Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 772
Filtrar
1.
Endocrinology ; 165(10)2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39248655

RESUMEN

Postprandial dyslipidemia is commonly present in people with type 2 diabetes and obesity and is characterized by overproduction of apolipoprotein B48-containing chylomicron particles from the intestine. Peripheral serotonin is emerging as a regulator of energy homeostasis with profound implications for obesity; however, its role in dietary fat absorption and chylomicron production is unknown. Chylomicron production was assessed in Syrian golden hamsters by administering an olive oil gavage and IP poloxamer to inhibit lipoprotein clearance. Administration of serotonin or selective serotonin reuptake inhibitor, fluoxetine, increased postprandial plasma triglyceride (TG) and TG-rich lipoproteins. Conversely, inhibiting serotonin synthesis pharmacologically by p-chlorophenylalanine (PCPA) led to a reduction in both the size and number of TG-rich lipoprotein particles, resulting in lower plasma TG and apolipoprotein B48 levels. The effects of PCPA occurred independently of gastric emptying and vagal afferent signaling. Inhibiting serotonin synthesis by PCPA led to increased TG within the intestinal lumen and elevated levels of TG and cholesterol in the stool when exposed to a high-fat/high-cholesterol diet. These findings imply compromised fat absorption, as evidenced by reduced lipase activity in the duodenum and lower levels of serum bile acids, which are indicative of intestinal bile acids. During the postprandial state, mRNA levels for serotonin receptors (5-HTRs) were upregulated in the proximal intestine. Administration of cisapride, a 5-HT4 receptor agonist, alleviated reductions in postprandial lipemia caused by serotonin synthesis inhibition, indicating that serotonin controls dietary fat absorption and chylomicron secretion via 5-HT4 receptor.


Asunto(s)
Quilomicrones , Grasas de la Dieta , Mesocricetus , Receptores de Serotonina 5-HT4 , Serotonina , Triglicéridos , Animales , Masculino , Quilomicrones/metabolismo , Serotonina/metabolismo , Receptores de Serotonina 5-HT4/metabolismo , Grasas de la Dieta/farmacología , Triglicéridos/metabolismo , Triglicéridos/sangre , Cricetinae , Fenclonina/farmacología , Absorción Intestinal/efectos de los fármacos , Fluoxetina/farmacología , Periodo Posprandial/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
2.
Am J Physiol Endocrinol Metab ; 327(3): E396-E410, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39082900

RESUMEN

Ghrelin is an appetite-stimulating hormone secreted from the gastric mucosa in the fasting state, and secretion decreases in response to food intake. After sleeve gastrectomy (SG), plasma concentrations of ghrelin decrease markedly. Whether this affects appetite and glucose tolerance postoperatively is unknown. We investigated the effects of ghrelin infusion on appetite and glucose tolerance in individuals with obesity before and 3 mo after SG. Twelve participants scheduled for SG were included. Before and 3 mo after surgery, a mixed-meal test followed by an ad libitum meal test was performed with concomitant infusions of acyl-ghrelin (1 pmol/kg/min) or placebo. Infusions began 60 min before meal intake to reach a steady state before the mixed-meal and were continued throughout the study day. Two additional experimental days with 0.25 pmol/kg/min and 10 pmol/kg/min of acyl-ghrelin infusions were conducted 3 mo after surgery. Both before and after SG, postprandial glucose concentrations increased dose dependently during ghrelin infusions compared with placebo. Ghrelin infusions inhibited basal and postprandial insulin secretion rates, resulting in lowered measures of ß-cell function, but no effect on insulin sensitivity was seen. Ad libitum meal intake was unaffected by the administration of ghrelin. In conclusion, ghrelin infusion increases postprandial plasma glucose concentrations and impairs ß-cell function before and after SG but has no effect on ad libitum meal intake. We speculate that the lower concentration of ghrelin after SG may impact glucose metabolism following this procedure.NEW & NOTEWORTHY Ghrelin's effect on glucose tolerance and food intake following sleeve gastrectomy (SG) was evaluated. Acyl-ghrelin was infused during a mixed-meal and ad libitum meals before and 3 mo after surgery. Postprandial glucose concentrations increased during ghrelin infusions, both before and after surgery, while insulin production was inhibited. However, ad libitum meal intake did not differ during ghrelin administration compared with placebo. The decreased ghrelin concentration following SG may contribute to the glycemic control after surgery.


Asunto(s)
Apetito , Glucemia , Ingestión de Alimentos , Gastrectomía , Ghrelina , Periodo Posprandial , Humanos , Ghrelina/sangre , Ghrelina/análogos & derivados , Masculino , Adulto , Femenino , Apetito/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Glucemia/metabolismo , Glucemia/efectos de los fármacos , Periodo Posprandial/efectos de los fármacos , Persona de Mediana Edad , Insulina/sangre , Obesidad Mórbida/cirugía , Obesidad Mórbida/metabolismo , Hormonas Gastrointestinales/metabolismo , Hormonas Gastrointestinales/sangre , Prueba de Tolerancia a la Glucosa , Resistencia a la Insulina/fisiología , Método Doble Ciego , Obesidad/cirugía , Obesidad/metabolismo
3.
Chem Senses ; 492024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-39046896

RESUMEN

Postprandial regulation of the gastric emptying (GE) rate plays an important role in food intake. Although oral sweetening with glucose may accelerate GE, the effects of different sweetness intensities of glucose (10% and 20%, w/v) and other energy sweeteners (e.g. fructose and sucrose) remain uncertain. The purpose of this study was to determine the effects of different glucose concentrations (Experiment 1) and different sugars with the same sweet taste intensity (Experiment 2) on postprandial GE. In both experiments, after ingesting a 200 kcal carbohydrate solution containing 50 g of maltodextrin, participants repeatedly sipped, but did not swallow, one of three (water, 10% and 20%, w/v glucose) or four (water and equally sweet 20%, w/v glucose, 12%, w/v fructose, and 14%, w/v sucrose) solutions for 1 min every 5 min over a 30 min period. GE was evaluated by measuring the temporal change in the cross-sectional area of the gastric antrum using ultrasound. In Experiment 1, oral stimulation with 20% (w/v) glucose resulted in greater GE than the control stimulus (i.e. water), but the effect of stimulation with 10% (w/v) glucose on GE was not different from that of the control stimulus. In Experiment 2, stimulation with 20% (w/v) glucose or 12% (w/v) fructose resulted in greater GE than the control stimulus. However, the effect of stimulation with 14% (w/v) sucrose on GE did not differ from that of the control stimulus. Consequently, oral stimulation with glucose or fructose solutions of moderate to high sweetness following a meal facilitates postprandial GE.


Asunto(s)
Fructosa , Vaciamiento Gástrico , Glucosa , Sacarosa , Humanos , Vaciamiento Gástrico/efectos de los fármacos , Fructosa/farmacología , Glucosa/farmacología , Glucosa/administración & dosificación , Masculino , Adulto , Sacarosa/farmacología , Femenino , Adulto Joven , Periodo Posprandial/efectos de los fármacos , Edulcorantes/farmacología , Administración Oral
4.
Eur J Endocrinol ; 191(2): 192-203, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39049802

RESUMEN

AIMS: Although metformin is widely used for treatment of type 2 diabetes (T2D), its glucose-lowering mechanism remains unclear. Using the glucagon-like peptide 1 (GLP-1) receptor (GLP-1R) antagonist exendin(9-39)NH2, we tested the hypothesis that postprandial GLP-1-mediated effects contribute to the glucose-lowering potential of metformin in T2D. METHODS: In a randomized, placebo-controlled, double-blind, crossover study, 15 individuals with T2D (median HbA1c 50 mmol/mol [6.7%], body mass index 30.1 kg/m2, age 71 years) underwent, in randomized order, 14 days of metformin and placebo treatment, respectively. Each treatment period was preceded by 14 days without any glucose-lowering medicine and concluded by two 4 h mixed meal tests performed in randomized order and separated by >24 h with either continuous intravenous exendin(9-39)NH2 or saline infusion. RESULTS: Compared to placebo, metformin treatment lowered fasting plasma glucose (mean of differences [MD] 1.4 mmol/L × min [95% CI 0.8-2.0]) as well as postprandial plasma glucose excursions during both saline infusion (MD 186 mmol/L × min [95% CI 64-307]) and exendin(9-39)NH2 infusion (MD 268 mmol/L × min [95% CI 108-427]). The metformin-induced improvement in postprandial glucose tolerance was unaffected by GLP-1R antagonization (MD 82 mmol/L × min [95% CI -6564-170]). Metformin treatment increased fasting plasma GLP-1 (MD 1.7 pmol/L × min [95% CI 0.39-2.9]) but did not affect postprandial GLP-1 responses (MD 820 pmol/L × min [95% CI -1750-111]). CONCLUSIONS: Using GLP-1R antagonization, we could not detect GLP-1-mediated postprandial glucose-lowering effect of metformin in individuals with T2D. We show that 2 weeks of metformin treatment increases fasting plasma GLP-1, which may contribute to metformin's beneficial effect on fasting plasma glucose in T2D. Trial registration: Clinicaltrials.gov NCT03246451.


Asunto(s)
Glucemia , Estudios Cruzados , Diabetes Mellitus Tipo 2 , Péptido 1 Similar al Glucagón , Hipoglucemiantes , Metformina , Periodo Posprandial , Humanos , Metformina/uso terapéutico , Metformina/administración & dosificación , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/sangre , Periodo Posprandial/efectos de los fármacos , Masculino , Femenino , Anciano , Péptido 1 Similar al Glucagón/sangre , Método Doble Ciego , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Hipoglucemiantes/uso terapéutico , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/farmacología , Persona de Mediana Edad , Receptor del Péptido 1 Similar al Glucagón/agonistas , Fragmentos de Péptidos
5.
Diabetes ; 73(10): 1605-1614, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38976482

RESUMEN

Treatment with glucagon-like peptide 1 receptor agonists reduces liver steatosis and cardiometabolic risk (CMR). Few data are available on lipid metabolism, and no information is available on the postprandial lipidomic profile. Thus, we investigated how exenatide treatment changes lipid metabolism and composition during fasting and after a mixed-meal tolerance test (MMTT) in adults with severe obesity without diabetes. Thirty individuals (26 females and 4 males, 30-60 years old, BMI >40 kg/m2, HbA1c 5.76%) were assigned (1:1) to diet with exenatide 10 µg twice daily treatment (n = 15) or without treatment as control (n = 15) for 3 months. Fasting and postprandial lipidomic profile (by liquid chromatography quadrupole time-of-flight mass spectrometry) and fatty acid metabolism (following a 6-h MMTT/tracer study) and composition (by gas chromatography-mass spectrometry) were evaluated before and after treatment. Both groups had slight weight loss (-5.5% vs. -1.9%, exenatide vs. control; P = 0.052). During fasting, exenatide, compared with control, reduced some ceramides (CERs) and lysophosphatidylcholines (LPCs) previously associated with CMR, while relatively increasing unsaturated phospholipid species (phosphatidylcholine [PC], LPC) with protective effects on CMR, although concentrations of total lipid species were unchanged. During MMTT, both groups showed suppressed lipolysis equal to baseline, but exenatide significantly lowered free fatty acid clearance and postprandial triacyclglycerol (TAG) concentrations, particularly saturated TAGs with 44-54 carbons. Exenatide also reduced some postprandial CERs, PCs, and LPCs previously linked to CMR. These changes in lipidomic profile remained statistically significant after adjusting for weight loss. Exenatide improved fasting and postprandial lipidomic profiles associated with CMR mainly by reducing saturated postprandial TAGs and CERs independently of weight loss and diabetes.


Asunto(s)
Exenatida , Ayuno , Receptor del Péptido 1 Similar al Glucagón , Lipidómica , Periodo Posprandial , Pérdida de Peso , Humanos , Masculino , Femenino , Periodo Posprandial/efectos de los fármacos , Persona de Mediana Edad , Adulto , Exenatida/uso terapéutico , Pérdida de Peso/efectos de los fármacos , Ayuno/sangre , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Hipoglucemiantes/uso terapéutico , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos
6.
Gut Microbes ; 14(1): 2003176, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34923903

RESUMEN

Non-fasting lipidemia (nFL), mainly contributed by postprandial lipidemia (PL), has recently been recognized as an important cardiovascular disease (CVD) risk as fasting lipidemia (FL). PL serves as a common feature of dyslipidemia in Type 2 Diabetes (T2D), albeit effective therapies targeting on PL were limited. In this study, we aimed to evaluate whether the therapy combining probiotics (Prob) and berberine (BBR), a proven antidiabetic and hypolipidemic regimen via altering gut microbiome, could effectively reduce PL in T2D and to explore the underlying mechanism. Blood PL (120 min after taking 100 g standard carbohydrate meal) was examined in 365 participants with T2D from the Probiotics and BBR on the Efficacy and Change of Gut Microbiota in Patients with Newly Diagnosed Type 2 Diabetes (PREMOTE study), a random, placebo-controlled, and multicenter clinical trial. Prob+BBR was superior to BBR or Prob alone in improving postprandial total cholesterol (pTC) and low-density lipoprotein cholesterol (pLDLc) levels with decrement of multiple species of postprandial lipidomic metabolites after 3 months follow-up. This effect was linked to the changes of fecal Bifidobacterium breve level responding to BBR alone or Prob+BBR treatment. Four fadD genes encoding long-chain acyl-CoA synthetase were identified in the genome of this B. breve strain, and transcriptionally activated by BBR. In vitro BBR treatment further decreased the concentration of FFA in the culture medium of B. breve compared to vehicle. Thus, the activation of fadD by BBR could enhance FFA import and mobilization in B. breve and diliminish the intraluminal lipids for absorption to mediate the effect of Prob+BBR on PL. Our study confirmed that BBR and Prob (B. breve) could exert a synergistic hypolipidemic effect on PL, acting as a gut lipid sink to achieve better lipidemia and CVD risk control in T2D.


Asunto(s)
Berberina/administración & dosificación , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hiperlipidemias/tratamiento farmacológico , Probióticos/administración & dosificación , Adulto , Animales , Colesterol/sangre , LDL-Colesterol/sangre , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/microbiología , Método Doble Ciego , Quimioterapia Combinada , Heces/microbiología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Hiperlipidemias/sangre , Hiperlipidemias/microbiología , Masculino , Persona de Mediana Edad , Periodo Posprandial/efectos de los fármacos
7.
Clin Nutr ; 41(1): 165-176, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34883305

RESUMEN

BACKGROUND & AIMS: Whilst the cardioprotective effects of blueberry intake have been shown in prospective studies and short-term randomized controlled trials (RCTs), it is unknown whether anthocyanin-rich blueberries can attenuate the postprandial, cardiometabolic dysfunction which follows energy-dense food intakes; especially in at-risk populations. We therefore examined whether adding blueberries to a high-fat/high-sugar meal affected the postprandial cardiometabolic response over 24 h. METHODS: A parallel, double-blind RCT (n = 45; age 63.4 ± 7.4 years; 64% male; BMI 31.4 ± 3.1 kg/m2) was conducted in participants with metabolic syndrome. After baseline assessments, an energy-dense drink (969 Kcals, 64.5 g fat, 84.5 g carbohydrate, 17.9 g protein) was consumed with either 26 g (freeze-dried) blueberries (equivalent to 1 cup/150 g fresh blueberries) or 26 g isocaloric matched placebo. Repeat blood samples (30, 60, 90, 120, 180, 360 min and 24 h), a 24 h urine collection and vascular measures (at 3, 6, and 24 h) were performed. Insulin and glucose, lipoprotein levels, endothelial function (flow mediated dilatation (FMD)), aortic and systemic arterial stiffness (pulse wave velocity (PWV), Augmentation Index (AIx) respectively), blood pressure (BP), and anthocyanin metabolism (serum and 24 h urine) were assessed. RESULTS: Blueberries favorably affected postprandial (0-24 h) concentrations of glucose (p < 0.001), insulin (p < 0.01), total cholesterol (p = 0.04), HDL-C, large HDL particles (L-HDL-P) (both p < 0.01), extra-large HDL particles (XL-HDL-P; p = 0.04) and Apo-A1 (p = 0.01), but not LDL-C, TG, or Apo-B. After a transient higher peak glucose concentration at 1 h after blueberry intake ([8.2 mmol/L, 95%CI: 7.7, 8.8] vs placebo [6.9 mmol/L, 95%CI: 6.4, 7.4]; p = 0.001), blueberries significantly attenuated 3 h glucose ([4.3 mmol/L, 95%CI: 3.8, 4.8] vs placebo [5.1 mmol/L, 95%CI: 4.6, 5.6]; p = 0.03) and insulin concentrations (blueberry: [23.4 pmol/L, 95%CI: 15.4, 31.3] vs placebo [52.9 pmol/L, 95%CI: 41.0, 64.8]; p = 0.0001). Blueberries also improved HDL-C ([1.12 mmol/L, 95%CI: 1.06, 1.19] vs placebo [1.08 mmol/L, 95%CI: 1.02, 1.14]; p = 0.04) at 90 min and XL-HDLP levels ([0.38 × 10-6, 95%CI: 0.35, 0.42] vs placebo [0.35 × 10-6, 95%CI: 0.32, 0.39]; p = 0.02) at 3 h. Likewise, significant improvements were observed 6 h after blueberries for HDL-C ([1.17 mmol/L, 95%CI: 1.11, 1.24] vs placebo [1.10 mmol/L, 95%CI: 1.03, 1.16]; p < 0.001), Apo-A1 ([1.37 mmol/L, 95%CI: 1.32, 1.41] vs placebo [1.31 mmol/L, 95%CI: 1.27, 1.35]; p = 0.003), L-HDLP ([0.70 × 10-6, 95%CI: 0.60, 0.81] vs placebo [0.59 × 10-6, 95%CI: 0.50, 0.68]; p = 0.003) and XL-HDLP ([0.44 × 10-6, 95%CI: 0.40, 0.48] vs placebo [0.40 × 10-6, 95%CI: 0.36, 0.44]; p < 0.001). Similarly, total cholesterol levels were significantly lower 24 h after blueberries ([4.9 mmol/L, 95%CI: 4.6, 5.1] vs placebo [5.0 mmol/L, 95%CI: 4.8, 5.3]; p = 0.04). Conversely, no effects were observed for FMD, PWV, AIx and BP. As anticipated, total anthocyanin-derived phenolic acid metabolite concentrations significantly increased in the 24 h after blueberry intake; especially hippuric acid (6-7-fold serum increase, 10-fold urinary increase). In exploratory analysis, a range of serum/urine metabolites were associated with favorable changes in total cholesterol, HDL-C, XL-HDLP and Apo-A1 (R = 0.43 to 0.50). CONCLUSIONS: For the first time, in an at-risk population, we show that single-exposure to the equivalent of 1 cup blueberries (provided as freeze-dried powder) attenuates the deleterious postprandial effects of consuming an energy-dense high-fat/high-sugar meal over 24 h; reducing insulinaemia and glucose levels, lowering cholesterol, and improving HDL-C, fractions of HDL-P and Apo-A1. Consequently, intake of anthocyanin-rich blueberries may reduce the acute cardiometabolic burden of energy-dense meals. CLINICAL TRIAL REGISTRY: NCT02035592 at www.clinicaltrials.gov.


