg/dl, p = 0.005). Body weight decreased slightly. At 6 months, alanine aminotransferase had decreased from 41 ± 3 IU/L to 29 ± 2 IU/L (p ≪0.001) and aspartate aminotransferase had decreased from 30 ± 2 IU/L to 23 ± 1 IU/L (p ≪0.001). Metabolomic analysis demonstrated that DMR had key lipid-lowering, insulin-sensitizing and anti-inflammatory effects, as well as increasing antioxidant capacity. Mean FIB-4 was also markedly decreased. CONCLUSION Hydrothermal ablation of the duodenum by DMR elicits a beneficial metabolic response in patients with T2DM. DMR also improves hepatic indices, potentially through an insulin-sensitizing mechanism. These encouraging data deserve further evaluation in randomized controlled trials. LAY SUMMARY Hydrothermal duodenal mucosal resurfacing (DMR) is an endoscopic technique designed to treat metabolic disease through ablation of the duodenal mucosa. DMR is a safe procedure which improves glycaemia and hepatic indices in patients with type 2 diabetes mellitus. DMR is an insulin-sensitizing intervention which can be complementary to lifestyle intervention approaches and pharmacological treatments aimed at preserving the pancreas and liver from failure. DMR is a potential therapeutic solution for patients with type 2 diabetes and fatty liver disease.
Collapse
Affiliation(s)
- Annieke C.G. van Baar
- Department of Gastroenterology & Hepatology, Amsterdam University Medical Centers, location Academic Medical Center, Amsterdam, The Netherlands
| | - Ulrich Beuers
- Department of Gastroenterology & Hepatology, Amsterdam University Medical Centers, location Academic Medical Center, Amsterdam, The Netherlands
| | - Kari Wong
- Metabolon, Inc., Morrisville, NC, United States
| | - Rehan Haidry
- Department of Gastroenterology, University College Hospital NHS Foundation Trust, London, United Kingdom
| | - Guido Costamagna
- Digestive Endoscopy Unit. Fondazione Policlinico Universitario A. Gemelli - IRCCS, Rome, Italy
- Università Cattolica del S. Cuore, Rome, Italy
| | - Alia Hafedi
- Department of Gastroenterology, Erasme University Hospital, Brussels, Belgium
| | - Jacques Deviere
- Department of Gastroenterology, Erasme University Hospital, Brussels, Belgium
| | | | | | | | | | - Arun Sanyal
- Department of Gastroenterology & Hepatology, Virginia Commonwealth University, Richmond, VA, United States
| | - Jacques J.G.H.M. Bergman
- Department of Gastroenterology & Hepatology, Amsterdam University Medical Centers, location Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Catalina MOS, Redondo PC, Granados MP, Cantonero C, Sanchez-Collado J, Albarran L, Lopez JJ. New Insights into Adipokines as Potential Biomarkers for Type-2 Diabetes Mellitus. Curr Med Chem 2019; 26:4119-4144. [PMID: 29210636 DOI: 10.2174/0929867325666171205162248] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 10/30/2017] [Accepted: 10/30/2017] [Indexed: 02/06/2023]
Abstract
A large number of studies have been focused on investigating serum biomarkers associated with risk or diagnosis of type-2 diabetes mellitus. In the last decade, promising studies have shown that circulating levels of adipokines could be used as a relevant biomarker for diabetes mellitus progression as well as therapeutic future targets. Here, we discuss the possible use of recently described adipokines, including apelin, omentin-1, resistin, FGF-21, neuregulin-4 and visfatin, as early biomarkers for diabetes. In addition, we also include recent findings of other well known adipokines such as leptin and adiponectin. In conclusion, further studies are needed to clarify the pathophysiological significance and clinical value of these biological factors as potential biomarkers in type-2 diabetes and related dysfunctions.
