1
|
Leyaro B, Howie L, McMahon K, Ali A, Carragher R. Weight loss outcomes and associated factors after metabolic bariatric surgery: Analysis of routine clinical data in Scotland. Am J Surg 2025; 241:116151. [PMID: 39719776 DOI: 10.1016/j.amjsurg.2024.116151] [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: 09/12/2024] [Revised: 12/03/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND Bariatric surgery is a cornerstone intervention for individuals with severe obesity, offering substantial and sustainable weight loss. METHODS This retrospective cohort study included 186 patients with obesity and Type2 diabetes who underwent sleeve gastrectomy (SG) or Roux-en-Y gastric bypass (RYGB) between 2009 and 2020 at University Hospital Ayr. Optimal clinical response weight loss was defined as excess weight loss (%EWL) ≥50 % or total weight loss (%TWL) ≥20 %. RESULTS At 2-years post-surgery, 43.6 % achieved ≥50 %EWL, and 44.1 % achieved ≥20%TWL, with 31.8 % maintaining this at 5-years. Depending on the definition used, between 11.2 % and 45.9 % of patients experienced recurrent weight gain. BMI had significant positive association with %TWL but negative with %EWL (p < 0.05). RYGB had significantly higher %TWL compared to SG (p < 0.05). CONCLUSION Most patients experienced weight loss which was maintained over time, however recurrent weight gain was noted. Pre-surgery BMI was significantly associated with weight changes.
Collapse
Affiliation(s)
- Beatrice Leyaro
- School of Computing, Engineering and Physical Sciences, University of West of Scotland, Scotland, UK; Institute of Public Health: Epidemiology and Biostatistics Department, Kilimanjaro Christian Medical University College, Moshi, Kilimanjaro, Tanzania.
| | - Lyz Howie
- School of Health and Life Sciences, University of the West of Scotland, Scotland, UK
| | - Kevin McMahon
- Department of General & Upper GI Surgery, University Hospital Ayr, Ayr, Scotland, UK
| | - Abdulmajid Ali
- Department of General & Upper GI Surgery, University Hospital Ayr, Ayr, Scotland, UK
| | - Raymond Carragher
- School of Computing, Engineering and Physical Sciences, University of West of Scotland, Scotland, UK
| |
Collapse
|
2
|
Çalık Başaran N, Dotan I, Dicker D. Post metabolic bariatric surgery weight regain: the importance of GLP-1 levels. Int J Obes (Lond) 2025; 49:412-417. [PMID: 38225284 PMCID: PMC11971041 DOI: 10.1038/s41366-024-01461-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/17/2024]
Abstract
Weight regain and insufficient weight loss are essential problems after metabolic bariatric surgery (MBS) in people living with obesity. Changes in the level of glucagon-like peptide-1 (GLP-1) secreted from the gut after bariatric surgery are one of the underlying mechanisms for successful initial weight loss. Studies and meta-analyses have revealed that postprandial GLP-1 levels increase after the Roux-en-Y gastric bypass and sleeve gastrectomy, but fasting GLP-1 levels do not increase significantly. Some observational studies have shown the relationship between higher postprandial GLP-1 levels and successful weight loss after bariatric surgery. There is growing evidence that GLP-1-receptor agonist (GLP-1-RA) use in patients who regained weight after bariatric surgery has resulted in significant weight loss. In this review, we aimed to summarize the changes in endogenous GLP-1 levels and their association with weight loss after MBS, describe the effects of GLP-1-RA use on weight loss after MBS, and emphasize metabolic adaptations in light of the recent literature. We hypothesized that maintaining higher basal-bolus GLP-1-RA levels may be a promising treatment choice in people with obesity who failed to lose weight after bariatric surgery.
Collapse
Affiliation(s)
- Nursel Çalık Başaran
- Hacettepe University, Faculty of Medicine, Department of Internal Medicine, General Internal Medicine, Ankara, Türkiye.
| | - Idit Dotan
- Rabin Medical Center, Beilinson Hospital, Department of Endocrinology and Obesity Clinic, Petah Tikva, Israel
- Tel Aviv University, Faculty of Medicine, Tel Aviv, Israel
| | - Dror Dicker
- Tel Aviv University, Faculty of Medicine, Tel Aviv, Israel
- Rabin Medical Center, Hasharon Hospital, Department of Internal Medicine and Obesity Clinic, Petah Tikva, Israel
| |
Collapse
|
3
|
Tiezzi M, Vieceli Dalla Sega F, Gentileschi P, Campanelli M, Benavoli D, Tremoli E. Effects of Weight Loss on Endothelium and Vascular Homeostasis: Impact on Cardiovascular Risk. Biomedicines 2025; 13:381. [PMID: 40002792 PMCID: PMC11853214 DOI: 10.3390/biomedicines13020381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Available knowledge shows that obesity is associated with an impaired endothelial function and an increase in cardiovascular risk, but the mechanisms of this association are not yet fully understood. Adipose tissue dysfunction, adipocytokines production, along with systemic inflammation and associated comorbidities (e.g., diabetes and hypertension), are regarded as the primary physiological and pathological factors. Various strategies are now available for the control of excess body weight. Dietary regimens alone, or in association with bariatric surgery when indicated, are now widely used. Of particular interest is the understanding of the effect of these interventions on endothelial homeostasis in relation to cardiovascular health. Substantial weight loss resulting from both diet and bariatric surgery decreases circulating biomarkers and improves endothelial function. Extensive clinical trials and meta-analyses show that bariatric surgery (particularly gastric bypass) has more substantial and long-lasting effect on weight loss and glucose regulation, as well as on distinct circulating biomarkers of cardiovascular risk. This review summarizes the current understanding of the distinct effects of diet-induced and surgery-induced weight loss on endothelial function, focusing on the key mechanisms involved in these effects.
Collapse
Affiliation(s)
- Margherita Tiezzi
- Dipartimento Cardiovascolare, Maria Cecilia Hospital GVM Care and Research, 48033 Cotignola, Italy;
| | | | - Paolo Gentileschi
- Dipartimento di Chirurgia Bariatrica e Metabolica, Maria Cecilia Hospital GVM Care and Research, 48033 Cotignola, Italy; (P.G.); (M.C.); (D.B.)
- Dipartimento di Scienze Chirurgiche, Università di Roma Tor Vergata, 00133 Roma, Italy
| | - Michela Campanelli
- Dipartimento di Chirurgia Bariatrica e Metabolica, Maria Cecilia Hospital GVM Care and Research, 48033 Cotignola, Italy; (P.G.); (M.C.); (D.B.)
| | - Domenico Benavoli
- Dipartimento di Chirurgia Bariatrica e Metabolica, Maria Cecilia Hospital GVM Care and Research, 48033 Cotignola, Italy; (P.G.); (M.C.); (D.B.)
| | - Elena Tremoli
- Dipartimento Cardiovascolare, Maria Cecilia Hospital GVM Care and Research, 48033 Cotignola, Italy;
| |
Collapse
|
4
|
Aukan MI, Rehfeld JF, Holst JJ, Martins C. Plasma concentration of gastrointestinal hormones and subjective appetite ratings after diet or bariatric surgery: 1-year results from the DISGAP study. Int J Obes (Lond) 2025; 49:306-314. [PMID: 39572763 DOI: 10.1038/s41366-024-01658-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 01/04/2025]
Abstract
OBJECTIVE Long-term weight loss outcomes are contrasting between bariatric surgery and dietary restriction alone. This is the first study to investigate changes in gastrointestinal (GI) hormones involved in appetite regulation, and subjective appetite feelings, at 1-year follow-up, after initial weight loss induced by a very-low energy (VLED) alone (controls), or with bariatric surgery. METHODS Patients scheduled for Sleeve Gastrectomy (SG) (n = 19) or Roux-en-Y gastric Bypass (RYGB) (n = 19), and controls (n = 16) were recruited. All groups underwent 10 weeks of a VLED (initial phase), followed by a 9-month maintenance phase. Body weight/composition, plasma concentrations of ghrelin, glucagon-like peptide 1 (GLP-1), peptide YY (PYY), cholecystokinin (CCK), and appetite ratings were measured before and after a meal, at baseline, week 11(W11), and 1Y follow-up. RESULTS Participants who completed all three follow ups were included in the analysis. Initial changes in body weight/composition were comparable across groups. SG (n = 11) and RYGB (n = 12) continued to lose weight from W11 to 1Y, whereas controls (n = 12) had regained weight. Postprandial GLP-1 increased over time post bariatric surgery and remained unchanged and lower in controls. Postprandial PYY increased in all groups, but greatest post-RYGB. Basal ghrelin decreased over time post-SG, while a small or marked increase was seen after RYGB and diet, respectively, with the control group exhibiting the greatest basal and postprandial concentrations at 1Y. A reduction in basal and postprandial CCK was seen in controls at 1Y, while no changes were observed post-bariatric surgery. Overall, small changes in subjective appetite ratings were seen over time. CONCLUSION Weight change at 1Y follow up after SG and RYGB is followed by a GI hormone profile favoring a lower drive to eat and increased satiety. The opposite is seen 1Y after WL induced by dietary restriction alone. CLINICAL TRIAL REGISTRATION clinicaltrials.gov NCT04051190.
Collapse
Affiliation(s)
- Marthe Isaksen Aukan
- Obesity Research Group, Department of Clinical and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- Centre of Obesity and Innovation (ObeCe), Clinic of Surgery, St. Olav University Hospital, Trondheim, Norway.
| | - Jens Frederik Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- The Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, University of Copenhagen, The Panum Institute, Copenhagen, Denmark
| | - Catia Martins
- Department of Nutrition Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| |
Collapse
|
5
|
Maxim M, Soroceanu RP, Vlăsceanu VI, Platon RL, Toader M, Miler AA, Onofriescu A, Abdulan IM, Ciuntu BM, Balan G, Trofin F, Timofte DV. Dietary Habits, Obesity, and Bariatric Surgery: A Review of Impact and Interventions. Nutrients 2025; 17:474. [PMID: 39940332 PMCID: PMC11820207 DOI: 10.3390/nu17030474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Eating behavior encompasses the psychological, physiological, and environmental factors influencing food intake. Dysregulation in eating behavior, such as emotional eating, binge eating, or loss of satiety signals, contributes to excessive caloric intake and weight gain. These behaviors are often linked to hormonal imbalances, stress, or genetic predisposition. Obesity is a chronic, multifactorial disease characterized by excessive body fat accumulation, with a body mass index (BMI) ≥ 30 kg/m2 often used for diagnosis. It is associated with significant morbidity, including type 2 diabetes, cardiovascular disease, and obstructive sleep apnea. Pathophysiological mechanisms underlying obesity include insulin resistance, leptin dysregulation, and altered gut microbiota, which perpetuate metabolic derangements. Lifestyle interventions remain first-line treatment, but sustained weight loss is challenging for many patients. Bariatric surgery is a therapeutic option for individuals with severe obesity (BMI ≥ 40 kg/m2 or ≥35 kg/m2 with comorbidities) who have failed conservative management. Procedures such as Roux-en-Y gastric bypass and sleeve gastrectomy alter gastrointestinal anatomy, promoting weight loss through restriction, malabsorption, and hormonal modulation (e.g., increased GLP-1 secretion). Bariatric surgery improves obesity-related comorbidities and enhances quality of life. However, it requires lifelong medical follow-up to address potential nutritional deficiencies and ensure sustainable outcomes.
Collapse
Affiliation(s)
- Mădălina Maxim
- “Grigore T. Popa” University of Medicine and Pharmacy, Faculty of Medicine, Str. Universitatii, No 16, 700115 Iasi, Romania; (M.M.); (R.P.S.); (V.I.V.); (R.L.P.); Romania; (M.T.); (A.A.M.); (A.O.); (B.-M.C.); (G.B.); (D.V.T.)
- Department of General Surgery, County Clinical Emergency Hospital St. Spiridon, 700111 Iasi, Romania
| | - Radu Petru Soroceanu
- “Grigore T. Popa” University of Medicine and Pharmacy, Faculty of Medicine, Str. Universitatii, No 16, 700115 Iasi, Romania; (M.M.); (R.P.S.); (V.I.V.); (R.L.P.); Romania; (M.T.); (A.A.M.); (A.O.); (B.-M.C.); (G.B.); (D.V.T.)
- Department of General Surgery, County Clinical Emergency Hospital St. Spiridon, 700111 Iasi, Romania
| | - Vlad Ionuț Vlăsceanu
- “Grigore T. Popa” University of Medicine and Pharmacy, Faculty of Medicine, Str. Universitatii, No 16, 700115 Iasi, Romania; (M.M.); (R.P.S.); (V.I.V.); (R.L.P.); Romania; (M.T.); (A.A.M.); (A.O.); (B.-M.C.); (G.B.); (D.V.T.)
- Department of General Surgery, County Clinical Emergency Hospital St. Spiridon, 700111 Iasi, Romania
| | - Răzvan Liviu Platon
- “Grigore T. Popa” University of Medicine and Pharmacy, Faculty of Medicine, Str. Universitatii, No 16, 700115 Iasi, Romania; (M.M.); (R.P.S.); (V.I.V.); (R.L.P.); Romania; (M.T.); (A.A.M.); (A.O.); (B.-M.C.); (G.B.); (D.V.T.)
- Department of General Surgery, County Clinical Emergency Hospital St. Spiridon, 700111 Iasi, Romania
| | - Mihaela Toader
- “Grigore T. Popa” University of Medicine and Pharmacy, Faculty of Medicine, Str. Universitatii, No 16, 700115 Iasi, Romania; (M.M.); (R.P.S.); (V.I.V.); (R.L.P.); Romania; (M.T.); (A.A.M.); (A.O.); (B.-M.C.); (G.B.); (D.V.T.)
| | - Ancuța Andreea Miler
- “Grigore T. Popa” University of Medicine and Pharmacy, Faculty of Medicine, Str. Universitatii, No 16, 700115 Iasi, Romania; (M.M.); (R.P.S.); (V.I.V.); (R.L.P.); Romania; (M.T.); (A.A.M.); (A.O.); (B.-M.C.); (G.B.); (D.V.T.)
| | - Alina Onofriescu
- “Grigore T. Popa” University of Medicine and Pharmacy, Faculty of Medicine, Str. Universitatii, No 16, 700115 Iasi, Romania; (M.M.); (R.P.S.); (V.I.V.); (R.L.P.); Romania; (M.T.); (A.A.M.); (A.O.); (B.-M.C.); (G.B.); (D.V.T.)
- Department of Diabetes and Metabolic Diseases, Clinical Emergency Hospital St. Spiridon, 700111 Iasi, Romania
| | - Irina Mihaela Abdulan
- Department of Medical Specialties I, “Grigore, T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Bogdan-Mihnea Ciuntu
- “Grigore T. Popa” University of Medicine and Pharmacy, Faculty of Medicine, Str. Universitatii, No 16, 700115 Iasi, Romania; (M.M.); (R.P.S.); (V.I.V.); (R.L.P.); Romania; (M.T.); (A.A.M.); (A.O.); (B.-M.C.); (G.B.); (D.V.T.)
- Department of General Surgery, County Clinical Emergency Hospital St. Spiridon, 700111 Iasi, Romania
| | - Gheorghe Balan
- “Grigore T. Popa” University of Medicine and Pharmacy, Faculty of Medicine, Str. Universitatii, No 16, 700115 Iasi, Romania; (M.M.); (R.P.S.); (V.I.V.); (R.L.P.); Romania; (M.T.); (A.A.M.); (A.O.); (B.-M.C.); (G.B.); (D.V.T.)
- Department of Gastroenterology, Clinical Emergency Hospital St. Spiridon, 700111 Iasi, Romania
| | - Felicia Trofin
- Department of Preventive Medicine and Interdisciplinarity—Microboology, “Grigore T. Popa” University of Medicine and Pharmacy, Str. Universitatii no 16, 700115 Iasi, Romania
| | - Daniel Vasile Timofte
- “Grigore T. Popa” University of Medicine and Pharmacy, Faculty of Medicine, Str. Universitatii, No 16, 700115 Iasi, Romania; (M.M.); (R.P.S.); (V.I.V.); (R.L.P.); Romania; (M.T.); (A.A.M.); (A.O.); (B.-M.C.); (G.B.); (D.V.T.)
