Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Am J Physiol Endocrinol Metab ; 325(5): E540-E551, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37755455

RESUMEN

Postprandial hypoglycemia is a complication of Roux-en-Y gastric bypass (RYGB), but the effects of postprandial exercise and meal glycemic index (GI) on postprandial glucose and glucoregulatory hormone responses are unknown. Ten RYGB-operated and 10 age and weight-matched unoperated women completed four test days in random order ingesting mixed meals with high GI (HGI, GI = 93) or low GI (LGI, GI = 54), but matched on energy and macronutrient content. Ten minutes after meal completion, participants rested or cycled for 30 min at 70% of maximum oxygen uptake (V̇o2max). Blood was collected for 4 h. Postprandial exercise did not lower plasma nadir glucose in RYGB after HGI (HGI/rest 3.7 ± 0.5 vs. HGI/Ex 4.1 ± 0.4 mmol/L, P = 0.070). Replacing HGI with LGI meals raised glucose nadir in RYGB (LGI/rest 4.1 ± 0.5 mmol/L, P = 0.034) and reduced glucose excursions (Δpeak-nadir) but less so in RYGB (-14% [95% CI: -27; -1]) compared with controls (-33% [-51; -14]). Insulin responses mirrored glucose concentrations. Glucagon-like peptide 1 (GLP-1) responses were greater in RYGB versus controls, and higher with HGI versus LGI. Glucose-dependent insulinotropic polypeptide (GIP) responses were greater after HGI versus LGI in both groups. Postexercise glucagon responses were lower in RYGB than controls, and noradrenaline responses tended to be lower in RYGB, whereas adrenaline responses were similar between groups. In conclusion, moderate intensity cycling shortly after meal intake did not increase the risk of postprandial hypoglycemia after RYGB. The low GI meal increased nadir glucose and reduced glucose excursions compared with the high GI meal. RYGB participants had lower postexercise glucagon responses compared with controls.NEW & NOTEWORTHY We investigate the effect of moderate exercise after a high or a low glycemic index meal on nadir glucose and glucoregulatory hormones in gastric bypass-operated individuals and in matched unoperated controls. Cycling shortly after meal intake did not increase the risk of hypoglycemia in operated individuals. The low glycemic index meal increased glucose nadir and reduced excursions compared with the high glycemic index meal. Operated individuals had lower postexercise glucagon responses compared with controls.


Asunto(s)
Derivación Gástrica , Hipoglucemia , Humanos , Femenino , Índice Glucémico , Glucemia , Glucagón/metabolismo , Consumo de Oxígeno , Oxígeno , Insulina , Comidas , Glucosa , Periodo Posprandial
2.
Diabetes ; 72(3): 336-347, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36478039

RESUMEN

Enhanced secretion of glucagon-like peptide 1 (GLP-1) seems to be essential for improved postprandial ß-cell function after Roux-en-Y gastric bypass (RYGB) but is less studied after sleeve gastrectomy (SG). Moreover, the role of the other major incretin hormone, glucose-dependent insulinotropic polypeptide (GIP), is relatively unexplored after bariatric surgery. We studied the effects of separate and combined GLP-1 receptor (GLP-1R) and GIP receptor (GIPR) blockade during mixed-meal tests in unoperated (CON), SG-operated, and RYGB-operated people with no history of diabetes. Postprandial GLP-1 concentrations were highest after RYGB but also higher after SG compared with CON. In contrast, postprandial GIP concentrations were lowest after RYGB. The effect of GLP-1R versus GIPR blockade differed between groups. GLP-1R blockade reduced ß-cell glucose sensitivity and increased or tended to increase postprandial glucose responses in the surgical groups but had no effect in CON. GIPR blockade reduced ß-cell glucose sensitivity and increased or tended to increase postprandial glucose responses in the CON and SG groups but had no effect in the RYGB group. Our results support that GIP is the most important incretin hormone in unoperated people, whereas GLP-1 and GIP are equally important after SG, and GLP-1 is the most important incretin hormone after RYGB.


Asunto(s)
Derivación Gástrica , Péptido 1 Similar al Glucagón , Humanos , Derivación Gástrica/métodos , Incretinas , Insulina , Glucemia , Polipéptido Inhibidor Gástrico , Glucosa , Gastrectomía/métodos
3.
Front Nutr ; 9: 889710, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35571890

