Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nutrients ; 13(10)2021 Sep 27.
Article in English | MEDLINE | ID: mdl-34684407

ABSTRACT

BACKGROUND: To investigate the potential synergistic effects of olive oil releasing 2-oleoylglycerol and hydrolyzed pine nut oil containing 20% pinolenic acid on GLP-1 secretion, glucose tolerance, insulin secretion and appetite in healthy individuals, when delivered to the small intestine as potential agonists of GPR119, FFA1 and FFA4. METHODS: Nine overweight/obese individuals completed three 6-h oral glucose tolerance tests (OGTTs) in a crossover design. At -30 min, participants consumed either: no oil, 6 g of hydrolyzed pine nut oil (PNO-FFA), or a combination of 3 g hydrolyzed pine nut oil and 3 g olive oil (PNO-OO) in delayed-release capsules. Repeated measures of glucose, insulin, C-peptide, GLP-1, GIP, ghrelin, subjective appetite and gastrointestinal tolerability were done. RESULTS: PNO-FFA augmented GLP-1 secretion from 0-360 min compared to no oil and PNO-OO (p < 0.01). GIP secretion was increased from 240-360 min after both PNO-FFA and PNO-OO versus no oil (p < 0.01). Both oil treatments suppressed subjective appetite by reducing hunger and prospective food consumption and increasing satiety (p < 0.05). CONCLUSIONS: In support of previous findings, 6 g of delayed-release hydrolyzed pine nut oil enhanced postprandial GLP-1 secretion and reduced appetite. However, no synergistic effect of combining hydrolyzed pine nut oil and olive oil on GLP-1 secretion was observed. These results need further evaluation in long-term studies including effects on bodyweight and insulin sensitivity.


Subject(s)
Appetite , Blood Glucose/metabolism , Dietary Fats, Unsaturated/administration & dosage , Glucagon-Like Peptide 1/blood , Incretins/blood , Olive Oil/administration & dosage , Plant Oils/administration & dosage , Cross-Over Studies , Delayed-Action Preparations , Dietary Supplements , Female , Glucose Tolerance Test , Humans , Insulin/blood , Male , Middle Aged , Nuts , Obesity/metabolism , Overweight/metabolism , Pinus , Postprandial Period
2.
Physiol Behav ; 234: 113315, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33460676

ABSTRACT

Triacylglycerol is the most abundant dietary lipid, and a strong stimulator of satiation. Absorption of triacylglycerol in the small intestine occurs in the form of free fatty acids and 2-monoacylglycerol, a process known to trigger not only the release of cholecystokinin (CCK) but also glucagon-like peptide 1 (GLP-1) and peptide YY (PYY). It remains controversial, however, whether endogenously released GLP-1 and PYY are required for fat-induced satiation. Using a self-administration model where mice are trained to self-administer Intralipid 30% intragastrically, we show that blocking the CCK1 receptors with intraperitoneal devazepide diminishes the post-oral satiation effect of ingested fat. Similarly, s.c. administration of a GLP-1 receptor antagonist with a prolonged half-life (Jant4-C16) also reduced the post-oral satiation effect of ingested fat. Importantly, coadministration of the GLP-1 antagonist together with devazepide increased fat self-infusions to a level equal to the combined blockade of each individual peptide action alone, indicating an additive effect of endogenous CCK and GLP-1 in fat satiation signaling. Blocking the PYY Y2 receptor did not further enhance the fat intake in devazepide-treated mice. Consistent with the above, we show that voluntary post-oral ingestion of fat increases CCK and GLP-1 plasma levels and is correlated positively with CCK and GLP-1 plasma concentrations. Taken together, our results support the role of endogenous GLP-1 in the regulation of fat intake and suggest that both CCK and GLP-1 are required for the fat satiation signaling.


Subject(s)
Cholecystokinin , Glucagon-Like Peptide 1 , Animals , Eating , Mice , Peptide YY , Receptors, Cholecystokinin , Satiation
3.
Clin Nutr ; 40(4): 2169-2179, 2021 04.
Article in English | MEDLINE | ID: mdl-33059911

ABSTRACT

BACGROUND & AIM: Pinolenic acid, a major component (~20%) of pine nut oil, is a dual agonist of the free fatty acid receptors, FFA1 and FFA4, which may regulate release of incretins and ghrelin from the gut. Here, we investigated the acute effects of hydrolyzed pine nut oil (PNO-FFA), delivered to the small intestine by delayed-release capsules, on glucose tolerance, insulin, incretin and ghrelin secretion, and appetite. METHODS: In two cross-over studies, we evaluated 3 g unhydrolyzed pine nut oil (PNO-TG) or 3 g PNO-FFA versus no oil in eight healthy, non-obese subjects (study 1), and 3 g PNO-FFA or 6 g PNO-FFA versus no oil in ten healthy, overweight/obese subjects (study 2) in both studies given in delayed-release capsules 30 min prior to a 4-h-oral glucose tolerance test (OGTT). Outcomes were circulating levels of glucose, insulin, GLP-1, GIP, ghrelin, appetite and gastrointestinal tolerability during OGTT. RESULTS: Both 3 g PNO-FFA in study 1 and 6 g PNO-FFA in study 2 markedly increased GLP-1 levels (p < 0.001) and attenuated ghrelin levels (p < 0.001) during the last 2 h of the OGTT compared with no oil. In study 2, these effects of PNO-FFA were accompanied by an increased satiety and fullness (p < 0.03), and decreased prospective food consumption (p < 0.05). PNO-FFA caused only small reductions in glucose and insulin levels during the first 2 h of the OGTT. CONCLUSIONS: Our results provide evidence that PNO-FFA delivered to the small intestine by delayed-release capsules may reduce appetite by augmenting GLP-1 release and attenuating ghrelin secretion in the late postprandial state. CLINICAL TRIAL REGISTRY NUMBERS: NCT03062592 and NCT03305367.


Subject(s)
Appetite/drug effects , Ghrelin/blood , Glucose Tolerance Test , Incretins/blood , Pinus , Plant Oils/administration & dosage , Adult , Aged , Blood Glucose/analysis , C-Peptide/blood , Cross-Over Studies , Delayed-Action Preparations , Diabetes Mellitus, Type 2/physiopathology , Female , Gastric Inhibitory Polypeptide/blood , Glucagon-Like Peptide 1/blood , Humans , Hydrolysis , Insulin/blood , Intestine, Small/drug effects , Linolenic Acids/administration & dosage , Male , Middle Aged , Plant Oils/chemistry , Seeds
4.
Mol Metab ; 42: 101103, 2020 12.
Article in English | MEDLINE | ID: mdl-33091626

ABSTRACT

OBJECTIVES: Long-chain fatty acids (LCFAs) released from adipocytes inhibit lipolysis through an unclear mechanism. We hypothesized that the LCFA receptor, FFAR4 (GPR120), which is highly expressed in adipocytes, may be involved in this feedback regulation. METHODS AND RESULTS: Liquid chromatography mass spectrometry (LC-MS) analysis of conditioned media from isoproterenol-stimulated primary cultures of murine and human adipocytes demonstrated that most of the released non-esterified free fatty acids (NEFAs) are known agonists for FFAR4. In agreement with this, conditioned medium from isoproterenol-treated adipocytes stimulated signaling strongly in FFAR4 transfected COS-7 cells as opposed to non-transfected control cells. In transfected 3T3-L1 cells, FFAR4 agonism stimulated Gi- and Go-mini G protein binding more strongly than Gq, effects which were blocked by the selective FFAR4 antagonist AH7614. In primary cultures of murine white adipocytes, the synthetic, selective FFAR4 agonist CpdA inhibited isoproterenol-induced intracellular cAMP accumulation in a manner similar to the antilipolytic control agent nicotinic acid acting through another receptor, HCAR2. In vivo, oral gavage with the synthetic, specific FFAR4 agonist CpdB decreased the level of circulating NEFAs in fasting lean mice to a similar degree as nicotinic acid. In agreement with the identified anti-lipolytic effect of FFAR4, plasma NEFAs and glycerol were increased in FFAR4-deficient mice as compared to littermate controls despite having elevated insulin levels, and cAMP accumulation in primary adipocyte cultures was augmented by treatment with the FFAR4 antagonist conceivably by blocking the stimulatory tone of endogenous NEFAs on FFAR4. CONCLUSIONS: In white adipocytes, FFAR4 functions as an NEFA-activated, autocrine, negative feedback regulator of lipolysis by decreasing cAMP though Gi-mediated signaling.


Subject(s)
Fatty Acids, Nonesterified/metabolism , Receptors, G-Protein-Coupled/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipocytes, White/metabolism , Adipose Tissue/metabolism , Adipose Tissue, White/metabolism , Animals , Autocrine Communication/physiology , Chromatography, Liquid/methods , Culture Media, Conditioned/pharmacology , Fatty Acids/metabolism , Feedback, Physiological/physiology , Female , Humans , Lipolysis/physiology , Male , Mass Spectrometry/methods , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/physiology
5.
Development ; 145(17)2018 09 03.
Article in English | MEDLINE | ID: mdl-30093553

ABSTRACT

Mutations in Hes1, a target gene of the Notch signalling pathway, lead to ectopic pancreas by a poorly described mechanism. Here, we use genetic inactivation of Hes1 combined with lineage tracing and live imaging to reveal an endodermal requirement for Hes1, and show that ectopic pancreas tissue is derived from the dorsal pancreas primordium. RNA-seq analysis of sorted E10.5 Hes1+/+ and Hes1-/- Pdx1-GFP+ cells suggested that upregulation of endocrine lineage genes in Hes1-/- embryos was the major defect and, accordingly, early pancreas morphogenesis was normalized, and the ectopic pancreas phenotype suppressed, in Hes1-/-Neurog3-/- embryos. In Mib1 mutants, we found a near total depletion of dorsal progenitors, which was replaced by an anterior Gcg+ extension. Together, our results demonstrate that aberrant morphogenesis is the cause of ectopic pancreas and that a part of the endocrine differentiation program is mechanistically involved in the dysgenesis. Our results suggest that the ratio of endocrine lineage to progenitor cells is important for morphogenesis and that a strong endocrinogenic phenotype without complete progenitor depletion, as seen in Hes1 mutants, provokes an extreme dysgenesis that causes ectopic pancreas.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Choristoma/genetics , Morphogenesis/genetics , Nerve Tissue Proteins/genetics , Pancreas/abnormalities , Pancreas/embryology , Transcription Factor HES-1/genetics , Animals , Endoderm/metabolism , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Ubiquitin-Protein Ligases/genetics
6.
Endocrinology ; 157(12): 4561-4569, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27779915

ABSTRACT

Triglycerides (TGs) are among the most efficacious stimulators of incretin secretion; however, the relative importance of FFA1 (G Protein-coupled Receptor [GPR] 40), FFA4 (GPR120), and GPR119, which all recognize TG metabolites, ie, long-chain fatty acid and 2-monoacylglycerol, respectively, is still unclear. Here, we find all 3 receptors to be highly expressed and highly enriched in fluorescence-activated cell sorting-purified GLP-1 and GIP cells isolated from transgenic reporter mice. In vivo, the TG-induced increase in plasma GIP was significantly reduced in FFA1-deficient mice (to 34%, mean of 4 experiments each with 8-10 animals), in GPR119-deficient mice (to 24%) and in FFA1/FFA4 double deficient mice (to 15%) but not in FFA4-deficient mice. The TG-induced increase in plasma GLP-1 was only significantly reduced in the GPR119-deficient and the FFA1/FFA4 double deficient mice, but not in the FFA1, and FFA4-deficient mice. In mouse colonic crypt cultures the synthetic FFA1 agonists, TAK-875 stimulated GLP-1 secretion to a similar extent as the prototype GLP-1 secretagogue neuromedin C; this, however, only corresponded to approximately half the maximal efficiency of the GPR119 agonist AR231453, whereas the GPR120 agonist Metabolex-209 had no effect. Importantly, when the FFA1 agonist was administered on top of appropriately low doses of the GPR119 agonist, a clear synergistic, ie, more than additive, effect was observed. It is concluded that the 2-monoacylglycerol receptor GPR119 is at least as important as the long-chain fatty acid receptor FFA1 in mediating the TG-induced secretion of incretins and that the 2 receptors act in synergy, whereas FFA4 plays a minor if any role.


Subject(s)
Colon/metabolism , Receptors, G-Protein-Coupled/metabolism , Triglycerides/metabolism , Animals , Benzofurans/pharmacology , Bombesin/pharmacology , Colon/drug effects , Dietary Fats , Glucagon-Like Peptide 1/metabolism , Mice , Mice, Knockout , Peptide Fragments/pharmacology , Receptors, G-Protein-Coupled/genetics , Sulfones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...