- Department of General Surgery, County Clinical Emergency Hospital St. Spiridon, 700111 Iasi, Romania
| |
Collapse
|
6
|
Vidmar AP, Batt CE, Moore JM. Assessment and medical management of weight regain after adolescent metabolic and bariatric surgery: a narrative review. Surg Obes Relat Dis 2025; 21:24-32. [PMID: 39609232 PMCID: PMC11729465 DOI: 10.1016/j.soard.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/02/2024] [Accepted: 10/05/2024] [Indexed: 11/30/2024]
Abstract
Metabolic and bariatric surgery (MBS) in adolescents results in durable treatment of severe obesity and related complications for most. However, substantial weight regain can undermine long-term health benefits. There is no evidence-based standard of care for the medical management of weight regain after MBS in pediatrics or adults. This narrative review summarizes current pediatric evidence pertaining to the assessment and medical management of post-MBS weight regain, identifies gaps, and offers recommendations. A PubMed search was conducted through March 2024 and focused on adolescents after sleeve gastrectomy or Roux-en-Y gastric bypass. Domains included nutrition, activity, mental health, antiobesity medications, type 2 diabetes, hypothalamic obesity, and transition of care. In total, 600 articles were screened and 61 were included in this review. Recent consensus definitions for post-MBS weight regain have been established for adults but have not been validated in pediatrics. Limited, high-quality evidence was identified in the nutrition domain, where targets that may mitigate weight regain include adequate protein intake (≥60 g/d), absence of loss-of-control eating, and micronutrient sufficiency. Emerging data for post-MBS antiobesity medications in adults with/without diabetes and in adolescents with persistent obesity are promising. Large gaps include post-MBS interventions focused on physical activity and mental health. The overall quality of pediatric-specific evidence for the assessment and medical management of post-MBS weight regain is low. A standard definition of weight regain associated with health outcomes in pediatrics would be valuable. Clarifying risk and protective factors for weight regain can guide more precise risk stratification and treatment.
Collapse
Affiliation(s)
- Alaina P Vidmar
- Division of Pediatric Endocrinology, Diabetes, and Metabolism, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California
| | - Courtney E Batt
- Division of Academic Pediatrics and Adolescent Medicine, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, Ohio
| | - Jaime M Moore
- Department of Pediatrics Section of Nutrition, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
7
|
Dagbasi A, Fuller A, Hanyaloglu AC, Carroll B, McLaughlin J, Frost G, Holliday A. The role of nutrient sensing dysregulation in anorexia of ageing: The little we know and the much we don't. Appetite 2024; 203:107718. [PMID: 39423861 DOI: 10.1016/j.appet.2024.107718] [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: 04/08/2024] [Revised: 08/01/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
The age-related decline in appetite and food intake - termed "anorexia of ageing" - is implicated in undernutrition in later life and hence provides a public health challenge for our ageing population. Eating behaviour is controlled, in part, by homeostatic mechanisms which sense nutrient status and provide feedback to appetite control regions of the brain. Such feedback signals, propagated by episodic gut hormones, are dysregulated in some older adults. The secretory responses of appetite-related gut hormones to feeding are amplified, inducing a more anorexigenic signal which is associated with reduced appetite and food intake. Such an augmented response would indicate an increase in gut sensitivity to nutrients. Consequently, this review explores the role of gastrointestinal tract nutrient sensing in age-related appetite dysregulation. We review and synthesise evidence for age-related alterations in nutrient sensing which may explain the observed hormonal dysregulation. Drawing on what is known regarding elements of nutrient sensing pathways in animal models, in other tissues of the body, and in certain models of disease, we identify potential causal mechanisms including alterations in enteroendocrine cell number and distribution, dysregulation of cell signalling pathways, and changes in the gut milieu. From identified gaps in evidence, we highlight interesting and important avenues for future research.
Collapse
Affiliation(s)
- Aygul Dagbasi
- Section of Nutrition, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London, W12 0NN, UK
| | - Amy Fuller
- Research Centre for Health and Life Sciences, Institute of Health and Wellbeing, Faculty of Health and Life Science, Coventry University, Coventry, CV1 5FB, UK
| | - Aylin C Hanyaloglu
- Institute of Reproductive and Developmental Biology (IRDB), Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Bernadette Carroll
- School of Biochemistry, University of Bristol, University Walk, Bristol, BS1 8TD, UK
| | - John McLaughlin
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester and Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK
| | - Gary Frost
- Section of Nutrition, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London, W12 0NN, UK
| | - Adrian Holliday
- School of Biomedical, Nutritional, and Sport Science, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK; Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK.
| |
Collapse
|
8
|
Björkman S, Höskuldsdóttir G, Mossberg K, Laurenius A, Engström M, Fändriks L, Eliasson B, Wallengren O, Larsson I. Impact of eating behavior on 24-month weight change after treatment of severe obesity-A clinical prospective cohort study. Obesity (Silver Spring) 2024; 32:2100-2110. [PMID: 39370375 DOI: 10.1002/oby.24131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/18/2024] [Accepted: 07/18/2024] [Indexed: 10/08/2024]
Abstract
OBJECTIVE This study aimed to evaluate the effects of self-reported baseline eating behaviors on 24-month weight change in adults with severe obesity. METHODS A prospective, nonrandomized clinical cohort study on surgical and medical obesity treatment included 971 adults (75% women) with a mean BMI of 42.0 (SD 4.9) kg/m2. To assess baseline eating behaviors and binge eating disorder, the Questionnaire on Eating and Weight Patterns-Revised and the Three-Factor Eating Questionnaire were used. RESULTS In analyses adjusted for treatment, age, sex, baseline weight, and BMI, those with nocturnal eating lost less weight (3.5 kg [95% CI: 0.02-6.9]; p < 0.05) at 24 months compared to those without nocturnal eating. Binge eating disorder was not significantly associated with weight loss over 24 months. Emotional eating was associated with less weight loss at 12 months: 1.16 kg per z score (95% CI: 0.37-1.95; p < 0.05). Compared with completers, dropout from medical obesity treatment was associated with emotional and uncontrolled eating at baseline (both p < 0.001). CONCLUSIONS The association between pretreatment eating behaviors and weight change was found to be generalizable and not restricted to any specific treatment. Certain eating behaviors may affect weight loss as well as attrition. Identifying eating behaviors that may impair treatment efficacy are suggested in the treatment of severe obesity.
Collapse
Affiliation(s)
- Sofia Björkman
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Gudrún Höskuldsdóttir
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Karin Mossberg
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Public Health and Community Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anna Laurenius
- Department of Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - My Engström
- Department of Surgery, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
- Institute of Health and Care Sciences, Sahlgrenska Academy, Gothenburg, Sweden
| | - Lars Fändriks
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Björn Eliasson
- Department of Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ola Wallengren
- Department of Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ingrid Larsson
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
9
|
Holst JJ. GLP-1 physiology in obesity and development of incretin-based drugs for chronic weight management. Nat Metab 2024; 6:1866-1885. [PMID: 39160334 DOI: 10.1038/s42255-024-01113-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024]
Abstract
The introduction of the highly potent incretin receptor agonists semaglutide and tirzepatide has marked a new era in the treatment of type 2 diabetes and obesity. With normalisation of glycated haemoglobin levels and weight losses around 15-25%, therapeutic goals that were previously unrealistic are now within reach, and clinical trials have documented that these effects are associated with reduced risk of cardiovascular events and premature mortality. Here, I review this remarkable development from the earliest observations of glucose lowering and modest weight losses with native glucagon-like peptide (GLP)-1 and short acting compounds, to the recent development of highly active formulations and new molecules. I will classify these agents as GLP-1-based therapies in the understanding that these compounds or combinations may have actions on other receptors as well. The physiology of GLP-1 is discussed as well as its mechanisms of actions in obesity, in particular, the role of sensory afferents and GLP-1 receptors in the brain. I provide details regarding the development of GLP-1 receptor agonists for anti-obesity therapy and discuss the possible mechanism behind their beneficial effects on adverse cardiovascular events. Finally, I highlight new pharmacological developments, including oral agents, and discuss important questions regarding maintenance therapy.
Collapse
Affiliation(s)
- Jens Juul Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences. Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
10
|
Phuong-Nguyen K, McGee SL, Aston-Mourney K, Mcneill BA, Mahmood MQ, Rivera LR. Yoyo Dieting, Post-Obesity Weight Loss, and Their Relationship with Gut Health. Nutrients 2024; 16:3170. [PMID: 39339770 PMCID: PMC11435324 DOI: 10.3390/nu16183170] [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: 08/21/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Excessive body weight is associated with many chronic metabolic diseases and weight loss, so far, remains the gold standard treatment. However, despite tremendous efforts exploring optimal treatments for obesity, many individuals find losing weight and maintaining a healthy body weight difficult. Weight loss is often not sustainable resulting in weight regain and subsequent efforts to lose weight. This cyclic pattern of weight loss and regain is termed "yoyo dieting" and predisposes individuals to obesity and metabolic comorbidities. How yoyo dieting might worsen obesity complications during the weight recurrence phase remains unclear. In particular, there is limited data on the role of the gut microbiome in yoyo dieting. Gut health distress, especially gut inflammation and microbiome perturbation, is strongly associated with metabolic dysfunction and disturbance of energy homeostasis in obesity. In this review, we summarise current evidence of the crosstalk between the gastrointestinal system and energy balance, and the effects of yoyo dieting on gut inflammation and gut microbiota reshaping. Finally, we focus on the potential effects of post-dieting weight loss in improving gut health and identify current knowledge gaps within the field, including gut-derived peptide hormones and their potential suitability as targets to combat weight regain, and how yoyo dieting and associated changes in the microbiome affect the gut barrier and the enteric nervous system, which largely remain to be determined.
Collapse
Affiliation(s)
- Kate Phuong-Nguyen
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Sean L McGee
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Kathryn Aston-Mourney
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Bryony A Mcneill
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Malik Q Mahmood
- School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Leni R Rivera
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| |
Collapse
|
11
|
Fredrickson G, Florczak K, Barrow F, Mahmud S, Dietsche K, Wang H, Parthiban P, Hakeem A, Almutlaq R, Adeyi O, Herman A, Bartolomucci A, Staley C, Dong X, Jahansouz C, Williams JW, Mashek DG, Ikramuddin S, Revelo XS. TREM2 macrophages mediate the beneficial effects of bariatric surgery against MASH. Hepatology 2024:01515467-990000000-01031. [PMID: 39292863 PMCID: PMC11913751 DOI: 10.1097/hep.0000000000001098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND AND AIMS For patients with obesity and metabolic syndrome, bariatric procedures such as vertical sleeve gastrectomy (VSG) have a clear benefit in ameliorating metabolic dysfunction-associated steatohepatitis (MASH). While the effects of bariatric surgeries have been mainly attributed to nutrient restriction and malabsorption, whether immuno-modulatory mechanisms are involved remains unclear. APPROACH AND RESULT Using murine models, we report that VSG ameliorates MASH progression in a weight loss-independent manner. Single-cell RNA sequencing revealed that hepatic lipid-associated macrophages (LAMs) expressing the triggering receptor expressed on myeloid cells 2 (TREM2) repress inflammation and increase their lysosomal activity in response to VSG. Remarkably, TREM2 deficiency in mice ablates the reparative effects of VSG, suggesting that TREM2 is required for MASH resolution. Mechanistically, TREM2 prevents the inflammatory activation of macrophages and is required for their efferocytic function. CONCLUSIONS Overall, our findings indicate that bariatric surgery improves MASH through a reparative process driven by TREM2+ macrophages, providing insights into the mechanisms of disease reversal that may result in new therapies and improved surgical interventions.
Collapse
Affiliation(s)
- Gavin Fredrickson
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kira Florczak
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Fanta Barrow
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Shamsed Mahmud
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
| | - Katrina Dietsche
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Haiguang Wang
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Preethy Parthiban
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Andrew Hakeem
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rawan Almutlaq
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Oyedele Adeyi
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Adam Herman
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alessandro Bartolomucci
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Christopher Staley
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Xiao Dong
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
| | - Cyrus Jahansouz
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jesse W Williams
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Douglas G Mashek
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sayeed Ikramuddin
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Xavier S Revelo
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
12
|
Lytvyak E, Zarrinpar A, Ore CD, Lee E, Yazdani-Boset K, Horgan S, Grunvald E. Stronger control of eating 3 months after sleeve gastrectomy predicts successful weight loss outcomes at one year. OBESITY PILLARS 2024; 11:100111. [PMID: 38770521 PMCID: PMC11103426 DOI: 10.1016/j.obpill.2024.100111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024]
Abstract
Background Weight loss response to sleeve gastrectomy (SG) is variable and predicting the effectiveness of surgery is challenging and elusive. The aim of our study was to assess and quantify the association between eating control and weight loss outcomes and identify the control of eating (CoE) attributes during the early postoperative period that might predict good vs. poor response to SG at one year. Methods A prospective longitudinal cohort study using the Control of Eating Questionnaire (CoEQ) was designed as a series before and at 3-, 6-, and 12-months post-SG. Primary outcomes were changes in CoE attributes and percent of total weight loss (%TWL) 12-months post-surgery. Subjects were categorized based on %TWL as good (GR, ≥25 %) or poor responders (PR, <25 %). A receiver operating characteristic and logistic regression analyses were performed. Results We included 41 participants (80.5% females, 51.2% Hispanic, mean age 41.7±10.6, median baseline body mass index (BMI) 43.6 kg/m2 [range 35.2-66.3]) who completed the CoEQ at all four timepoints. The "Difficulty to control eating" score at 3 months revealed the highest area under the curve (AUC) (AUC 0.711; 95%CI 0.524-0.898; p=0.032). In a trade-off between a high Youden index and high sensitivity, the "Difficulty to control eating" score of 7 at 3 months was identified as the optimal cut-off for distinguishing between GRs and PRs. Score ≤7 at 3 months was strongly independently associated with a successful weight loss target of 25%TWL at one-year post-SG (Relative Risk 4.43; 95%CI 1.06-18.54; p=0.042). Conclusion "Difficulty to control eating" score at 3 months post-SG is an independent early predictor of optimal response (achieving a successful TWL target of ≥25 % at one-year post-SG). Our results support the utility of this easy-to-administer validated tool for predicting the effectiveness of SG and may assist in identifying individuals with suboptimal response early and helping them with interventions to attain optimal weight loss targets.