RESUMEN

Background and aims: The metabolic consequences after Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG) are often studied using a liquid mixed meal. However, liquid meals may not be representative of the patients' everyday diet. We therefore examined postprandial glucose and gut hormone responses using mixed meals differing only with respect to meal texture. Methods: Twelve RYGB-operated, 12 SG-operated, and 12 unoperated individuals (controls) were enrolled in the study. Participants were matched on age, sex, and body mass index. In randomized order, each participant underwent a liquid and a solid 4-h mixed meal test on separate days. The meals were isocaloric (309 kcal), and with identical macronutrient composition (47 E% carbohydrate, 18 E% protein, 32 E% fat, and 3 E% dietary fibers). The liquid meal was blended to create a smooth liquid texture while the other meal retained its solid components. Results: Postprandial glucose concentrations (peak and incremental area under curve, iAUC) did not differ between the two meal textures in any group. In the control group, peak C-peptide was higher after the liquid meal compared with the solid meal (p = 0.04), whereas iAUCs of C-peptide were similar between the two meals in all groups. Peak of glucagon-like peptide-1 (GLP-1) was higher after the liquid meal compared with the solid meal in RYGB- and SG-operated individuals (RYGB p = 0.02; SG p < 0.01), but iAUC of GLP-1 did not differ between meal textures within any group. Peak of glucose-dependent insulin tropic polypeptide (GIP) was higher after the liquid meal in the SG and control groups (SG p = 0.02; controls p < 0.01), but iAUCs of GIP were equal between meals. There were no differences in total AUC of ghrelin between the liquid and solid meals within any of the groups. Conclusion: A liquid and a solid meal with identical macronutrient composition result in similar postprandial glucose responses, both in operated and unoperated individuals. Small differences were observed for the postprandial peaks of C-peptide, GLP-1, and GIP concentrations. Overall, a liquid meal is suitable for evaluating glucose tolerance, ß-cell function, and gut hormones responses, both after RYGB and SG and in unoperated individuals. Clinical Trial Registration: [www.clinicaltrials.gov], identifier [NCT04082923].

4.
Obes Surg ; 32(4): 998-1004, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35083702

RESUMEN

PURPOSE: We aimed to estimate the total cost of bariatric surgery in Denmark. MATERIALS AND METHODS: The study population included all Danish citizens ≥ 18 years who had received bariatric surgery, identified in the Danish National Patient Register in the period from 2002 to 2018. Patients who had received bariatric surgery were matched with three controls on gender, year of birth, and region of residence. A difference-in-difference approach was used to estimate the healthcare costs attributable to bariatric surgery from 3 years before to 5 years after surgery. RESULTS: Total healthcare costs for cases receiving bariatric surgery during the first 5 years following surgery amounted to EUR 32,899, and EUR 16,651 for their matched controls. Thereby, the difference in total healthcare costs (EUR 16,248) between persons receiving bariatric surgery and their matched controls was 2.2 times the DRG rate for the surgery itself (EUR 7387). Moreover, the results suggest that receiving bariatric surgery led to a total increase in gross earnings of EUR 5970 (5%) and a total reduction in receipt of transfer payments of EUR 4488 (12%) in the period up until 5 years after surgery. CONCLUSION: The results showed a significant and persistent increase in healthcare costs for people with obesity receiving bariatric surgery during the first 5 years after surgery. We also found that bariatric surgery was associated with increased attachment to the labor market.


Asunto(s)
Cirugía Bariátrica , Obesidad Mórbida , Costos de la Atención en Salud , Humanos , Renta , Obesidad/cirugía , Obesidad Mórbida/cirugía
5.
Neurogastroenterol Motil ; 34(1): e14210, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34378827

RESUMEN

OBJECTIVE: Neurotensin (NT) is released from enteroendocrine cells and lowers food intake in rodents. We evaluated postprandial NT secretion in humans after surgeries associated with accelerated small intestinal nutrient delivery, and after Roux-en-Y gastric bypass (RYGB) when glucagon-like peptide-1 (GLP-1) signalling and dipeptidyl peptidase 4 (DPP-4) were inhibited, and during pharmacological treatments influencing entero-pancreatic functions. METHODS: We measured NT concentrations in plasma from meal studies: (I) after truncal vagotomy with pyloroplasty (TVP), cardia resection +TVP (CTVP), and matched controls (n = 10); (II) after RYGB, sleeve gastrectomy (SG), and in matched controls (n = 12); (III) after RYGB (n = 11) with antagonism of GLP-1 signalling using exendin(9-39) and DPP-4 inhibition using sitagliptin; (IV) after RYGB (n = 11) during a run-in period and subsequent treatment with, sitagliptin, liraglutide (GLP-1 receptor agonist), verapamil (calcium antagonist), acarbose (alpha glucosidase inhibitor), and pasireotide (somatostatin analogue), respectively. RESULTS: (I) NT secretion was similar after TVP/CTVP (p = 0.9), but increased vs. controls (p < 0.0001). (II) NT secretion was increased after RYGB vs. SG and controls (p < 0.0001). NT responses were similar in SG and controls (p = 0.3), but early postprandial NT concentrations were higher after SG (p < 0.05). (III) Exendin (9-39) and sitagliptin did not change NT responses vs placebo (p > 0.2), but responses were lower during sitagliptin vs. exendin(9-39) (p = 0.03). (IV) Pasireotide suppressed NT secretion (p = 0.004). Sitagliptin tended to lower NT secretion (p = 0.08). Liraglutide, verapamil, and acarbose had no effect (p > 0.9). CONCLUSION: Neurotensin secretion is increased after surgeries associated with accelerated gastric emptying and lowered by pasireotide.