Asunto(s)
Antocianinas/administración & dosificación , Arándanos Azules (Planta) , Ingestión de Energía/efectos de los fármacos , Comidas/efectos de los fármacos , Síndrome Metabólico/metabolismo , Anciano , Antocianinas/sangre , Antocianinas/orina , Glucemia/metabolismo , Presión Sanguínea/efectos de los fármacos , Dieta de Carga de Carbohidratos/efectos adversos , Dieta Alta en Grasa/efectos adversos , Método Doble Ciego , Endotelio Vascular/efectos de los fármacos , Femenino , Humanos , Insulina/sangre , Lipoproteínas/sangre , Masculino , Persona de Mediana Edad , Periodo Posprandial/efectos de los fármacos , Análisis de la Onda del Pulso , Rigidez Vascular/efectos de los fármacos
8.
Biomed Res Int ; 2021: 3532864, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34950731

RESUMEN

Curcumin is a bioactive compound derived from Curcuma longa L. root, extensively studied due to its antioxidant and anti-inflammatory properties. This study evaluates the effects of different doses of powdered C. longa root on antioxidant capacity in healthy men. In a pilot randomized, double-blinded, crossover experiment, we acutely administered a low dose (1.5 g, LCG), moderate dose (3.0 g, MCG), and high dose (6.0 g, HCG) of C. longa to nine healthy men. There were no differences in plasma curcumin levels (p = 0.593) and antioxidant capacity (p = 0.473) for time × group interactions. Plasma curcumin levels increased in all groups after 20 and 90 min of C. longa intake (p < 0.05). HCG had a lower postprandial incremental area under the antioxidant capacity curve than LCG or MCG (p < 0.01). A low dose of C. longa increased the antioxidant capacity in healthy men. However, plasma curcumin levels were not dose dependently affected.


Asunto(s)
Antioxidantes/farmacología , Curcuma/química , Curcumina/farmacología , Extractos Vegetales/farmacología , Antiinflamatorios/farmacología , Estudios Cruzados , Método Doble Ciego , Humanos , Proyectos Piloto , Periodo Posprandial/efectos de los fármacos , Polvos/farmacología
9.
Nutrients ; 13(11)2021 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-34836252

RESUMEN

Postprandial lipemia (PPL) is an important risk factor for cardiovascular disease. Inter-individual variation in the dietary response to a meal is known to be influenced by genetic factors, yet genes that dictate variation in postprandial lipids are not completely characterized. Genetic studies of the plasma lipidome can help to better understand postprandial metabolism by isolating lipid molecular species which are more closely related to the genome. We measured the plasma lipidome at fasting and 6 h after a standardized high-fat meal in 668 participants from the Genetics of Lipid-Lowering Drugs and Diet Network study (GOLDN) using ultra-performance liquid chromatography coupled to (quadrupole) time-of-flight mass spectrometry. A total of 413 unique lipids were identified. Heritable and responsive lipid species were examined for association with single-nucleotide polymorphisms (SNPs) genotyped on the Affymetrix 6.0 array. The most statistically significant SNP findings were replicated in the Amish Heredity and Phenotype Intervention (HAPI) Heart Study. We further followed up findings from GOLDN with a regional analysis of cytosine-phosphate-guanine (CpGs) sites measured on the Illumina HumanMethylation450 array. A total of 132 lipids were both responsive to the meal challenge and heritable in the GOLDN study. After correction for multiple testing of 132 lipids (α = 5 × 10-8/132 = 4 × 10-10), no SNP was statistically significantly associated with any lipid response. Four SNPs in the region of a known lipid locus (fatty acid desaturase 1 and 2/FADS1 and FADS2) on chromosome 11 had p < 8.0 × 10-7 for arachidonic acid FA(20:4). Those SNPs replicated in HAPI Heart with p < 3.3 × 10-3. CpGs around the FADS1/2 region were associated with arachidonic acid and the relationship of one SNP was partially mediated by a CpG (p = 0.005). Both SNPs and CpGs from the fatty acid desaturase region on chromosome 11 contribute jointly and independently to the diet response to a high-fat meal.


Asunto(s)
Genómica , Hipolipemiantes/farmacología , Lipidómica , Periodo Posprandial/efectos de los fármacos , Periodo Posprandial/genética , Adulto , Anciano , delta-5 Desaturasa de Ácido Graso/genética , Ácido Graso Desaturasas/genética , Femenino , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Lípidos , Masculino , Comidas , Persona de Mediana Edad , Fenotipo , Plasma , Polimorfismo de Nucleótido Simple
10.
Am J Physiol Endocrinol Metab ; 321(5): E728-E736, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34658253

RESUMEN

Type 2 diabetes is a disease characterized by impaired insulin secretion and defective glucagon suppression in the postprandial period. We examined the effect of impaired glucagon suppression on glucose concentrations and endogenous glucose production (EGP) at different degrees of insulin secretory impairment. The contribution of anthropometric characteristics, peripheral, and hepatic insulin action to this variability was also examined. To do so, we studied 54 nondiabetic subjects on two occasions in which endogenous hormone secretion was inhibited by somatostatin, with glucagon infused at a rate of 0.65 ng/kg/min, at 0 min to prevent a fall in glucagon (nonsuppressed day) or at 120 min to create a transient fall in glucagon (suppressed day). Subjects received glucose (labeled with [3-3H]-glucose) infused to mimic the systemic appearance of 50-g oral glucose. Insulin was infused to mimic a prandial insulin response in 18 subjects, another 18 received 80% of the dose, and the remaining 18 received 60%. EGP was measured using the tracer-dilution technique. Decreased prandial insulin resulted in greater % increase in peak glucose but not in integrated glucose concentrations attributable to nonsuppressed glucagon. The % change in integrated EGP was unaffected by insulin dose. Multivariate regression analysis, adjusted for age, sex, weight, and insulin dose, did not show a relationship between the EGP response to impaired suppression of glucagon and insulin action as measured at the time of screening by oral glucose tolerance. A similar analysis for hepatic insulin action also did not show a relationship with the EGP response. These data indicate that the effect of impaired glucagon suppression on EGP is independent of anthropometric characteristics and insulin action.NEW & NOTEWORTHY In prediabetes, anthropometric characteristics as well as insulin action do not alter the hepatic response to glucagon. The postprandial suppression or lack of suppression of glucagon secretion is an important factor governing postprandial glucose tolerance independent of insulin secretion.


Asunto(s)
Glucagón/metabolismo , Glucosa/metabolismo , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Somatostatina/farmacología , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Femenino , Glucagón/antagonistas & inhibidores , Glucagón/farmacología , Prueba de Tolerancia a la Glucosa , Voluntarios Sanos , Humanos , Insulina/farmacología , Secreción de Insulina/fisiología , Islotes Pancreáticos/metabolismo , Masculino , Persona de Mediana Edad , Periodo Posprandial/efectos de los fármacos , Periodo Posprandial/fisiología
11.
Neurosci Lett ; 764: 136230, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34500001

RESUMEN

S-Adenosylmethionine (SAM) is the major endogenous methyl donor for methyltransferase reactions, while 5-Azacytidine (AZA) is a synthetic drug inhibiting DNA methyltransferase activity. Both molecules can thus influence DNA methylation patterns in an organism and thereby affect gene expression and ultimately behavior in the long-term. Whether or not effects on behavior are exerted on a shorter time scale is unclear. The goal of this study was to explore the direct effects of SAM and AZA on appetite regulation, using broiler chicken and Japanese quail as the animal models. Fed or 180 min-fasted broilers (at day 4 post-hatch) or 360 min-fasted quail (at day 7 post-hatch) were intracerebroventricularly injected with SAM or AZA and food intake was measured for 360 min. For broilers, there was no effect of AZA, at any dose, on food intake in either fed or fasted chicks at any time point. In contrast, 1 and 10 µg doses of SAM reduced food intake in fed chicks at 60 min post-injection. In fasted chicks, although there were no differences for the first 30 min post-injection, SAM suppressed food intake during the second 30-min period. For quail, however, AZA (25 µg dose) decreased food intake at 60 and 150-360 min post-injection in fasted birds. A reduction in food intake was also observed at 120- and 360-min post-injection in fed quail in response to 5 and 25 µg doses of AZA, respectively. SAM had no effect when quail were fasted, whereas 1 µg dose of SAM suppressed food consumption in fed quail during the third 30-min period. Thus, when administered directly into the central nervous system, SAM may act as a transient appetite suppressant in both broilers and quail, whereas the direct inhibitory effect of AZA on food consumption depends on species and nutritional states.


Asunto(s)
Apetito/efectos de los fármacos , Azacitidina/administración & dosificación , Ingestión de Alimentos/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , S-Adenosilmetionina/administración & dosificación , Animales , Apetito/genética , Pollos/fisiología , Coturnix/fisiología , Metilación de ADN/efectos de los fármacos , Ingestión de Alimentos/genética , Ayuno , Femenino , Inyecciones Intraventriculares , Masculino , Modelos Animales , Periodo Posprandial/efectos de los fármacos , Periodo Posprandial/genética , Especificidad de la Especie
12.
Nutrients ; 13(8)2021 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-34444773

RESUMEN

Structural differences in dietary fatty acids modify their rate of oxidation and effect on satiety, endpoints that may influence the development of obesity. This study tests the hypothesis that meals containing fat sources with elevated unsaturated fats will result in greater postprandial energy expenditure, fat oxidation, and satiety than meals containing fats with greater saturation. In a randomized, 5-way crossover design, healthy men and women (n = 23; age: 25.7 ± 6.6 years; BMI: 27.7 ± 3.8 kg/m2) consumed liquid meals containing 30 g of fat from heavy cream (HC), olive oil (OO), sunflower oil (SFO), flaxseed oil (FSO), and fish oil (FO). Energy expenditure and diet-induced thermogenesis (DIT) were determined by metabolic rate over a 240 min postprandial period. Serum concentrations of ghrelin, glucose, insulin, and triacylglycerol (TAG) were assessed. DIT induced by SFO was 5% lower than HC and FO (p = 0.04). Energy expenditure and substrate oxidation did not differ between fat sources. Postprandial TAG concentrations were significantly affected by fat source (p = 0.0001). Varying fat sources by the degree of saturation and PUFA type modified DIT but not satiety responses in normal to obese adult men and women.


Asunto(s)
Grasas de la Dieta/farmacología , Ácidos Grasos Insaturados/farmacología , Ácidos Grasos/farmacología , Saciedad/efectos de los fármacos , Termogénesis/efectos de los fármacos , Adolescente , Adulto , Estudios Cruzados , Metabolismo Energético/efectos de los fármacos , Grasas/química , Grasas/metabolismo , Grasas/farmacología , Ácidos Grasos/química , Ácidos Grasos/metabolismo , Ácidos Grasos Monoinsaturados/farmacología , Ácidos Grasos Insaturados/química , Femenino , Humanos , Masculino , Comidas , Persona de Mediana Edad , Obesidad/metabolismo , Aceite de Oliva/farmacología , Oxidación-Reducción , Periodo Posprandial/efectos de los fármacos , Respuesta de Saciedad/efectos de los fármacos , Adulto Joven
13.
Front Endocrinol (Lausanne) ; 12: 696977, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34220720