Collapse
Affiliation(s)
| | - Pedro C Redondo
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003-Caceres, Spain
| | - Maria P Granados
- Aldea Moret's Medical Center, Extremadura Health Service, 10195-Caceres, Spain
| | - Carlos Cantonero
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003-Caceres, Spain
| | - Jose Sanchez-Collado
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003-Caceres, Spain
| | - Letizia Albarran
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003-Caceres, Spain
| | - Jose J Lopez
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003-Caceres, Spain
| |
Collapse
|
31
|
Dynamic changes of muscle insulin sensitivity after metabolic surgery. Nat Commun 2019; 10:4179. [PMID: 31519890 PMCID: PMC6744497 DOI: 10.1038/s41467-019-12081-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 08/15/2019] [Indexed: 02/07/2023] Open
Abstract
The mechanisms underlying improved insulin sensitivity after surgically-induced weight loss are still unclear. We monitored skeletal muscle metabolism in obese individuals before and over 52 weeks after metabolic surgery. Initial weight loss occurs in parallel with a decrease in muscle oxidative capacity and respiratory control ratio. Persistent elevation of intramyocellular lipid intermediates, likely resulting from unrestrained adipose tissue lipolysis, accompanies the lack of rapid changes in insulin sensitivity. Simultaneously, alterations in skeletal muscle expression of genes involved in calcium/lipid metabolism and mitochondrial function associate with subsequent distinct DNA methylation patterns at 52 weeks after surgery. Thus, initial unfavorable metabolic changes including insulin resistance of adipose tissue and skeletal muscle precede epigenetic modifications of genes involved in muscle energy metabolism and the long-term improvement of insulin sensitivity. Surgical weight-loss interventions improve insulin sensitivity via incompletely understood mechanisms. Here the authors assess skeletal muscle epigenetic changes in individuals with obesity following metabolic surgery and compare them with data from individuals without obesity.
Collapse
|
32
|
Katsogiannos P, Kamble PG, Boersma GJ, Karlsson FA, Lundkvist P, Sundbom M, Pereira MJ, Eriksson JW. Early Changes in Adipose Tissue Morphology, Gene Expression, and Metabolism After RYGB in Patients With Obesity and T2D. J Clin Endocrinol Metab 2019; 104:2601-2613. [PMID: 30689903 DOI: 10.1210/jc.2018-02165] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 01/18/2019] [Indexed: 02/13/2023]
Abstract
CONTEXT Roux-en-Y gastric bypass (RYGB) surgery effectively prevents or treats type 2 diabetes (T2D). Adipose tissue (AT) mechanisms may be of importance. OBJECTIVE To assess the relationship between early changes in whole-body and AT metabolism in surgically treated patients with T2D. DESIGN AND SETTING A randomized single-center study. PATIENTS Nineteen patients with T2D with body mass index 30 to 45 kg/m2. INTERVENTIONS Thirteen patients were assessed at baseline and 4 and 24 weeks after RYGB (preceded by a 4-week low-calorie diet) and compared with 6 control patients continuing standard medical treatment: oral glucose tolerance test, subcutaneous AT biopsies for gene expression, adipocyte size, glucose uptake, lipolysis, and insulin action. RESULTS At 4 and 24 weeks post-RYGB, all patients but one had stopped diabetes medication. Fasting glucose, HbA1c, and insulin levels decreased and the Matsuda index increased compared with baseline (P < 0.01 for all), indicating improved whole-body insulin sensitivity. Mean adipocyte size significantly reduced, more at 4 than at 24 weeks; at 4 weeks, glucose uptake per adipocyte was lowered, and isoproterenol-stimulated lipolysis tended to increase, whereas the fold insulin effects on glucose uptake and lipolysis were unchanged. Expression of genes involved in fatty acid oxidation, CPT1b and adiponectin, was increased at 4 weeks, whereas leptin and E2F1 (involved in cell proliferation) were reduced (P < 0.05 for all). CONCLUSION Glycemic control and in vivo insulin sensitivity improved 4 weeks after RYGB, but adipocyte insulin sensitivity did not change despite a marked reduction in adipocyte size. Thus, mechanisms for a rapid improvement of T2D after RYGB may occur mainly in other tissues than adipose.