Asunto(s)
Gastrectomía , Derivación Gástrica , Neurotensina/sangre , Obesidad/cirugía , Vagotomía Troncal , Glucemia , Péptido 1 Similar al Glucagón/sangre , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Liraglutida/administración & dosificación , Liraglutida/uso terapéutico , Obesidad/sangre , Obesidad/tratamiento farmacológico , Periodo Posprandial
6.
Int J Obes (Lond) ; 44(9): 1872-1883, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32317753

RESUMEN

BACKGROUND/OBJECTIVES: Bile acids in plasma are elevated after bariatric surgery and may contribute to metabolic improvements, but underlying changes in bile flow are poorly understood. We assessed bilio-enteric flow of bile and plasma bile concentrations in individuals with Roux-en-Y gastric bypass (RYGB) or sleeve gastrectomy (SG) surgery compared with matched non-surgical controls (CON). SUBJECTS/METHODS: Fifteen RYGB, 10 SG and 15 CON underwent 99Tc-mebrofenin cholescintigraphy combined with intake of a high-fat 111In-DTPA-labelled meal and frequent blood sampling. A 75Se-HCAT test was used to assess bile acid retention. RESULTS: After RYGB, gallbladder filling was decreased (p = 0.045 versus CON), basal flow of bile into the small intestine increased (p = 0.005), bile acid retention augmented (p = 0.021) and basal bile acid plasma concentrations elevated (p = 0.009). During the meal, foods passed unimpeded through the gastric pouch resulting in almost instant postprandial mixing of bile and foods, but the postprandial rise in plasma bile acids was brief and associated with decreased overall release of fibroblast growth factor-19 (FGF-19) compared with CON (p = 0.033). After SG, bile flow and retention were largely unaltered (p > 0.05 versus CON), but gastric emptying was accelerated (p < 0.001) causing earlier mixture of bile and foods also in this group. Neither basal nor postprandial bile acid concentrations differed between SG and CON. CONCLUSIONS: Bilio-enteric bile flow is markedly altered after RYGB resulting in changes in plasma concentrations of bile acids and FGF-19, whereas bile flow and plasma concentrations are largely unaltered after SG.


Asunto(s)
Ácidos y Sales Biliares/sangre , Ácidos y Sales Biliares/metabolismo , Gastrectomía/estadística & datos numéricos , Derivación Gástrica/estadística & datos numéricos , Adulto , Conductos Biliares/metabolismo , Femenino , Factores de Crecimiento de Fibroblastos/sangre , Humanos , Masculino , Persona de Mediana Edad , Obesidad Mórbida/cirugía , Periodo Posprandial/fisiología
7.
Sci Rep ; 9(1): 15154, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31641146

RESUMEN

To describe glucose metabolism in the late, weight stable phase after Roux-en-Y Gastric Bypass (RYGB) in patients with and without preoperative type 2 diabetes we invited 55 RYGB-operated persons from two existing cohorts to participate in a late follow-up study. 44 (24 with normal glucose tolerance (NGT)/20 with type 2 diabetes (T2D) before surgery) accepted the invitation (median follow-up 2.7 [Range 2.2-5.0 years]). Subjects were examined during an oral glucose stimulus and results compared to preoperative and 1-year (1 y) post RYGB results. Glucose tolerance, insulin resistance, beta-cell function and incretin hormone secretion were evaluated. 1 y weight loss was maintained late after surgery. Glycemic control, insulin resistance, beta-cell function and GLP-1 remained improved late after surgery in both groups. In NGT subjects, nadir glucose decreased 1 y after RYGB, but did not change further. In T2D patients, relative change in weight from 1 y to late after RYGB correlated with relative change in fasting glucose and HbA1c, whereas relative changes in glucose-stimulated insulin release correlated inversely with relative changes in postprandial glucose excursions. In NGT subjects, relative changes in postprandial nadir glucose correlated with changes in beta-cell glucose sensitivity. Thus, effects of RYGB on weight and glucose metabolism are maintained late after surgery in patients with and without preoperative T2D. Weight loss and improved beta-cell function both contribute to maintenance of long-term glycemic control in patients with type 2 diabetes, and increased glucose stimulated insulin secretion may contribute to postprandial hypoglycemia in NGT subjects.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Derivación Gástrica , Glucosa/metabolismo , Adulto , Peso Corporal , Femenino , Humanos , Incretinas/metabolismo , Resistencia a la Insulina , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Persona de Mediana Edad , Pérdida de Peso
8.
Am J Physiol Endocrinol Metab ; 316(4): E615-E621, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30721097