Collapse
Affiliation(s)
- Ellina Lytvyak
- Division of Preventive Medicine, Department of Medicine, University of Alberta, 5-30 University Terrace, 8303 112 Street, Edmonton, Alberta, T6G 2T4, Canada
| | - Amir Zarrinpar
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Cecilia Dalle Ore
- School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Euyhyun Lee
- Altman Clinical and Translational Research Institute, University of California San Diego, La Jolla, CA, USA
| | | | - Santiago Horgan
- Bariatric and Metabolic Institute, Division of Minimally Invasive Surgery, University of California San Diego, 4303 La Jolla Village Drive, Suite 2110, San Diego, CA, 92122, USA
| | - Eduardo Grunvald
- School of Medicine, University of California San Diego, La Jolla, CA, USA
- Division of General Internal Medicine, University of California San Diego, La Jolla, CA, USA
- Bariatric and Metabolic Institute, Division of Minimally Invasive Surgery, University of California San Diego, 4303 La Jolla Village Drive, Suite 2110, San Diego, CA, 92122, USA
| |
Collapse
|
13
|
Saiyalam C, Shantavasinkul PC, Chirnaksorn S, Rattanakaemakorn P, Taonam N, Rodphech V, Putadechakum S, Rattanasiri S, Sirivarasai J, Ongphiphadhanakul B, Sumritpradit P. Effects of Very Low-Calorie Diet versus Roux-en-Y Gastric Bypass Surgery on Body Composition in Patients with Obesity. Nutrients 2024; 16:2407. [PMID: 39125287 PMCID: PMC11313779 DOI: 10.3390/nu16152407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
Roux-en-Y gastric bypass (RYGB) is the most effective treatment for severe obesity. A very low-calorie diet (VLCD) is another effective dietary intervention to treat obesity. This study evaluated the effect of a VLCD versus RYGB on weight reduction, changes in body composition and the resolution of comorbidities during a 12-week period. Individuals with obesity at the obesity clinic, Ramathibodi Hospital, Mahidol University, Thailand with a body mass index (BMI) ≥ 37.5 kg/m2 or ≥32.5 kg/m2 with obesity-related complications were recruited. Treatment options, either RYGB or VLCD, were assigned depending on patients' preferences and physicians' judgment. The analysis included 16 participants in the RYGB group and 15 participants in the VLCD group. Baseline characteristics were similar between groups; nevertheless, the participants in the VLCD group were significantly younger than those in the RYGB group. The number of patients with type 2 diabetes (T2D) was slightly higher in the RYGB group (43.8% vs. 33.3%, p = 0.552). Additionally, patients in the RYGB group had a longer duration of T2D and were treated with anti-diabetic agents, while VLCD patients received only lifestyle modifications. At 12 weeks, total and percentage weight loss in the RYGB and VLCD groups, respectively, were as follows: -17.6 ± 6.0 kg vs. -15.6 ± 5.1 kg (p = 0.335) and -16.2% ± 4.3% vs. -14.1% ± 3.6% (p = 0.147). Changes in biochemical data and the resolution of comorbidities were similar between the groups at 12 weeks. A 12-week VLCD resulted in similar weight loss and metabolic improvement compared with RYGB. Large-scale studies with long follow-up periods are needed to elucidate whether VLCD is a viable alternative treatment to bariatric surgery.
Collapse
Affiliation(s)
- Chanawit Saiyalam
- Doctor of Philosophy Program in Nutrition, Faculty of Medicine Ramathibodi Hospital and Institute of Nutrition, Mahidol University, Bangkok 10400, Thailand;
| | - Prapimporn Chattranukulchai Shantavasinkul
- Division of Nutrition and Biochemical Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (N.T.); (V.R.); (S.P.)
- Graduate Program in Nutrition, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Supphamat Chirnaksorn
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Ploysyne Rattanakaemakorn
- Division of Dermatology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Naphat Taonam
- Division of Nutrition and Biochemical Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (N.T.); (V.R.); (S.P.)
| | - Vorachat Rodphech
- Division of Nutrition and Biochemical Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (N.T.); (V.R.); (S.P.)
| | - Supanee Putadechakum
- Division of Nutrition and Biochemical Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (N.T.); (V.R.); (S.P.)
| | - Sasivimol Rattanasiri
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Jintana Sirivarasai
- Graduate Program in Nutrition, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Boonsong Ongphiphadhanakul
- Division of Endocrine and Metabolism, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Preeda Sumritpradit
- Department of Surgery, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| |
Collapse
|
14
|
Dagbasi A, Byrne C, Blunt D, Serrano-Contreras JI, Becker GF, Blanco JM, Camuzeaux S, Chambers E, Danckert N, Edwards C, Bernal A, Garcia MV, Hanyaloglu A, Holmes E, Ma Y, Marchesi J, Martinez-Gili L, Mendoza L, Tashkova M, Perez-Moral N, Garcia-Perez I, Robles AC, Sands C, Wist J, Murphy KG, Frost G. Diet shapes the metabolite profile in the intact human ileum, which affects PYY release. Sci Transl Med 2024; 16:eadm8132. [PMID: 38896603 DOI: 10.1126/scitranslmed.adm8132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 05/24/2024] [Indexed: 06/21/2024]
Abstract
The human ileum contains a high density of enteroendocrine L-cells, which release the appetite-suppressing hormones glucagon-like peptide-1 (GLP-1) and peptide tyrosine tyrosine (PYY) in response to food intake. Recent evidence highlighted the potential role of food structures in PYY release, but the link between food structures, ileal metabolites, and appetite hormone release remains unclear owing to limited access to intact human ileum. In a randomized crossover trial (ISRCTN11327221; isrctn.com), we investigated the role of human ileum in GLP-1 and PYY release by giving healthy volunteers diets differing in fiber and food structure: high-fiber (intact or disrupted food structures) or low-fiber disrupted food structures. We used nasoenteric tubes to sample chyme from the intact distal ileum lumina of humans in the fasted state and every 60 min for 480 min postprandially. We demonstrate the highly dynamic, wide-ranging molecular environment of the ileum over time, with a substantial decrease in ileum bacterial numbers and bacterial metabolites after food intake. We also show that high-fiber diets, independent of food structure, increased PYY release compared with a low-fiber diet during 0 to 240 min postprandially. High-fiber diets also increased ileal stachyose, and a disrupted high-fiber diet increased certain ileal amino acids. Treatment of human ileal organoids with ileal fluids or an amino acid and stachyose mixture stimulated PYY expression in a similar profile to blood PYY concentrations, confirming the role of ileal metabolites in PYY release. Our study demonstrates the diet-induced changes over time in the metabolite environment of intact human ileum, which play a role in PYY release.
Collapse
Affiliation(s)
- Aygul Dagbasi
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Claire Byrne
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Dominic Blunt
- Department of Imaging, Charing Cross Hospital, Imperial NHS Trust, London W6 8RF, UK
| | - Jose Ivan Serrano-Contreras
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Georgia Franco Becker
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Jesus Miguens Blanco
- Division of Digestive Diseases, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Stephane Camuzeaux
- National Phenome Centre, Imperial College London, Hammersmith Hospital Campus, London W12 0HS, UK
| | - Edward Chambers
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Nathan Danckert
- Division of Digestive Diseases, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | | | - Andres Bernal
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Maria Valdivia Garcia
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Aylin Hanyaloglu
- Institute of Reproductive and Development Biology (IRDB), Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Hammersmith Hospital, London W12 0NN, UK
| | - Elaine Holmes
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Yue Ma
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Julian Marchesi
- Division of Digestive Diseases, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Laura Martinez-Gili
- Division of Digestive Diseases, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
- Section of Bioinformatics, Division of Systems Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London W12 0NN, UK
| | - Lilian Mendoza
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Martina Tashkova
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | | | - Isabel Garcia-Perez
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Andres Castillo Robles
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Caroline Sands
- National Phenome Centre, Imperial College London, Hammersmith Hospital Campus, London W12 0HS, UK
| | - Julien Wist
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
- Chemistry Department, Universidad del Valle, Cali 76001, Colombia
| | - Kevin G Murphy
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Gary Frost
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| |
Collapse
|
15
|
Holst JJ, Madsbad S, Bojsen-Møller KN, Dirksen C, Svane M. New Lessons from the gut: Studies of the role of gut peptides in weight loss and diabetes resolution after gastric bypass and sleeve gastrectomy. Peptides 2024; 176:171199. [PMID: 38552903 DOI: 10.1016/j.peptides.2024.171199] [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: 01/29/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
It has been known since 2005 that the secretion of several gut hormones changes radically after gastric bypass operations and, although more moderately, after sleeve gastrectomy but not after gastric banding. It has therefore been speculated that increased secretion of particularly GLP-1 and Peptide YY (PYY), which both inhibit appetite and food intake, may be involved in the weight loss effects of surgery and for improvements in glucose tolerance. Experiments involving inhibition of hormone secretion with somatostatin, blockade of their actions with antagonists, or blockade of hormone formation/activation support this notion. However, differences between results of bypass and sleeve operations indicate that distinct mechanisms may also be involved. Although the reductions in ghrelin secretion after sleeve gastrectomy would seem to provide an obvious explanation, experiments with restoration of ghrelin levels pointed towards effects on insulin secretion and glucose tolerance rather than on food intake. It seems clear that changes in GLP-1 secretion are important for insulin secretion after bypass and appear to be responsible for postbariatric hypoglycemia in glucose-tolerant individuals; however, with time the improvements in insulin sensitivity, which in turn are secondary to the weight loss, may be more important. Changes in bile acid metabolism do not seem to be of particular importance in humans.
Collapse
Affiliation(s)
- Jens Juul Holst
- The NovoNordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Denmark.
| | - Sten Madsbad
- Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Denmark
| | | | - Carsten Dirksen
- Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Denmark
| | - Maria Svane
- Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Denmark
| |
Collapse
|
16
|
Gan HW, Cerbone M, Dattani MT. Appetite- and Weight-Regulating Neuroendocrine Circuitry in Hypothalamic Obesity. Endocr Rev 2024; 45:309-342. [PMID: 38019584 PMCID: PMC11074800 DOI: 10.1210/endrev/bnad033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Since hypothalamic obesity (HyOb) was first described over 120 years ago by Joseph Babinski and Alfred Fröhlich, advances in molecular genetic laboratory techniques have allowed us to elucidate various components of the intricate neurocircuitry governing appetite and weight regulation connecting the hypothalamus, pituitary gland, brainstem, adipose tissue, pancreas, and gastrointestinal tract. On a background of an increasing prevalence of population-level common obesity, the number of survivors of congenital (eg, septo-optic dysplasia, Prader-Willi syndrome) and acquired (eg, central nervous system tumors) hypothalamic disorders is increasing, thanks to earlier diagnosis and management as well as better oncological therapies. Although to date the discovery of several appetite-regulating peptides has led to the development of a range of targeted molecular therapies for monogenic obesity syndromes, outside of these disorders these discoveries have not translated into the development of efficacious treatments for other forms of HyOb. This review aims to summarize our current understanding of the neuroendocrine physiology of appetite and weight regulation, and explore our current understanding of the pathophysiology of HyOb.
Collapse
Affiliation(s)
- Hoong-Wei Gan
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Manuela Cerbone
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Mehul Tulsidas Dattani
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
17
|
Lytvyak E, Zarrinpar A, Dalle Ore C, Lee E, Yazdani-Boset K, Horgan S, Grunvald E. Control of Eating Attributes and Weight Loss Outcomes over One Year After Sleeve Gastrectomy. Obes Surg 2024; 34:1618-1629. [PMID: 38502520 DOI: 10.1007/s11695-024-07158-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/21/2024]
Abstract
PURPOSE Sleeve gastrectomy (SG) is a commonly performed metabolic-bariatric surgery, but its effectiveness is variable and difficult to predict. Our study aimed to compare control of eating (CoE) attributes pre- and post-SG depending on the achievement of optimal weight loss target at 1 year post-SG. MATERIALS AND METHODS A prospective longitudinal cohort study using the CoE Questionnaire, pre-SG, and postoperatively at 3, 6, and 12 months was conducted. Total weight loss (TWL) ≥ 25% at 12 months post-SG was set as an optimal weight loss target. RESULTS Forty-one patients (80.5% females, mean age 41.7 ± 10.6) were included. Sex, age, baseline weight, BMI, and smoking status were all comparable. At 3 months post-SG, "difficulty to control eating" score became significantly different between ≥ 25%TWL and < 25%TWL groups (7 [0-50] vs. 17 [5-63], p = 0.042). At 6 months, it was followed by "feeling of fullness," "frequency of food cravings," and "difficulty to resist cravings" demonstrating significant differences between ≥ 25%TWL and < 25%TWL groups. At 12 months, significant differences between groups were observed in "feeling hungry," "difficulty to resist cravings," "eating in response to cravings," and difficulty to control eating scores. CONCLUSION Individuals with obesity who achieved a target of ≥ 25%TWL at 1 year post-SG have an early improvement in overall eating control at 3 months that steadily persists at 6 and 12 months. Improvements in other aspects tend to follow later, at 6 and 12 months. These findings may assist in identifying individuals with inadequate response to help attain optimal weight loss targets and developing a prognostic model to identify predictors of successful weight loss.
Collapse
Affiliation(s)
- Ellina Lytvyak
- Division of Preventive Medicine, Department of Medicine, University of Alberta, 5-30 University Terrace, 8303 112 Street, Edmonton, AB, T6G 2T4, Canada.
| | - Amir Zarrinpar
- Division of Gastroenterology, University of California San Diego, 9500 Gilman Dr, La Jolla, San Diego, CA, 92093, USA
- School of Medicine, University of California San Diego, 9500 Gilman Dr, La Jolla, San Diego, CA, 92093, USA
| | - Cecilia Dalle Ore
- School of Medicine, University of California San Diego, 9500 Gilman Dr, La Jolla, San Diego, CA, 92093, USA
| | - Euyhyun Lee
- Altman Clinical and Translational Research Institute, University of California San Diego, 9452 Medical Ctr Dr, La Jolla, San Diego, CA, 92037, USA
| | - Keila Yazdani-Boset
- University of California San Diego, 9500 Gilman Dr, La Jolla, San Diego, CA, 92093, USA
| | - Santiago Horgan
- Division of Minimally Invasive Surgery, Bariatric and Metabolic Institute, University of California San Diego, 4303 La Jolla Village Drive, Suite 2110, San Diego, CA, 92122, USA
| | - Eduardo Grunvald
- School of Medicine, University of California San Diego, 9500 Gilman Dr, La Jolla, San Diego, CA, 92093, USA
- Division of General Internal Medicine, University of California San Diego, 9500 Gilman Dr, La Jolla, San Diego, CA, 92093, USA
- Division of Minimally Invasive Surgery, Bariatric and Metabolic Institute, University of California San Diego, 4303 La Jolla Village Drive, Suite 2110, San Diego, CA, 92122, USA
| |
Collapse
|
18
|
Simoneau M, McKay B, Brooks E, Doucet É, Baillot A. Gut peptides before and following Roux-En-Y gastric bypass: A systematic review and meta-analysis. Obes Rev 2024; 25:e13702. [PMID: 38327045 DOI: 10.1111/obr.13702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 02/09/2024]
Abstract
A systematic search was conducted in Medline Ovid, Embase, Scopus, and Cochrane Central Register of Controlled Trials up until March 2021 following PRISMA guidelines. Studies included evaluated ghrelin, GLP-1, PYY or appetite sensation via visual analogue scales (VASs) before and after Roux-en-Y gastric bypass (RYGB) in adults. A multilevel model with random effects for study and follow-up time points nested in study was fit to the data. The model included kcal consumption as a covariate and time points as moderators. Among the 2559 articles identified, k = 47 were included, among which k = 19 evaluated ghrelin, k = 40 GLP-1, k = 22 PYY, and k = 8 appetite sensation. Our results indicate that fasting ghrelin levels are decreased 2 weeks post-RYGB (p = 0.005) but do not differ from baseline from 6 weeks to 1-year post-RYGB. Postprandial ghrelin and fasting GLP-1 levels were not different from pre-surgical values. Postprandial levels of GLP-1 increased significantly from 1 week (p < 0.001) to 2 years post-RYGB (p < 0.01) compared with pre-RYGB. Fasting PYY increased at 6 months (p = 0.034) and 1 year (p = 0.029) post-surgery; also, postprandial levels increased up to 1 year (p < 0.01). Insufficient data on appetite sensation were available to be meta-analyzed.
Collapse
Affiliation(s)
- Mylène Simoneau
- School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Brad McKay
- Department of kinesiology, University of McMaster, Hamilton, Ontario, Canada
| | - Emma Brooks
- School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Éric Doucet
- School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Aurélie Baillot
- Department of nursing, University of Québec en Outaouais, Gatineau, Quebec, Canada
| |
Collapse
|
19
|
Nedelcut S, Axer S, Olbers T. The risk and benefit of revisional vs. primary metabolic- bariatric surgery and drug therapy - A narrative review. Metabolism 2024; 154:155799. [PMID: 38281557 DOI: 10.1016/j.metabol.2024.155799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 01/30/2024]
Abstract
Metabolic and bariatric surgery (MBS) leads to long-term weight loss, reduced risk of cardiovascular events and cancer, and reduced mortality. Sleeve gastrectomy and Roux-en-Y gastric bypass are currently the most common surgical techniques. Weight loss after MBS was previously believed to work through restriction and malabsorption, however, mechanistic studies show that MBS techniques with long term efficacy instead alter physiological signaling between the gut and the brain. In revisional MBS, the initial surgical technique is corrected, modified, or converted to a new one. The indication for revisional MBS can be to achieve further weight loss or improvement in obesity comorbidity, but it may be necessary due to complications (e.g., gastroesophageal reflux or obstruction). Revisional MBS is associated with an increased risk of surgical complications and often less weight loss compared to the results following primary surgery. This narrative review summarizes data from revisional MBS where information is often presented with inconsistent definitions for indications and outcomes, making comparison between strategies difficult. In summary, we suggest careful weighing of potential benefits and risks with revisional MBS, bearing in mind the option of add-on therapy with new anti-obesity drugs.