Collapse
MESH Headings
- Adipocytes/metabolism
- Adult
- Biopsy
- Blood Glucose/analysis
- Blood Glucose/metabolism
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/surgery
- Female
- Follow-Up Studies
- Gastric Bypass
- Humans
- Insulin/metabolism
- Insulin Resistance
- Male
- Middle Aged
- Obesity, Morbid/blood
- Obesity, Morbid/complications
- Obesity, Morbid/metabolism
- Obesity, Morbid/surgery
- Subcutaneous Fat, Abdominal/cytology
- Subcutaneous Fat, Abdominal/metabolism
- Subcutaneous Fat, Abdominal/pathology
- Treatment Outcome
Collapse
Affiliation(s)
| | - Prasad G Kamble
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Gretha J Boersma
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Per Lundkvist
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Magnus Sundbom
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Maria J Pereira
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
33
|
Goldstein DS. How does homeostasis happen? Integrative physiological, systems biological, and evolutionary perspectives. Am J Physiol Regul Integr Comp Physiol 2019; 316:R301-R317. [PMID: 30649893 DOI: 10.1152/ajpregu.00396.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Homeostasis is a founding principle of integrative physiology. In current systems biology, however, homeostasis seems almost invisible. Is homeostasis a key goal driving body processes, or is it an emergent mechanistic fact? In this perspective piece, I propose that the integrative physiological and systems biological viewpoints about homeostasis reflect different epistemologies, different philosophies of knowledge. Integrative physiology is concept driven. It attempts to explain biological phenomena by continuous formation of theories that experimentation or observation can test. In integrative physiology, "function" refers to goals or purposes. Systems biology is data driven. It explains biological phenomena in terms of "omics"-i.e., genomics, gene expression, epigenomics, proteomics, and metabolomics-it depicts the data in computer models of complex cascades or networks, and it makes predictions from the models. In systems biology, "function" refers more to mechanisms than to goals. The integrative physiologist emphasizes homeostasis of internal variables such as Pco2 and blood pressure. The systems biologist views these emphases as teleological and unparsimonious in that the "regulated variable" (e.g., arterial Pco2 and blood pressure) and the "regulator" (e.g., the "carbistat" and "barostat") are unobservable constructs. The integrative physiologist views systems biological explanations as not really explanations but descriptions that cannot account for phenomena we humans believe exist, although they cannot be observed directly, such as feelings and, ultimately, the conscious mind. This essay reviews the history of the two epistemologies, emphasizing autonomic neuroscience. I predict rapprochement of integrative physiology with systems biology. The resolution will avoid teleological purposiveness, transcend pure mechanism, and incorporate adaptiveness in evolution, i.e., "Darwinian medicine."
Collapse
Affiliation(s)
- David S Goldstein
- Clinical Neurocardiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health , Bethesda, Maryland
| |
Collapse
|
34
|
Andersson DP, Dahlman I, Eriksson Hogling D, Bäckdahl J, Toft E, Qvisth V, Näslund E, Thorell A, Rydén M, Arner P. Improved metabolism and body composition beyond normal levels following gastric bypass surgery: a longitudinal study. J Intern Med 2019; 285:92-101. [PMID: 30141528 DOI: 10.1111/joim.12824] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The cardiometabolic risk profile improves following bariatric surgery. However, the degree of improvement in relation to weight-stable control subjects is unknown. OBJECTIVES To study the differences in cardiometabolic risk profile between formerly obese patients following Roux-en-Y gastric bypass (RYGB) surgery and control subjects. METHODS Subjects undergoing RYGB and reaching a BMI <30 kg m-2 2 years postsurgery were matched with control subjects regarding age, sex and BMI. The following examinations were performed: insulin sensitivity measured by hyperinsulinaemic-euglycaemic clamp, insulin clearance, homeostatic model assessment of insulin resistance (HOMA-IR), lipid profile, inflammatory marker levels, dual-energy X-ray absorptiometry and subcutaneous adipose tissue cellularity (fat cell size and number). RESULTS Sixty-nine subjects undergoing RYGB were matched to a control subject. Insulin sensitivity measured by hyperinsulinaemic-euglycaemic clamp, blood pressure, inflammatory status and glucose, triglyceride and HDL cholesterol levels were comparable to values of control subjects. However, HOMA-IR (1.0 ± 0.5 vs. 1.3 ± 0.7, P = 0.005), insulin clearance (0.38 ± 0.08 vs. 0.34 ± 0.08 μL m-2 min-1 , P < 0.0001) and circulating levels of insulin (31 ± 15 vs. 37 ± 17 pmol L-1 , P = 0.008), total cholesterol (4.1 ± 0.7 vs. 4.8 ± 0.9 mmol L-1 , P < 0.0001) and LDL cholesterol (2.1 ± 0.6 vs. 2.9 ± 0.8 mmol L-1 , P < 0.0001) were improved beyond the levels in matched control subjects. Furthermore, formerly obese subjects had higher lean and lower fat mass as well as a more benign type of adipose cellularity (hyperplasia with many small fat cells) compared to control subjects. CONCLUSIONS Subjects who underwent RYGB and reached a postobese state demonstrated a beneficial body composition, slightly increased insulin sensitivity as indirectly measured by HOMA-IR and higher insulin clearance, lower atherogenic lipid/lipoprotein levels and benign adipocyte morphology compared with control subjects who had never been obese. In line with previous results, our findings may in part explain why RYGB confers long-term protection against metabolic complications.
Collapse
Affiliation(s)
- D P Andersson
- Department of Medicine (H7), Karolinska University Hospital, Stockholm, Sweden
| | - I Dahlman
- Department of Medicine, Ersta Hospital, Stockholm, Sweden
| | - D Eriksson Hogling
- Department of Medicine (H7), Karolinska University Hospital, Stockholm, Sweden
| | - J Bäckdahl
- Department of Medicine (H7), Karolinska University Hospital, Stockholm, Sweden
| | - E Toft
- Department of Medicine, Ersta Hospital, Stockholm, Sweden
| | - V Qvisth
- Department of Medicine, Ersta Hospital, Stockholm, Sweden
| | - E Näslund
- Department of Clinical Sciences, Danderyd Hospital, Stockholm, Sweden
| | - A Thorell
- Department of Surgery, Karolinska Institutet, Ersta Hospital, Stockholm, Sweden
| | - M Rydén
- Department of Medicine (H7), Karolinska University Hospital, Stockholm, Sweden
| | - P Arner
- Department of Medicine (H7), Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
35
|
Laferrère B, Pattou F. Weight-Independent Mechanisms of Glucose Control After Roux-en-Y Gastric Bypass. Front Endocrinol (Lausanne) 2018; 9:530. [PMID: 30250454 PMCID: PMC6140402 DOI: 10.3389/fendo.2018.00530] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 08/22/2018] [Indexed: 12/14/2022] Open
Abstract
Roux-en-Y gastric bypass results in large and sustained weight loss and resolution of type 2 diabetes in 60% of cases at 1-2 years. In addition to calorie restriction and weight loss, various gastro-intestinal mediated mechanisms, independent of weight loss, also contribute to glucose control. The anatomical re-arrangement of the small intestine after gastric bypass results in accelerated nutrient transit, enhances the release of post-prandial gut hormones incretins and of insulin, alters the metabolism and the entero-hepatic cycle of bile acids, modifies intestinal glucose uptake and metabolism, and alters the composition and function of the microbiome. The amelioration of beta cell function after gastric bypass in individuals with type 2 diabetes requires enteric stimulation. However, beta cell function in response to intravenous glucose stimulus remains severely impaired, even in individuals in full clinical diabetes remission. The permanent impairment of the beta cell may explain diabetes relapse years after surgery.
Collapse
Affiliation(s)
- Blandine Laferrère
- Division of Endocrinology, New York Obesity Nutrition Research Center, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - François Pattou
- Translational Research on Diabetes, UMR 1190, Inserm, Université Lille, Lille, France
- Endocrine and Metabolic Surgery, CHU Lille, Lille, France
| |
Collapse
|
36
|
One-year impact of bariatric surgery on serum anti-Mullerian-hormone levels in severely obese women. J Assist Reprod Genet 2018; 35:1317-1324. [PMID: 29754264 DOI: 10.1007/s10815-018-1196-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 04/25/2018] [Indexed: 01/27/2023] Open
Abstract
INTRODUCTION Although bariatric surgery seems to increase spontaneous fertility by improving ovulatory function in young women, its impact on ovarian reserve remains largely unknown. OBJECTIVE To evaluate changes in serum anti-Mullerian hormone (AMH) levels in reproductive-age severely obese women after bariatric surgery (BS). METHODS AMH levels were measured retrospectively in 39 women (mean age 34.6 ± 1.1 years, range 18-45) that underwent a sleeve gastrectomy or Roux-en-Y gastric bypass (RYGB) at baseline, and 6 and 12 months after BS. Metabolic and micronutrient status, including fasting plasma insulin and glucose, HOMA-IR, leptin, adiponectin, calcium, albumin, transthyretin, ferritin, vitamins (B9, B12, B1, A, E, D), zinc, and selenium, were assessed in all patients before and 1 year after BS. RESULTS Of the patients, 79% had class-3 obesity. At 6 and 12 months, mean total weight losses (TWL) were 26 and 30%; mean excess weight losses (EWL) were 61.7 and 70.2%. Compared to baseline, AMH levels significantly decreased by 18% at 6 months, and 32% at 12 months post-operatively (p = 0.010 and p = 0.001, respectively). There was no correlation between AMH variation and changes in metabolic parameters or micronutrient levels. Remarkably, changes in AMH levels did not differ between sleeve and RYGB patients and were not correlated with EWL. CONCLUSION This pilot study shows a drastic reduction in AMH levels at 1 year after BS in reproductive-age severely obese women, which was not related to weight loss: this suggests a negative impact of BS on ovarian reserve, at least in the short term.
Collapse
|
37
|
Meister KM, Schauer PR, Brethauer SA, Aminian A. Effect of Gastrogastric Fistula Closure in Type 2 Diabetes. Obes Surg 2017; 28:1086-1090. [PMID: 29090378 DOI: 10.1007/s11695-017-2976-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Roux-en-Y gastric bypass (RYGB) has been shown to significantly improve glucose control in patients with type 2 diabetes (T2DM). The formation of a gastrogastric fistula (GGF) allows nutrients to pass through the native route, rather than bypassing the duodenum in typical RYGB configuration. We sought to evaluate the effect of revisional bariatric surgery for known GGF on control of diabetes. METHODS A retrospective chart review of a single academic institution was performed to identify patients who had T2DM at the time of corrective surgery for a GGF. Baseline characteristics, and postoperative outcomes including changes in body mass index (BMI), glycated hemoglobin, fasting blood glucose (FBG), and diabetes medications were assessed. RESULTS Ten patients were identified with GGF who had T2DM at the time of corrective surgery. Patients had a male-to-female ratio of 2:3, a mean age of 59.2 ± 10 years, a mean baseline BMI of 38.1 ± 17.6 kg/m2, and a median duration of 9 years (interquartile range 6-14) from initial RYGB to revision. At a mean follow-up of 14.9 ± 8.5 months, a mean reduction in BMI of 4.9 ± 6 kg/m2 was associated with a significant mean reduction in FBG (167.1 ± 88.2 vs. 106.1 ± 20.4 mg/dL, p = 0.04) and number of diabetes medications (1.4 ± 0.8 vs. 0.7 ± 0.7, p = 0.04). CONCLUSION In patients with diabetes and GGF, a corrective surgery for closure of fistula and restoration of bypass anatomy results in improvement of glucose control and status of diabetes medications. This finding can highlight the potential metabolic significance of duodenal exclusion.
Collapse
Affiliation(s)
- Katherine M Meister
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, 9500 Euclid Avenue, M61, Cleveland, OH, 44195, USA
| | - Philip R Schauer
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, 9500 Euclid Avenue, M61, Cleveland, OH, 44195, USA
| | - Stacy A Brethauer
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, 9500 Euclid Avenue, M61, Cleveland, OH, 44195, USA
| | - Ali Aminian
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, 9500 Euclid Avenue, M61, Cleveland, OH, 44195, USA.
| |
Collapse
|
38
|
Ramachandran D, Clara R, Fedele S, Hu J, Lackzo E, Huang JY, Verdin E, Langhans W, Mansouri A. Intestinal SIRT3 overexpression in mice improves whole body glucose homeostasis independent of body weight. Mol Metab 2017; 6:1264-1273. [PMID: 29031725 PMCID: PMC5641632 DOI: 10.1016/j.molmet.2017.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/11/2017] [Accepted: 07/14/2017] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Intestinal metabolism might play a greater role in regulating whole body metabolism than previously believed. We aimed to enhance enterocyte metabolism in mice and investigate if it plays a role in diet-induced obesity (DIO) and its comorbidities. METHODS Using the cre-loxP system, we overexpressed the mitochondrial NAD+ dependent protein deacetylase SIRT3 in enterocytes of mice (iSIRT3 mice). We chronically fed iSIRT3 mice and floxed-SIRT3 control (S3fl) mice a low-fat, control diet (CD) or a high-fat diet (HFD) and then phenotyped the mice. RESULTS There were no genotype differences in any of the parameters tested when the mice were fed CD. Also, iSIRT3 mice were equally susceptible to the development of DIO as S3fl mice when fed HFD. They were, however, better able than S3fl mice to regulate their blood glucose levels in response to exogenous insulin and glucose, indicating that they were protected from developing insulin resistance. This improved glucose homeostasis was accompanied by an increase in enterocyte metabolic activity and an upregulation of ketogenic gene expression in the small intestine. CONCLUSION Enhancing enterocyte oxidative metabolism can improve whole body glucose homeostasis.
Collapse
Affiliation(s)
| | - Rosmarie Clara
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Shahana Fedele
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Junmin Hu
- Functional Genomics Center Zurich (FGCZ), ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Endre Lackzo
- Functional Genomics Center Zurich (FGCZ), ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Jing-Yi Huang
- Gladstone Institute of Virology and Immunology, University of California, San Francisco, CA, USA
| | - Eric Verdin
- Gladstone Institute of Virology and Immunology, University of California, San Francisco, CA, USA
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Abdelhak Mansouri
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
39
|
Michaud A, Grenier-Larouche T, Caron-Dorval D, Marceau S, Biertho L, Simard S, Richard D, Tchernof A, Carpentier AC. Biliopancreatic diversion with duodenal switch leads to better postprandial glucose level and beta cell function than sleeve gastrectomy in individuals with type 2 diabetes very early after surgery. Metabolism 2017; 74:10-21. [PMID: 28764844 DOI: 10.1016/j.metabol.2017.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/16/2017] [Accepted: 06/17/2017] [Indexed: 12/29/2022]
Abstract
OBJECTIVE The aim of this study was to compare the short-term effect of sleeve gastrectomy (SG) and biliopancreatic diversion with duodenal switch (DS) in order to determine if exclusion of the upper gastrointestinal tract confers greater metabolic improvement, independent of weight loss. METHODS Standard meals were administered before and on day 3 and 4 after SG to assess insulin sensitivity, β-cell function and gastrointestinal hormone responses in matched normoglycemic (NG) and type 2 diabetes (T2D) participants. A third group of matched T2D participants who underwent DS with the same meal test administered prior to and 3days after surgery was also recruited. RESULTS Despite significant metabolic improvement, T2D participants failed to fully normalize insulin resistance and β-cell dysfunction 3 and 4days after SG. Our results demonstrate the superiority of DS over SG in terms of short-term improvement in postprandial glucose excursion and β-cell function 3days after the surgery, with similar improvement in hepatic insulin sensitivity. CONCLUSION Our findings support the notion that caloric restriction represents an important mechanism to explain the very early anti-diabetic effects observed after bariatric surgery. However, exclusion of the upper gastrointestinal tract also provides further metabolic improvements, possibly mediated by gastrointestinal hormonal responses and altered postprandial glucose absorption.
Collapse
Affiliation(s)
- Andréanne Michaud
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Thomas Grenier-Larouche
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada; Department of Medicine, Division of Endocrinology, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Québec, Canada
| | - Dominique Caron-Dorval
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
| | - Simon Marceau
- Department of Surgery, Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec, Canada
| | - Laurent Biertho
- Department of Surgery, Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec, Canada
| | - Serge Simard
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
| | - Denis Richard
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
| | - André Tchernof
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada.
| | - André C Carpentier
- Department of Medicine, Division of Endocrinology, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Québec, Canada.
| |
Collapse
|
40
|
Deem JD, Muta K, Scarlett JM, Morton GJ, Schwartz MW. How Should We Think About the Role of the Brain in Glucose Homeostasis and Diabetes? Diabetes 2017; 66:1758-1765. [PMID: 28603139 PMCID: PMC5482090 DOI: 10.2337/dbi16-0067] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 02/25/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Jennifer D Deem
- Department of Medicine, University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Kenjiro Muta
- Department of Medicine, University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Jarrad M Scarlett
- Department of Medicine, University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Gregory J Morton
- Department of Medicine, University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Michael W Schwartz
- Department of Medicine, University of Washington Diabetes Institute, University of Washington, Seattle, WA
| |
Collapse
|
41
|
Droz BA, Sneed BL, Jackson CV, Zimmerman KM, Michael MD, Emmerson PJ, Coskun T, Peterson RG. Correlation of disease severity with body weight and high fat diet in the FATZO/Pco mouse. PLoS One 2017. [PMID: 28640904 PMCID: PMC5480996 DOI: 10.1371/journal.pone.0179808] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Obesity in many current pre-clinical animal models of obesity and diabetes is mediated by monogenic mutations; these are rarely associated with the development of human obesity. A new mouse model, the FATZO mouse, has been developed to provide polygenic obesity and a metabolic pattern of hyperglycemia and hyperinsulinemia, that support the presence of insulin resistance similar to metabolic disease in patients with insulin resistance/type 2 diabetes. The FATZO mouse resulted from a cross of C57BL/6J and AKR/J mice followed by selective inbreeding for obesity, increased insulin and hyperglycemia. Since many clinical studies have established a close link between higher body weight and the development of type 2 diabetes, we investigated whether time to progression to type 2 diabetes or disease severity in FATZO mice was dependent on weight gain in young animals. Our results indicate that lighter animals developed metabolic disturbances much slower and to a lesser magnitude than their heavier counterparts. Consumption of a diet containing high fat, accelerated weight gain in parallel with disease progression. A naturally occurring and significant variation in the body weight of FATZO offspring enables these mice to be identified as low, mid and high body weight groups at a young age. These weight groups remain into adulthood and correspond to slow, medium and accelerated development of type 2 diabetes. Thus, body weight inclusion criteria can optimize the FATZO model for studies of prevention, stabilization or treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Brian A. Droz
- Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Bria L. Sneed
- Ball State University, Muncie, Indiana, United States of America
| | - Charles V. Jackson
- Crown Bioscience - Indiana, Indianapolis, Indiana, United States of America
| | - Karen M. Zimmerman
- Crown Bioscience - Indiana, Indianapolis, Indiana, United States of America
| | - M. Dodson Michael
- Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Paul J. Emmerson
- Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Tamer Coskun
- Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Richard G. Peterson
- Crown Bioscience - Indiana, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
42
|
Affiliation(s)
- Jian-Ping Weng
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Gang Hu
- Pennington Biomedical Research Center, Baton Rouge, LA
| |
Collapse
|
43
|
Small Intestinal Bypass Induces a Persistent Weight-Loss Effect and Improves Glucose Tolerance in Obese Rats. Obes Surg 2017; 27:1859-1866. [DOI: 10.1007/s11695-017-2571-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|