RESUMEN

Bariatric surgery results in marked body weight loss and improves type 2 diabetes in most patients with obesity. The growth differentiation factor 15 (GDF15) has recently emerged as a novel satiety factor. To begin to understand whether GDF15 is involved in mediating the effects of bariatric surgery on body weight and glycemia in humans, we measured plasma GDF15 in patients with obesity ( n = 25) and in patients with obesity and diabetes ( n = 22) before and after Roux-en-Y gastric bypass (RYGB) surgery. GDF15 was increased 1 wk after RYGB compared with before surgery (689 ± 45 vs. 487 ± 28 pg/ml, P < 0.001) and GDF15 remained elevated at 3 mo (554 ± 37 pg/ml, P < 0.05), at 1 yr (566 ± 37 pg/ml, P < 0.05), and at 2.5-4 yr (630 ± 50 pg/ml, P < 0.001) after RYGB surgery. Both age and insulin sensitivity correlated with GDF15 before the surgery ( r = 0.46, P < 0.0001 and r = 0.34, P < 0.001, respectively). These correlations disappeared at 2.5-4 yr following the surgery. Conversely, weight loss magnitude correlated with GDF15, measured 2.5-4 yr postsurgery ( r = 0.21, P < 0.0055). In summary, circulating GDF15 increases and correlates with body weight loss following RYGB surgery.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Derivación Gástrica , Factor 15 de Diferenciación de Crecimiento/sangre , Obesidad/cirugía , Adulto , Factores de Edad , Cirugía Bariátrica , Diabetes Mellitus Tipo 2/complicaciones , Femenino , Estudios de Seguimiento , Humanos , Resistencia a la Insulina , Masculino , Persona de Mediana Edad , Obesidad/sangre , Obesidad/complicaciones , Pérdida de Peso
9.
Cell Syst ; 7(6): 601-612.e3, 2018 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-30528273

RESUMEN

Obesity-related diseases affect half of the global population, and bariatric surgery is one of the few interventions with long-lasting weight loss and cardio-metabolic effects. Here, we investigated the effect of Roux-en-Y gastric bypass surgery on the plasma proteome, hypothesizing that specific proteins or protein patterns may serve as key mediators and markers of the metabolic response. We performed mass spectrometry (MS)-based proteomics on two longitudinal studies encompassing 47 morbidly obese patients, generating quantitative information on more than 1,700 proteins. A global correlation matrix incorporating about 200,000 relationships revealed functional connections between proteins and assigned them to physiological processes. The main classes of significantly altered proteins were markers of systemic inflammation and those involved in lipid metabolism. Our data highlight robust correlative and anti-correlative behaviors of circulating proteins to each other and to clinical parameters. A group of inflammation-related proteins showed distinct inverse relationships to proteins consistently associated with insulin sensitivity.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Derivación Gástrica , Metabolismo de los Lípidos , Obesidad Mórbida/sangre , Obesidad Mórbida/cirugía , Mapas de Interacción de Proteínas , Adulto , Biomarcadores/sangre , Biomarcadores/metabolismo , Femenino , Derivación Gástrica/métodos , Humanos , Inflamación/sangre , Inflamación/metabolismo , Resistencia a la Insulina , Masculino , Persona de Mediana Edad , Obesidad Mórbida/metabolismo , Proteoma/metabolismo , Proteómica
10.
Cell Rep ; 21(6): 1452-1460, 2017 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-29117552

RESUMEN

Glucagon is secreted from pancreatic α cells, and hypersecretion (hyperglucagonemia) contributes to diabetic hyperglycemia. Molecular heterogeneity in hyperglucagonemia is poorly investigated. By screening human plasma using high-resolution-proteomics, we identified several glucagon variants, among which proglucagon 1-61 (PG 1-61) appears to be the most abundant form. PG 1-61 is secreted in subjects with obesity, both before and after gastric bypass surgery, with protein and fat as the main drivers for secretion before surgery, but glucose after. Studies in hepatocytes and in ß cells demonstrated that PG 1-61 dose-dependently increases levels of cAMP, through the glucagon receptor, and increases insulin secretion and protein levels of enzymes regulating glycogenolysis and gluconeogenesis. In rats, PG 1-61 increases blood glucose and plasma insulin and decreases plasma levels of amino acids in vivo. We conclude that glucagon variants, such as PG 1-61, may contribute to glucose regulation by stimulating hepatic glucose production and insulin secretion.


Asunto(s)
Glucemia/análisis , Insulina/análisis , Fallo Renal Crónico/patología , Proglucagón/sangre , Animales , Células COS , Estudios de Casos y Controles , Células Cultivadas , Chlorocebus aethiops , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Receptor del Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Receptor del Péptido 1 Similar al Glucagón/genética , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Gluconeogénesis/efectos de los fármacos , Humanos , Insulina/sangre , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Fallo Renal Crónico/sangre , Fallo Renal Crónico/metabolismo , Masculino , Ratones , Fosforilasa Quinasa/genética , Fosforilasa Quinasa/metabolismo , Proglucagón/farmacología , Ratas , Ratas Wistar , Receptores de Glucagón/genética , Receptores de Glucagón/metabolismo
11.
Am J Physiol Regul Integr Comp Physiol ; 312(5): R797-R805, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28202439

RESUMEN

Roux-en-Y gastric bypass (RYGB) induces weight loss and improves insulin sensitivity when evaluated by the hyperinsulinemic-euglycemic clamp (HEC). Surrogate indices of insulin sensitivity calculated from insulin and glucose concentrations at fasting or after an oral glucose tolerance test (OGTT) are frequently used, but have not been validated after RYGB. Our aim was to evaluate whether surrogate indices reliably estimate changes in insulin sensitivity after RYGB. Four fasting surrogates (inverse-HOMA-IR, HOMA2-%S, QUICKI, revised-QUICKI) and three OGTT-derived surrogates (Matsuda, Gutt, OGIS) were compared with HEC-estimated peripheral insulin sensitivity (Rd or Rd/I, depending on how the index was originally validated) and the tracer-determined hepatic insulin sensitivity index (HISI) in patients with preoperative type 2 diabetes (n = 10) and normal glucose tolerance (n = 10) 1 wk, 3 mo, and 1 yr postoperatively. Post-RYGB changes in inverse-HOMA-IR and HOMA2-%S did not correlate with changes in Rd at any visit, but were comparable to changes in HISI at 1 wk. Changes in QUICKI and revised-QUICKI correlated with Rd/I after surgery. Changes in the Matsuda and Gutt indices did not correlate with changes in Rd/I and Rd, respectively, whereas OGIS changes correlated with Rd changes at 1 yr post-RYGB. In conclusion, surrogate measures of insulin sensitivity may not reflect results obtained with gold standard methodology after RYGB, underscoring the importance of critical reflection when surrogate endpoints are used. Fasting surrogate indices may be particularly affected by post-RYGB changes in insulin clearance, whereas the validity of OGTT-derived surrogates may be compromised by surgical rearrangements of the gut.


Asunto(s)
Derivación Gástrica , Técnica de Clampeo de la Glucosa/métodos , Prueba de Tolerancia a la Glucosa/métodos , Resistencia a la Insulina , Insulina/sangre , Obesidad Mórbida/sangre , Obesidad Mórbida/cirugía , Adulto , Glucemia/análisis , Femenino , Humanos , Masculino , Obesidad Mórbida/diagnóstico , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Resultado del Tratamiento
12.
Obes Surg ; 26(9): 2263-2267, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27138601

RESUMEN

Postprandial hyperinsulinemia characterizes Roux-en-Y gastric bypass (RYGB) and sometimes leads to reactive hypoglycemia. We prospectively evaluated changes in beta cell function in seven RYGB-operated patients with a median follow-up of 2.9 years with hyperglycemic clamps and oral glucose tolerance tests (OGTTs). Three years after RYGB, weight loss was 26 % and insulin sensitivity had improved. Insulin secretion during clamp experiments was largely unchanged compared to before surgery. In contrast, insulin secretion in response to the OGTTs doubled when evaluated by the disposition index and 2-h plasma glucose declined to a mean of 3.3 ± 0.3 mmol/l postoperatively. Our findings indicate that intrinsic beta cell function remains unchanged in glucose-tolerant patients even years after RYGB, while altered gut-islet regulation drive risk of postprandial hyperinsulinemic hypoglycemia.


Asunto(s)
Derivación Gástrica/estadística & datos numéricos , Hipoglucemia/epidemiología , Islotes Pancreáticos/fisiopatología , Glucemia , Humanos , Obesidad Mórbida/cirugía , Periodo Posprandial , Estudios Prospectivos
13.
Am J Physiol Endocrinol Metab ; 310(7): E505-14, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26786780

RESUMEN

Exaggerated secretion of glucagon-like peptide 1 (GLP-1) is important for postprandial glucose tolerance after Roux-en-Y gastric bypass (RYGB), whereas the role of glucose-dependent insulinotropic polypeptide (GIP) remains to be resolved. We aimed to explore the relative importance of endogenously secreted GLP-1 and GIP on glucose tolerance and ß-cell function after RYGB. We used DPP-4 inhibition to enhance concentrations of intact GIP and GLP-1 and the GLP-1 receptor antagonist exendin-(9-39) (Ex-9) for specific blockage of GLP-1 actions. Twelve glucose-tolerant patients were studied after RYGB in a randomized, placebo-controlled, 4-day crossover study with standard mixed-meal tests and concurrent administration of placebo, oral sitagliptin, Ex-9 infusion, or combined Ex-9-sitagliptin. GLP-1 receptor antagonism increased glucose excursions, clearly attenuated ß-cell function, and aggravated postprandial hyperglucagonemia compared with placebo, whereas sitagliptin had no effect despite two- to threefold increased concentrations of intact GLP-1 and GIP. Similarly, sitagliptin did not affect glucose tolerance or ß-cell function during GLP-1R blockage. This study confirms the importance of GLP-1 for glucose tolerance after RYGB via increased insulin and attenuated glucagon secretion in the postprandial state, whereas amplification of the GIP signal (or other DPP-4-sensitive glucose-lowering mechanisms) did not appear to contribute to the improved glucose tolerance seen after RYGB.


Asunto(s)
Glucemia/metabolismo , Péptido C/metabolismo , Derivación Gástrica , Polipéptido Inhibidor Gástrico/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Glucagón/metabolismo , Obesidad/cirugía , Adulto , Glucemia/efectos de los fármacos , Estudios Cruzados , Ensayo de Inmunoadsorción Enzimática , Femenino , Glucagón/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Prueba de Tolerancia a la Glucosa/métodos , Humanos , Hipoglucemiantes/farmacología , Modelos Lineales , Masculino , Obesidad/metabolismo , Fragmentos de Péptidos/farmacología , Periodo Posprandial , Método Simple Ciego , Fosfato de Sitagliptina/farmacología
14.
Am J Clin Nutr ; 102(3): 600-7, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26245808