Collapse
Affiliation(s)
| | - Stephan Axer
- Faculty of Health and Medicine, Örebro University, Campus USÖ, 701 82 Örebro, Sweden; Department of General Surgery, Torsby Hospital, Box 502, 685 29 Torsby, Sweden
| | - Torsten Olbers
- Department of Biomedical and Clinical Sciences, Department of Surgery, Linköping University, Norrköping, Sweden
| |
Collapse
|
20
|
Hamamah S, Hajnal A, Covasa M. Influence of Bariatric Surgery on Gut Microbiota Composition and Its Implication on Brain and Peripheral Targets. Nutrients 2024; 16:1071. [PMID: 38613104 PMCID: PMC11013759 DOI: 10.3390/nu16071071] [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: 03/14/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Obesity remains a significant global health challenge, with bariatric surgery remaining as one of the most effective treatments for severe obesity and its related comorbidities. This review highlights the multifaceted impact of bariatric surgery beyond mere physical restriction or nutrient malabsorption, underscoring the importance of the gut microbiome and neurohormonal signals in mediating the profound effects on weight loss and behavior modification. The various bariatric surgery procedures, such as Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG), act through distinct mechanisms to alter the gut microbiome, subsequently impacting metabolic health, energy balance, and food reward behaviors. Emerging evidence has shown that bariatric surgery induces profound changes in the composition of the gut microbiome, notably altering the Firmicutes/Bacteroidetes ratio and enhancing populations of beneficial bacteria such as Akkermansia. These microbiota shifts have far-reaching effects beyond gut health, influencing dopamine-mediated reward pathways in the brain and modulating the secretion and action of key gut hormones including ghrelin, leptin, GLP-1, PYY, and CCK. The resultant changes in dopamine signaling and hormone levels contribute to reduced hedonic eating, enhanced satiety, and improved metabolic outcomes. Further, post-bariatric surgical effects on satiation targets are in part mediated by metabolic byproducts of gut microbiota like short-chain fatty acids (SCFAs) and bile acids, which play a pivotal role in modulating metabolism and energy expenditure and reducing obesity-associated inflammation, as well as influencing food reward pathways, potentially contributing to the regulation of body weight and reduction in hedonic eating behaviors. Overall, a better understanding of these mechanisms opens the door to developing non-surgical interventions that replicate the beneficial effects of bariatric surgery on the gut microbiome, dopamine signaling, and gut hormone regulation, offering new avenues for obesity treatment.
Collapse
Affiliation(s)
- Sevag Hamamah
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 9176, USA;
| | - Andras Hajnal
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA;
| | - Mihai Covasa
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 9176, USA;
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 7200229 Suceava, Romania
| |
Collapse
|
21
|
Zhang L, Wang P, Huang J, Xing Y, Wong FS, Suo J, Wen L. Gut microbiota and therapy for obesity and type 2 diabetes. Front Endocrinol (Lausanne) 2024; 15:1333778. [PMID: 38596222 PMCID: PMC11002083 DOI: 10.3389/fendo.2024.1333778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/06/2024] [Indexed: 04/11/2024] Open
Abstract
There has been a major increase in Type 2 diabetes and obesity in many countries, and this will lead to a global public health crisis, which not only impacts on the quality of life of individuals well but also places a substantial burden on healthcare systems and economies. Obesity is linked to not only to type 2 diabetes but also cardiovascular diseases, musculoskeletal disorders, and certain cancers, also resulting in increased medical costs and diminished quality of life. A number of studies have linked changes in gut in obesity development. Dysbiosis, a deleterious change in gut microbiota composition, leads to altered intestinal permeability, associated with obesity and Type 2 diabetes. Many factors affect the homeostasis of gut microbiota, including diet, genetics, circadian rhythms, medication, probiotics, and antibiotics. In addition, bariatric surgery induces changes in gut microbiota that contributes to the metabolic benefits observed post-surgery. Current obesity management strategies encompass dietary interventions, exercise, pharmacotherapy, and bariatric surgery, with emerging treatments including microbiota-altering approaches showing promising efficacy. While pharmacotherapy has demonstrated significant advancements in recent years, bariatric surgery remains one of the most effective treatments for sustainable weight loss. However, access to this is generally limited to those living with severe obesity. This underscores the need for non-surgical interventions, particularly for adolescents and mildly obese patients. In this comprehensive review, we assess longitudinal alterations in gut microbiota composition and functionality resulting from the two currently most effective anti-obesity treatments: pharmacotherapy and bariatric surgery. Additionally, we highlight the functions of gut microbiota, focusing on specific bacteria, their metabolites, and strategies for modulating gut microbiota to prevent and treat obesity. This review aims to provide insights into the evolving landscape of obesity management and the potential of microbiota-based approaches in addressing this pressing global health challenge.
Collapse
Affiliation(s)
- Luyao Zhang
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Pai Wang
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Juan Huang
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha, Hunan, China
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanpeng Xing
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - F Susan Wong
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Jian Suo
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Li Wen
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
22
|
Patil M, Casari I, Warne LN, Falasca M. G protein-coupled receptors driven intestinal glucagon-like peptide-1 reprogramming for obesity: Hope or hype? Biomed Pharmacother 2024; 172:116245. [PMID: 38340396 DOI: 10.1016/j.biopha.2024.116245] [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: 09/18/2023] [Revised: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
'Globesity' is a foremost challenge to the healthcare system. The limited efficacy and adverse effects of available oral pharmacotherapies pose a significant obstacle in the fight against obesity. The biology of the leading incretin hormone glucagon-like-peptide-1 (GLP-1) has been highly captivated during the last decade owing to its multisystemic pleiotropic clinical outcomes beyond inherent glucoregulatory action. That fostered a pharmaceutical interest in synthetic GLP-1 analogues to tackle type-2 diabetes (T2D), obesity and related complications. Besides, mechanistic insights on metabolic surgeries allude to an incretin-based hormonal combination strategy for weight loss that emerged as a forerunner for the discovery of injectable 'unimolecular poly-incretin-agonist' therapies. Physiologically, intestinal enteroendocrine L-cells (EECs) are the prominent endogenous source of GLP-1 peptide. Despite comprehending the potential of various G protein-coupled receptors (GPCRs) to stimulate endogenous GLP-1 secretion, decades of translational GPCR research have failed to yield regulatory-approved endogenous GLP-1 secretagogue oral therapy. Lately, a dual/poly-GPCR agonism strategy has emerged as an alternative approach to the traditional mono-GPCR concept. This review aims to gain a comprehensive understanding by revisiting the pharmacology of a few potential GPCR-based complementary avenues that have drawn attention to the design of orally active poly-GPCR agonist therapy. The merits, challenges and recent developments that may aid future poly-GPCR drug discovery are critically discussed. Subsequently, we project the mechanism-based therapeutic potential and limitations of oral poly-GPCR agonism strategy to augment intestinal GLP-1 for weight loss. We further extend our discussion to compare the poly-GPCR agonism approach over invasive surgical and injectable GLP-1-based regimens currently in clinical practice for obesity.
Collapse
Affiliation(s)
- Mohan Patil
- Metabolic Signalling Group, Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Ilaria Casari
- Metabolic Signalling Group, Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Leon N Warne
- Little Green Pharma, West Perth, Western Australia 6872, Australia
| | - Marco Falasca
- University of Parma, Department of Medicine and Surgery, Via Volturno 39, 43125 Parma, Italy.
| |
Collapse
|
23
|
Huber H, Schieren A, Holst JJ, Simon MC. Dietary impact on fasting and stimulated GLP-1 secretion in different metabolic conditions - a narrative review. Am J Clin Nutr 2024; 119:599-627. [PMID: 38218319 PMCID: PMC10972717 DOI: 10.1016/j.ajcnut.2024.01.007] [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: 09/07/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/15/2024] Open
Abstract
Glucagon-like peptide 1 (GLP-1), a gastrointestinal peptide and central mediator of glucose metabolism, is secreted by L cells in the intestine in response to food intake. Postprandial secretion of GLP-1 is triggered by nutrient-sensing via transporters and G-protein-coupled receptors (GPCRs). GLP-1 secretion may be lower in adults with obesity/overweight (OW) or type 2 diabetes mellitus (T2DM) than in those with normal glucose tolerance (NGT), but these findings are inconsistent. Because of the actions of GLP-1 on stimulating insulin secretion and promoting weight loss, GLP-1 and its analogs are used in pharmacologic preparations for the treatment of T2DM. However, physiologically stimulated GLP-1 secretion through the diet might be a preventive or synergistic method for improving glucose metabolism in individuals who are OW, or have impaired glucose tolerance (IGT) or T2DM. This narrative review focuses on fasting and postprandial GLP-1 secretion in individuals with different metabolic conditions and degrees of glucose intolerance. Further, the influence of relevant diet-related factors (e.g., specific diets, meal composition, and size, phytochemical content, and gut microbiome) that could affect fasting and postprandial GLP-1 secretion are discussed. Some studies showed diminished glucose- or meal-stimulated GLP-1 response in participants with T2DM, IGT, or OW compared with those with NGT, whereas other studies have reported an elevated or unchanged GLP-1 response in T2DM or IGT. Meal composition, especially the relationship between macronutrients and interventions targeting the microbiome can impact postprandial GLP-1 secretion, although it is not clear which macronutrients are strong stimulants of GLP-1. Moreover, glucose tolerance, antidiabetic treatment, grade of overweight/obesity, and sex were important factors influencing GLP-1 secretion. The results presented in this review highlight the potential of nutritional and physiologic stimulation of GLP-1 secretion. Further research on fasting and postprandial GLP-1 concentrations and the resulting metabolic consequences under different metabolic conditions is needed.
Collapse
Affiliation(s)
- Hanna Huber
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Institute of Neuroscience and Physiology, Mölndal, Sweden; Department Nutrition and Microbiota, University of Bonn, Institute of Nutrition and Food Science, Bonn, Germany
| | - Alina Schieren
- Department Nutrition and Microbiota, University of Bonn, Institute of Nutrition and Food Science, Bonn, Germany
| | - Jens Juul Holst
- Department of Biomedical Sciences, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark; The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Marie-Christine Simon
- Department Nutrition and Microbiota, University of Bonn, Institute of Nutrition and Food Science, Bonn, Germany.
| |
Collapse
|
24
|
Salazar J, Duran P, Garrido B, Parra H, Hernández M, Cano C, Añez R, García-Pacheco H, Cubillos G, Vasquez N, Chacin M, Bermúdez V. Weight Regain after Metabolic Surgery: Beyond the Surgical Failure. J Clin Med 2024; 13:1143. [PMID: 38398456 PMCID: PMC10888585 DOI: 10.3390/jcm13041143] [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: 12/11/2023] [Revised: 01/20/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Patients undergoing metabolic surgery have factors ranging from anatomo-surgical, endocrine metabolic, eating patterns and physical activity, mental health and psychological factors. Some of the latter can explain the possible pathophysiological neuroendocrine, metabolic, and adaptive mechanisms that cause the high prevalence of weight regain in postbariatric patients. Even metabolic surgery has proven to be effective in reducing excess weight in patients with obesity; some of them regain weight after this intervention. In this vein, several studies have been conducted to search factors and mechanisms involved in weight regain, to stablish strategies to manage this complication by combining metabolic surgery with either lifestyle changes, behavioral therapies, pharmacotherapy, endoscopic interventions, or finally, surgical revision. The aim of this revision is to describe certain aspects and mechanisms behind weight regain after metabolic surgery, along with preventive and therapeutic strategies for this complication.
Collapse
Affiliation(s)
- Juan Salazar
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Pablo Duran
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Bermary Garrido
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Heliana Parra
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Marlon Hernández
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Clímaco Cano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Roberto Añez
- Departamento de Endocrinología y Nutrición, Hospital Quirónsalud, 28009 Madrid, Spain
| | - Henry García-Pacheco
- Facultad de Medicina, Departamento de Cirugía, Universidad del Zulia, Hospital General del Sur, Dr. Pedro Iturbe, Maracaibo 4004, Venezuela
- Unidad de Cirugía para Obesidad y Metabolismo (UCOM), Maracaibo 4004, Venezuela
| | | | | | - Maricarmen Chacin
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 080001, Colombia
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla 080001, Colombia
| | - Valmore Bermúdez
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 080001, Colombia
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla 080001, Colombia
| |
Collapse
|
25
|
Papamargaritis D, le Roux CW, Holst JJ, Davies MJ. New therapies for obesity. Cardiovasc Res 2024; 119:2825-2842. [PMID: 36448672 PMCID: PMC10874276 DOI: 10.1093/cvr/cvac176] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/16/2022] [Accepted: 10/19/2022] [Indexed: 02/17/2024] Open
Abstract
Obesity is a chronic disease associated with serious complications and increased mortality. Weight loss (WL) through lifestyle changes results in modest WL long-term possibly due to compensatory biological adaptations (increased appetite and reduced energy expenditure) promoting weight gain. Bariatric surgery was until recently the only intervention that consistently resulted in ≥ 15% WL and maintenance. Our better understanding of the endocrine regulation of appetite has led to the development of new medications over the last decade for the treatment of obesity with main target the reduction of appetite. The efficacy of semaglutide 2.4 mg/week-the latest glucagon-like peptide-1 (GLP-1) receptor analogue-on WL for people with obesity suggests that we are entering a new era in obesity pharmacotherapy where ≥15% WL is feasible. Moreover, the WL achieved with the dual agonist tirzepatide (GLP-1/glucose-dependent insulinotropic polypeptide) for people with type 2 diabetes and most recently also obesity, indicate that combining the GLP-1 with other gut hormones may lead to additional WL compared with GLP-1 receptor analogues alone and in the future, multi-agonist molecules may offer the potential to bridge further the efficacy gap between bariatric surgery and the currently available pharmacotherapies.
Collapse
Affiliation(s)
- Dimitris Papamargaritis
- Diabetes Research Centre, Leicester General Hospital, University of Leicester College of Medicine Biological Sciences and Psychology, Leicester LE5 4PW, UK
| | - Carel W le Roux
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Dublin 4, Ireland
- Diabetes Research Centre, Ulster University, Coleraine BT52 1SA, UK
| | - Jens J Holst
- Department of Biomedical Sciences and the NNF Center for Basic Metabolic Research, University of Copenhagen Panum Institute, Copenhagen 2200, Denmark
| | - Melanie J Davies
- Diabetes Research Centre, Leicester General Hospital, University of Leicester College of Medicine Biological Sciences and Psychology, Leicester LE5 4PW, UK
| |
Collapse
|
26
|
Nymo S, Lundanes J, Eriksen K, Aukan M, Rehfeld JF, Holst JJ, Johnsen G, Græslie H, Kulseng B, Sandvik J, Martins C. Suboptimal Weight Loss 13 Years After Roux-en-Y Gastric Bypass Is Associated with Blunted Appetite Response. Obes Surg 2024; 34:592-601. [PMID: 38159146 PMCID: PMC10811108 DOI: 10.1007/s11695-023-07028-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
PURPOSE Bariatric surgery remains the most efficient treatment to achieve a sustained weight loss. However, a large proportion of patients experience suboptimal weight loss (SWL). The exact mechanisms involved remain to be fully elucidated, but the homeostatic appetite control system seems to be involved. The aim of this study was, therefore, to compare the plasma concentration of gastrointestinal hormones, and appetite ratings, between those experiencing SWL and optimal weight loss (OWL) after Roux-en-Y gastric bypass (RYGB). MATERIALS AND METHODS Fifty participants from the Bariatric Surgery Observation Study (BAROBS) experiencing either SWL or OWL (< or ≥ 50% of excess weight loss (EWL), respectively) > 13 years post-RYGB were compared to 25 non-surgical controls. Plasma concentrations of acylated ghrelin (AG), total glucagon-like peptide-1 (GLP-1), total peptide YY (PYY), cholecystokinin (CCK), and subjective ratings of hunger, fullness, desire to eat (DTE), and prospective food consumption (PFC) were assessed in the fasting and postprandial (area under the curve (AUC)) states. RESULTS Those experiencing OWL presented with higher basal AG and GLP-1 iAUC, and lower AG iAUC compared with SWL and controls. Additionally, both bariatric groups presented with higher PYY and CCK iAUC compared to controls. PFC tAUC was also lower in OWL compared to the SWL group. Total weight loss was positively correlated with GLP-1 tAUC and negatively correlated with fasting and tAUC DTE and PFC tAUC. CONCLUSIONS SWL > 13 years post-RYGB is associated with lower basal ghrelin, as well as a weaker satiety response to a meal. Future studies should investigate the causality of these associations.
Collapse
Affiliation(s)
- Siren Nymo
- Obesity Research Group, Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Forsyningssenteret, Prinsesse Kristinas gate 5, 7030, Trondheim, Norway.
- Centre for Obesity and Innovation (ObeCe), Clinic of Surgery, St. Olav University Hospital, Trondheim, Norway.
- Nord-Trøndelag Hospital Trust, Clinic of Surgery, Namsos Hospital, Namsos, Norway.
| | - Julianne Lundanes
- Obesity Research Group, Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Forsyningssenteret, Prinsesse Kristinas gate 5, 7030, Trondheim, Norway
- Nord-Trøndelag Hospital Trust, Clinic of Surgery, Namsos Hospital, Namsos, Norway
| | - Kevin Eriksen
- Obesity Research Group, Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Forsyningssenteret, Prinsesse Kristinas gate 5, 7030, Trondheim, Norway
| | - Marthe Aukan
- Obesity Research Group, Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Forsyningssenteret, Prinsesse Kristinas gate 5, 7030, Trondheim, Norway
- Centre for Obesity and Innovation (ObeCe), Clinic of Surgery, St. Olav University Hospital, Trondheim, Norway
| | - Jens Frederik Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Gjermund Johnsen
- Obesity Research Group, Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Forsyningssenteret, Prinsesse Kristinas gate 5, 7030, Trondheim, Norway
| | - Hallvard Græslie
- Nord-Trøndelag Hospital Trust, Clinic of Surgery, Namsos Hospital, Namsos, Norway
| | - Bård Kulseng
- Obesity Research Group, Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Forsyningssenteret, Prinsesse Kristinas gate 5, 7030, Trondheim, Norway
- Centre for Obesity and Innovation (ObeCe), Clinic of Surgery, St. Olav University Hospital, Trondheim, Norway
| | - Jorunn Sandvik
- Obesity Research Group, Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Forsyningssenteret, Prinsesse Kristinas gate 5, 7030, Trondheim, Norway
- Centre for Obesity and Innovation (ObeCe), Clinic of Surgery, St. Olav University Hospital, Trondheim, Norway
- Møre and Romsdal Hospital Trust, Clinic of Surgery, Ålesund Hospital, Ålesund, Norway
| | - Catia Martins
- Obesity Research Group, Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Forsyningssenteret, Prinsesse Kristinas gate 5, 7030, Trondheim, Norway
- Centre for Obesity and Innovation (ObeCe), Clinic of Surgery, St. Olav University Hospital, Trondheim, Norway
- Department of Nutrition Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| |
Collapse
|
27
|
Zeng Y, Wu Y, Zhang Q, Xiao X. Crosstalk between glucagon-like peptide 1 and gut microbiota in metabolic diseases. mBio 2024; 15:e0203223. [PMID: 38055342 PMCID: PMC10790698 DOI: 10.1128/mbio.02032-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023] Open
Abstract
Gut microbiota exert influence on gastrointestinal mucosal permeability, bile acid metabolism, short-chain fatty acid synthesis, dietary fiber fermentation, and farnesoid X receptor/Takeda G protein-coupled receptor 5 (TGR5) signal transduction. The incretin glucagon-like peptide 1 (GLP-1) is mainly produced by L cells in the gut and regulates postprandial blood glucose. Changes in gut microbiota composition and function have been observed in obesity and type 2 diabetes (T2D). Meanwhile, the function and rhythm of GLP-1 have also been affected in subjects with obesity or T2D. Therefore, it is necessary to discuss the link between the gut microbiome and GLP-1. In this review, we describe the interaction between GLP-1 and the gut microbiota in metabolic diseases. On the one hand, gut microbiota metabolites stimulate GLP-1 secretion, and gut microbiota affect GLP-1 function and rhythm. On the other hand, the mechanism of action of GLP-1 on gut microbiota involves the inflammatory response. Additionally, we discuss the effects and mechanism of various interventions, such as prebiotics, probiotics, antidiabetic drugs, and bariatric surgery, on the crosstalk between gut microbiota and GLP-1. Finally, we stress that gut microbiota can be used as a target for metabolic diseases, and the clinical application of GLP-1 receptor agonists should be individualized.
Collapse
Grants
- 81870545, 81870579, 82170854, 81570715, 81170736 MOST | National Natural Science Foundation of China (NSFC)
- 7202163 Natural Science Foundation of Beijing Municipality (Beijing Natural Science Foundation)
- Z201100005520011 Beijing Municipal Science and Technology Commission, Adminitrative Commission of Zhongguancun Science Park
- 2017YFC1309603, 2021YFC2501700, 2016YFA0101002, 2018YFC2001100 MOST | National Key Research and Development Program of China (NKPs)
- 2019DCT-M-05 Beijing Municipal Human Resources and Social Security Bureau (BMHRSSB)
- 2017PT31036, 2018PT31021 Chinese Academy of Medical Sciences (CAMS)
- 2017PT32020, 2018PT32001 Chinese Academy of Medical Sciences (CAMS)
- CIFMS2017-I2M-1-008, CIFMS2021-I2M-1-002 Chinese Academy of Medical Sciences (CAMS)
- 2022-PUMCH- C-019, 2022-PUMCH-B-121 National High Level Hospital Clinical Research Funding
Collapse
Affiliation(s)
- Yuan Zeng
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yifan Wu
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qian Zhang
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinhua Xiao
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
28
|
Hurtado A, Chandar AK, Perez JA, Casselberry R, Martin SA, DeLano K, Abbas M, Chak A. Reduced risk of de novo Barrett esophagus after bariatric surgery: a national database study. Surg Obes Relat Dis 2024; 20:40-45. [PMID: 37722939 PMCID: PMC10843733 DOI: 10.1016/j.soard.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 06/27/2023] [Accepted: 08/05/2023] [Indexed: 09/20/2023]
Abstract
BACKGROUND Bariatric surgery is an effective treatment for obesity and may decrease the morbidity and mortality of obesity-associated cancers. OBJECTIVE We investigated the risk of a new diagnosis of Barrett esophagus (BE) following bariatric surgery compared to screening colonoscopy controls. SETTING Large national database including patients who received care in inpatient, outpatient, and specialty care services. METHODS A national healthcare database (TriNetX, LLC) was used for this analysis. Cases included adults (aged ≥18 yr) who had undergone either sleeve gastrectomy (SG) or Roux-en-Y gastric bypass (RYGB). Controls included adults undergoing screening colonoscopy and an esophagoduodenoscopy on the same day and had never undergone bariatric surgery. Cases and controls were propensity-matched for confounders. The risk of de novo diagnosis of BE at least 1 year after bariatric surgery was compared between cases and controls. Secondary analyses examined the effect of bariatric surgery on metabolic outcomes such as weight loss and body mass index (BMI). The risk of de novo diagnosis of BE in SG was compared with RYGB. Odds ratios (OR) and 95% CI were used to report on these associations. RESULTS In the propensity-matched analysis, patients who had undergone a bariatric surgical procedure showed a significantly reduced risk of de novo BE when compared with screening colonoscopy controls (.67 [.48, .94]). There was substantial reduction in weight and BMI in the bariatric surgery group when compared with baseline. There was no significant difference in de novo BE diagnosis between the propensity-matched SG and RYGB groups (.77 [.5, 1.2]). CONCLUSION Patients who underwent bariatric surgery (RYGB or SG) had a lower risk of being diagnosed with BE compared with screening colonoscopy controls who did not receive bariatric surgery. This effect appears to be largely mediated by reduction in weight and BMI.
Collapse
Affiliation(s)
- Alexander Hurtado
- Case Western Reserve University School of Medicine, Cleveland, Ohio.
| | - Apoorva K Chandar
- Case Western Reserve University School of Medicine, Cleveland, Ohio; University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | | | - Scott A Martin
- University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | - Mujjahid Abbas
- University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Amitabh Chak
- Case Western Reserve University School of Medicine, Cleveland, Ohio; University Hospitals Cleveland Medical Center, Cleveland, Ohio
| |
Collapse
|
29
|
Kashihara H, Okikawa S, Morine Y, Yoshikawa K, Tokunaga T, Nishi M, Takasu C, Nishiyama M, Zushi M, Shimada M. Impact of Daikenchuto (TU-100) on the early postoperative period in duodenal-jejunal bypass. THE JOURNAL OF MEDICAL INVESTIGATION 2024; 71:210-218. [PMID: 39462554 DOI: 10.2152/jmi.71.210] [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] [Indexed: 10/29/2024]
Abstract
INTRODUCTION We investigated the effect of Daikenchuto (TU-100) on the early postoperative period in duodenal-jejunal bypass (DJB). METHODS Study 1:The effect of TU-100 on diabetic rats was investigated. Rats were sacrificed after receiving TU-100 for one week. Study 2:The effect of TU-100 on DJB was investigated. Rats in the DJB and TU-100 treated DJB groups were sacrificed 24 hours postoperation to evaluate blood glucose, cytokine expression, and gut microbiome. RESULTS Study 1:TU-100 did not affect glucose or body weight. TU-100 suppressed intestinal inflammation and modified the gut microbiome. Specifically, Bifidobacterium and Blautia were increased, and Turicibacter were decreased in this group. Study 2:Both DJB and TU-100 treated DJB rats showed lower blood glucose at 24 hours postoperation than at preoperation. Cytokine expression in the liver and small intestine of the TU-100 treated DJB group was significantly lower than that of the DJB group. The gut microbiome composition in TU-100 treated DJB rats was altered. In particular, Bifidobacterium and Blautia were increased in this group. CONCLUSION DJB suppressed blood glucose during the early postoperative period. TU-100 may enhance the anti-diabetic effect of metabolic surgery by changing the gut microbiome and suppressing inflammation in the early postoperative period. J. Med. Invest. 71 : 210-218, August, 2024.
Collapse
Affiliation(s)
| | - Shohei Okikawa
- Department of Surgery, Tokushima University, Tokushima, Japan
| | - Yuji Morine
- Department of Surgery, Tokushima University, Tokushima, Japan
| | - Kozo Yoshikawa
- Department of Surgery, Tokushima University, Tokushima, Japan
| | - Takuya Tokunaga
- Department of Surgery, Tokushima University, Tokushima, Japan
| | - Masaaki Nishi
- Department of Surgery, Tokushima University, Tokushima, Japan
| | - Chie Takasu
- Department of Surgery, Tokushima University, Tokushima, Japan
| | - Mitsue Nishiyama
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ami, Ibaraki, Japan
| | - Makoto Zushi
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ami, Ibaraki, Japan
| | - Mitsuo Shimada
- Department of Surgery, Tokushima University, Tokushima, Japan
| |
Collapse
|
30
|
Engin A. Bariatric Surgery in Obesity: Metabolic Quality Analysis and Comparison of Surgical Options. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:697-726. [PMID: 39287870 DOI: 10.1007/978-3-031-63657-8_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Obesity is a constantly growing health problem which reduces quality of life and life expectancy. Bariatric surgery (BS) for obesity is considered when all other conservative treatment modalities have failed. Comparison of the multidisciplinary programs with BS regarding to the weight loss showed that substantial and durable weight reduction have been achieved only with bariatric surgical treatments. Although laparoscopic sleeve gastrectomy is the most popular BS, it has high long-term failure rates, and it is claimed that one of every three patients will undergo another bariatric procedure within a 10-year period. Although BS provides weight loss and improvement of metabolic comorbidities, in long-term follow-up, weight gain is observed in half of the patients, while decrease in bone mass and nutritional deficiencies occur in up to 90%. Moreover, despite significant weight loss, several psychological aspects of patients are worsened in comparison to preoperative levels. Nearly one-fifth of postoperative patients with "Loss-of-eating control" meet food addiction criteria. Therefore, the benefits of weight loss following bariatric procedures alone are still debated in terms of the proinflammatory and metabolic profile of obesity.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
31
|
Revelo X, Fredrickson G, Florczak K, Barrow F, Dietsche K, Wang H, Parthiban P, Almutlaq R, Adeyi O, Herman A, Bartolomucci A, Staley C, Jahansouz C, Williams J, Mashek D, Ikramuddin S. Hepatic lipid-associated macrophages mediate the beneficial effects of bariatric surgery against MASH. RESEARCH SQUARE 2023:rs.3.rs-3446960. [PMID: 37961666 PMCID: PMC10635378 DOI: 10.21203/rs.3.rs-3446960/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
For patients with obesity and metabolic syndrome, bariatric procedures such as vertical sleeve gastrectomy (VSG) have a clear benefit in ameliorating metabolic dysfunction-associated steatohepatitis (MASH). While the effects of bariatric surgeries have been mainly attributed to nutrient restriction and malabsorption, whether immuno-modulatory mechanisms are involved remains unclear. Here we report that VSG ameliorates MASH progression in a weight loss-independent manner. Single-cell RNA sequencing revealed that hepatic lipid-associated macrophages (LAMs) expressing the triggering receptor expressed on myeloid cells 2 (TREM2) increase their lysosomal activity and repress inflammation in response to VSG. Remarkably, TREM2 deficiency in mice ablates the reparative effects of VSG, suggesting that TREM2 is required for MASH resolution. Mechanistically, TREM2 prevents the inflammatory activation of macrophages and is required for their efferocytotic function. Overall, our findings indicate that bariatric surgery improves MASH through a reparative process driven by hepatic LAMs, providing insights into the mechanisms of disease reversal that may result in new therapies and improved surgical interventions.
Collapse
|
32
|
Huang S, Huang Y, Gu Y, Chen H, Lv R, Wu S, Song P, Zhao D, Hu L, Yuan C. Adherence to 24-Hour Movement Guidelines in Relation to the Risk of Overweight and Obesity Among Children and Adolescents. J Adolesc Health 2023; 73:887-895. [PMID: 37565981 DOI: 10.1016/j.jadohealth.2023.06.009] [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: 10/25/2022] [Revised: 05/03/2023] [Accepted: 06/09/2023] [Indexed: 08/12/2023]
Abstract
PURPOSE Adherence to overall 24-hour Movement Guidelines (24HGs) has been associated with childhood obesity in cross-sectional studies. However, few longitudinal studies have examined such associations, especially in China. We aimed to explore prospective associations between adherence to recommendations of 24HGs and risks of developing overweight and obesity among children and adolescents. METHODS We included participants (aged 6-17 years) without overweight and obesity at enrollment from the China Health and Nutrition Survey in 2004-2011 surveys and followed them till 2015. We assigned one point each to the adherence of guidelines for moderate-to-vigorous physical activity, recreational screen time and sleep, and summed them up to indicate the overall level of adherence to 24HGs (range: 0-3 points). The primary outcome was the first occurrence of overweight or obesity. Multivariable cox proportional hazard models were used to evaluate the corresponding associations. RESULTS Among 1,382 participants (mean age: 10.3 ± 3.2 years; 48.4% girls), a total of 152 (11%) individuals were identified as incident overweight and obesity during an average of 4.7 years of follow-up. Compared with participants nonadherent to any of the guidelines, those adhering to one (hazard ratio [HR] = 0.38, 95% confidence interval [CI]: 0.21-0.71, p < .01), two (HR = 0.49, 95% CI: 0.28-0.88, p = .02), and three (HR = 0.40, 95% CI: 0.17-0.91, p = .03) recommendations had significantly lower risks of developing overweight and obesity. DISCUSSION Children and adolescents who met any recommendations of 24HGs had significantly lower risks of developing subsequent overweight and obesity. Setting achievable goals such as adopting at least one recommendation could be considered in future public health recommendations to accelerate progress in childhood obesity prevention.
Collapse
Affiliation(s)
- Siyi Huang
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Sports Science, College of Education, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuhui Huang
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuxuan Gu
- Department of Social Security, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Hui Chen
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Rongxia Lv
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shiyi Wu
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Peige Song
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Dong Zhao
- Department of Nutrition and Food Safety, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Liang Hu
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Changzheng Yuan
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.
| |
Collapse
|
33
|
Bojsen-Møller KN, Svane MS, Martinussen C, Dirksen C, Jørgensen NB, Jensen JEB, Jensen CZ, Torekov SS, Kristiansen VB, Rehfeld JF, Bork-Jensen J, Grarup N, Hansen T, Hartmann B, Holst JJ, Madsbad S. Primary weight loss failure after Roux-en-Y gastric bypass is characterized by impaired gut-hormone mediated regulation of food intake. Int J Obes (Lond) 2023; 47:1143-1151. [PMID: 37653071 PMCID: PMC10599997 DOI: 10.1038/s41366-023-01372-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND/OBJECTIVES After Roux-en-Y gastric bypass (RYGB) a subset of patients never obtain excess BMI loss (EBMIL) > 50% and are categorized as having primary weight loss (WL) failure. We hypothesized that postprandial concentrations of glucagon-like peptide 1 (GLP-1) and peptide YY (PYY) would be lower in patients with primary WL failure compared with patients with successfully maintained WL. Furthermore, that inhibition of gut hormone secretions would increase ad libitum food intake less in patients with primary WL failure. SUBJECTS/METHODS Twenty women with primary WL failure (LowEBMIL < 50%) were individually matched to twenty women with successful WL (HighEBMIL > 60%) on age, preoperative BMI and time from RYGB. On separate days performed in a random order, patient-blinded subcutaneous injections of octreotide or saline (placebo) were followed by a fixed breakfast and an ad libitum lunch with blood sampling for appetite regulating hormones and Visual-Analogue-Scale (VAS)-scoring of hunger/satiety. Furthermore, participants underwent gene variant analysis for GLP-1, PYY and their receptors, indirect calorimetry, dual-energy X-ray absorptiometry (DXA)-scans, 4-days at-home food registration and 14-days step counting. RESULTS On placebo days, postprandial GLP-1, PYY and cholecystokinin (CCK) concentrations were similar between groups after breakfast. Fasting ghrelin was lower in LowEBMIL, but the postprandial suppression was similar. LowEBMIL had lower satiety VAS-scores and less suppression of hunger VAS-scores. Gene variants did not differ between groups. Octreotide diminished GLP-1, PYY, CCK and ghrelin concentrations in both groups. Octreotide did not affect ad libitum food intake in LowEBMIL (-1% [-13, 12], mean [95%CI]), while food intake increased in HighEBMIL (+23% [2,44]). CONCLUSIONS Primary WL failure after RYGB was not characterized by impaired secretions of appetite regulating gut hormones. Interestingly, inhibition of gut hormone secretions with octreotide only increased food intake in patients with successful WL post-RYGB. Thus, an impaired central anorectic response to gut hormones may contribute to primary WL failure after RYGB.
Collapse
Affiliation(s)
- Kirstine Nyvold Bojsen-Møller
- Dept. of Endocrinology, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark.
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| | - Maria Saur Svane
- Dept. of Endocrinology, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Christoffer Martinussen
- Dept. of Endocrinology, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Carsten Dirksen
- Dept. of Endocrinology, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Dept. of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Nils Bruun Jørgensen
- Dept. of Endocrinology, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jens-Erik Beck Jensen
- Dept. of Endocrinology, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
- Dept. of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Christian Zinck Jensen
- Dept. of Endocrinology, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Signe Sørensen Torekov
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Dept. of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Jette Bork-Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Dept. of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Dept. of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sten Madsbad
- Dept. of Endocrinology, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Dept. of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
34
|
Mok J, Adeleke MO, Brown A, Magee CG, Firman C, Makahamadze C, Jassil FC, Marvasti P, Carnemolla A, Devalia K, Fakih N, Elkalaawy M, Pucci A, Jenkinson A, Adamo M, Omar RZ, Batterham RL, Makaronidis J. Safety and Efficacy of Liraglutide, 3.0 mg, Once Daily vs Placebo in Patients With Poor Weight Loss Following Metabolic Surgery: The BARI-OPTIMISE Randomized Clinical Trial. JAMA Surg 2023; 158:1003-1011. [PMID: 37494014 PMCID: PMC10372755 DOI: 10.1001/jamasurg.2023.2930] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/08/2023] [Indexed: 07/27/2023]
Abstract
Importance Metabolic surgery leads to weight loss and improved health, but these outcomes are highly variable. Poor weight loss is associated with lower circulating levels of glucagon-like peptide-1 (GLP-1). Objective To assess the efficacy and safety of the GLP-1 receptor agonist, liraglutide, 3.0 mg, on percentage body weight reduction in patients with poor weight loss and suboptimal GLP-1 response after metabolic surgery. Design, Setting, and Participants The Evaluation of Liraglutide 3.0 mg in Patients With Poor Weight Loss and a Suboptimal Glucagon-Like Peptide-1 Response (BARI-OPTIMISE) randomized placebo-controlled trial recruited adult patients at least 1 year after metabolic surgery who had experienced 20% or less body weight loss from the day of surgery and a suboptimal nutrient-stimulated GLP-1 response from 2 hospitals in London, United Kingdom, between October 2018 and November 2019. Key exclusion criteria were type 1 diabetes; severe concomitant psychiatric, gastrointestinal, cardiac, kidney or metabolic disease; and use of insulin, GLP-1 receptor analogues, and medication that can affect weight. The study period was 24 weeks followed by a 4-week follow-up period. Last participant follow-up was completed in June 2020. All participants and clinical study personnel were blinded to treatment allocation. Of 154 assessed for eligibility, 70 met trial criteria and were included in the study, and 57 completed follow-up. Interventions Liraglutide, 3.0 mg, once daily or placebo as an adjunct to lifestyle intervention with a 500-kcal daily energy deficit for 24 weeks, on a 1:1 allocation by computer-generated randomization sequence, stratified by surgery type (Roux-en-Y gastric bypass [RYGB] or sleeve gastrectomy [SG]) and type 2 diabetes status. Main Outcome and Measures The primary outcome was change in percentage body weight from baseline to the end of the 24-week study period based on an intention-to-treat analysis. Participant safety was assessed through monitoring of biochemical parameters, including kidney and liver function, physical examination, and assessment for adverse events. Results A total of 70 participants (mean [SD] age, 47.6 [10.7] years; 52 [74%] female) with a poor weight loss response following RYGB or SG were randomized to receive 3.0-mg liraglutide (n = 35) or placebo (n = 35). All participants received at least 1 dose of the trial drug. Eight participants discontinued treatment (4 per group), and 2 in the 3.0-mg liraglutide group and 1 in the placebo group were lost to follow-up. Due to COVID-19 restrictions, 3 participants in the 3.0-mg liraglutide group and 7 in the placebo group were unable to attend their final in-person assessment. Estimated change in mean (SD) percentage body weight from baseline to week 24 was -8.82 (4.94) with liraglutide, 3.0 mg (n = 31), vs -0.54 (3.32) with placebo (n = 26). The mean difference in percentage body weight change for liraglutide, 3.0 mg, vs placebo was -8.03 (95% CI, -10.39 to -5.66; P < .001). Adverse events, predominantly gastrointestinal, were more frequent with liraglutide, 3.0 mg (28 events [80%]), than placebo (20 events [57%]). There were no serious adverse events and no treatment-related deaths. Conclusion and Relevance These findings support the use of adjuvant liraglutide, 3.0 mg, for weight management in patients with poor weight loss and suboptimal GLP-1 response after metabolic surgery. Trial Registration ClinicalTrials.gov Identifier: NCT03341429.
Collapse
Affiliation(s)
- Jessica Mok
- Division of Medicine, University College London Centre for Obesity Research, Rayne Institute, London, United Kingdom
- Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospitals, National Health Service Foundation Trust, London, United Kingdom
- National Institute for Health and Care Research, University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Mariam O. Adeleke
- Department of Statistical Science, University College London, London, United Kingdom
| | - Adrian Brown
- Division of Medicine, University College London Centre for Obesity Research, Rayne Institute, London, United Kingdom
- Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospitals, National Health Service Foundation Trust, London, United Kingdom
- National Institute for Health and Care Research, University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Cormac G. Magee
- Division of Medicine, University College London Centre for Obesity Research, Rayne Institute, London, United Kingdom
- Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospitals, National Health Service Foundation Trust, London, United Kingdom
- National Institute for Health and Care Research, University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Chloe Firman
- Division of Medicine, University College London Centre for Obesity Research, Rayne Institute, London, United Kingdom
- Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospitals, National Health Service Foundation Trust, London, United Kingdom
- National Institute for Health and Care Research, University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Christwishes Makahamadze
- Division of Medicine, University College London Centre for Obesity Research, Rayne Institute, London, United Kingdom
- Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospitals, National Health Service Foundation Trust, London, United Kingdom
| | - Friedrich C. Jassil
- Division of Medicine, University College London Centre for Obesity Research, Rayne Institute, London, United Kingdom
- Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospitals, National Health Service Foundation Trust, London, United Kingdom
| | - Parastou Marvasti
- Division of Medicine, University College London Centre for Obesity Research, Rayne Institute, London, United Kingdom
- National Institute for Health and Care Research, University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Alisia Carnemolla
- Division of Medicine, University College London Centre for Obesity Research, Rayne Institute, London, United Kingdom
| | - Kalpana Devalia
- Bariatric Surgery Department Homerton University Hospital National Health Service Trust, London, United Kingdom
| | - Naim Fakih
- Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospitals, National Health Service Foundation Trust, London, United Kingdom
| | - Mohamed Elkalaawy
- Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospitals, National Health Service Foundation Trust, London, United Kingdom
| | - Andrea Pucci
- Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospitals, National Health Service Foundation Trust, London, United Kingdom
| | - Andrew Jenkinson
- Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospitals, National Health Service Foundation Trust, London, United Kingdom
| | - Marco Adamo
- Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospitals, National Health Service Foundation Trust, London, United Kingdom
| | - Rumana Z. Omar
- Department of Statistical Science, University College London, London, United Kingdom
| | - Rachel L. Batterham
- Division of Medicine, University College London Centre for Obesity Research, Rayne Institute, London, United Kingdom
- Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospitals, National Health Service Foundation Trust, London, United Kingdom
- National Institute for Health and Care Research, University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Janine Makaronidis
- Division of Medicine, University College London Centre for Obesity Research, Rayne Institute, London, United Kingdom
- Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospitals, National Health Service Foundation Trust, London, United Kingdom
- National Institute for Health and Care Research, University College London Hospitals Biomedical Research Centre, London, United Kingdom
| |
Collapse
|
35
|
Tolvanen L, Christenson A, Bonn SE, Surkan PJ, Lagerros YT. Patients' Perspectives on Dietary Patterns and Eating Behaviors During Weight Regain After Gastric Bypass Surgery. Obes Surg 2023; 33:2517-2526. [PMID: 37402891 PMCID: PMC10345057 DOI: 10.1007/s11695-023-06718-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/24/2023] [Accepted: 06/28/2023] [Indexed: 07/06/2023]
Abstract
PURPOSE Food quality, energy intake, and various eating-related problems have been highlighted as some of the components influencing weight after bariatric surgery. This study aimed to increase our knowledge of patients' perspectives on dietary patterns and eating behaviors during weight regain after bariatric surgery. MATERIALS AND METHODS We recruited 4 men and 12 women with obesity and the experience of weight regain after bariatric surgery at an obesity clinic in Stockholm, Sweden. Data were collected during 2018-2019. We conducted a qualitative study, carried out individual semi-structured interviews, and analyzed the recorded and transcribed interview data with thematic analysis. RESULTS Participants had regained 12 to 71% from their lowest weight after gastric bypass surgery performed 3 to 15 years before. They perceived their dietary challenges as overwhelming and had not expected weight management, meal patterns, increasing portion sizes, and appealing energy-dense foods to be problematic after surgery. In addition, difficulties with disordered eating patterns, emotional eating, and increased alcohol intake further contributed to the weight management hurdles. Insufficient nutritional knowledge and lack of support limited participants' ability to avoid weight regain, leading to restrictive eating and dieting without sustained weight loss. CONCLUSION Eating behavior and dietary factors such as lack of nutritional knowledge, emotional eating, or disorganized meal patterns contribute to difficulties with weight management after gastric bypass surgery. Improved counseling may help patients prepare for possible weight regain and remaining challenges with food and eating. The results highlight the importance of regular medical nutrition therapy after gastric bypass surgery.
Collapse
Affiliation(s)
- Liisa Tolvanen
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Maria Aspmans gata 30A, Stockholm, SE-171 64 Sweden
- Center for Obesity, Academic Specialist Center, Stockholm, Sweden
| | - Anne Christenson
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Maria Aspmans gata 30A, Stockholm, SE-171 64 Sweden
- Center for Obesity, Academic Specialist Center, Stockholm, Sweden
| | - Stephanie E. Bonn
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Maria Aspmans gata 30A, Stockholm, SE-171 64 Sweden
| | - Pamela J. Surkan
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | - Ylva Trolle Lagerros
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Maria Aspmans gata 30A, Stockholm, SE-171 64 Sweden
- Center for Obesity, Academic Specialist Center, Stockholm, Sweden
| |
Collapse
|
36
|
Masse KE, Lu VB. Short-chain fatty acids, secondary bile acids and indoles: gut microbial metabolites with effects on enteroendocrine cell function and their potential as therapies for metabolic disease. Front Endocrinol (Lausanne) 2023; 14:1169624. [PMID: 37560311 PMCID: PMC10407565 DOI: 10.3389/fendo.2023.1169624] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/05/2023] [Indexed: 08/11/2023] Open
Abstract
The gastrointestinal tract hosts the largest ecosystem of microorganisms in the body. The metabolism of ingested nutrients by gut bacteria produces novel chemical mediators that can influence chemosensory cells lining the gastrointestinal tract. Specifically, hormone-releasing enteroendocrine cells which express a host of receptors activated by these bacterial metabolites. This review will focus on the activation mechanisms of glucagon-like peptide-1 releasing enteroendocrine cells by the three main bacterial metabolites produced in the gut: short-chain fatty acids, secondary bile acids and indoles. Given the importance of enteroendocrine cells in regulating glucose homeostasis and food intake, we will also discuss therapies based on these bacterial metabolites used in the treatment of metabolic diseases such as diabetes and obesity. Elucidating the mechanisms gut bacteria can influence cellular function in the host will advance our understanding of this fundamental symbiotic relationship and unlock the potential of harnessing these pathways to improve human health.
Collapse
Affiliation(s)
| | - Van B. Lu
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| |
Collapse
|
37
|
Mourougavelou V, Chowdhury TA. Management of hyperglycaemia in people with obesity. Clin Med (Lond) 2023; 23:364-371. [PMID: 38614651 PMCID: PMC10541039 DOI: 10.7861/clinmed.2023-0135] [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] [Indexed: 08/02/2023]
Abstract
Diabetes and obesity are closely interlinked. Obesity is a major risk factor for the development of type 2 diabetes mellitus and appears to be an important risk factor for diabetic micro- and macrovascular complications. Management of hyperglycaemia in people with diabetes is important to reduce diabetes-related complications. Previously, there was a significant tension between management of hyperglycaemia and mitigating weight gain. Older drugs, such as sulfonylureas, glitazones, and insulin, although effective antihyperglycaemic agents, tend to induce weight gain. There is now an increasing recognition in people with obesity and diabetes that the focus should be on aiding weight loss, initially with improvements in diet and physical activity, possibly with the use of low-calorie diet programmes. Subsequent addition of metformin and newer agents, such as sodium-glucose transporter-2 inhibitors and glucagon-like peptide-1 analogues, will aid glucose control and weight reduction, and offer cardiovascular and renal protection. These drugs are now much higher in the therapeutic pathway in many national and international guidelines. Bariatric surgery may also be an effective way to manage hyperglycaemia or induce remission in individuals with both obesity and diabetes.
Collapse
|
38
|
Salman MA, Abelsalam A, Nashed GA, Yacoub M, Abdalla A. Long Biliopancreatic Limb Roux-En-Y Gastric Bypass Versus One-Anastomosis Gastric Bypass: a Randomized Controlled Study. Obes Surg 2023; 33:1966-1973. [PMID: 37178225 PMCID: PMC10289940 DOI: 10.1007/s11695-023-06631-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 04/29/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND Roux-en-Y gastric bypass (RYGB) is the gold standard in bariatric surgery. The one-anastomosis gastric bypass (OAGB) procedure, first introduced by Dr. Rutledge, has demonstrated a 25% greater weight loss efficiency than the traditional Roux-en-Y gastric bypass (RYGB) procedure due to the substantially longer biliopancreatic limb (BPL). AIM OF THE STUDY The current work aimed to compare the outcomes of OAGB and long BPL RYGB regarding weight loss and comorbidity resolution. PATIENTS AND METHODS This randomized controlled trial was done at our institution between September 2019 and January 2021. Patients who were candidates for bariatric surgery were randomly and equally allocated to two groups. Group A underwent OAGB, while group B underwent long BPL RYGB. Patients were followed up for 6 months postoperatively. RESULTS This study included 62 patients equally allocated to OAGB or long BPL RYGB, with no dropouts during follow-up. At 6 months, there was no statistically significant difference between the two groups regarding postoperative BMI (P = 0.313) and the EBWL (P = 0.238). There was comparable remission of diabetes mellitus (P = 0.708), hypertension (P = 0.999), OSA (P = 0.999), joint pain (P = 0.999), and low back pain (P = 0.999). Seven patients in the OAGB group experienced reflux symptoms (P = 0.011), which were managed by proton pump inhibitors. CONCLUSION Extending the BPL in RYGB provides weight loss and comorbidity remission comparable to that of OAGB. Some OAGB-related reflux cases remain a concern. However, they were sufficiently controlled with PPIs. Due to OAGB superior technical simplicity, long BPL RYGB should be preserved for cases whom are more risky for bile reflux.
Collapse
|
39
|
Lammert M, Medawar E, Hartmann H, Grasser L, Dietrich A, Fenske W, Horstmann A. Distinct adaptations of endocrine and cognitive functions may contribute to high variability in long-term weight loss outcome after bariatric surgery. Physiol Behav 2023:114279. [PMID: 37356514 DOI: 10.1016/j.physbeh.2023.114279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 05/08/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023]
Abstract
BACKGROUND Bariatric surgery has been widely recognized as the most efficient long-term treatment method in severe obesity, yet therapy success shows considerable interindividual variability. Postoperative metabolic adaptations, including improved gut hormone secretion (GLP-1, PYY and ghrelin), and restored executive function may play an explanatory role in weight loss, yet causes for poor success in individual patients remain unknown. This study investigates gut-hormonal and cognitive characteristics in extreme weight loss responders to bariatric surgery. METHODS Patients (n=47) with high or low excessive weight loss (EWL) at least 2 years after Roux-en-Y-gastric bypass or sleeve gastrectomy were allocated into good responders (GR, EWL 82.4 ± 11.6%) and poor responders (PR, EWL 24.0 ± SD 12.8%) to study differences in postprandial secretion of GLP-1, PYY, ghrelin and in working memory (WM). RESULTS Mean BMI was 47.1 ± 6.2 kg/m² in PR (n=21) and 28.9 ± 3.1 kg/m² in GR (n=26, p < 0.001). Fasted GLP-1 and PYY were comparable for GR and PR (p > 0.2) and increased strongly after a standardized test meal (300 kcal liquid meal) with a peak at 15 to 30 minutes. The increase was stronger in GR compared to PR (GLP-1, PYY: Time x Group p < 0.05). Plasma ghrelin levels already differed between groups at fasted state, showing significantly higher levels for GR (p < 0.05). Postprandially, ghrelin secretion was suppressed in both groups, but suppression was higher in GR (Time x Group p < 0.05). GR showed significantly higher WM scores than PR (p < 0.05). Postprandial ghrelin (iAUC), but not GLP-1 or PYY plasma levels, significantly mediated the relationship between EWL and a WM subscore (IS score, CI = 0.07 - 1.68), but not WM main score (MIS score, CI = -0.07 - 1.54), in mediation analyses. CONCLUSION Excess weight loss success after bariatric surgical procedures is associated with distinct profiles of gut-hormones at fasted and postprandial state, and differences in working memory. Better working memory performance in GR might be mediated by higher postprandial reduction in ghrelin plasma levels. Future studies need to integrate longitudinal data, larger samples and more sensitive cognitive tests.
Collapse
Affiliation(s)
- Mathis Lammert
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, 04103 Leipzig, Germany; Leipzig University Medical Centre, IFB Adiposity Diseases, 04103 Leipzig, Germany; Leipzig University Medical Centre, Collaborative Research Centre 1052-A5, 04103 Leipzig, Germany.
| | - Evelyn Medawar
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, 04103 Leipzig, Germany.
| | - Hendrik Hartmann
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, 04103 Leipzig, Germany; Leipzig University Medical Centre, Collaborative Research Centre 1052-A5, 04103 Leipzig, Germany; Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Linda Grasser
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, 04103 Leipzig, Germany; Leipzig University Medical Centre, IFB Adiposity Diseases, 04103 Leipzig, Germany.
| | - Arne Dietrich
- Department of Obesity, Metabolic and Endocrine Surgery, University Hospital Leipzig, Liebigstraße 18, 04103 Leipzig, Germany.
| | - Wiebke Fenske
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany.
| | - Annette Horstmann
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, 04103 Leipzig, Germany; Leipzig University Medical Centre, IFB Adiposity Diseases, 04103 Leipzig, Germany; Leipzig University Medical Centre, Collaborative Research Centre 1052-A5, 04103 Leipzig, Germany; Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| |
Collapse
|
40
|
Lim JJ, Liu Y, Lu LW, Sequeira IR, Poppitt SD. No Evidence That Circulating GLP-1 or PYY Are Associated with Increased Satiety during Low Energy Diet-Induced Weight Loss: Modelling Biomarkers of Appetite. Nutrients 2023; 15:nu15102399. [PMID: 37242282 DOI: 10.3390/nu15102399] [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: 02/27/2023] [Revised: 04/04/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Bariatric surgery and pharmacology treatments increase circulating glucagon-like peptide-1 (GLP-1) and peptide YY (PYY), in turn promoting satiety and body weight (BW) loss. However, the utility of GLP-1 and PYY in predicting appetite response during dietary interventions remains unsubstantiated. This study investigated whether the decrease in hunger observed following low energy diet (LED)-induced weight loss was associated with increased circulating 'satiety peptides', and/or associated changes in glucose, glucoregulatory peptides or amino acids (AAs). In total, 121 women with obesity underwent an 8-week LED intervention, of which 32 completed an appetite assessment via a preload challenge at both Week 0 and Week 8, and are reported here. Visual analogue scales (VAS) were administered to assess appetite-related responses, and blood samples were collected over 210 min post-preload. The area under the curve (AUC0-210), incremental AUC (iAUC0-210), and change from Week 0 to Week 8 (∆) were calculated. Multiple linear regression was used to test the association between VAS-appetite responses and blood biomarkers. Mean (±SEM) BW loss was 8.4 ± 0.5 kg (-8%). Unexpectedly, the decrease in ∆AUC0-210 hunger was best associated with decreased ∆AUC0-210 GLP-1, GIP, and valine (p < 0.05, all), and increased ∆AUC0-210 glycine and proline (p < 0.05, both). The majority of associations remained significant after adjusting for BW and fat-free mass loss. There was no evidence that changes in circulating GLP-1 or PYY were predictive of changes in appetite-related responses. The modelling suggested that other putative blood biomarkers of appetite, such as AAs, should be further investigated in future larger longitudinal dietary studies.
Collapse
Affiliation(s)
- Jia Jiet Lim
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland 1024, New Zealand
- Riddet Institute, Palmerston North 4442, New Zealand
| | - Yutong Liu
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland 1024, New Zealand
- Department of Medicine, University of Auckland, Auckland 1010, New Zealand
| | - Louise W Lu
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland 1024, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1010, New Zealand
| | - Ivana R Sequeira
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland 1024, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1010, New Zealand
| | - Sally D Poppitt
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland 1024, New Zealand
- Riddet Institute, Palmerston North 4442, New Zealand
- Department of Medicine, University of Auckland, Auckland 1010, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1010, New Zealand
| |
Collapse
|
41
|
Brzozowska MM, Isaacs M, Bliuc D, Baldock PA, Eisman JA, White CP, Greenfield JR, Center JR. Effects of bariatric surgery and dietary intervention on insulin resistance and appetite hormones over a 3 year period. Sci Rep 2023; 13:6032. [PMID: 37055514 PMCID: PMC10102182 DOI: 10.1038/s41598-023-33317-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 04/11/2023] [Indexed: 04/15/2023] Open
Abstract
To examine an impact of three types of bariatric surgery compared with dietary intervention (DIET), on concurrent changes in Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) and appetite hormones over 3 years. Fifty-five adults were studied during phase of weight loss (0-12 months) and during weight stability (12-36 months) post intervention. Measurements of HOMA-IR, fasting and postprandial PYY and GLP1, adiponectin, CRP, RBP4, FGF21 hormones and dual-Xray absorptiometry were performed throughout the study. All surgical groups achieved significant reductions in HOMA-IR with greatest difference between Roux-en-Y gastric bypass and DIET (- 3.7; 95% CI - 5.4, - 2.1; p = 0.001) at 12-36 months. Initial (0-12 months) HOMA-IR values were no different to DIET after adjustment for the lost weight. During 12-36 months, after controlling for treatment procedure and weight, for every twofold increase in postprandial PYY and adiponectin, HOMA-IR decreased by 0.91 (95% CI - 1.71, - 0.11; p = 0.030) and by 0.59 (95% CI - 1.10, - 0.10; p = 0.023) respectively. Initial, non-sustained changes in RBP4 and FGF21 were not associated with HOMA-IR values. While initial rapid weight loss reduces insulin resistance, the enhanced secretions of PYY and adiponectin may contribute to weight-independent improvements in HOMA-IR during weight stability.Clinical trial registration: Australian New Zealand Clinical Trials Registry (ANZCTR): ACTRN12613000188730.
Collapse
Affiliation(s)
- Malgorzata M Brzozowska
- Endocrinology, The Sutherland Hospital, Caringbah, Australia.
- Faculty of Medicine, UNSW Sydney, Sydney, Australia.
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia.
| | - Michelle Isaacs
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Endocrinology, St Vincent's Hospital Sydney, Darlinghurst, Australia
| | - Dana Bliuc
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia
| | - Paul A Baldock
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia
- School of Medicine, The University of Notre Dame Australia, Darlinghurst, Australia
| | - John A Eisman
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia
- Endocrinology, St Vincent's Hospital Sydney, Darlinghurst, Australia
- School of Medicine, The University of Notre Dame Australia, Darlinghurst, Australia
| | - Chris P White
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Prince of Wales Hospital, NSW Health Pathology, Randwick, Australia
- Endocrinology, Prince of Wales Hospital, Randwick, Australia
| | - Jerry R Greenfield
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia
- Endocrinology, St Vincent's Hospital Sydney, Darlinghurst, Australia
| | - Jacqueline R Center
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia
- Endocrinology, St Vincent's Hospital Sydney, Darlinghurst, Australia
- School of Medicine, The University of Notre Dame Australia, Darlinghurst, Australia
| |
Collapse
|
42
|
Elhag W, Lock M, El Ansari W. When Definitions Differ, are Comparisons Meaningful? Definitions of Weight Regain After Bariatric Surgery and Their Associations with Patient Characteristics and Clinical Outcomes - A Need for a Revisit? Obes Surg 2023; 33:1390-1400. [PMID: 36995562 PMCID: PMC10156838 DOI: 10.1007/s11695-023-06528-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/17/2023] [Accepted: 02/23/2023] [Indexed: 03/31/2023]
Abstract
INTRODUCTION Definitions and prevalence of weight regain (WR) after bariatric surgery remains inconsistent and their clinical significance unclear. OBJECTIVES To assess WR five years after sleeve gastrectomy (LSG), employing six definitions; and appraise their association with patient characteristics/clinical outcomes. METHODS Consecutive patients (N = 589) who underwent LSG were followed up for 5 years. WR prevalence was calculated yearly employing six definitions. Regression analysis assessed associations between WR at 5 years, and patient characteristics (age, sex, preop BMI, number of follow-up visits, number of comorbidities) and remission of comorbidities (type 2 diabetes, hypertension, and dyslipidemia). RESULTS Sample's mean age and BMI were 34 ± 11.6 years and 43.13 ± 5.77 kg/m2, and 64% were females. Percentage of patients with WR at 2, 3, 4, and 5 years fluctuated between 2.53% and 94.18%, subject to definition, and time point. The definition "Any WR" generated the highest prevalence of WR (86-94%) across all time points. At 5 years, for patient characteristics, preoperative BMI was associated with three definitions (P 0.49 to < 0.001), sex was associated with two (P < 0.026-0.032), and number of comorbidities was associated with one definition (P = 0.01). In terms of comorbidities, only hypertension was associated with WR (one definition, P = 0.025). No other definitions of WR were associated with any of the variables under examination. CONCLUSION Weight regain is reasonably expected after BMS. WR definitions were of minor clinical significance due to weak associations with limited comorbidities. Dichotomous definitions might offer some guidance while managing individual patients. However, its utility as a comparator metric across patients/procedures requires refinements.
Collapse
Affiliation(s)
- Wahiba Elhag
- Department of Bariatric Surgery/Bariatric Medicine, Hamad Medical Corporation, Doha, Qatar
- Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Merilyn Lock
- Division of Exercise Science, Health and Epidemiology, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Walid El Ansari
- Weill Cornell Medicine-Qatar, Doha, Qatar.
- Department of Surgery, Hamad Medical Corporation, Doha, Qatar.
- College of Medicine, Qatar University, Doha, Qatar.
| |
Collapse
|
43
|
Brethvad AO, Zakariassen HL, Holt J, Lundgren JR, Jakobsen A, Hartmann B, Lehmann EW, Kissow H, Holst JJ, Madsbad S, Torekov SS, Holst B. Increased meal-induced neurotensin response predicts successful maintenance of weight loss - Data from a randomized controlled trial. Metabolism 2023; 143:155534. [PMID: 36933790 DOI: 10.1016/j.metabol.2023.155534] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/28/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND The gut derived anorexigenic hormone neurotensin (NT) is upregulated after bariatric surgery which may contribute to the sustained weight loss. In contrast, diet-induced weight loss is most often followed by weight regain. We therefore investigated whether diet-induced weight loss impacts levels of circulating NT in mice and humans and whether NT levels predicts body weight change after weight loss in humans. METHODS In vivo mice study: Obese mice were fed ad-libitum or a restricted diet (40-60 % of average food intake) for 9 days to obtain similar weight loss as observed in the human study. At termination, intestinal segments, the hypothalamus and plasma were collected for histological, real time PCR, and radioimmunoassay (RIA) analysis. CLINICAL TRIAL Plasma samples from 42 participants with obesity, completing an 8-week low-calorie diet in a randomized controlled trial, were analyzed. Plasma NT was measured by RIA at fasting and during a meal test before and after diet-induced weight loss and after one year of intended weight maintenance. RESULTS In obese mice, food restriction-induced body weight loss of 14 % was associated with a 64 % reduction in fasting plasma NT (p < 0.0001). In the mouse duodenum (p = 0.07) and jejunum (p < 0.05), NT tissue concentration was decreased without tissue atrophy indicative of a physiological downregulation. In the mouse hypothalamus a downregulation of Pomc (p < 0.01) along with upregulation of Npy (p < 0.001) and Agrp (p < 0.0001) expression was found after restricted feeding in support of increased hunger after diet-induced weight loss. Therefore, we investigated the NT response in humans undergoing weight loss maintenance. In humans, similar to the mice, the low-calorie diet induced weight loss of 13 % body weight was associated with 40 % reduction in fasting plasma NT levels (p < 0.001). Meal-induced NT peak responses were greater in humans who lost additional weight during the 1 year maintenance phase compared to participants who regained weight (p < 0.05). CONCLUSION Diet-induced weight loss decreased fasting plasma NT levels in both humans and mice with obesity, and regulated hunger-associated hypothalamic gene expression in mice. Meal-induced NT responses were greater in humans who lost additional weight during the 1 year maintenance phase compared to participants who regained weight. This indicates that increased peak secretion of NT after weight loss may contribute to successful maintenance of weight loss. CLINICAL TRIAL REGISTRATION NUMBER NCT02094183.
Collapse
Affiliation(s)
| | - Hannah Louise Zakariassen
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 12.4., 2200 Copenhagen, Denmark.
| | - Joachim Holt
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 12.4., 2200 Copenhagen, Denmark
| | - Julie Rehné Lundgren
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 12.4., 2200 Copenhagen, Denmark
| | - Alexander Jakobsen
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 12.4., 2200 Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 12.4., 2200 Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Eva Winning Lehmann
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 12.4., 2200 Copenhagen, Denmark
| | - Hannelouise Kissow
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 12.4., 2200 Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 12.4., 2200 Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Hvidovre University Hospital, Hvidovre, Denmark
| | - Signe Sørensen Torekov
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 12.4., 2200 Copenhagen, Denmark.
| | - Birgitte Holst
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 12.4., 2200 Copenhagen, Denmark
| |
Collapse
|
44
|
Unhapipatpong C, Hiranyatheb P, Phanachet P, Warodomwichit D, Sriphrapradang C, Shantavasinkul PC. Postprandial hypoglycemia after ileocolic interposition and Billroth-II gastrojejunostomy: A case report. Clin Case Rep 2023; 11:e7060. [PMID: 36950668 PMCID: PMC10025248 DOI: 10.1002/ccr3.7060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/03/2023] [Accepted: 02/22/2023] [Indexed: 03/24/2023] Open
Abstract
Postprandial reactive hypoglycemia, or late dumping syndrome, is a common but underrecognized complication from bypass surgery. We report an unusual case of postprandial reactive hypoglycemia in a patient with a severe esophageal stricture from corrosive agent ingestion who underwent ileocolic interposition and an antecolic Billroth-II gastrojejunostomy. A 22-year-old male patient with a one-year history of corrosive ingestion was referred to the hospital for a surgical correction of severe esophageal stricture. After the patient underwent ileocolic interposition and an antecolic Billroth-II gastrojejunostomy, he experienced multiple episodes of gastroesophageal refluxsymptoms during nasogastric feeding and had onset of hypoglycemic symptoms. His plasma glucose level was 59 mg/dL. After we had intraoperatively re-inserted a jejunostomy tube bypassing the ileocolic interposition, and reintroduced enteral nutrition, his hypoglycemic symptoms resolved. We performed a mixed meal tolerance test by nasogastric tube, but the results did not show postprandial hypoglycemia. Although the specific mechanism is unclear, this case suggests gastroesophageal reflux to the ileal interposition may have caused a state of exaggerated hyperinsulinemic response and rebound hypoglycemia. To the best of our knowledge, we are the first to report case of postprandial hypoglycemia after ileocolic interposition, which may have been caused by exaggerated hyperinsulinemic response due to gastroesophageal reflux to the ileal interposition. This syndrome should be considered in the patient who has had ileocolic interposition surgery and has developed postprandial hypoglycemia.
Collapse
Affiliation(s)
- Chanita Unhapipatpong
- Department of Medicine, Division of Clinical NutritionKhon Kaen HospitalKhon KaenThailand
| | - Pitichote Hiranyatheb
- Department of Surgery, Division of General Surgery, Faculty of Medicine Ramathibodi HospitalMahidol UniversityBangkokThailand
| | - Pariya Phanachet
- Department of Medicine, Division of Nutrition and Biochemical Medicine, Faculty of Medicine Ramathibodi HospitalMahidol UniversityBangkokThailand
| | - Daruneewan Warodomwichit
- Department of Medicine, Division of Nutrition and Biochemical Medicine, Faculty of Medicine Ramathibodi HospitalMahidol UniversityBangkokThailand
| | - Chutintorn Sriphrapradang
- Department of Medicine, Division of Endocrinology and Metabolism, Faculty of Medicine Ramathibodi HospitalMahidol UniversityBangkokThailand
| | - Prapimporn Chattranukulchai Shantavasinkul
- Department of Medicine, Division of Nutrition and Biochemical Medicine, Faculty of Medicine Ramathibodi HospitalMahidol UniversityBangkokThailand
- Graduate Program in Nutrition, Faculty of Medicine Ramathibodi HospitalMahidol UniversityBangkokThailand
| |
Collapse
|
45
|
Alabduljabbar K, Al-Najim W, le Roux CW. Food preferences after bariatric surgery: a review update. Intern Emerg Med 2023; 18:351-358. [PMID: 36478323 DOI: 10.1007/s11739-022-03157-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/20/2022] [Indexed: 12/13/2022]
Abstract
Obesity is a serious and global health problem. The multiple complications of obesity reduce quality of life and increase mortality. Bariatric surgery is one of the best treatment options for obesity management. Bariatric surgery helps people reduce their caloric intake by treating the disease of obesity effectively, in part by increasing signaling from the gut to the brain. The most frequent surgical options are Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG). There is controversy regarding changes in food preferences and selection after bariatric surgery. In this review, we aim to outline the changes in food intake and selection, clarify the behavior changes in food intake, and assess the potential mechanisms responsible for these changes in patients after bariatric surgery.
Collapse
Affiliation(s)
- Khaled Alabduljabbar
- Department of Family Medicine and Polyclinics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Dublin, Ireland
| | - Werd Al-Najim
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Dublin, Ireland
| | - Carel W le Roux
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Dublin, Ireland.
| |
Collapse
|
46
|
Association Between Gut Hormones and Weight Change After Bariatric Arterial Embolization: Results from the BEAT Obesity Trial. Cardiovasc Intervent Radiol 2023; 46:220-228. [PMID: 36138191 DOI: 10.1007/s00270-022-03280-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 09/02/2022] [Indexed: 02/04/2023]
Abstract
PURPOSE To evaluate associations of ghrelin, glucagon-like peptide 1 (GLP-1), and peptide YY 3-36 (PYY3-36) with weight change after bariatric arterial embolization (BAE). MATERIALS AND METHODS Subgroup analysis of data collected during the BEAT Obesity Trial involving 7 participants with BMI > 40 who were embolized with 300- to 500-μm Embosphere Microspheres. Three participants were characterized as "responders" (top tertile of weight loss at each visit) and 4 as "non-responders" (bottom tertile of weight loss at each visit). Mean ± standard deviation participant age was 44 ± 11 years, and 6 of 7 participants were women. Participants were evaluated at baseline, 2 weeks, and 1, 3, 6, and 12 months after BAE. After fasting, participants consumed a mixed meal test at each visit; blood samples were collected at 0, 15, 30, 60, 120, 180, and 240 min. Study outcome measures were changes in weight from baseline and plasma serum hormone levels. RESULTS Percentage change in ghrelin decreased significantly in non-responders at 60 and 120 min at 1 and 12 months (estimated difference between 60 vs. 0 min at 1 month: 69% [95% CI - 126%, - 13%]; estimated difference between 120 vs. 0 min at 12 months: - 131% (95% CI - 239%, - 23%]). Responders had significantly lower ghrelin and greater weight loss than non-responders at 6 and 12 months. GLP-1 and PYY3-36 levels did not differ between groups. CONCLUSION Participants with consistent weight loss throughout follow-up had lower ghrelin than non-responders, supporting decreased ghrelin as a mechanism underlying BAE-induced weight loss. LEVEL OF EVIDENCE I High-quality randomized trial or prospective study; testing of previously developed diagnostic criteria on consecutive patients; sensible costs and alternatives; values obtained from many studies with multiway sensitivity analyses; systematic review of Level I RCTs and Level I studies.
Collapse
|
47
|
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: 18] [Impact Index Per Article: 9.0] [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
|
48
|
Patel D, Borrelli N, Patey O, Johnson M, DI Salvo G, Savvidou MD. Effect of bariatric surgery on maternal cardiovascular system. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2023; 61:207-214. [PMID: 36722427 PMCID: PMC10107918 DOI: 10.1002/uog.26042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/12/2022] [Accepted: 07/14/2022] [Indexed: 05/27/2023]
Abstract
OBJECTIVE Bariatric surgery is a successful treatment for sustainable weight loss and has been associated with improvement in cardiovascular function. Pregnancy after bariatric surgery is becoming increasingly common; however, little is known about the maternal cardiovascular system postsurgery. The aim of this study was to investigate maternal cardiovascular adaptation to pregnancy in women with previous bariatric surgery, compared with that in women with no history of weight-loss surgery and an early-pregnancy body mass index (BMI) similar to the presurgery BMI of the postbariatric women. METHODS This was a prospective, observational, longitudinal study conducted from April 2018 to June 2020 including 30 pregnant women who had undergone bariatric surgery and 30 who had not, matched for presurgery BMI. Participants were seen at three timepoints during pregnancy: 12-14, 20-24 and 30-32 weeks' gestation. At all visits, maternal blood pressure (BP) was measured and cardiac geometry and function were assessed using two-dimensional (2D) transthoracic echocardiography. On a subset of patients (15 in each group), 2D speckle tracking was performed to assess global longitudinal and circumferential strain. Offline analysis was performed, and multilevel linear mixed-effects models were used for all comparisons. RESULTS Compared with the no-surgery group, and across all trimesters, pregnant women with previous bariatric surgery had lower BP, heart rate and cardiac output and higher peripheral vascular resistance (P < 0.01 for all). Similarly, the postbariatric group demonstrated more favorable cardiac geometry and diastolic indices, including lower left ventricular mass, left atrial volume and relative wall thickness, together with higher E-wave/A-wave flow velocity across the mitral valve and higher mitral velocity (E') at the lateral and medial annulus on tissue Doppler imaging (P < 0.01 for all). There was no difference in ejection fraction, although global longitudinal strain was lower in postbariatric women (P < 0.01), indicating better systolic function. CONCLUSION Our findings indicate better maternal cardiovascular adaptation in women with previous bariatric surgery compared with presurgery BMI-matched pregnant women with no history of weight-loss surgery. © 2022 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- D. Patel
- Academic Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital, Department of MetabolismDigestion and Reproduction, Imperial College LondonLondonUK
| | - N. Borrelli
- Royal Brompton Hospital, Royal Brompton and Harefield NHS Foundation TrustLondonUK
| | - O. Patey
- Royal Brompton Hospital, Royal Brompton and Harefield NHS Foundation TrustLondonUK
| | - M. Johnson
- Academic Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital, Department of MetabolismDigestion and Reproduction, Imperial College LondonLondonUK
| | - G. DI Salvo
- Royal Brompton Hospital, Royal Brompton and Harefield NHS Foundation TrustLondonUK
| | - M. D. Savvidou
- Academic Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital, Department of MetabolismDigestion and Reproduction, Imperial College LondonLondonUK
- Fetal Medicine Unit, Chelsea and Westminster HospitalLondonUK
| |
Collapse
|
49
|
Ebadinejad A, Shahshahani M, Hosseinpanah F, Ghazy F, Khalaj A, Mahdavi M, Valizadeh M, Barzin M. Comparison of hypertension remission and relapse after sleeve gastrectomy and one-anastomosis gastric bypass: a prospective cohort study. Hypertens Res 2023; 46:1287-1296. [PMID: 36670229 DOI: 10.1038/s41440-023-01180-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 11/27/2022] [Accepted: 12/28/2022] [Indexed: 01/21/2023]
Abstract
This study aimed to assess the remission and relapse of hypertension (HTN) in hypertensive individuals who underwent sleeve gastrectomy (SG) and one-anastomosis gastric bypass (OAGB) and identify the predictors of HTN remission and relapse following bariatric surgery. A prospective cohort study with a follow-up of 3 years was conducted on severely obese patients between 2013 and 2018. Hypertension remission was defined as the normalization of blood pressure (BP) with the discontinuation of medical treatment, and HTN relapse was defined as the need for the onset of antihypertensive drugs or the occurrence of BP impairment. A total of 787 hypertensive patients were included in this study. The cumulative incidence of HTN remission and relapse were 83.9% (95% CI: 81.6-86.5) and 31.4% (95% CI: 25.6-38.2), respectively. Remission and relapse were not significantly different among the patients undergoing either surgery (SG or OAGB). A higher remission rate was linked to a younger age and the use of fewer antihypertensive medications pre-operation. However, failure to successfully lose weight during the first year postoperative and weight regain predicted a higher risk of HTN relapse after 3 years. Following bariatric surgery, BP drops initially but then gradually rises. These alterations are responsible for about 31% relapse after 3 years in those who initially achieve remission. Patients who are younger and use less antihypertensive medications before surgery may benefit the most from bariatric surgery in terms of HTN. First-year successful weight loss and control of weight regain may prevent HTN relapse in the following years.
Collapse
Affiliation(s)
- Amir Ebadinejad
- Obesity Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahbod Shahshahani
- Obesity Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farhad Hosseinpanah
- Obesity Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faranak Ghazy
- Obesity Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Khalaj
- Tehran Obesity Treatment Center, Department of Surgery, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Maryam Mahdavi
- Obesity Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Valizadeh
- Obesity Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Barzin
- Obesity Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
50
|
Feris F, McRae A, Kellogg TA, McKenzie T, Ghanem O, Acosta A. Mucosal and hormonal adaptations after Roux-en-Y gastric bypass. Surg Obes Relat Dis 2023; 19:37-49. [PMID: 36243547 PMCID: PMC9797451 DOI: 10.1016/j.soard.2022.08.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 01/12/2023]
Abstract
The aim of this study was to perform a comprehensive literature review regarding the relevant hormonal and histologic changes observed after Roux-en-Y gastric bypass (RYGB). We aimed to describe the relevant hormonal (glucagon-like peptides 1 and 2 [GLP-1 and GLP-2], peptide YY [PYY], oxyntomodulin [OXM], bile acids [BA], cholecystokinin [CCK], ghrelin, glucagon, gastric inhibitory polypeptide [GIP], and amylin) profiles, as well as the histologic (mucosal cellular) adaptations happening after patients undergo RYGB. Our review compiles the current evidence and furthers the understanding of the rationale behind the food intake regulatory adaptations occurring after RYGB surgery. We identify gaps in the literature where the potential for future investigations and therapeutics may lie. We performed a comprehensive database search without language restrictions looking for RYGB bariatric surgery outcomes in patients with pre- and postoperative blood work hormonal profiling and/or gut mucosal biopsies. We gathered the relevant study results and describe them in this review. Where human findings were lacking, we included animal model studies. The amalgamation of physiologic, metabolic, and cellular adaptations following RYGB is yet to be fully characterized. This constitutes a fundamental aspiration for enhancing and individualizing obesity therapy.
Collapse
Affiliation(s)
- Fauzi Feris
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Alison McRae
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Todd A Kellogg
- Division of Endocrine and Metabolic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Travis McKenzie
- Division of Endocrine and Metabolic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Omar Ghanem
- Division of Endocrine and Metabolic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|