RESUMEN

Purpose: Elevated postprandial glycaemia [PPG] increases the risk of cardiometabolic complications in insulin-resistant, centrally obese individuals. Therefore, strategies that improve PPG are of importance for this population. Consuming large doses of whey protein [WP] before meals reduces PPG by delaying gastric emptying and stimulating the secretion of the incretin peptides, glucose-dependent insulinotropic polypeptide [GIP] and glucagon-like peptide 1 [GLP-1]. It is unclear if these effects are observed after smaller amounts of WP and what impact central adiposity has on these gastrointestinal processes. Methods: In a randomised-crossover design, 12 lean and 12 centrally obese adult males performed two 240 min mixed-meal tests, ~5-10 d apart. After an overnight fast, participants consumed a novel, ready-to-drink WP shot (15 g) or volume-matched water (100 ml; PLA) 10 min before a mixed-nutrient meal. Gastric emptying was estimated by oral acetaminophen absorbance. Interval blood samples were collected to measure glucose, insulin, GIP, GLP-1, and acetaminophen. Results: WP reduced PPG area under the curve [AUC0-60] by 13 and 18.2% in the centrally obese and lean cohorts, respectively (both p <0.001). In both groups, the reduction in PPG was accompanied by a two-three-fold increase in GLP-1 and delayed gastric emptying. Despite similar GLP-1 responses during PLA, GLP-1 secretion during the WP trial was ~27% lower in centrally obese individuals compared to lean (p = 0.001). In lean participants, WP increased the GLP-1ACTIVE/TOTAL ratio comparative to PLA (p = 0.004), indicative of reduced GLP-1 degradation. Conversely, no treatment effects for GLP-1ACTIVE/TOTAL were seen in obese subjects. Conclusion: Pre-meal ingestion of a novel, ready-to-drink WP shot containing just 15 g of dietary protein reduced PPG in lean and centrally obese males. However, an attenuated GLP-1 response to mealtime WP and increased incretin degradation might impact the efficacy of nutritional strategies utilising the actions of GLP-1 to regulate PPG in centrally obese populations. Whether these defects are caused by an individual's insulin resistance, their obese state, or other obesity-related ailments needs further investigation. Clinical Trial Registration: ISRCTN.com, identifier [ISRCTN95281775]. https://www.isrctn.com/.


Asunto(s)
Glucemia/metabolismo , Hormonas Gastrointestinales/metabolismo , Obesidad Abdominal/dietoterapia , Proteína de Suero de Leche/farmacología , Adulto , Glucemia/efectos de los fármacos , Péptido C/sangre , Estudios Cruzados , Ingestión de Alimentos , Inglaterra , Alimentos Formulados , Vaciamiento Gástrico/fisiología , Polipéptido Inhibidor Gástrico/sangre , Polipéptido Inhibidor Gástrico/efectos de los fármacos , Glucagón/sangre , Péptido 1 Similar al Glucagón/sangre , Péptido 1 Similar al Glucagón/efectos de los fármacos , Humanos , Insulina/sangre , Masculino , Persona de Mediana Edad , Obesidad Abdominal/sangre , Obesidad Abdominal/metabolismo , Periodo Posprandial/efectos de los fármacos , Delgadez/sangre , Delgadez/metabolismo , Proteína de Suero de Leche/administración & dosificación , Adulto Joven
14.
Nutrients ; 13(7)2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-34202167

RESUMEN

In a recent study, we showed that konjac glucomannan (KGM) inhibits rice gruel-induced postprandial increases in plasma glucose and insulin levels. To extend this research, we investigated the effects of KGM addition to rice gruel on pre- and postprandial concentrations of circulating lipoprotein lipase (LPL), glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 (GPIHBP1), hepatic triglyceride lipase (HTGL), free fatty acids (FFA), and triglycerides (TG). A total of 13 Japanese men, without diabetes, dyslipidemia, or gastrointestinal diseases, interchangeably ingested rice gruel containing no KGM (0%G), rice gruel supplemented with 0.4% KGM (0.4%G), and rice gruel supplemented with 0.8% KGM (0.8%G), every Sunday for 3 weeks. Blood samples were obtained at baseline and at 30, 60, and 120 min after ingestion to measure the abovementioned lipid parameters. Lipid parameters showed small, but significant, changes. Significant reductions were found in circulating FFA levels among all participants. Circulating TG levels significantly declined at 30 min and then remained nearly constant in the 0.8%G group but exhibited no significant difference in the 0%G and 0.4%G groups. Although circulating levels of LPL and GPIHBP1 significantly decreased in the 0%G and 0.4%G groups, they increased at 120 min in the 0.8%G group. Participants in the 0%G and 0.4%G groups showed significant decreases in circulating HTGL levels, which was not observed in the 0.8%G group. Our results demonstrate the novel pleiotropic effects of KGM. Supplementation of rice gruel with KGM powder led to TG reduction accompanied by LPL and GPIHBP1 elevation and HTGL stabilization, thereby attenuating TG metabolism.


Asunto(s)
Suplementos Dietéticos , Grano Comestible , Mananos , Oryza , Triglicéridos/sangre , Adulto , Estudios Cruzados , Método Doble Ciego , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Lipoproteína Lipasa/sangre , Masculino , Persona de Mediana Edad , Periodo Posprandial/efectos de los fármacos , Polvos , Receptores de Lipoproteína/sangre
15.
Mol Nutr Food Res ; 65(17): e2100487, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34288404

RESUMEN

SCOPE: Despite its beneficial properties, higher adiponectin concentrations are paradoxically associated with mortality in advanced age. Several mechanisms are being discussed. However, little is known about postprandial regulation of adiponectin in older adults. We assessed age-specific differences of the adiponectin response to different test meals considering potential determinants. METHODS AND RESULTS: Older (n = 20) and younger (n = 22) women are randomized to a dextrose (DEX) or high-fat (HF) dietary challenge. Postprandial adiponectin and fibroblast growth factor 21 (FGF21) concentrations are measured before and 60, 120, 240 min after ingestion. We assessed postprandial changes and group differences using linear mixed models controlled for possible determinants. In younger women, postprandial adiponectin remains stable after both test meals. In contrast, adiponectin increases following DEX and decreases after HF in older women, irrespective of control variables. Postprandial adiponectin is positively associated with malondialdehyde and inversely associated with interleukin-6 following DEX and also negatively associated with metabolic parameters after both test meals. In older women, elevated postprandial FGF21 concentrations are associated with a higher adiponectin response (ß = 30.7, 95% CI 10.6-50.8, p = 0.007). CONCLUSIONS: Adiponectin response is associated with type of dietary challenge, age, and FGF21 response. Age-group differences are partly attributable to metabolic parameters and oxidative stress.


Asunto(s)
Adiponectina/sangre , Dieta Alta en Grasa/efectos adversos , Periodo Posprandial/fisiología , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Ayuno , Femenino , Factores de Crecimiento de Fibroblastos/sangre , Glucosa/efectos adversos , Humanos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Periodo Posprandial/efectos de los fármacos
16.
Nutrients ; 13(5)2021 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-34064996

RESUMEN

(1) Background: Whey protein lowers postprandial blood glucose in health and type 2 diabetes, by stimulating insulin and incretin hormone secretion and slowing gastric emptying. The branched-chain amino acids, leucine, isoleucine and valine, abundant in whey, may mediate the glucoregulatory effects of whey. We investigated the comparative effects of intragastric administration of leucine, isoleucine and valine on the plasma glucose, C-peptide and glucagon responses to and gastric emptying of a mixed-nutrient drink in healthy men. (2) Methods: 15 healthy men (27 ± 3 y) received, on four separate occasions, in double-blind, randomised fashion, either 10 g of leucine, 10 g of isoleucine, 10 g of valine or control, intragastrically, 30 min before a mixed-nutrient drink. Plasma glucose, C-peptide and glucagon concentrations were measured before, and for 2 h following, the drink. Gastric emptying of the drink was quantified using 13C-acetate breath-testing. (3) Results: Amino acids alone did not affect plasma glucose or C-peptide, while isoleucine and valine, but not leucine, stimulated glucagon (p < 0.05), compared with control. After the drink, isoleucine and leucine reduced peak plasma glucose compared with both control and valine (all p < 0.05). Neither amino acid affected early (t = 0-30 min) postprandial C-peptide or glucagon. While there was no effect on overall gastric emptying, plasma glucose at t = 30 min correlated with early gastric emptying (p < 0.05). (4) Conclusion: In healthy individuals, leucine and isoleucine lower postprandial blood glucose, at least in part by slowing gastric emptying, while valine does not appear to have an effect, possibly due to glucagon stimulation.


Asunto(s)
Aminoácidos de Cadena Ramificada/farmacología , Glucemia/metabolismo , Péptido C/sangre , Vaciamiento Gástrico/efectos de los fármacos , Glucagón/sangre , Isoleucina/farmacología , Leucina/farmacología , Valina/farmacología , Adulto , Diabetes Mellitus Tipo 2 , Método Doble Ciego , Polipéptido Inhibidor Gástrico/sangre , Humanos , Insulina , Masculino , Persona de Mediana Edad , Periodo Posprandial/efectos de los fármacos , Proteína de Suero de Leche/farmacología , Adulto Joven
17.
Nutrients ; 13(5)2021 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-34065380

RESUMEN

Fasting and postprandial hypertriglyceridemia are causal risk factors for atherosclerosis. The prevalence of hypertriglyceridemia is approximately 25-30% and most hypertriglyceridemic patients suffer from mild to moderate hypertriglyceridemia. Data regarding dietary interventions on postprandial triglyceride metabolism of mildly to moderately hypertriglyceridemic patients is, however, sparse. In a randomized controlled trial, eight mildly hypertriglyceridemic patients and five healthy, normolipidemic controls received three separate standardized fat-meals containing either saturated fatty acids (SFA), mono-unsaturated fatty acids (MUFA), or medium-chain fatty acids (MCFA) in a randomized order. Fasting and postprandial lipid parameters were determined over a 10 h period and the (incremental) area under the curve (AUC/iAUC) for plasma triglycerides and other parameters were determined. MCFA do not lead to a significant elevation of postprandial total plasma triglycerides and other triglyceride parameters, while both SFA (patients: p = 0.003, controls: p = 0.03 compared to MCFA) and MUFA (patients: p = 0.001; controls: p = 0.14 compared to MCFA) do lead to such an increase. Patients experienced a significantly more pronounced increase of plasma triglycerides than controls (SFA: patients iAUC = 1006 mg*h/dL, controls iAUC = 247 mg*h/dL, p = 0.02; MUFA: patients iAUC = 962 mg*h/dL, controls iAUC = 248 mg*h/dL, p = 0.05). Replacing SFA with MCFA may be a treatment option for mildly to moderately hypertriglyceridemic patients as it prevents postprandial hypertriglyceridemia.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Hipertrigliceridemia/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Comidas/fisiología , Periodo Posprandial/efectos de los fármacos , Adulto , Ayuno/sangre , Ácidos Grasos/administración & dosificación , Ácidos Grasos Monoinsaturados/administración & dosificación , Femenino , Humanos , Lípidos/sangre , Masculino , Persona de Mediana Edad , Triglicéridos/sangre
18.
Am J Physiol Endocrinol Metab ; 320(6): E1119-E1137, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33938234

RESUMEN

Global prevalence of type 2 diabetes (T2D) is rising and may affect 700 million people by 2045. Totum-63 is a polyphenol-rich natural composition developed to reduce the risk of T2D. We first investigated the effects of Totum-63 supplementation in high-fat diet (HFD)-fed mice for up to 16 wk and thereafter assessed its safety and efficacy (2.5 g or 5 g per day) in 14 overweight men [mean age 51.5 yr, body mass index (BMI) 27.6 kg·m-2] for 4 wk. In HFD-fed mice, Totum-63 reduced body weight and fat mass gain, whereas lean mass was unchanged. Moreover, fecal energy excretion was higher in Totum-63-supplemented mice, suggesting a reduction of calorie absorption in the digestive tract. In the gut, metagenomic analyses of fecal microbiota revealed a partial restoration of HFD-induced microbial imbalance, as shown by principal coordinate analysis of microbiota composition. HFD-induced increase in HOMA-IR score was delayed in supplemented mice, and insulin response to an oral glucose tolerance test was significantly reduced, suggesting that Totum-63 may prevent HFD-related impairments in glucose homeostasis. Interestingly, these improvements could be linked to restored insulin signaling in subcutaneous adipose tissue and soleus muscle. In the liver, HFD-induced steatosis was reduced by 40% (as shown by triglyceride content). In the subsequent study in men, Totum-63 (5 g·day-1) improved glucose and insulin responses to a high-carbohydrate breakfast test (84% kcal carbohydrates). It was well tolerated, with no clinically significant adverse events reported. Collectively, these data suggest that Totum-63 could improve glucose homeostasis in both HFD-fed mice and overweight individuals, presumably through a multitargeted action on different metabolic organs.NEW & NOTEWORTHY Totum-63 is a novel polyphenol-rich natural composition developed to reduce the risk of T2D. Totum-63 showed beneficial effects on glucose homeostasis in HFD-fed mice, presumably through a multitargeted action on different metabolic organs. Totum-63 was well tolerated in humans and improved postprandial glucose and insulin responses to a high-carbohydrate breakfast test.


Asunto(s)
Glucemia/efectos de los fármacos , Hiperglucemia/prevención & control , Extractos Vegetales/farmacología , Adulto , Animales , Glucemia/metabolismo , Chrysanthemum/química , Cynara scolymus/química , Control Glucémico/métodos , Homeostasis/efectos de los fármacos , Humanos , Hiperglucemia/sangre , Hiperglucemia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Olea/química , Sobrepeso/sangre , Sobrepeso/tratamiento farmacológico , Sobrepeso/metabolismo , Proyectos Piloto , Piper nigrum/química , Extractos Vegetales/química , Extractos Vegetales/uso terapéutico , Periodo Posprandial/efectos de los fármacos , Investigación Biomédica Traslacional , Vaccinium myrtillus/química
19.
Food Funct ; 12(12): 5399-5406, 2021 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-33988204

RESUMEN

Inhibition of glucose uptake in the intestine through sodium-dependent glucose transporter 1 (SGLT1) or glucose transporter 2 (GLUT2) may be beneficial in controlling postprandial blood glucose levels. Gallic acid and ten of its derivatives were identified in the active fractions of Terminalia chebula Retz. fructus immaturus, a popular edible plant fruit which has previously been associated with the inhibition of glucose uptake. Gallic acid derivatives (methyl gallate, ethyl gallate, pentyl gallate, 3,4,6-tri-O-galloyl-ß-d-glucose, and corilagin) showed good glucose transport inhibition with inhibitory rates of 72.1 ± 1.6%, 71.5 ± 1.4%, 79.9 ± 1.2%, 44.7 ± 1.2%, and 75.0 ± 0.7% at 5 mM d-glucose and/or 56.3 ± 2.3, 52.1 ± 3.2%, 70.2 ± 1.7%, 15.6 ± 1.6%, and 37.1 ± 0.8% at 25 mM d-glucose. However, only 3,4,6-tri-O-galloyl-ß-d-glucose and corilagin were confirmed GLUT2-specific inhibitors. Whilst some tea flavonoids demonstrated minimal glucose transport inhibition, their gallic acid derivatives strongly inhibited transport effect with GLUT2 specificity. This suggests that gallic acid structures are crucial for glucose transport inhibition. Plants, such as T. chebula, which contain high levels of gallic acid and its derivatives, show promise as natural functional ingredients for inclusion in foods and drinks designed to control postprandial glucose levels.


Asunto(s)
Transporte Biológico/efectos de los fármacos , Ácido Gálico/química , Ácido Gálico/farmacología , Glucosa/metabolismo , Extractos Vegetales/farmacología , Periodo Posprandial/efectos de los fármacos , Células CACO-2 , Flavonoides , Frutas/química , Ácido Gálico/análogos & derivados , Transportador de Glucosa de Tipo 2 , Glucósidos , Humanos , Taninos Hidrolizables , Intestinos , Transportador 1 de Sodio-Glucosa , Terminalia/efectos de los fármacos
20.
J Diabetes Investig ; 12(12): 2162-2171, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34022121

RESUMEN

AIMS/INTRODUCTION: Differences in the glucose-lowering mechanisms of glucagon-like peptide-1 receptor agonists (GLP-1RAs) have been noted. Clarifying these differences could facilitate the choice of optimal drugs for individuals with type 2 diabetes and requires investigation in a clinical setting. MATERIALS AND METHODS: A single-arm, prospective, observational study was conducted to evaluate the effects of various GLP-1RAs on postprandial glucose excursion, secretions of insulin and glucagon as well as on the gastric emptying rate. Participants were subjected to meal tolerance tests before and 2 weeks and 12 weeks after GLP-1RA initiation. Effects on postprandial secretions of glucose-dependent insulinotropic polypeptide (GIP) and apolipoprotein B48 were also investigated. RESULTS: Eighteen subjects with type 2 diabetes received one of three GLP-1RAs, i.e., lixisenatide, n = 7; liraglutide, n = 6; or dulaglutide, n = 5. While 12-week administration of all of the GLP-1RAs significantly reduced HbA1c, only lixisenatide and liraglutide, but not dulaglutide, significantly reduced body weight. Postprandial glucose elevation was improved by all of the GLP-1RAs. Postprandial insulin levels were suppressed by lixisenatide, while insulin levels were enhanced by liraglutide. Postprandial glucagon levels were suppressed by lixisenatide. The gastric emptying rate was significantly delayed by lixisenatide, while liraglutide and dulaglutide had limited effects on gastric emptying. GIP secretion was suppressed by lixisenatide and liraglutide. Apolipoprotein B48 secretion was suppressed by all of the GLP-1RAs. CONCLUSIONS: All of the GLP-1RAs were found to improve HbA1c in a 12-week prospective observational study in Japanese individuals with type 2 diabetes. However, differences in the mechanisms of the glucose-lowering effects and body weight reduction were observed.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Vaciamiento Gástrico/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/agonistas , Hipoglucemiantes/farmacología , Adulto , Apolipoproteína B-48/metabolismo , Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/sangre , Femenino , Polipéptido Inhibidor Gástrico/metabolismo , Glucagón/efectos de los fármacos , Péptidos Similares al Glucagón/análogos & derivados , Péptidos Similares al Glucagón/farmacología , Humanos , Fragmentos Fc de Inmunoglobulinas/farmacología , Insulina/sangre , Japón , Liraglutida/farmacología , Masculino , Persona de Mediana Edad , Péptidos/farmacología , Periodo Posprandial/efectos de los fármacos , Estudios Prospectivos , Proteínas Recombinantes de Fusión/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA