Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Eur J Nutr ; 60(8): 4439-4452, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34081167

ABSTRACT

PURPOSE: This study investigated metabolic benefits of protein hydrolysates from the macroalgae Palmaria palmata, previously shown to inhibit dipeptidylpeptidase-4 (DPP-4) activity in vitro. METHODS: Previously, Alcalase/Flavourzyme-produced P. palmata protein hydrolysate (PPPH) improved glycaemia and insulin production in streptozotocin-induced diabetic mice. Here the PPPH, was compared to alternative Alcalase, bromelain and Promod-derived hydrolysates and an unhydrolysed control. All PPPH's underwent simulated gastrointestinal digestion (SGID) to establish oral bioavailability. PPPH's and their SGID counterparts were tested in pancreatic, clonal BRIN-BD11 cells to assess their insulinotropic effect and associated intracellular mechanisms. PPPH actions on the incretin effect were assessed via measurement of DPP-4 activity, coupled with GLP-1 and GIP release from GLUTag and STC-1 cells, respectively. Acute in vivo effects of Alcalase/Flavourzyme PPPH administration on glucose tolerance and satiety were assessed in overnight-fasted mice. RESULTS: PPPH's (0.02-2.5 mg/ml) elicited varying insulinotropic effects (p < 0.05-0.001). SGID of the unhydrolysed protein control, bromelain and Promod PPPH's retained, or improved, bioactivity regarding insulin secretion, DPP-4 inhibition and GIP release. Insulinotropic effects were retained for all SGID-hydrolysates at higher PPPH concentrations. DPP-4 inhibitory effects were confirmed for all PPPH's and SGID counterparts (p < 0.05-0.001). PPPH's were shown to directly influence the incretin effect via upregulated GLP-1 and GIP (p < 0.01-0.001) secretion in vitro, largely retained after SGID. Alcalase/Flavourzyme PPPH produced the greatest elevation in cAMP (p < 0.001, 1.7-fold), which was fully retained post-SGID. This hydrolysate elicited elevations in intracellular calcium (p < 0.01) and membrane potential (p < 0.001). In acute in vivo settings, Alcalase/Flavourzyme PPPH improved glucose tolerance (p < 0.01-0.001) and satiety (p < 0.05-0.001). CONCLUSION: Bioavailable PPPH peptides may be useful for the management of T2DM and obesity.


Subject(s)
Diabetes Mellitus, Experimental , Glucagon-Like Peptide 1 , Animals , Blood Glucose , Gastric Inhibitory Polypeptide , Incretins , Insulin/metabolism , Mice , Protein Hydrolysates , Up-Regulation
2.
Peptides ; 100: 219-228, 2018 02.
Article in English | MEDLINE | ID: mdl-29412822

ABSTRACT

Nine structurally modified apelin-13 analogues were assessed for their in vitro and acute in vivo antidiabetic potential. Stability was assessed in mouse plasma and insulinotropic efficacy tested in cultured pancreatic BRIN-BD11 cells and isolated mouse pancreatic islets. Intracellular Ca2+ and cAMP production in BRIN-BD11 cells was determined, as was glucose uptake in 3T3-L1 adipocytes. Acute antihyperglycemic effects of apelin analogues were assessed following i.p. glucose tolerance tests (ipGGT, 18 mmol/kg) in normal and diet-induced-obese (DIO) mice and on food intake in normal mice. Apelin analogues all showed enhanced in vitro stability (up to 5.8-fold, t½â€¯= 12.8 h) in mouse plasma compared to native apelin-13 (t½â€¯= 2.1 h). Compared to glucose controls, stable analogues exhibited enhanced insulinotropic responses from BRIN-BD11 cells (up to 4.7-fold, p < 0.001) and isolated mouse islets (up to 5.3-fold) for 10-7 M apelin-13 amide (versus 7.6-fold for 10-7 M GLP-1). Activation of APJ receptors on BRIN-BD11 cells increased intracellular Ca2+ (up to 3.0-fold, p < 0.001) and cAMP (up to 1.7-fold, p < 0.01). Acute ipGTT showed improved insulinotropic and glucose disposal responses in normal and DIO mice (p < 0.05 and p < 0.01, respectively). Apelin-13 amide and (pGlu)apelin-13 amide were the most effective analogues exhibiting acute, dose-dependent and persistent biological actions. Both analogues stimulated insulin-independent glucose uptake by differentiated adipocytes (2.9-3.3-fold, p < 0.05) and inhibited food intake (26-33%, p < 0.001), up to 180 min in mice, versus saline. In contrast, (Ala13)apelin-13 and (Val13)apelin-13 inhibited insulin secretion, suppressed beta-cell signal transduction and stimulated food intake in mice. Thus, stable analogues of apelin-13 have potential for diabetes/obesity therapy.


Subject(s)
Hypoglycemic Agents/administration & dosage , Insulin/metabolism , Intercellular Signaling Peptides and Proteins/administration & dosage , Obesity/drug therapy , 3T3-L1 Cells , Animals , Blood Glucose/drug effects , Cyclic AMP/metabolism , Gastric Inhibitory Polypeptide/chemistry , Glucagon/metabolism , Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Glucose Tolerance Test , Humans , Hypoglycemic Agents/chemistry , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Intercellular Signaling Peptides and Proteins/chemistry , Mice , Mice, Obese , Obesity/metabolism , Obesity/pathology , Rats , Receptors, Glucagon/metabolism
3.
Diabetes Obes Metab ; 18(10): 1013-24, 2016 10.
Article in English | MEDLINE | ID: mdl-27357054

ABSTRACT

AIMS: To investigate the antidiabetic actions of three dogfish glucagon peptide analogues [known glucagon-like peptide-1 and glucagon receptor co-agonists] after chronic administration in diet-induced high-fat-diet-fed diabetic mice. MATERIALS AND METHODS: National Institutes of Health Swiss mice were pre-conditioned to a high-fat diet (45% fat) for 100 days, and control mice were fed a normal diet (10% fat). Normal diet control and high-fat-fed control mice received twice-daily intraperitoneal (i.p.) saline injections, while the high-fat-fed treatment groups (n = 8) received twice-daily injections of exendin-4(1-39), [S2a]dogfish glucagon, [S2a]dogfish glucagon exendin-4(31-39) or [S2a]dogfish glucagon-Lys(30) -γ-glutamyl-PAL (25 nmol/kg body weight) for 51 days. RESULTS: After dogfish glucagon analogue treatment, there was a rapid and sustained decrease in non-fasting blood glucose and an associated insulinotropic effect (analysis of variance, p < .05 to <.001) compared with saline-treated high-fat-fed controls. All peptide treatments significantly improved i.p. and oral glucose tolerance with concomitant increased insulin secretion compared with saline-treated high-fat-fed controls (p <.05 to <.001). After chronic treatment, no receptor desensitization was observed but insulin sensitivity was enhanced for all peptide-treated groups (p < .01 to <.001) except [S2a]dogfish glucagon. Both exendin-4 and [S2a]dogfish glucagon exendin-4(31-39) significantly reduced plasma triglyceride concentrations compared with those found in lean controls (p = .0105 and p = .0048, respectively). Pancreatic insulin content was not affected by peptide treatments but [S2a]dogfish glucagon and [S2a]dogfish glucagon exendin-4(31-39) decreased pancreatic glucagon by 28%-34% (p = .0221 and p = .0075, respectively). The percentage of ß-cell area within islets was increased by exendin-4 and peptide analogue treatment groups compared with high-fat-fed controls and the ß-cell area decreased (p < .05 to <.01). CONCLUSIONS: Overall, dogfish glucagon co-agonist analogues had several beneficial metabolic effects, showing therapeutic potential for type 2 diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Glucagon/pharmacology , Hyperglycemia/prevention & control , Insulin/metabolism , Insulin/physiology , Obesity/metabolism , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/complications , Diet, High-Fat , Dogfish/metabolism , Glucagon/analogs & derivatives , Glucagon/metabolism , Glucose Tolerance Test , Hyperglycemia/complications , Insulin Resistance , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Obese , Obesity/etiology
4.
Mol Cell Endocrinol ; 431: 133-44, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27179756

ABSTRACT

The antidiabetic potential of thirteen novel dogfish glucagon derived analogues were assessed in vitro and in acute in vivo studies. Stable peptide analogues enhanced insulin secretion from BRIN-BD11 ß-cells (p < 0.001) and reduced acute glycaemic responses following intraperitoneal glucose (25 nmol/kg) in healthy NIH Swiss mice (p < 0.05-p<0.001). The in vitro insulinotropic actions of [S2a]dogfish glucagon, [S2a]dogfish glucagon-exendin-4(31-39) and [S2a]dogfish glucagon-Lys(30)-γ-glutamyl-PAL, were blocked (p < 0.05-p<0.001) by the specific GLP-1 and glucagon receptor antagonists, exendin-4(9-39) and (desHis(1)Pro(4)Glu(9))glucagon amide but not by (Pro(3))GIP, indicating lack of GIP receptor involvement. These analogues dose-dependently stimulated cAMP production in GLP-1 and glucagon (p < 0.05-p<0.001) but not GIP-receptor transfected cells. They improved acute glycaemic and insulinotropic responses in high-fat fed diabetic mice and in wild-type C57BL/6J and GIPR-KO mice (p < 0.05-p<0.001), but not GLP-1R-KO mice, confirming action on GLP-1 but not GIP receptors. Overall, dogfish glucagon analogues have potential for diabetes therapy, exerting beneficial metabolic effects via GLP-1 and glucagon receptors.


Subject(s)
Dogfish/metabolism , Glucagon/metabolism , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Peptides/pharmacology , Animals , Cell Line , Cricetinae , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , HEK293 Cells , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/metabolism
5.
J Endocrinol ; 229(3): 319-30, 2016 06.
Article in English | MEDLINE | ID: mdl-27098830

ABSTRACT

Ablation of glucagon receptor signaling represents a potential treatment option for type 2 diabetes (T2DM). Additionally, activation of glucose-dependent insulinotropic polypeptide (GIP) receptor signaling also holds therapeutic promise for T2DM. Therefore, this study examined both independent and combined metabolic actions of desHis(1)Pro(4)Glu(9)(Lys(12)PAL)-glucagon (glucagon receptor antagonist) and d-Ala(2)GIP (GIP receptor agonist) in diet-induced obese mice. Glucagon receptor binding has been linked to alpha-helical structure and desHis(1)Pro(4)Glu(9)(Lys(12)PAL)-glucagon displayed enhanced alpha-helical content compared with native glucagon. In clonal pancreatic BRIN-BD11 beta-cells, desHis(1)Pro(4)Glu(9)(Lys(12)PAL)-glucagon was devoid of any insulinotropic or cAMP-generating actions, and did not impede d-Ala(2)GIP-mediated (P<0.01 to P<0.001) effects on insulin and cAMP production. Twice-daily injection of desHis(1)Pro(4)Glu(9)(Lys(12)PAL)-glucagon or d-Ala(2)GIP alone, and in combination, in high-fat-fed mice failed to affect body weight or energy intake. Circulating blood glucose levels were significantly (P<0.05 to P<0.01) decreased by all treatments regimens, with plasma and pancreatic insulin elevated (P<0.05 to P<0.001) in all mice receiving d-Ala(2)GIP. Interestingly, plasma glucagon concentrations were decreased (P<0.05) by sustained glucagon inhibition (day 28), but increased (P<0.05) by d-Ala(2)GIP therapy, with a combined treatment resulting in glucagon concentration similar to saline controls. All treatments improved (P<0.01) intraperitoneal and oral glucose tolerance, and peripheral insulin sensitivity. d-Ala(2)GIP-treated mice showed increased glucose-induced insulin secretion in response to intraperitoneal and oral glucose. Metabolic rate and ambulatory locomotor activity were increased (P<0.05 to P<0.001) in all desHis(1)Pro(4)Glu(9)(Lys(12)PAL)-glucagon-treated mice. These studies highlight the potential of glucagon receptor inhibition alone, and in combination with GIP receptor activation, for T2DM treatment.


Subject(s)
Gastric Inhibitory Polypeptide/agonists , Obesity/drug therapy , Receptors, Glucagon/antagonists & inhibitors , Animals , Cell Line , Cyclic AMP/biosynthesis , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/physiopathology , Diet, High-Fat/adverse effects , Energy Metabolism/drug effects , Gastric Inhibitory Polypeptide/administration & dosage , Gastric Inhibitory Polypeptide/analogs & derivatives , Glucagon/administration & dosage , Glucagon/analogs & derivatives , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Male , Mice , Mice, Obese , Motor Activity/drug effects , Obesity/etiology , Obesity/physiopathology , Rats
6.
Diabetes Obes Metab ; 16(12): 1214-22, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25060150

ABSTRACT

AIMS: To examine the effect of two novel, enzymatically stable, glucagon receptor peptide antagonists, on metabolic control in two mouse models of obesity/diabetes. METHOD: The effects of twice daily i.p. administration of desHis(1)Pro(4)Glu(9)-glucagon or desHis(1)Pro(4)Glu(9)Lys(12)FA-glucagon for 10 days on metabolic control in high-fat-fed (HFF; 45% fat) and obese diabetic (ob/ob) mice were compared with saline-treated controls. RESULTS: Neither analogue altered body weight or food intake in either model over 10 days; however, treatment with each peptide restored non-fasting blood glucose towards normal control values in HFF mice. Basal glucose was also reduced (p < 0.01) in desHis(1)Pro(4)Glu(9)Lys(12)FA-glucagon treated ob/ob mice by day 10, coinciding with increases (p < 0.001) in circulating insulin. At the end of the treatment period, both analogues significantly (p < 0.05-0.01) improved oral and i.p. glucose tolerance (p < 0.05) and peripheral insulin sensitivity, increased pancreatic insulin and glucagon content (p < 0.05-0.01) and decreased (p < 0.05) cholesterol levels in HFF mice. Similarly beneficial metabolic effects on oral glucose tolerance (p < 0.01) and pancreatic insulin content (p < 0.05) were observed in ob/ob mice, especially after desHis(1)Pro(4)Glu(9)Lys(12)FA-glucagon treatment. No significant differences in circulating triglycerides or aspects of indirect calorimetry were noted between peptide treatment groups and respective control HFF and ob/ob mice. Finally, glucagon-mediated elevations of glucose and insulin were significantly (p < 0.05-0.01) annulled after 10 days of desHis(1)Pro(4)Glu(9)-glucagon or desHis(1)Pro(4)Glu(9)Lys(12)FA-glucagon treatment in both animal models. CONCLUSION: These data indicate that peptide-based glucagon receptor antagonists can reverse aspects of genetically and dietary-induced obesity-related diabetes.


Subject(s)
Blood Glucose/drug effects , Body Weight/drug effects , Diabetes Mellitus, Experimental/drug therapy , Eating/drug effects , Glucagon/analogs & derivatives , Hypoglycemic Agents/pharmacology , Receptors, Glucagon/antagonists & inhibitors , Animals , Diabetes Mellitus, Experimental/etiology , Diet, High-Fat , Drug Administration Schedule , Glucagon/administration & dosage , Glucagon/pharmacology , Hypoglycemic Agents/administration & dosage , Mice , Mice, Obese
7.
Mol Cell Endocrinol ; 381(1-2): 26-34, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-23891841

ABSTRACT

Acute in vitro and in vivo biological activities of four novel structural analogues of glucagon were tested. desHis(1)Pro(4)-glucagon, desHis(1)Pro(4)Glu(9)-glucagon, desHis(1)Pro(4)Glu(9)Lys(12)FA-glucagon and desHis(1)Pro(4)Glu(9)Lys(30)FA-glucagon were stable to DPP-4 degradation and dose-dependently inhibited glucagon-mediated cAMP production (p<0.05 to p<0.001). None stimulated insulin secretion in vitro above basal levels, but all inhibited glucagon-induced insulin secretion (p<0.01 to p<0.001). In normal mice all analogues antagonised acute glucagon-mediated elevations of blood glucose (p<0.05 to p<0.001) and blocked corresponding insulinotropic responses. In high-fat fed mice, glucagon-induced increases in plasma insulin (p<0.05 to p<0.001) and glucagon-induced hyperglycaemia were blocked (p<0.05 to p<0.01) by three analogues. In obese diabetic (ob/ob) mice only desHis(1)Pro(4)Glu(9)-glucagon effectively (p<0.05 to p<0.01) inhibited both glucagon-mediated glycaemic and insulinotropic responses. desHis(1)Pro(4)-glucagon and desHis(1)Pro(4)Glu(9)-glucagon were biologically ineffective when administered 8h prior to glucagon, whereas desHis(1)Pro(4)Glu(9)Lys(12)FA-glucagon retained efficacy (p<0.01) for up to 24h. Such peptide-derived glucagon receptor antagonists have potential for type 2 diabetes therapy.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucagon/analogs & derivatives , Glucagon/pharmacology , Hypoglycemic Agents/pharmacology , Receptors, Glucagon/antagonists & inhibitors , Animals , Blood Glucose , Cell Line , Cyclic AMP/biosynthesis , Diabetes Mellitus, Type 2/blood , Dipeptidyl Peptidase 4/chemistry , Drug Evaluation, Preclinical , Glucagon/chemistry , HEK293 Cells , Humans , Insulin/metabolism , Insulin Secretion , Male , Mice , Mice, Obese , Proteolysis
8.
Int J Obes (Lond) ; 37(8): 1058-63, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23164696

ABSTRACT

OBJECTIVE: Compromise of gastric inhibitory polypeptide (GIP) receptor action and activation of cholecystokinin (CCK) receptors represent mechanistically different approaches to the possible treatment of obesity-related diabetes. In the present study, we have compared the individual and combined effects of (Pro(3))GIP[mPEG] and (pGlu-Gln)-CCK-8 as an enzymatically stable GIP receptor antagonist and CCK receptor agonist molecule, respectively. RESULTS: Twice-daily injections of (pGlu-Gln)-CCK-8 alone and in combination with (Pro(3))GIP[mPEG] in high-fat-fed mice for 34 days significantly decreased the energy intake throughout the entire study (P<0.05 to P<0.01). Body weights were significantly depressed (P<0.05 to P<0.01) in all treatment groups from day 18 onwards. Administration of (pGlu-Gln)-CCK-8, (Pro(3))GIP[mPEG] or a combination of both peptides significantly (P<0.01 to P<0.001) decreased the overall glycaemic excursion in response to both oral and intraperitoneal glucose challenge when compared with the controls. Furthermore, oral glucose tolerance returned to lean control levels in all treatment groups. The beneficial effects on glucose homeostasis were not associated with altered insulin levels in any of the treatment groups. In keeping with this, the estimated insulin sensitivity was restored to control levels by twice-daily treatment with (pGlu-Gln)-CCK-8, (Pro(3))GIP[mPEG] or a combination of both peptides. The blood lipid profile on day 34 was not significantly different between the high-fat controls and all treated mice. CONCLUSION: These studies highlight the potential of (pGlu-Gln)-CCK-8 and (Pro(3))GIP[mPEG] in the treatment of obesity-related diabetes, but there was no evidence of a synergistic effect of the combined treatment.


Subject(s)
Anti-Obesity Agents/administration & dosage , Diabetes Mellitus, Type 2/metabolism , Obesity/metabolism , Receptors, Cholecystokinin/agonists , Receptors, Gastrointestinal Hormone/agonists , Sincalide/analogs & derivatives , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Diabetes Mellitus, Type 2/drug therapy , Diet, High-Fat , Drug Therapy, Combination , Energy Intake/drug effects , Insulin Resistance , Male , Mice , Obesity/drug therapy , Sincalide/administration & dosage , Time Factors
9.
Diabetologia ; 55(10): 2747-2758, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22814764

ABSTRACT

AIMS/HYPOTHESIS: Cholecystokinin (CCK) is a rapidly degraded gastrointestinal peptide that stimulates satiety and insulin secretion. We aimed to investigate the beneficial weight-lowering and metabolic effects of the novel N-terminally modified CCK analogue, (pGlu-Gln)-CCK-8. METHODS: The biological actions of (pGlu-Gln)-CCK-8 were comprehensively evaluated in pancreatic clonal BRIN BD11 cells and in vivo in high-fat-fed and ob/ob mice. RESULTS: (pGlu-Gln)-CCK-8 was completely resistant to enzymatic degradation and its satiating effects were significantly (p < 0.05 to p < 0.001) more potent than CCK-8. In BRIN-BD11 cells, (pGlu-Gln)-CCK-8 exhibited enhanced (p < 0.01 to p < 0.001) insulinotropic actions compared with CCK-8. When administered acutely to high-fat-fed or ob/ob mice, (pGlu-Gln)-CCK-8 improved glucose homeostasis. Sub-chronic twice daily injections of (pGlu-Gln)-CCK-8 in high-fat-fed mice for 28 days significantly decreased body weight (p < 0.05 to p < 0.001), accumulated food intake (p < 0.05 to p < 0.001), non-fasting glucose (p < 0.05) and triacylglycerol deposition in pancreatic (p < 0.01), adipose (p < 0.05) and liver (p < 0.001) tissue, and improved oral (p < 0.05) and i.p. (p < 0.05) glucose tolerance and insulin sensitivity (p < 0.001). Similar observations were noted in ob/ob mice given twice daily injections of (pGlu-Gln)-CCK-8. In addition, these beneficial effects were not reproduced by simple dietary restriction and were not associated with changes in energy expenditure. There was no evidence for development of tolerance to (pGlu-Gln)-CCK-8, and analysis of histology or blood-borne markers for pancreatic, liver and renal function in mice treated with (pGlu-Gln)-CCK-8 suggested little abnormal pathology. CONCLUSIONS/INTERPRETATION: These studies emphasise the potential of (pGlu-Gln)-CCK-8 for the alleviation of obesity and insulin resistance.


Subject(s)
Cholecystokinin/agonists , Diabetes Mellitus/prevention & control , Obesity/prevention & control , Sincalide/therapeutic use , Animals , Comorbidity , Diabetes Mellitus/metabolism , Diabetes Mellitus/physiopathology , Disease Models, Animal , Glucose/metabolism , Homeostasis/drug effects , Homeostasis/physiology , Insulin Resistance/physiology , Male , Mice , Mice, Obese , Obesity/metabolism , Obesity/physiopathology , Sincalide/pharmacology
10.
Diabetes Res Clin Pract ; 91(3): e68-70, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21194775

ABSTRACT

We examined the postprandial glucose regulators nateglinide and GLP-1, separately and in combination, in people with type 2 diabetes. Nateglinide inhibited DPP-4 activity, reduced GLP-1 degradation and enhanced its insulinotropic and blood glucose lowering effect. Combining nateglinide and GLP-1 derivatives may effectively control postprandial glycaemia.


Subject(s)
Cyclohexanes/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide 1/pharmacology , Phenylalanine/analogs & derivatives , Aged , Blood Glucose/drug effects , Cross-Over Studies , Drug Synergism , Drug Therapy, Combination , Female , Humans , Hypoglycemic Agents , Male , Nateglinide , Phenylalanine/pharmacology , Postprandial Period , Treatment Outcome
11.
Diabet Med ; 26(6): 649-54, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19538242

ABSTRACT

AIMS: Glucagon-like peptide-1 (GLP-1) is an insulinotropic hormone and major component of the enteroinsular axis. Its therapeutic potential in human diabetes is limited by rapid degradation and inactivation by the enzyme dipeptidylpeptidase-4 (DPP-4). We investigated the acute effects of metformin with and without food on DPP-4 activity in Type 2 diabetes. METHODS: Ten subjects with Type 2 diabetes (6 male/4 female, age 65.8 +/- 2.6 years, body mass index 30.0 +/- 1.2 kg/m2, glycated haemoglobin (HbA(1c)) 6.3 +/- 0.2%, mean +/- SEM) received metformin 1 g orally or placebo together with a standard mixed meal (SMM) in a random crossover design. Six subjects re-attended fasting and received metformin 1 g without a SMM. RESULTS: Following SMM (n = 10), DPP-4 activity was not suppressed by metformin compared with placebo [area under curve (AUC)(0-4 h) 1574 +/- 4 vs. 1581 +/- 8 micromol/ml/min, respectively]. Plasma glucose, insulin and active GLP-1 were not different. However, DPP-4 activity was suppressed with metformin following fasting compared with a SMM (n = 6) (AUC(0-4 h) 1578 +/- 4 vs. 1494 +/- 9 micromol/min, P < 0.02). Metformin serum levels were significantly lower (P < 0.001) after SMM than fasting (AUC(0-4 h) 350 +/- 66 vs. 457 +/- 55 mg/ml/min). CONCLUSION: Metformin inhibits DPP-4 activity in Type 2 diabetic patients in the fasting state but not when taken with a standard mixed meal. Metformin serum concentrations are lower if the drug is taken with food. These findings should be taken into account in establishing how to maximize efficacy of the drug.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Adult , Aged , Aged, 80 and over , Cross-Over Studies , Diabetes Mellitus, Type 2/blood , Dipeptidyl-Peptidase IV Inhibitors/blood , Eating/physiology , Fasting/blood , Female , Humans , Hypoglycemic Agents/pharmacology , Male , Metformin/pharmacology , Middle Aged
12.
Diabetologia ; 50(7): 1532-40, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17486314

ABSTRACT

AIMS/HYPOTHESIS: Ablation of gastric inhibitory polypeptide (GIP) receptor action is reported to protect against obesity and associated metabolic abnormalities. The aim of this study was to use prediabetic ob/ob mice to examine whether 60 days of chemical GIP receptor ablation with (Pro(3))GIP is able to counter the development of genetic obesity-related diabetes. MATERIALS AND METHODS: Young (5-7 weeks) ob/ob mice received once daily i.p. injections of either saline vehicle or (Pro(3))GIP (25 nmol kg(-1) day(-1)) over a 60 day period. Food intake, body weight and circulating glucose and insulin were measured at frequent intervals. At 60 days, glucose tolerance, response to native GIP, postprandial responses, insulin sensitivity, HbA(1c), circulating hormones and plasma lipids were assessed. RESULTS: Body weight and food intake in (Pro(3))GIP-treated mice did not differ from ob/ob controls. GIP receptor blockade significantly improved non-fasting glucose (p < 0.001), HbA(1c) (p < 0.05), glucose tolerance (p < 0.001), meal tolerance (p < 0.001) and insulin sensitivity (p < 0.05). Remarkably, (Pro(3))GIP treatment prevented the age-related development of diabetes, as none of these parameters differed significantly between treated ob/ob mice and normal age-matched lean controls. Circulating levels of glucagon, corticosterone, adiponectin and total cholesterol were unchanged by (Pro(3))GIP, while levels of triacylglycerol, LDL-cholesterol and resistin were decreased (p < 0.05) compared with those in control ob/ob mice. Plasma and pancreatic insulin concentrations were generally lower after (Pro(3))GIP treatment than in control ob/ob mice (p < 0.01), but plasma insulin levels remained substantially raised (p < 0.001) compared with those observed in lean controls. CONCLUSIONS/INTERPRETATION: These data indicate that sustained GIP receptor antagonism provides an effective means of preventing the development of many of the metabolic abnormalities of obesity-driven diabetes.


Subject(s)
Diabetes Mellitus/prevention & control , Gastric Inhibitory Polypeptide/administration & dosage , Obesity/genetics , Animals , Body Weight , Feeding Behavior , Female , Glucose Tolerance Test , Insulin/metabolism , Male , Mice , Mice, Obese , Obesity/prevention & control , Pancreas/metabolism , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Time Factors
13.
J Endocrinol ; 191(1): 93-100, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17065392

ABSTRACT

Glucose-dependent insulinotrophic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) are important enteroendocrine hormones that are rapidly degraded by an ubiquitous enzyme dipeptidyl peptidase IV to yield truncated metabolites GIP(3-42) and GLP-1(9-36)amide. In this study, we investigated the effects of sub-chronic exposure to these major circulating forms of GIP and GLP-1 on blood glucose control and endocrine pancreatic function in obese diabetic (ob/ob) mice. A once daily injection of either peptide for 14 days had no effect on body weight, food intake or pancreatic insulin content or islet morphology. GLP-1(9-36)amide also had no effect on plasma glucose homeostasis or insulin secretion. Mice receiving GIP(3-42) exhibited small but significant improvements in non-fasting plasma glucose, glucose tolerance and glycaemic response to feeding. Accordingly, plasma insulin responses were unchanged suggesting that the observed enhancement of insulin sensitivity was responsible for the improvement in glycaemic control. These data indicate that sub-chronic exposure to GIP and GLP-1 metabolites does not result in physiological impairment of insulin secretion or blood glucose control. GIP(3-42) might exert an overall beneficial effect by improving insulin sensitivity through extrapancreatic action.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Glucagon-Like Peptide 1/analogs & derivatives , Insulin/metabolism , Obesity/metabolism , Peptide Fragments/pharmacology , Peptides/pharmacology , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Animals , Diabetes Mellitus/physiopathology , Eating , Glucagon-Like Peptide 1/pharmacology , Glucose Tolerance Test , Homeostasis , Immunohistochemistry/methods , Insulin/analysis , Insulin/blood , Insulin Resistance , Insulin Secretion , Insulin-Secreting Cells/chemistry , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Mice , Mice, Obese , Obesity/physiopathology , Organ Size/drug effects
14.
Horm Metab Res ; 38(2): 94-7, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16523409

ABSTRACT

There is mounting evidence that elevated circulating concentrations of glycated insulin play a role in insulin resistance in type 2 diabetes. This study evaluated the secretion of glycated insulin in response to enteral stimulation in type 2 diabetic subjects. Following a mixed meal (450 kcal; 44 % carbohydrate; 40 % fat; 16 % protein), glycated insulin rose 10-fold to peak (60 min) at 104.5 +/- 25.0 pmol/l (p < 0.001), representing 22 % total circulating insulin. The response paralleled early rises in insulin and C-peptide, which peaked at 90 min and were more protracted. Maximum glucose concentrations were observed at 50 min. These data indicate that type 2 diabetic subjects exhibit a rapid meal-induced release of glycated insulin from readily releasable pancreatic beta-cell stores, which might contribute to impaired glucose homeostasis following enteral nutrition.


Subject(s)
C-Peptide/blood , Diabetes Mellitus, Type 2/blood , Food , Insulin/blood , Blood Glucose/analysis , Female , Homeostasis , Humans , Insulin-Secreting Cells/metabolism , Male , Time Factors
15.
Diabetes Obes Metab ; 7(5): 563-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16050949

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is an important insulinotropic hormone with potential in the treatment of type 2 diabetes. However, the short biological half-life of the peptide after cleavage by dipeptidylpeptidase IV (DPP IV) is a major limitation. Inhibition of DPP IV activity and the development of resistant GLP-1 analogues is the subject of ongoing research. In this study, we determined cell growth, insulin content, insulin accumulation and insulin secretory function of a insulin-secreting cell line cultured for 3 days with either GLP-1, GLP-1 plus the DPP IV inhibitor diprotin A (DPA) or stable N-acetyl-GLP-1. Native GLP-1 was rapidly degraded by DPP IV during culture with accumulation of the inactive metabolite GLP-1(9-36)amide. Inclusion of DPA or use of the DPP IV-resistant analogue, N-acetyl-GLP-1, improved cellular function compared to exposure to GLP-1 alone. Most notably, basal and accumulated insulin secretion was enhanced, and glucose responsiveness was improved. However, prolonged GLP-1 treatment resulted in GLP-1 receptor desensitization regardless of DPP IV status. The results indicate that prevention of DPP IV action is necessary for beneficial effects of GLP-1 on pancreatic beta cells and that prolonged exposure to GLP-1(9-36)amide may be detrimental to insulin secretory function. These observations also support the ongoing development of DPP-IV-resistant forms of GLP-1, such as N-acetyl-GLP-1.


Subject(s)
Dipeptidyl Peptidase 4/physiology , Glucagon-Like Peptide 1/pharmacology , Insulin-Secreting Cells/drug effects , Insulin/metabolism , Animals , Cell Division/drug effects , Cell Line , Dipeptidyl Peptidase 4/metabolism , Dose-Response Relationship, Drug , Glucagon-Like Peptide 1/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Oligopeptides/pharmacology , Protease Inhibitors/pharmacology , Rats
16.
Diabetes Obes Metab ; 7(5): 595-604, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16050953

ABSTRACT

AIM: The two major incretin hormones--glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP)--are being actively researched by the pharmaceutical industry because of their glucose-lowering and potential anti-diabetic properties. Unfortunately, the inactivation of GLP-1 and GIP in the circulation brought about by dipeptidyl-peptidase-IV (DPP-IV) degradation makes their biological actions short-lived. This study directly compares the cellular and biological properties of GLP-1, GIP and their N-terminally modified counterparts, with glucitol extension at positions His7 and Tyr1 respectively, to confer DPP-IV resistance. METHODS: Using both the glucose-responsive pancreatic beta-cell line, BRIN BD11, and the obese diabetic (ob/ob) mouse, we assessed adenosine 3',5'-cyclic monophosphate (cAMP) production and insulinotropic action in vitro as well as in vivo glucose-lowering and insulin-releasing actions. RESULTS: The results reveal that glycation of the N-terminus of GLP-1 or GIP stabilized both peptides against DPP-IV degradation. However, N-glucitol-GLP-1 displayed reduced cAMP production, insulinotropic activity and glucose-lowering potency, compared to native GLP-1. By contrast, N-glucitol-GIP exhibited substantially improved biological activities, compared to native GIP, and possessed similar or enhanced in vivo potency to GLP-1. N-terminal extension by means of glucitol addition is more beneficial to bioactivity of GIP than it is to GLP-1. CONCLUSIONS: N-terminal glycation generates a super GIP agonist, which possesses acute in vivo glucose-lowering and insulinotropic actions superior to native GLP-1. Therefore, N-glucitol-GIP is a particularly attractive potential candidate molecule for drug therapy of type 2 diabetes.


Subject(s)
Dipeptidyl Peptidase 4/pharmacology , Gastric Inhibitory Polypeptide/pharmacology , Glucagon-Like Peptide 1/pharmacology , Insulin-Secreting Cells/drug effects , Sorbitol/pharmacology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Cell Culture Techniques , Cell Line , Cyclic AMP/biosynthesis , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Dose-Response Relationship, Drug , Gastric Inhibitory Polypeptide/therapeutic use , Glucagon-Like Peptide 1/therapeutic use , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Mice , Mice, Obese , Sorbitol/therapeutic use , Spectrometry, Mass, Electrospray Ionization
17.
J Endocrinol ; 185(2): 307-17, 2005 May.
Article in English | MEDLINE | ID: mdl-15845923

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is a potent insulinotropic hormone proposed to play a role in both the pathophysiology and treatment of type 2 diabetes. This study has employed the GLP-1 receptor antagonist, exendin-4(9-39)amide (Ex(9-39)) to evaluate the role of endogenous GLP-1 in genetic obesity-related diabetes and related metabolic abnormalities using ob/ob and normal mice. Acute in vivo antagonistic potency of Ex(9-39) was confirmed in ob/ob mice by blockade of the insulin-releasing and anti-hyperglycaemic actions of intraperitoneal GLP-1. In longer term studies, ob/ob mice were given once daily injections of Ex(9-39) or vehicle for 11 days. Feeding activity, body weight, and both basal and glucose-stimulated insulin secretion were not significantly affected by chronic Ex(9-39) treatment. However, significantly elevated basal glucose concentrations and impaired glucose tolerance were evident at 11 days. These disturbances in glucose homeostasis were independent of changes of insulin sensitivity and reversed by discontinuation of the Ex(9-39) for 9 days. Similar treatment of normal mice did not affect any of the parameters measured. These findings illustrate the physiological extrapancreatic glucose-lowering actions of GLP-1 in ob/ob mice and suggest that the endogenous hormone plays a minor role in the metabolic abnormalities associated with obesity-related diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucagon/metabolism , Obesity/metabolism , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Precursors/metabolism , Receptors, Glucagon/antagonists & inhibitors , Animals , Blood Glucose/analysis , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/genetics , Female , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Glucose Tolerance Test , Insulin/blood , Insulin/metabolism , Male , Mice , Mice, Obese , Obesity/genetics , Time Factors
18.
Diabet Med ; 22(5): 654-7, 2005 May.
Article in English | MEDLINE | ID: mdl-15842525

ABSTRACT

AIMS: Glucagon-like peptide-1 (GLP-1) and gastric inhibitory polypeptide (GIP) are important insulinotropic hormones that enhance the insulin secretory response to feeding. Their potential for treating Type 2 diabetes is limited by short biological half-life owing to degradation by dipeptidyl peptidase IV (DPP IV). We investigated the acute effects of metformin on DPP IV activity in Type 2 diabetes to elucidate inhibition of DPP IV as a possible mechanism of action. METHODS: Eight fasting subjects with Type 2 diabetes (5M/3F, age 53.1+/-4.2 years, BMI 36.8+/-1.8 kg/m2, glucose 8.9+/-1.2 mmol/l, HbA1c 7.8+/-0.6%) received placebo or metformin 1 g orally 1 week apart in a random, crossover design. RESULTS: Following metformin, DPP IV activity was suppressed compared with placebo (AUC0-6 h 3230+/-373 vs. 5764+/-504 nmol ml/l, respectively, P=0.001). Circulating glucose, insulin and total GLP-1 were unchanged. Metformin also concentration-dependently inhibited endogenous DPP IV activity in vitro in plasma from Type 2 diabetic subjects. CONCLUSION: Oral metformin effectively inhibits DPP IV activity in Type 2 diabetic patients, suggesting that the drug may have potential for future combination therapy with incretin hormones.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Dipeptidyl Peptidase 4/blood , Hypoglycemic Agents/administration & dosage , Metformin/administration & dosage , Administration, Oral , Blood Glucose/metabolism , Cross-Over Studies , Female , Gastric Inhibitory Polypeptide/metabolism , Glucagon/metabolism , Glucagon-Like Peptide 1 , Humans , Male , Middle Aged , Peptide Fragments , Protein Precursors
19.
Arch Biochem Biophys ; 428(2): 136-43, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15246869

ABSTRACT

The two major incretin hormones, glucagon-like peptide-1 (GLP-1), and glucose-dependent insulinotropic polypeptide (GIP), are currently being considered as prospective drug candidates for treatment of type 2 diabetes. Interest in these gut hormones was initially spurred by their potent insulinotropic activities, but a number of other antihyperglycaemic actions are now established. One of the foremost barriers in progressing GLP-1 and GIP to the clinic concerns their rapid degradation and inactivation by the ubiquitous enzyme, dipeptidyl peptidase IV (DPP IV). Here, we compare the DPP IV resistance and biological properties of Abu8/Abu2 (2-aminobutyric acid) substituted analogues of GLP-1 and GIP engineered to impart DPP IV resistance. Whereas (Abu8)GLP-1 was completely stable to human plasma (half-life >12 h), GLP-1, GIP, and (Abu2)GIP were rapidly degraded (half-lives: 6.2, 6.0, and 7.1 h, respectively). Native GIP, GLP-1, and particularly (Abu8)GLP-1 elicited significant adenylate cyclase and insulinotropic activity, while (Abu2)GIP was less effective. Similarly, in obese diabetic (ob/ob) mice, GIP, GLP-1, and (Abu8)GLP-1 displayed substantial glucose-lowering and insulin-releasing activities, whereas (Abu2)GIP was only weakly active. These studies illustrate divergent effects of penultimate amino acid Ala8/Ala2 substitution with Abu on the biological properties of GLP-1 and GIP, suggesting that (Abu8)GLP-1 represents a potential candidate for future therapeutic development.


Subject(s)
Aminobutyrates/pharmacology , Diabetes Mellitus/drug therapy , Gastric Inhibitory Polypeptide/pharmacology , Glucagon/pharmacology , Insulin/metabolism , Peptide Fragments/pharmacology , Protein Precursors/pharmacology , Adenylyl Cyclases/metabolism , Alanine/chemistry , Animals , Cells, Cultured , Chromatography, High Pressure Liquid , Cyclic AMP/metabolism , Dipeptidyl Peptidase 4/chemistry , Dose-Response Relationship, Drug , Gastric Inhibitory Polypeptide/chemistry , Glucagon/chemistry , Glucagon-Like Peptide 1 , Glucose/metabolism , Humans , Insulin Secretion , Islets of Langerhans/metabolism , Mice , Mice, Obese , Models, Chemical , Peptide Fragments/chemistry , Peptides/chemistry , Protein Precursors/chemistry , Protein Structure, Tertiary , Spectrometry, Mass, Electrospray Ionization , Time Factors
20.
J Endocrinol ; 180(3): 379-88, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15012592

ABSTRACT

Glucagon-like peptide-1(7-36)amide (GLP-1) possesses several unique and beneficial effects for the potential treatment of type 2 diabetes. However, the rapid inactivation of GLP-1 by dipeptidyl peptidase IV (DPP IV) results in a short half-life in vivo (less than 2 min) hindering therapeutic development. In the present study, a novel His(7)-modified analogue of GLP-1, N-pyroglutamyl-GLP-1, as well as N-acetyl-GLP-1 were synthesised and tested for DPP IV stability and biological activity. Incubation of GLP-1 with either DPP IV or human plasma resulted in rapid degradation of native GLP-1 to GLP-1(9-36)amide, while N-acetyl-GLP-1 and N-pyroglutamyl-GLP-1 were completely resistant to degradation. N-acetyl-GLP-1 and N-pyroglutamyl-GLP-1 bound to the GLP-1 receptor but had reduced affinities (IC(50) values 32.9 and 6.7 nM, respectively) compared with native GLP-1 (IC(50) 0.37 nM). Similarly, both analogues stimulated cAMP production with EC(50) values of 16.3 and 27 nM respectively compared with GLP-1 (EC(50) 4.7 nM). However, N-acetyl-GLP-1 and N-pyroglutamyl-GLP-1 exhibited potent insulinotropic activity in vitro at 5.6 mM glucose (P<0.05 to P<0.001) similar to native GLP-1. Both analogues (25 nM/kg body weight) lowered plasma glucose and increased plasma insulin levels when administered in conjunction with glucose (18 nM/kg body weight) to adult obese diabetic (ob/ob) mice. N-pyroglutamyl-GLP-1 was substantially better at lowering plasma glucose compared with the native peptide, while N-acetyl-GLP-1 was significantly more potent at stimulating insulin secretion. These studies indicate that N-terminal modification of GLP-1 results in DPP IV-resistant and biologically potent forms of GLP-1. The particularly powerful antihyperglycaemic action of N-pyroglutamyl-GLP-1 shows potential for the treatment of type 2 diabetes.


Subject(s)
Hypoglycemic Agents/chemical synthesis , Peptide Fragments/metabolism , Animals , Cell Line, Transformed , Cricetinae , Cyclic AMP/metabolism , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Dipeptidyl Peptidase 4/metabolism , Glucagon/analogs & derivatives , Glucagon/chemical synthesis , Glucagon/metabolism , Glucagon/therapeutic use , Glucagon-Like Peptide 1 , Glucagon-Like Peptides , Hypoglycemic Agents/analysis , Hypoglycemic Agents/therapeutic use , Insulin/metabolism , Mesocricetus , Mice , Mice, Obese , Peptide Fragments/chemical synthesis , Peptide Fragments/therapeutic use , Protein Precursors/metabolism , Pyrrolidonecarboxylic Acid/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL
...