RESUMEN

BACKGROUND: Roux-en-Y gastric bypass (RYGB) involves exclusion of major parts of the stomach and changes in admixture of gastro-pancreatic enzymes, which could have a major impact on protein digestion and amino acid absorption. OBJECTIVE: We investigated the effect of RYGB on amino acid appearance in the systemic circulation from orally ingested protein and from endogenous release. DESIGN: Nine obese glucose-tolerant subjects, with a mean body mass index (in kg/m(2)) of 39.2 (95% CI: 35.2, 43.3) and mean glycated hemoglobin of 5.3% (95% CI: 4.9%, 5.6%), were studied before and 3 mo after RYGB. Leucine and phenylalanine kinetics were determined under basal conditions and during 4 postprandial hours by intravenous infusions of [3,3,3-(2)H3]-leucine and [ring-(2)D5]-phenylalanine combined with ingestion of [1-(13)C]-leucine intrinsically labeled caseinate as the sole protein source of the meal. Changes in body composition were assessed by dual-energy X-ray absorptiometry. RESULTS: After RYGB, basal plasma leucine concentration did not change, but marked changes were seen postprandially with 1.7-fold increased peak concentrations (before­mean: 217 µmol/L; 95% CI: 191, 243 µmol/L; 3 mo­mean: 377 µmol/L; 95% CI: 252, 502 µmol/L; P = 0.012) and 2-fold increased incremental AUC (before-mean: 4.1 mmol ∙ min/L; 95% CI: 2.7, 5.5 mmol ∙ min/L; 3 mo-mean: 9.5 mmol ∙ min/L; 95% CI: 4.9, 14.2 mmol ∙ min/L; P = 0.032). However, the postprandial hyperleucinemia was transient, and concentrations were below basal concentrations in the fourth postprandial hour. These concentration differences were mainly caused by changes in leucine appearance rate from orally ingested caseinate: peak rate increased nearly 3-fold [before­mean: 0.5 µmol/(kg fat-free mass ∙ min); 95% CI: 0.4, 0.5 µmol/(kg fat-free mass ∙ min); 3 mo­mean 1.4 µmol/(kg fat-free mass ∙ min); 95% CI: 0.8, 1.9 µmol/(kg fat-free mass ∙ min); P = 0.002], and time to peak was much shorter (before­mean: 173 min; 95% CI: 137, 209 min; 3 mo­mean: 65 min; 95% CI: 46, 84 min; P < 0.001). Only minor changes were seen in endogenous leucine release after RYGB. CONCLUSIONS: RYGB accelerates caseinate digestion and amino acid absorption, resulting in faster and higher but more transient postprandial elevation of plasma amino acids. Changes are likely mediated by accelerated intestinal nutrient entry and clearly demonstrate that protein digestion is not impaired after RYGB. This trial was registered at clinicaltrials.gov as NCT01559792.


Asunto(s)
Derivación Gástrica , Leucina/sangre , Fenilalanina/sangre , Proteolisis , Absorciometría de Fotón , Adolescente , Adulto , Anciano , Glucemia/metabolismo , Índice de Masa Corporal , Peso Corporal , Caseínas/administración & dosificación , Proteínas en la Dieta/administración & dosificación , Femenino , Hemoglobina Glucada/metabolismo , Humanos , Absorción Intestinal , Leucina/administración & dosificación , Masculino , Comidas , Persona de Mediana Edad , Obesidad/cirugía , Fenilalanina/administración & dosificación , Periodo Posprandial , Adulto Joven
15.
Am J Physiol Endocrinol Metab ; 308(6): E535-44, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25628424

RESUMEN

Roux-en-Y gastric bypass surgery (RYGB) in patients with type 2 diabetes often leads to early disease remission, and it is unknown to what extent this involves improved pancreatic ß-cell function per se and/or enhanced insulin- and non-insulin-mediated glucose disposal (glucose effectiveness). We studied 30 obese patients, including 10 with type 2 diabetes, 8 with impaired glucose tolerance, and 12 with normal glucose tolerance before, 1 wk, and 3 mo after RYGB, using an intravenous glucose tolerance test (IVGTT) to estimate first-phase insulin response, insulin sensitivity (Si), and glucose effectiveness with Bergman's minimal model. In the fasting state, insulin sensitivity was estimated by HOMA-S and ß-cell function by HOMA-ß. Moreover, mixed-meal tests and oral GTTs were performed. In patients with type 2 diabetes, glucose levels normalized after RYGB, first-phase insulin secretion in response to iv glucose increased twofold, and HOMA-ß already improved 1 wk postoperatively, with further enhancements at 3 mo. Insulin sensitivity increased in the liver (HOMA-S) at 1 wk and at 3 mo in peripheral tissues (Si), whereas glucose effectiveness did not improve significantly. During oral testing, GLP-1 responses and insulin secretion increased regardless of glucose tolerance. Therefore, in addition to increased insulin sensitivity and exaggerated postprandial GLP-1 levels, diabetes remission after RYGB involves early improvement of pancreatic ß-cell function per se, reflected in enhanced first-phase insulin secretion to iv glucose and increased HOMA-ß. A major role for improved glucose effectiveness after RYGB was not supported by this study.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/cirugía , Derivación Gástrica , Insulina/metabolismo , Adulto , Peso Corporal/fisiología , Ayuno/metabolismo , Derivación Gástrica/rehabilitación , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/cirugía , Prueba de Tolerancia a la Glucosa , Humanos , Secreción de Insulina , Persona de Mediana Edad , Obesidad/metabolismo , Obesidad/cirugía , Periodo Posoperatorio
16.
J Clin Endocrinol Metab ; 100(3): E396-406, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25536209

RESUMEN

CONTEXT: Bile acids and fibroblast growth factor 19 (FGF19) have been suggested as key mediators of the improvements in glucose metabolism after Roux-en-Y gastric bypass (RYGB). OBJECTIVE: To describe fasting and postprandial state total bile acid (TBA) and FGF19 concentrations before and after RYGB and relate them to parameters of glucose metabolism, glucagon-like peptide-1, cholecystokinin, and cholesterol fractions. DESIGN AND SETTING: A prospective descriptive study was performed at the Department of Endocrinology, Hvidovre Hospital, Hvidovre, Denmark. PATIENTS: Thirteen type 2 diabetic (T2D) patients and 12 normal glucose tolerant (NGT) subjects participated in the study. INTERVENTION: A 4-hour liquid meal test was performed before and 1 week, 3 months, and 1 year after RYGB. MAIN OUTCOME MEASURES: We measured fasting and postprandial TBA and FGF19 concentrations. RESULTS: Fasting TBA concentrations decreased in NGT subjects (P < .001) and were unchanged in T2D patients 1 week after surgery, but then increased gradually in both groups with time from surgery (ANOVA Ptime < .001). Area under the curve (AUC) TBA was decreased in NGT subjects 1 week after RYGB (before surgery, 567 mmol * min/L [interquartile range, 481-826]; 1 wk, 419 [381-508]; P = .009) and was unchanged in T2D patients (894 [573-1002]; 695 [349-1147]; P = .97) but then increased with time from surgery in both groups (Ptime < .001). Fasting FGF19 concentrations were unchanged acutely after RYGB (NGT, 140 pg/mL [100-162], 134 [119-204], P = .42; T2D, 162 [130-196], 154 [104-164], P = .68) and remained unchanged throughout the follow-up period. AUC FGF19 increased gradually with time after surgery (Ptime < .001), resembling the changes seen with AUC TBA. One week after RYGB, glucose metabolism improved, low-density lipoprotein-cholesterol and high-density lipoprotein-cholesterol decreased, and cholecystokinin and glucagon-like peptide-1 secretion increased, whereas FFA concentrations were unchanged. CONCLUSION: TBA and FGF19 do not explain acute changes in glucose metabolism, cholesterol fractions, and gut hormone secretion after RYGB.


Asunto(s)
Ácidos y Sales Biliares/sangre , Glucemia/metabolismo , Factores de Crecimiento de Fibroblastos/sangre , Derivación Gástrica , Obesidad Mórbida/metabolismo , Obesidad Mórbida/cirugía , Adulto , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Regulación hacia Arriba
17.
Diabetes ; 63(5): 1725-37, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24241533

RESUMEN

Roux-en-Y gastric bypass (RYGB) improves glycemic control within days after surgery, and changes in insulin sensitivity and ß-cell function are likely to be involved. We studied 10 obese patients with type 2 diabetes (T2D) and 10 obese glucose-tolerant subjects before and 1 week, 3 months, and 1 year after RYGB. Participants were included after a preoperative diet-induced total weight loss of -9.2 ± 1.2%. Hepatic and peripheral insulin sensitivity were assessed using the hyperinsulinemic- euglycemic clamp combined with the glucose tracer technique, and ß-cell function was evaluated in response to an intravenous glucose-glucagon challenge as well as an oral glucose load. Within 1 week, RYGB reduced basal glucose production, improved basal hepatic insulin sensitivity, and increased insulin clearance, highlighting the liver as an important organ responsible for early effects on glucose metabolism after surgery. Insulin-mediated glucose disposal and suppression of fatty acids did not improve immediately after surgery but increased at 3 months and 1 year; this increase likely was related to the reduction in body weight. Insulin secretion increased after RYGB only in patients with T2D and only in response to oral glucose, underscoring the importance of the changed gut anatomy.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Resistencia a la Insulina/fisiología , Insulina/metabolismo , Hígado/metabolismo , Obesidad/metabolismo , Adulto , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/cirugía , Femenino , Derivación Gástrica , Técnica de Clampeo de la Glucosa , Humanos , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Persona de Mediana Edad , Obesidad/complicaciones , Obesidad/cirugía , Periodo Posprandial
18.
Obes Res Clin Pract ; 7(4): e269-74, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24306154

RESUMEN

Experience with Roux-en-Y gastric bypass in patients with type 1 diabetes is very limited, despite an increasing prevalence of obesity also in this population. We describe changes in anthropometric measures, insulin dose, HbA1c, blood pressure, lipid status, and metabolic response to a liquid mixed meal throughout the first year after RYGB in an obese patient with type 1 diabetes. No change in HbA1c was observed, but a 48% reduction in weight-adjusted insulin dose and improvements in cardiovascular risk factors was seen 1 year after surgery. Exaggerated secretions of anorexigenic gut hormones were seen during the meals.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Diabetes Mellitus Tipo 1/cirugía , Derivación Gástrica , Células Secretoras de Insulina/citología , Insulina/administración & dosificación , Obesidad Mórbida/cirugía , Adulto , Diabetes Mellitus Tipo 1/sangre , Femenino , Humanos , Insulina/sangre , Obesidad Mórbida/sangre , Factores de Riesgo
19.
Diabetologia ; 56(12): 2679-87, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24048673

RESUMEN

AIMS/HYPOTHESIS: Roux-en-Y gastric bypass (RYGB) improves glycaemic control in part by increasing postprandial insulin secretion through exaggerated glucagon-like peptide (GLP)-1 release. However, it is unknown whether islet cell responsiveness to i.v. glucose, non-glucose (arginine) and incretin hormones, including GLP-1, is altered. METHODS: Eleven severely obese glucose-tolerant individuals underwent three hyperglycaemic clamps with arginine bolus and co-infusion of either GLP-1, glucose-dependent insulinotropic polypeptide (GIP) or saline before, and at 1 week and 3 months after RYGB. In addition, an OGTT was performed before and 3 months after surgery. RESULTS: After RYGB, insulin sensitivity improved at 1 week and 3 months, while insulin stimulation and glucagon suppression in response to the clamp with saline co-infusion were largely unaltered. The influence of i.v. GLP-1 and GIP on insulin and glucagon secretion was also unchanged postoperatively. In response to the postoperative OGTT at 3 months, insulin and GLP-1, but not GIP, secretion increased. Furthermore, the glucose profile during the OGTT was altered, with a substantial reduction in 2 h plasma glucose and a paradoxical hypersecretion of glucagon. CONCLUSIONS/INTERPRETATION: After RYGB, insulin hypersecretion is linked to the oral, but not the i.v., route of administration and is associated with exaggerated release and preserved insulinotropic action of GLP-1, while both the secretion and action of GIP are unchanged. The results highlight the importance of increased GLP-1 secretion for improving postoperative glucose metabolism. TRIAL REGISTRATION: ClinicalTrials.gov NCT01559779.


Asunto(s)
Glucemia/metabolismo , Derivación Gástrica , Polipéptido Inhibidor Gástrico/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Insulina/metabolismo , Obesidad Mórbida/metabolismo , Páncreas/metabolismo , Fragmentos de Péptidos/metabolismo , Adulto , Femenino , Glucagón/metabolismo , Técnica de Clampeo de la Glucosa/métodos , Prueba de Tolerancia a la Glucosa , Humanos , Hiperglucemia/metabolismo , Resistencia a la Insulina , Secreción de Insulina , Masculino , Obesidad Mórbida/fisiopatología , Obesidad Mórbida/cirugía , Periodo Posprandial , Resultado del Tratamiento
20.
Diabetologia ; 56(10): 2250-4, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23893303

RESUMEN

AIMS/HYPOTHESIS: Roux-en-Y gastric bypass surgery (RYGB) improves glucose tolerance in patients with type 2 diabetes, but also changes the glucose profile in response to a meal in glucose-tolerant individuals. We hypothesised that the driving force for the changed postprandial glucose profiles after RYGB is rapid entry of glucose into the systemic circulation due to modified gastrointestinal anatomy, causing hypersecretion of insulin and other hormones influencing glucose disappearance and endogenous glucose production. METHODS: We determined glucose absorption and metabolism and the rate of lipolysis before and 3 months after RYGB in obese glucose-tolerant individuals using the double-tracer technique during a mixed meal. RESULTS: After RYGB, the postprandial plasma glucose profile changed, with a higher peak glucose concentration followed by a faster return to lower than basal levels. These changes were brought about by changes in glucose kinetics: (1) a more rapid appearance of ingested glucose in the systemic circulation, and a concomitant increase in insulin and glucagon-like peptide-1 secretion; (2) postprandial glucose disappearance was maintained at a high rate for a longer time after RYGB. Endogenous glucose production was similar before and after surgery. Postoperative glucagon secretion increased and showed a biphasic response after RYGB. Adipose tissue basal rate of lipolysis was higher after RYGB. CONCLUSIONS/INTERPRETATION: A rapid rate of absorption of ingested glucose into the systemic circulation, followed by increased insulin secretion and glucose disappearance appears to drive the changes in the glucose profile observed after RYGB, while endogenous glucose production remains unchanged. TRIAL REGISTRATION: ClinicalTrials.gov NCT01559792. FUNDING: The study was part of the UNIK program: Food, Fitness & Pharma for Health and Disease (see www.foodfitnesspharma.ku.dk ). Funding was received from the Novo Nordisk foundation and the Strategic Research Counsel for the Capital Area and Danish Research Agency. The primary investigator received a PhD scholarship from the University of Copenhagen, which was one-third funded by Novo Nordisk.


Asunto(s)
Derivación Gástrica , Absorciometría de Fotón , Índice de Masa Corporal , Femenino , Glucosa/metabolismo , Humanos , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA