Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Bone Miner Res ; 38(5): 733-748, 2023 05.
Article in English | MEDLINE | ID: mdl-36850034

ABSTRACT

Due to aging of the population, bone frailty is dramatically increasing worldwide. Although some therapeutic options exist, they do not fully protect or prevent against the occurrence of new fractures. All current drugs approved for the treatment of bone fragility target bone mass. However, bone resistance to fracture is not solely due to bone mass but relies also on bone extracellular matrix (ECM) material properties, i.e., the quality of the bone matrix component. Here, we introduce the first-in-class unimolecular dual glucose-dependent insulinotropic polypeptide/glucagon-like peptide-2 (GIP/GLP-2) analogue, GL-0001, that activates simultaneously the glucose-dependent insulinotropic polypeptide receptor (GIPr) and the glucagon-like peptide-2 receptor (GLP-2r). GL-0001 acts synergistically through a cyclic adenosine monophosphate-lysyl oxidase pathway to enhance collagen maturity. Furthermore, bilateral ovariectomy was performed in 32 BALB/c mice at 12 weeks of age prior to random allocation to either saline, dual GIP/GLP-2 analogues (GL-0001 or GL-0007) or zoledronic acid groups (n = 8/group). Treatment with dual GIP/GLP-2 analogues was initiated 4 weeks later for 8 weeks. At the organ level, GL-0001 modified biomechanical parameters by increasing ultimate load, postyield displacement, and energy-to-fracture of cortical bone. GL-0001 also prevented excess trabecular bone degradation at the appendicular skeleton and enhanced bone ECM material properties in cortical bone through a reduction of the mineral-to-matrix ratio and augmentation in enzymatic collagen cross-linking. These results demonstrate that targeting bone ECM material properties is a viable option to enhance bone strength and opens an innovative pathway for the treatment of patients suffering from bone fragility. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Fractures, Bone , Glucagon-Like Peptide 1 , Animals , Mice , Bone and Bones/metabolism , Bone Density , Fractures, Bone/drug therapy , Gastric Inhibitory Polypeptide/analogs & derivatives , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism
2.
Peptides ; 122: 170155, 2019 12.
Article in English | MEDLINE | ID: mdl-31539554

ABSTRACT

This paper describes the early history of Gastric Inhibitory Polypeptide, better referred to simply as GIP, from its isolation by purification from a crude preparation of CCK-PZ (cholecystokinin/pancreozymin) to its recognition as a key player in the pathogenesis of obesity and other metabolic disorders far removed from the enterogastrone properties by which it was originally identified. Augmentation of glucose mediated insulin release, the incretin effect, was discovered soon after GIP was first isolated and only much later was its important role in the pathogenesis of obesity, through mechanism other than insulin secretion, appreciated. Immunoassay - the only method by which the concentration of GIP was measured in plasma until quite recently - was found to be flawed and to depend upon which specific epitope of the hormone an assay detected. This was especially true if it was an amino-acid sequence specific to porcine rather than human GIP. A further confounder was the discovery that much of the GIP measured by immunoassay was its biological antagonist produced by cleavage of its two N-terminal amino-acids in the circulation by the same dipeptidyl-peptidase as de-activates GLP-1. Potential use of synthetic agonistic and antagonistic GIP analogues in therapeutics was barely alluded to before year 2000.


Subject(s)
Gastric Inhibitory Polypeptide/genetics , Glucose/metabolism , Insulin/genetics , Obesity/genetics , Cholecystokinin/metabolism , Epitopes/genetics , Gastric Inhibitory Polypeptide/analogs & derivatives , Gastric Inhibitory Polypeptide/blood , Gastric Inhibitory Polypeptide/therapeutic use , Gastrointestinal Hormones/metabolism , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide Receptors/genetics , Glucose/genetics , Humans , Incretins/genetics , Insulin/metabolism , Obesity/blood , Obesity/pathology , Peptides/metabolism
3.
Mol Metab ; 20: 51-62, 2019 02.
Article in English | MEDLINE | ID: mdl-30578168

ABSTRACT

OBJECTIVE: Structurally-improved GIP analogs were developed to determine precisely whether GIP receptor (GIPR) agonism or antagonism lowers body weight in obese mice. METHODS: A series of peptide-based GIP analogs, including structurally diverse agonists and a long-acting antagonist, were generated and characterized in vitro using functional assays in cell systems overexpressing human and mouse derived receptors. These analogs were characterized in vivo in DIO mice following acute dosing for effects on glycemic control, and following chronic dosing for effects on body weight and food intake. Pair-feeding studies and indirect calorimetry were used to survey the mechanism for body weight lowering. Congenital Gipr-/- and Glp1r-/- DIO mice were used to investigate the selectivity of the agonists and to ascribe the pharmacology to effects mediated by the GIPR. RESULTS: Non-acylated, Aib2 substituted analogs derived from human GIP sequence showed full in vitro potency at human GIPR and subtly reduced in vitro potency at mouse GIPR without cross-reactivity at GLP-1R. These GIPR agonists lowered acute blood glucose in wild-type and Glp1r-/- mice, and this effect was absent in Gipr-/- mice, which confirmed selectivity towards GIPR. Chronic treatment of DIO mice resulted in modest yet consistent, dose-dependent decreased body weight across many studies with diverse analogs. The mechanism for body weight lowering is due to reductions in food intake, not energy expenditure, as suggested by pair-feeding studies and indirect calorimetry assessment. The weight lowering effect was preserved in DIO Glp-1r-/- mice and absent in DIO Gipr-/- mice. The body weight lowering efficacy of GIPR agonists was enhanced with analogs that exhibit higher mouse GIPR potency, with increased frequency of administration, and with fatty-acylated peptides of extended duration of action. Additionally, a fatty-acylated, N-terminally truncated GIP analog was shown to have high in vitro antagonism potency for human and mouse GIPR without cross-reactive activity at mouse GLP-1R or mouse glucagon receptor (GcgR). This acylated antagonist sufficiently inhibited the acute effects of GIP to improve glucose tolerance in DIO mice. Chronic treatment of DIO mice with high doses of this acylated GIPR antagonist did not result in body weight change. Further, co-treatment of this acylated GIPR antagonist with liraglutide, an acylated GLP-1R agonist, to DIO mice did not result in increased body weight lowering relative to liraglutide-treated mice. Enhanced body weight lowering in DIO mice was evident however following co-treatment of long-acting selective individual agonists for GLP-1R and GIPR, consistent with previous data. CONCLUSIONS: We conclude that peptide-based GIPR agonists, not peptide-based GIPR antagonists, that are suitably optimized for receptor selectivity, cross-species activity, and duration of action consistently lower body weight in DIO mice, although with moderate efficacy relative to GLP-1R agonists. These preclinical rodent pharmacology results, in accordance with recent clinical results, provide definitive proof that systemic GIPR agonism, not antagonism, is beneficial for body weight loss.


Subject(s)
Anti-Obesity Agents/pharmacology , Gastric Inhibitory Polypeptide/analogs & derivatives , Obesity/drug therapy , Peptide Fragments/pharmacology , Receptors, Gastrointestinal Hormone/agonists , Weight Loss/drug effects , Animals , Anti-Obesity Agents/chemistry , Anti-Obesity Agents/therapeutic use , Blood Glucose/analysis , Diet, High-Fat/adverse effects , HEK293 Cells , Humans , Liraglutide/pharmacology , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Peptide Fragments/chemistry , Peptide Fragments/therapeutic use , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/metabolism
4.
Expert Opin Ther Targets ; 22(7): 615-628, 2018 07.
Article in English | MEDLINE | ID: mdl-29911915

ABSTRACT

INTRODUCTION: Incretin hormones, glucose-dependent insulinotropic polypeptide (GIP), and glucagon-like peptide-1 (GLP-1) exert pleiotropic effects on endocrine pancreas and nervous system. Expression of GIP and GIP receptor (GIPR) in neurons, their roles in neurogenesis, synaptic plasticity, neurotransmission, and neuromodulation uniquely position GIPR for therapeutic applications in neurodegenerative disorders. GIP analogs acting as GIPR agonists attenuate neurobehavioral and neuropathological sequelae of neurodegenerative disorders in preclinical models, e.g. Alzheimer's disease (AD), Parkinson's disease (PD), and cerebrovascular disorders. Modulation of GIPR signaling offers an unprecedented approach for disease modification by arresting neuronal viability decline, enabling neuronal regeneration, and reducing neuroinflammation. Growth-promoting effects of GIP signaling and broad-based neuroprotection highlight the therapeutic potential of GIPR agonists. Areas covered: This review focuses on the role of GIPR-mediated signaling in the central nervous system in neurophysiological and neuropathological conditions. In context of neurodegeneration, the article summarizes potential of targeting GIPR signaling for neurodegenerative conditions such as AD, PD, traumatic brain injury, and cerebrovascular disorders. Expert opinion: GIPR represents a validated therapeutic target for neurodegenerative disorders. GIPR agonists impart symptomatic improvements, slowed neurodegeneration, and enhanced neuronal regenerative capacity in preclinical models. Modulation of GIPR signaling is potentially a viable therapeutic approach for disease modification in neurodegenerative disorders.


Subject(s)
Gastric Inhibitory Polypeptide/analogs & derivatives , Neurodegenerative Diseases/physiopathology , Receptors, Gastrointestinal Hormone/agonists , Animals , Drug Development/methods , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Incretins/metabolism , Neurodegenerative Diseases/drug therapy , Receptors, Gastrointestinal Hormone/metabolism , Signal Transduction/drug effects
5.
Diabet Med ; 35(1): 33-40, 2018 01.
Article in English | MEDLINE | ID: mdl-29044772

ABSTRACT

The incretin hormones glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) have attracted considerable scientific and clinical interest due largely to their insulin-releasing and glucose-lowering properties. Indeed, GLP-1-based therapies are now key treatment options for many people with diabetes worldwide. In contrast, GIP-based agents have yet to reach the clinic based primarily on the impaired insulinotropic action of GIP observed in people with diabetes. Nevertheless, GIP is a key physiological regulator of insulin secretion and stable forms of GIP show much promise in rodent models to alleviate diabetes-obesity. Recent studies suggest that GIP may have an important role to play in a combination therapeutic approach or bioengineered with other gut peptides. Moreover, recent experimental studies indicate that incretins also exert pleiotropic effects in regions of the brain associated with learning and memory, thereby supporting preclinical data demonstrating that incretin-based drugs improve cognitive function. This review article, based on the RD Lawrence Lecture presented at Diabetes UK Annual Professional Conference (2017), provides a brief overview of incretins with a major focus on GIP, the development of designer GIP analogues, and how these molecules can improve cognition. Thus, incretins can be considered as 'the intelligent hormones' and may hold the key to successfully treating the alarming rise in neurodegenerative disorders.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Gastric Inhibitory Polypeptide/physiology , Glucagon-Like Peptide 1/physiology , Hypoglycemic Agents/therapeutic use , Incretins/physiology , Alzheimer Disease , Animals , Cognition/drug effects , Disease Models, Animal , Gastric Inhibitory Polypeptide/analogs & derivatives , Gastric Inhibitory Polypeptide/pharmacology , Gastric Inhibitory Polypeptide/therapeutic use , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide 1/therapeutic use , Humans , Incretins/pharmacology , Incretins/therapeutic use , Learning/drug effects , Memory/drug effects , Nootropic Agents/pharmacology
6.
Diabetes Obes Metab ; 20(1): 60-68, 2018 01.
Article in English | MEDLINE | ID: mdl-28598027

ABSTRACT

AIM: To investigate the effects of the novel glucose-dependent insulinotropic polypeptide (GIP) analogue, ZP4165, on body weight and glycaemic control in rodents, and to investigate if ZP4165 modulates the anti-obesity and anti-hyperglycaemic effects of a glucagon-like peptide-1 (GLP-1) agonist (liraglutide). METHODS: The acute insulinotropic effect of ZP4165 was investigated in rats during an oral glucose tolerance test. The long-term effects of ZP4165 on body weight and glycaemic control, either alone or in combination with liraglutide, were assessed in diet-induced obese mice and diabetic db/db mice. RESULTS: ZP4165 showed insulinotropic action in rats. The GIP analogue did not alter the body weight of obese mice but enhanced GLP-1-induced weight loss. In diabetic mice, 4 weeks' dosing with ZP4165 reduced glycated haemoglobin levels vs vehicle by an extent similar to the GLP-1 agonist. CONCLUSIONS: ZP4165 potentiated the anti-obesity effect of a GLP-1 agonist in obese mice and improved glycaemic control in diabetic mice. These studies support further investigation of dual-incretin therapy as a more effective treatment option than mono GLP-1 medication for type 2 diabetes mellitus and obesity.


Subject(s)
Anti-Obesity Agents/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Gastric Inhibitory Polypeptide/analogs & derivatives , Gastric Inhibitory Polypeptide/therapeutic use , Hyperglycemia/prevention & control , Hypoglycemic Agents/therapeutic use , Obesity/drug therapy , Receptors, Gastrointestinal Hormone/agonists , Animals , Anti-Obesity Agents/blood , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/pharmacology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Drug Design , Drug Therapy, Combination , Gastric Inhibitory Polypeptide/pharmacokinetics , Gastric Inhibitory Polypeptide/pharmacology , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , HEK293 Cells , Half-Life , Humans , Hypoglycemic Agents/blood , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Incretins/pharmacology , Incretins/therapeutic use , Liraglutide/pharmacology , Liraglutide/therapeutic use , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Obesity/blood , Obesity/metabolism , Rats, Sprague-Dawley , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Weight Loss/drug effects
7.
Eur J Pharmacol ; 797: 162-172, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-27913104

ABSTRACT

Type 2 diabetes mellitus (T2DM) is a risk factor for Parkinson's disease (PD). Therefore, treatment to improve insulin resistance in T2DM may be useful for PD patients. Glucose dependent insulinotropic polypeptide (GIP) is a member of the incretin hormone family that can promote insulin release and improve insulin resistance. Several GIP analogues have been developed as potential treatments for T2DM. We had shown previously that D-Ala2-GIP-glu-PAL, a novel long-acting GIP analogue, can play a neuroprotective role in the PD mouse model induced by acute MPTP injection. The drug reduced damage to the dopaminergic neurons and increased CREB-mediated Bcl-2 expression to prevent apoptosis and reduced chronic inflammation in the brain. In the present study, we further tested the effects of chronic treatment by D-Ala2-GIP-glu-PAL in a chronic PD mouse model induced by MPTP (25mg/kg ip.) combination with probenecid (250mg/kg ip.) injection for 5 weeks. The results demonstrated that chronic treatment with D-Ala2-GIP-glu-PAL inhibits MPTP -induced Parkinsonism-like motor disorders in mice, and that the drug prevents dopaminergic neuronal loss in the substantia nigra pars compacta (SNpc). Moreover, D-Ala2-GIP-glu-PAL also inhibited the increased levels of expression of α-synuclein in the SNpc and striatum induced by MPTP. Furthermore, drug treatment reduced chronic neuroinflammation, oxidative stress and lipid peroxidation, and increased the expression of BDNF. These findings show that GIP signaling is neuroprotective and holds promise as a novel treatment of PD.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Gastric Inhibitory Polypeptide/analogs & derivatives , Gastric Inhibitory Polypeptide/pharmacology , Gene Expression Regulation/drug effects , Lipid Peroxidation/drug effects , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Cell Count , Chronic Disease , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Gastric Inhibitory Polypeptide/therapeutic use , Inflammation/drug therapy , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Motor Activity/drug effects , Neostriatum/drug effects , Neostriatum/metabolism , Neostriatum/pathology , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Pars Compacta/drug effects , Pars Compacta/metabolism , Pars Compacta/pathology , alpha-Synuclein/metabolism
8.
Morphologie ; 101(332): 9-18, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27423214

ABSTRACT

Bone is a tissue with multiple functions that is built from the molecular to anatomical levels to resist and adapt to mechanical strains. Among all the factors that might control the bone organization, a role for several gut hormones called "incretins" has been suspected. The present review summarizes the current evidences on the effects of glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) in bone physiology.


Subject(s)
Bone and Bones/physiology , Enteroendocrine Cells/metabolism , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Incretins/metabolism , Animals , Bone and Bones/cytology , Bone and Bones/metabolism , Bone and Bones/pathology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Gastric Inhibitory Polypeptide/analogs & derivatives , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Incretins/therapeutic use , Osteoblasts/metabolism , Osteoclasts/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Stress, Mechanical
9.
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
10.
Br J Pharmacol ; 173(1): 27-38, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26359804

ABSTRACT

BACKGROUND AND PURPOSE: Specific, high potency receptor antagonists are valuable tools when evaluating animal and human physiology. Within the glucose-dependent, insulinotropic polypeptide (GIP) system, considerable attention has been given to the presumed GIP receptor antagonist, (Pro3)GIP, and its effect in murine studies. We conducted a pharmacological analysis of this ligand including interspecies differences between the rodent and human GIP system. EXPERIMENTAL APPROACH: Transiently transfected COS-7 cells were assessed for cAMP accumulation upon ligand stimulation and assayed in competition binding using (125) I-human GIP. Using isolated perfused pancreata both from wild type and GIP receptor-deficient rodents, insulin-releasing, glucagon-releasing and somatostatin-releasing properties in response to species-specific GIP and (Pro3)GIP analogues were evaluated. KEY RESULTS: Human (Pro3)GIP is a full agonist at human GIP receptors with similar efficacy (Emax ) for cAMP production as human GIP, while both rat and mouse(Pro3)GIP were partial agonists on their corresponding receptors. Rodent GIPs are more potent and efficacious at their receptors than human GIP. In perfused pancreata in the presence of 7 mM glucose, both rodent (Pro3)GIP analogues induced modest insulin, glucagon and somatostatin secretion, corresponding to the partial agonist activities observed in cAMP production. CONCLUSIONS AND IMPLICATIONS: When evaluating new compounds, it is important to consider interspecies differences both at the receptor and ligand level. Thus, in rodent models, human GIP is a comparatively weak partial agonist. Human (Pro3)GIP was not an antagonist at human GIP receptors, so there is still a need for a potent antagonist in order to elucidate the physiology of human GIP.


Subject(s)
Drug Partial Agonism , Gastric Inhibitory Polypeptide/pharmacology , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Animals , Binding, Competitive/drug effects , COS Cells , Chlorocebus aethiops , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Gastric Inhibitory Polypeptide/analogs & derivatives , Glucagon/metabolism , Humans , Insulin/metabolism , Iodine Radioisotopes/metabolism , Male , Mice , Pancreas/metabolism , Radioligand Assay , Rats , Somatostatin/metabolism , Species Specificity
11.
Rev Neurosci ; 27(1): 61-70, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26351802

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is a member of the incretin hormones and growth factors. Neurons express the GIP receptor, and GIP and its agonists can pass through the blood brain barrier and show remarkable neuroprotective effects by protecting synapse function and numbers, promoting neuronal proliferation, reducing amyloid plaques in the cortex and reducing the chronic inflammation response of the nervous system. Long-acting analogues of GIP that are protease resistant had been developed as a treatment for type 2 diabetes. It has been found that such GIP analogues show good protective effects in animal models of Alzheimer's disease. Novel dual agonist peptides that activate the GIP receptor and another incretin receptor, glucagon-like peptide -1 (GLP-1), are under development that show superior effects in diabetic patients compared to single GLP-1 agonists. The dual agonists also show great promise in treating neurodegenerative disorders, and there are currently several clinical trials ongoing, testing GLP-1 mimetics in people with Alzheimer's or Parkinson's disease.


Subject(s)
Alzheimer Disease/drug therapy , Gastric Inhibitory Polypeptide/analogs & derivatives , Neuroprotective Agents/therapeutic use , Alzheimer Disease/prevention & control , Animals , Gastric Inhibitory Polypeptide/pharmacology , Glucagon-Like Peptide-1 Receptor/agonists , Humans , Neuroprotective Agents/pharmacology , Receptors, Gastrointestinal Hormone/agonists , Signal Transduction , Synapses/drug effects
12.
Mol Cell Endocrinol ; 401: 120-9, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25449420

ABSTRACT

Compromise of gastric inhibitory polypeptide (GIP) receptor signalling represents a possible therapeutic strategy for the treatment of obesity-related diabetes. This study has characterised and evaluated the C-terminally fatty acid derivatised GIP analogues, GIP(3-30)Cex-K(40)[Pal] and Pro(3)GIP(3-30)Cex-K(40)[Pal], as potential GIP inhibitors. Both GIP analogues lack the two N-terminal amino acids cleaved by DPP-4 and have addition of nine amino acids from the C-terminal of exendin(1-39), Cex. GIP(3-30)Cex-K(40)[Pal] and Pro(3)GIP(3-30)Cex-K(40)[Pal] effectively (p < 0.01 to p < 0.001) inhibited GIP-induced cAMP production and insulin secretion in vitro. In normal mice, GIP(3-30)Cex-K(40)[Pal] and Pro(3)GIP(3-30)Cex-K(40)[Pal] displayed a significant (p < 0.05 to p < 0.001) and prolonged inhibitory effect on GIP-induced glucose-lowering and insulin-releasing actions. When injected once daily for 21 days in obese-diabetic high fat fed mice, both GIP(3-30)Cex-K(40)[Pal] and Pro(3)GIP(3-30)Cex-K(40)[Pal] significantly reduced body weight (p < 0.01 to p < 0.001) and lowered circulating glucose (p < 0.001) and insulin (p < 0.01 to p < 0.001) concentrations. The observed beneficial changes were independent of effects on energy intake, locomotor activity or metabolic rate. Oral and intraperitoneal glucose tolerance were significantly (p < 0.05 to p < 0.001) improved in both treatment groups at the end of the study, despite reduced glucose-induced plasma insulin concentrations. This improvement of metabolic control was accompanied by enhanced (p < 0.05 to p < 0.01) insulin sensitivity compared with high fat controls. These data demonstrate the potential offered by GIP(3-30)Cex-K(40)[Pal] and Pro(3)GIP(3-30)Cex-K(40)[Pal] for the treatment of obesity-related diabetes.


Subject(s)
Anti-Obesity Agents/administration & dosage , Diet, High-Fat/adverse effects , Gastric Inhibitory Polypeptide/analogs & derivatives , Gastric Inhibitory Polypeptide/antagonists & inhibitors , Obesity/drug therapy , Administration, Oral , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Drug Administration Schedule , Gastric Inhibitory Polypeptide/chemistry , Glucose Tolerance Test , Injections, Intraperitoneal , Insulin/blood , Lipoylation , Mice , Obesity/chemically induced
13.
Diabetes Obes Metab ; 16(1): 75-85, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23859463

ABSTRACT

AIM: Glucose-dependent insulinotropic peptide (GIP) is an incretin hormone that is released from intestinal K cells in response to nutrient ingestion. We aimed to investigate the therapeutic potential of the novel N- and C-terminally modified GIP analogue AC163794. METHODS: AC163794 was synthesized by solid-phase peptide synthesis. Design involved the substitution of the C-terminus tail region of the dipeptidyl peptidase IV (DPP-IV)-resistant GIP analogue [d-Ala(2) ]GIP(1-42) with the unique nine amino acid tail region of exenatide. The functional activity and binding of AC163794 to the GIP receptor were evaluated in RIN-m5F ß-cells. In vitro metabolic stability was tested in human plasma and kidney membrane preparations. Acute insulinotropic effects were investigated in isolated mouse islets and during an intravenous glucose tolerance test in normal and diabetic Zucker fatty diabetic (ZDF) rats. The biological actions of AC163794 were comprehensively assessed in normal, ob/ob and high-fat-fed streptozotocin (STZ)-induced diabetic mice. Acute glucoregulatory effects of AC163794 were tested in diet-induced obese mice treated subchronically with AC3174, the exendatide analogue [Leu(14) ] exenatide. Human GIP or [d-Ala(2) ]GIP(1-42) were used for comparison. RESULTS: AC163794 exhibited nanomolar functional GIP receptor potency in vitro similar to GIP and [d-Ala(2) ]GIP(1-42). AC163794 was metabolically more stable in vitro and displayed longer duration of insulinotropic action in vivo versus GIP and [d-Ala(2) ]GIP(1-42). In diabetic mice, AC163794 improved HbA1c through enhanced insulinotropic action, partial restoration of pancreatic insulin content and improved insulin sensitivity with no adverse effects on fat storage and metabolism. AC163794 provided additional baseline glucose-lowering when injected to mice treated with AC3174. CONCLUSIONS: These studies support the potential use of a novel GIP analogue AC163794 for the treatment of type 2 diabetes.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Gastric Inhibitory Polypeptide/analogs & derivatives , Gastric Inhibitory Polypeptide/metabolism , Hypoglycemic Agents/pharmacology , Incretins/pharmacology , Obesity/metabolism , Animals , Chemistry, Pharmaceutical , Diabetes Mellitus, Experimental/drug therapy , Female , Gastric Inhibitory Polypeptide/chemical synthesis , Gastric Inhibitory Polypeptide/drug effects , Gastric Inhibitory Polypeptide/pharmacology , Male , Mice , Mice, Obese , Obesity/drug therapy , Rats , Rats, Sprague-Dawley , Rats, Zucker
14.
Biochim Biophys Acta ; 1830(6): 3407-13, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23518200

ABSTRACT

BACKGROUND: Rapid enzymatic degradation of the incretin hormone, glucose-dependent insulinotropic polypeptide (GIP), limits therapeutic use of the native peptide for diabetes. However, enzymatically stable analogues of GIP, such as (d-Ala(2))GIP, have been generated, but are still susceptible to renal filtration. METHODS: The present study examines the in vitro and in vivo biological actions of a novel, acylated GIP analogue, (d-Ala(2))GIP[Lys(37)PAL]. RESULTS: In BRIN-BD11 cells, (d-Ala(2))GIP[Lys(37)PAL] concentration-dependently stimulated (p<0.05 to p<0.001) insulin secretion at 5.6 and 16.7mM glucose. Intraperitoneal administration of (d-Ala(2))GIP[Lys(37)PAL] to normal mice 8h prior to a glucose load significantly reduced (p<0.05) the overall glycaemic excursion compared to controls, and increased (p<0.001) the insulinotropic response compared to (d-Ala(2))GIP and saline treated high fat control mice. Once daily administration of (d-Ala(2))GIP[Lys(37)PAL] for 21days in high fat fed mice did not affect energy intake, body weight or fat deposition. However, circulating blood glucose was significantly lower (p<0.05) accompanied by increased (p<0.05) insulin concentrations by day 21. In addition, (d-Ala(2))GIP[Lys(37)PAL] treatment significantly (p<0.01) reduced the overall glycaemic excursion and increased pancreatic insulin content (p<0.05) and the insulinotropic response (p<0.01) to an exogenous glucose challenge on day 21. Chronic treatment with (d-Ala(2))GIP[Lys(37)PAL] did not result in resistance to the metabolic effects of a bolus injection of native GIP. Finally, insulin sensitivity was significantly improved (p<0.001) in (d-Ala(2))GIP[Lys(37)PAL] treated mice compared to high fat controls. CONCLUSIONS: These data confirm that (d-Ala(2))GIP[Lys(37)PAL] is a stable, long-acting potent GIP agonist. GENERAL SIGNIFICANCE: (d-Ala(2))GIP[Lys(37)PAL] may be suitable for further evaluation and future clinical development.


Subject(s)
Body Fat Distribution , Diabetes Mellitus, Experimental/drug therapy , Gastric Inhibitory Polypeptide/analogs & derivatives , Gastric Inhibitory Polypeptide/pharmacology , Hypoglycemic Agents/pharmacology , Animals , Diabetes Mellitus, Experimental/metabolism , Gastric Inhibitory Polypeptide/agonists , Gastric Inhibitory Polypeptide/chemical synthesis , Gastric Inhibitory Polypeptide/chemistry , Gastrointestinal Agents/chemical synthesis , Gastrointestinal Agents/chemistry , Gastrointestinal Agents/pharmacology , Glucose/metabolism , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/chemistry , Insulin/metabolism , Male , Mice
15.
Diabetes Obes Metab ; 15(7): 650-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23388064

ABSTRACT

AIM: The incretin hormones, glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) and cholecystokinin (CCK) are gastrointestinal peptides with important physiological effects. However, rapid enzymatic degradation results in short-lived biological actions. METHODS: This study has examined metabolic actions of exendin-4, GIP[mPEG] and a novel CCK-8 analogue, (pGlu-Gln)-CCK-8 as enzymatically stable forms of GLP-1, GIP and CCK, respectively. RESULTS: All peptides significantly (p < 0.01-p < 0.001) stimulated insulin secretion from BRIN BD11 cells, and acute in vivo experiments confirmed prominent antihyperglycaemic and insulinotropic responses to GLP-1 or GIP receptor activation in normal mice. Twice daily injection of (pGlu-Gln)-CCK-8 alone and in combination with exendin-4 or GIP[mPEG] in high fat-fed mice significantly decreased accumulated food intake (p < 0.05-p < 0.01), body weight gain (p < 0.05-p < 0.01) and improved (p < 0.05) insulin sensitivity in high fat-fed mice. However, there was no evidence for superior effects compared to (pGlu-Gln)-CCK-8 alone. Combined treatment of (pGlu-Gln)-CCK-8 and exendin-4 resulted in significantly (p < 0.05) lowered circulating glucose levels and improved (p < 0.05) intraperitoneal glucose tolerance. These effects were superior to either treatment regime alone but not associated with altered insulin concentrations. A single injection of (pGlu-Gln)-CCK-8, or combined with exendin-4, significantly (p < 0.05) lowered blood glucose levels 24 h post injection in untreated high fat-fed mice. CONCLUSION: This study highlights the potential of (pGlu-Gln)-CCK-8 alone and in combination with incretin hormones for the treatment of type 2 diabetes.


Subject(s)
Anti-Obesity Agents/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Gastric Inhibitory Polypeptide/analogs & derivatives , Hypoglycemic Agents/therapeutic use , Obesity/drug therapy , Peptides/therapeutic use , Sincalide/analogs & derivatives , Venoms/therapeutic use , Animals , Anti-Obesity Agents/pharmacology , Appetite Regulation/drug effects , Cell Line , Diabetes Mellitus, Type 2/complications , Diet, High-Fat/adverse effects , Drug Therapy, Combination , Exenatide , Gastric Inhibitory Polypeptide/pharmacology , Gastric Inhibitory Polypeptide/therapeutic use , Hyperglycemia/prevention & control , Hypoglycemic Agents/pharmacology , Incretins/pharmacology , Incretins/therapeutic use , Insulin/agonists , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred Strains , Obesity/complications , Obesity/etiology , Peptides/pharmacology , Rats , Sincalide/pharmacology , Sincalide/therapeutic use , Time Factors , Venoms/pharmacology
16.
Neuroscience ; 228: 294-300, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-23103794

ABSTRACT

Type 2 diabetes mellitus has been identified as a risk factor for Alzheimer's disease (AD). Insulin is a neuroprotective growth factor, and an impairment of insulin signalling has been found in AD brains. Glucose-dependent insulinotropic polypeptide (GIP), an incretin hormone, normalises insulin signalling and also acts as a neuroprotective growth factor. GIP plays an important role in memory formation, synaptic plasticity and cell proliferation. We have shown previously that the long-lasting incretin hormone analogue D-Ala(2)GIP protects memory formation and synaptic plasticity, reduces plaques, normalises the proliferation of stem cells, reduces the activation of microglia, and prevents the loss of synapses in the cortex of the APPswe/PS1deltaE9 mouse model of Alzheimer's disease. D-Ala(2)GIP was injected for 35 days at 25 nmol/kg i.p. once daily in APP/PS1 male mice and wild-type (WT) littermates aged 6, 12 and 19 months. In a follow-up study, we analysed plaque load, the activation of astrocytes as a means of chronic inflammation in the brain, and oxidative stress in the brains of these mice (8-oxoguanine levels). D-Ala(2)GIP reduced the amyloid plaque load in 12- and 19-month-old mice, and the inflammation response as shown in the reduction of activated astrocytes in 12- and 19-month old APP/PS1 mice. Chronic oxidative stress in the brain was reduced in 12- and 19-month-old mice as shown in the reduction of 8-oxoguanine levels in the cortex of D-Ala(2)GIP-injected APP/PS1 mice. The results demonstrate that D-Ala(2)GIP has neuroprotective properties on key markers found in Alzheimer's disease. This finding shows that novel GIP analogues have the potential to be developed as novel therapeutics for Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Gastric Inhibitory Polypeptide/therapeutic use , Gliosis/metabolism , Oxidative Stress/physiology , Plaque, Amyloid/metabolism , Presenilin-1/genetics , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Female , Gastric Inhibitory Polypeptide/analogs & derivatives , Gastric Inhibitory Polypeptide/pharmacology , Gliosis/drug therapy , Gliosis/pathology , Incretins/pharmacology , Incretins/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidative Stress/drug effects , Plaque, Amyloid/drug therapy , Plaque, Amyloid/pathology
17.
Eur J Pharmacol ; 674(2-3): 294-306, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22115896

ABSTRACT

Type 2 diabetes is a risk factor for Alzheimer's disease. Insulin receptor desensitisation has been found in Alzheimer brains, which may be the underlying link. Glucose-dependent insulinotropic polypeptide (GIP), an incretin hormone, normalises insulin signalling in diabetes. GIP and the GIP receptors are widely expressed in the brain, and GIP has been shown to have growth factor and neuroprotective properties. Here we investigate the potential therapeutic properties of different doses of the protease resistant long-lasting GIP receptor agonist D-Ala2GIP and the antagonist (Pro3)GIP in C57Bl/6 mice. We found that after acute injection, D-Ala2GIP had few effects on general behaviour in the open field at any dose tested (2.5, 25, 100, or 250 nmol/kg i.p.). In memory tests, no change was observed, whilst (Pro3)GIP at 25 nmol/kg i.p. impaired memory formation. In a chronic study over 4 weeks, mice injected with D-Ala2GIP (2.5 or 25 nmol/kg i.p.) and (Pro3)GIP (25 nmol/kg i.p.) learned a water maze task and object recognition task without impairment. In LTP recording in area CA1, both (Pro3)GIP as well as D-Ala2GIP enhanced LTP formation. In addition, the proliferation of neuronal progenitor cells in the dentate gyrus was increased both by D-Ala2GIP and (Pro3)GIP. The results show that the antagonist (Pro3)GIP has agonistic effects in chronic use, and both (Pro3)GIP and the agonist D-Ala2GIP are safe to use in wt mice and induces no major behavioural side effects nor impairments in learning whilst enhancing LTP and neuronal progenitor cell proliferation, which may be useful in treating neurodegenerative diseases.


Subject(s)
Cognition/drug effects , Gastric Inhibitory Polypeptide/analogs & derivatives , Gastric Inhibitory Polypeptide/pharmacology , Neurogenesis/drug effects , Neuronal Plasticity/drug effects , Synapses/drug effects , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/physiology , Cell Proliferation/drug effects , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Dentate Gyrus/physiology , Exploratory Behavior/drug effects , Gastric Inhibitory Polypeptide/administration & dosage , Long-Term Potentiation/drug effects , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Receptors, Gastrointestinal Hormone/agonists , Synapses/metabolism , Time Factors
18.
Peptides ; 32(5): 938-45, 2011 May.
Article in English | MEDLINE | ID: mdl-21334410

ABSTRACT

GIP receptor knockout mice were shown to be protected from the development of obesity on a high fat diet, suggesting a role of GIP in the development of obesity. In our study we aimed to test the hypothesis if excess of GIP could accelerate development of obesity and to identify GIP gene targets in adipose tissue. Therefore, mice were kept on a chow or a high fat diet and during the last 2 weeks D-Ala(2)-GIP or PBS injections were performed. Afterwards, serum LPL activity and several biochemical parameters (TG, FFA, cholesterol, glucose, insulin, resistin, IL-6, IL-1ß, TNFα, GIP) were measured. Fat tissue was isolated and QPCR was performed for a set of genes involved in energy metabolism and inflammation. A DNA-microarray was used to identify GIP gene targets in adipose tissue of the chow diet group. We found that the D-Ala(2)-GIP injections caused a significant decrease in both body weight and LPL activity compared to controls. Serum biochemical parameters were not affected by D-Ala(2)-GIP, with an exception for resistin and insulin. The set of inflammatory genes were significantly decreased in adipose tissue in the D-Ala(2)-GIP injected animals on a chow diet. A DNA-microarray revealed that APO-genes and CYP-genes were affected by D-Ala(2)-GIP treatment in adipose tissue. These results suggest that the body weight-reducing effect of D-Ala(2)-GIP may be explained by lower LPL activity and insulin serum level. Moreover, the identified GIP candidate gene targets in adipose tissue link GIP action to lipid metabolism exerted by APO and CYP genes.


Subject(s)
Body Weight/drug effects , Gastric Inhibitory Polypeptide/pharmacology , Lipoprotein Lipase/blood , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Dietary Fats/toxicity , Gastric Inhibitory Polypeptide/analogs & derivatives , Gastric Inhibitory Polypeptide/blood , Insulin/blood , Interleukin-6/blood , Lipid Metabolism/drug effects , Lipoprotein Lipase/metabolism , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Resistin/blood , Tumor Necrosis Factor-alpha/blood
19.
Expert Opin Investig Drugs ; 19(9): 1039-48, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20698813

ABSTRACT

IMPORTANCE TO THE FIELD: Glucose-dependent insulinotropic polypeptide (GIP) is an incretin hormone that potentiates nutrient-induced insulin release. To date, the physiological importance of GIP has received much less attention than its younger sister incretin hormone glucagon-like peptide-1. Thus, it is worthwhile to refocus on this important and somewhat neglected incretin hormone. AREAS COVERED IN THIS REVIEW: The potential role of GIP as a treatment option for type 2 diabetes is highlighted. Furthermore, the use of GIP as a new therapeutic option for obesity, osteoporosis and cognitive impairment is also considered. WHAT THE READER WILL GAIN: Long-acting GIP receptor agonists offer a potential new class of antidiabetic drugs. Furthermore, recent observations suggest an as yet untapped potential for GIP agonists in the treatment of osteoporosis and cognitive impairment. In addition, GIP is known to play a role in lipid metabolism and fat deposition. Accordingly, both genetic and chemical ablation of GIP signalling in mice with obesity-diabetes can protect against, or reverse, many of the obesity-associated metabolic disturbances. This review focuses on preclinical data generated to date. TAKE HOME MESSAGE: GIP-based therapeutics have potential for the treatment of type 2 diabetes and obesity, with the possibility of further beneficial actions in osteoporosis and cognitive decline.


Subject(s)
Alzheimer Disease , Diabetes Mellitus, Type 2 , Gastric Inhibitory Polypeptide , Obesity , Osteoporosis , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Cell Line , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/physiopathology , Gastric Inhibitory Polypeptide/agonists , Gastric Inhibitory Polypeptide/analogs & derivatives , Gastric Inhibitory Polypeptide/metabolism , Gastric Inhibitory Polypeptide/therapeutic use , Humans , Hypoglycemic Agents/therapeutic use , Obesity/drug therapy , Obesity/metabolism , Osteoporosis/drug therapy , Osteoporosis/metabolism , Osteoporosis, Postmenopausal/drug therapy , Osteoporosis, Postmenopausal/metabolism , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Gastrointestinal Hormone/metabolism
20.
Recent Pat CNS Drug Discov ; 5(2): 109-17, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20337586

ABSTRACT

Analogues of the incretins Glucagon-like peptide 1 (GLP-1) and Glucose-dependent insulinotropic peptide (GIP) have been developed to treat type 2 diabetes mellitus. They are protease resistant and have a longer biological half life than the native peptides. Some of these novel analogues can cross the blood-brain barrier, have neuroprotective effects, activate neuronal stem cells in the brain, and can improve cognition. The receptors for GIP and GLP-1 are expressed in neurons, and both GIP and GLP-1 are expressed and released as transmitters by neurons. GIP analogues such as DAla(2)GIP and GLP-1 analogues such as liraglutide enhance synaptic plasticity in the brain and also reverse the betaamyloid induced impairment of synaptic plasticity. In mouse models of Alzheimer's disease, GLP-1 analogues Val(8)GLP-1 and liraglutide prevent memory impairment and the block of synaptic plasticity in the brain. Since two GLP- 1 analogues exendin-4 (Exenatide, Byetta) and liraglutide (Victoza) are already on the market as treatments for Type 2 diabetes, and others are in late stage clinical trials, these drugs show promise as treatments for neurodegenerative diseases such as Alzheimer's disease. Currently, there are three patents covering native GLP-1 and different GLP-1 analogues and one patent for the use of GIP and different GIP analogues for the treatment of neurodegenerative diseases.


Subject(s)
Alzheimer Disease/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Gastric Inhibitory Polypeptide/therapeutic use , Glucagon-Like Peptide 1/therapeutic use , Incretins/therapeutic use , Neuroprotective Agents/therapeutic use , Amino Acid Sequence , Animals , Blood-Brain Barrier/metabolism , Gastric Inhibitory Polypeptide/analogs & derivatives , Gastric Inhibitory Polypeptide/pharmacology , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/pharmacology , Humans , Models, Neurological , Molecular Sequence Data , Neuroprotective Agents/pharmacology , Patents as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...