Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Br J Pharmacol ; 180(13): 1674-1689, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36683195

RESUMO

BACKGROUND AND PURPOSE: Glucagon-like peptide-2 (GLP-2) is secreted postprandially by enteroendocrine L-cells and stimulates growth of the gut and bone. One GLP-2 analogue is approved for short bowel syndrome (SBS). To improve therapeutic efficacy, we developed biased GLP-2 receptor (GLP-2R) agonists through N-terminal modifications. EXPERIMENTAL APPROACH: Variants with Ala and Trp substitutions of the first seven positions of GLP-2(1-33) were studied in vitro for affinity, G protein activation (cAMP accumulation), recruitment of ß-arrestin 1 and 2, and internalization of the human and mouse GLP-2R. The intestinotrophic actions of the most efficacious (cAMP) biased variant were examined in mice. KEY RESULTS: Ala substitutions had more profound effects than Trp substitutions. For both, alterations at positions 1, 3 and 6 most severely impaired activity. ß-arrestin recruitment was more affected than cAMP accumulation. Among Ala substitutions, [H1A], [D3A] and [F6A] impaired potency (EC50 ) for cAMP-accumulation >20-fold and efficacy (Emax ) to 48%-87%, and were unable to recruit arrestins. The Trp substitutions, [A2W], [D3W] and [G4W] were partial agonists (Emax of 46%-59%) with 1.7-12-fold decreased potencies in cAMP and diminished ß-arrestin recruitment. The biased variants, [F6A], [F6W] and [S7W] induced less GLP-2R internalization compared with GLP-2, which induced internalization in a partly arrestin-independent manner. In mice, [S7W] enhanced gut trophic actions with increased weight of the small intestine, increased villus height and crypt depth compared with GLP-2. CONCLUSION AND IMPLICATIONS: G protein-biased GLP-2R agonists with diminished receptor desensitization have superior intestinotrophic effects and may represent improved treatment of intestinal insufficiency including SBS.


Assuntos
Arrestina , Peptídeo 2 Semelhante ao Glucagon , Camundongos , Humanos , Animais , Peptídeo 2 Semelhante ao Glucagon/farmacologia , Arrestina/metabolismo , Proteínas de Ligação ao GTP/metabolismo , beta-Arrestinas/metabolismo , Arrestinas , beta-Arrestina 1/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo
2.
Br J Pharmacol ; 179(18): 4486-4499, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35710141

RESUMO

BACKGROUND AND PURPOSE: The incretin hormone, gastric inhibitory peptide/glucose-dependent insulinotropic polypeptide (GIP), secreted by the enteroendocrine K-cells in the proximal intestine, may regulate lipid metabolism and adiposity, but its exact role in these processes is unclear. EXPERIMENTAL APPROACH: We characterized in vitro and in vivo antagonistic properties of a novel GIP analogue, mGIPAnt-1. We further assessed the in vivo pharmacokinetic profile of this antagonist, as well as its ability to affect high-fat diet (HFD)-induced body weight gain in ovariectomised mice during an 8-week treatment period. KEY RESULTS: mGIPAnt-1 showed competitive antagonistic properties to the GIP receptor in vitro as it inhibited GIP-induced cAMP accumulation in COS-7 cells. Furthermore, mGIPAnt-1 was capable of inhibiting GIP-induced glucoregulatory and insulinotropic effects in vivo and has a favourable pharmacokinetic profile with a half-life of 7.2 h in C57Bl6 female mice. Finally, sub-chronic treatment with mGIPAnt-1 in ovariectomised HFD mice resulted in a reduction of body weight and fat mass. CONCLUSION AND IMPLICATIONS: mGIPAnt-1 successfully inhibited acute GIP-induced effects in vitro and in vivo and sub-chronically induces resistance to HFD-induced weight gain in ovariectomised mice. Our results support the development of GIP antagonists for the therapy of obesity.


Assuntos
Dieta Hiperlipídica , Receptores dos Hormônios Gastrointestinais , Animais , Glicemia/metabolismo , Peso Corporal , Feminino , Polipeptídeo Inibidor Gástrico/metabolismo , Polipeptídeo Inibidor Gástrico/farmacologia , Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/tratamento farmacológico , Obesidade/etiologia , Obesidade/metabolismo , Aumento de Peso
3.
Br J Pharmacol ; 179(18): 4473-4485, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35523760

RESUMO

BACKGROUND AND PURPOSE: To fully elucidate the regulatory role of the GLP-2 system in the gut and the bones, potent and selective GLP-2 receptor (GLP-2R) antagonists are needed. Searching for antagonist activity, we performed systematic N-terminal truncations of human GLP-2(1-33). EXPERIMENTAL APPROACH: COS-7 cells were transfected with the human GLP-2R and assessed for cAMP accumulation or competition binding using 125 I-GLP-2(1-33)[M10Y]. To examine selectivity, COS-7 cells expressing human GLP-1 or GIP receptors were assessed for cAMP accumulation. KEY RESULTS: Affinity of the N-terminally truncated GLP-2 peptides for the GLP-2 receptor decreased with reduced N-terminal peptide length (Ki 6.5-871 nM), while increasing antagonism appeared with inhibitory potencies (IC50 ) values from 79 to 204 nM for truncation up to GLP-2(4-33) and then declined. In contrast, truncation-dependent increases in intrinsic activity were observed from an Emax of only 20% for GLP-(2-33) up to 46% for GLP-2(6-33) at 1 µM, followed by a decline. GLP-2(9-33) had the highest intrinsic efficacy (Emax 65%) and no antagonistic properties. Moreover, with truncations up to GLP-2(8-33), a gradual loss in selectivity for the GLP-2 receptor appeared with increasing GLP-1 receptor (GLP-1R) inhibition (up to 73% at 1 µM). Lipidation of the peptides improved antagonism (IC50 down to 7.9 nM) for both the GLP-2 and the GLP-1R. CONCLUSION AND IMPLICATIONS: The N-terminus of GLP-2 is crucial for GLP-2R activity and selectivity. Our observations form the basis for the development of tool compounds for further characterization of the GLP-2 system.


Assuntos
Peptídeo 1 Semelhante ao Glucagon , Receptor do Peptídeo Semelhante ao Glucagon 1 , Animais , Células COS , Chlorocebus aethiops , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 2 , Humanos , Peptídeos/química
4.
Pharmacol Res ; 176: 106058, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34995796

RESUMO

The intestinal hormones glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-2 (GLP-2) are key regulators of postprandial bone turnover in humans. We hypothesized that GIP and GLP-2 co-administration would provide stronger effect on bone turnover than administration of the hormones separately, and tested this using subcutaneous injections of GIP and GLP-2 alone or in combination in humans. Guided by these findings, we designed series of GIPR-GLP-2R co-agonists as template for new osteoporosis treatment. The clinical experiment was a randomized cross-over design including 10 healthy men administered subcutaneous injections of GIP and GLP-2 alone or in combination. The GIPR-GLP-2R co-agonists were characterized in terms of binding and activation profiles on human and rodent GIP and GLP-2 receptors, and their pharmacokinetic (PK) profiles were improved by dipeptidyl peptidase-4 protection and site-directed lipidation. Co-administration of GIP and GLP-2 in humans resulted in an additive reduction in bone resorption superior to each hormone individually. The GIPR-GLP-2R co-agonists, designed by combining regions of importance for cognate receptor activation, obtained similar efficacies as the two native hormones and nanomolar potencies on both human receptors. The PK-improved co-agonists maintained receptor activity along with their prolonged half-lives. Finally, we found that the GIPR-GLP-2R co-agonists optimized toward the human receptors for bone remodeling are not feasible for use in rodent models. The successful development of potent and efficacious GIPR-GLP-2R co-agonists, combined with the improved effect on bone metabolism in humans by co-administration, support these co-agonists as a future osteoporosis treatment.


Assuntos
Remodelação Óssea/efeitos dos fármacos , Polipeptídeo Inibidor Gástrico/farmacologia , Peptídeo 2 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 2/agonistas , Receptores dos Hormônios Gastrointestinais/agonistas , Adulto , Animais , Células COS , Chlorocebus aethiops , Estudos Cross-Over , Feminino , Polipeptídeo Inibidor Gástrico/sangue , Polipeptídeo Inibidor Gástrico/farmacocinética , Peptídeo 2 Semelhante ao Glucagon/sangue , Peptídeo 2 Semelhante ao Glucagon/farmacocinética , Receptor do Peptídeo Semelhante ao Glucagon 2/genética , Humanos , Masculino , Camundongos Endogâmicos C57BL , Osteoporose/tratamento farmacológico , Receptores dos Hormônios Gastrointestinais/genética , Método Simples-Cego , Adulto Jovem
5.
Basic Clin Pharmacol Toxicol ; 126 Suppl 6: 122-132, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31299132

RESUMO

In patients with type 2 diabetes mellitus (T2DM), the insulinotropic action of the GIP system is desensitized, whereas this is not the case for the GLP-1 system. This has raised an interesting discussion of whether GIP agonists or antagonists are most suitable for future treatment of T2DM together with GLP-1-based therapies. Homozygous carriers of the GIP receptor (GIPR) variant, [E354Q], display lower bone mineral density, increased bone fracture risk and slightly increased blood glucose. Here, we present an in-depth molecular pharmacological phenotyping of GIPR-[E354Q]. In silico modelling suggested similar interaction of the endogenous agonist GIP(1-42) to [E354Q] as to GIPR wt. This was supported by homologous competition binding in COS-7 cells revealing GIPR wt-like affinities of GIP(1-42) with Kd values of ~2 nmol/L and wt-like agonist association rates (Kon ). In contrast, the dissociation rates (Koff ) were slower, resulting in 25% higher agonist residence time for GIPR-[E354Q]. Moreover, in Gαs signalling (cAMP production) GIP(1-42) was ~2-fold more potent and more efficacious on GIPR-[E354Q] compared to wt with 17.5% higher basal activity. No difference from GIPR wt was found in the recruitment of ß-arrestin 2, whereas the agonist-induced internalization rate was 2.1- to 2.3-fold faster for [E354Q]. Together with the previously described impaired recycling of [E354Q], our findings with enhanced signalling and internalization rate possibly explained by an altered ligand-binding kinetics will lead to receptor desensitization and down-regulation. This could explain the long-term functional impairment of the GIP system in bone metabolism and blood sugar maintenance for [E354Q] carriers and may shed light on the desensitization of the insulinotropic action of GIP in patients with T2DM.


Assuntos
Polipeptídeo Inibidor Gástrico/metabolismo , Animais , Células COS , Chlorocebus aethiops , Polipeptídeo Inibidor Gástrico/agonistas , Polipeptídeo Inibidor Gástrico/química , Células HEK293 , Humanos , Estrutura Molecular , Transdução de Sinais , beta-Arrestinas
6.
Peptides ; 125: 170224, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31809770

RESUMO

Enzymatic cleavage of endogenous peptides is a commonly used principle to initiate, modulate and terminate action for instance among cytokines and peptide hormones. The incretin hormones, glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), and the related hormone glucagon-like peptide-2 (GLP-2) are all rapidly N-terminally truncated with severe loss of intrinsic activity. The most abundant circulating form of full length GIP(1-42) is GIP(3-42) (a dipeptidyl peptidase-4 (DPP-4) product). GIP(1-30)NH2 is another active form resulting from prohormone convertase 2 (PC2) cleavage of proGIP. Like GIP(1-42), GIP(1-30)NH2 is a substrate for DPP-4 generating GIP(3-30)NH2 which, compared to GIP(3-42), binds with higher affinity and very efficiently inhibits GIP receptor (GIPR) activity with no intrinsic activity. Here, we review the action of these four and multiple other N- and C-terminally truncated forms of GIP with an emphasis on molecular pharmacology, i.e. ligand binding, subsequent receptor activation and desensitization. Our overall conclusion is that the N-terminus is essential for receptor activation as GIP N-terminal truncation leads to decreased/lost intrinsic activity and antagonism (similar to GLP-1 and GLP-2), whereas the C-terminal extension of GIP(1-42), as compared to GLP-1, GLP-2 and glucagon (29-33 amino acids), has no apparent impact on the GIPR in vitro, but may play a role for other properties such as stability and tissue distribution. A deeper understanding of the molecular interaction of naturally occurring and designed GIP-based peptides, and their impact in vivo, may contribute to a future therapeutic targeting of the GIP system - either with agonists or with antagonists, or both.


Assuntos
Polipeptídeo Inibidor Gástrico/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores dos Hormônios Gastrointestinais/metabolismo , Animais , Polipeptídeo Inibidor Gástrico/química , Humanos , Fragmentos de Peptídeos/química , Receptores dos Hormônios Gastrointestinais/química , Relação Estrutura-Atividade
7.
Cell Metab ; 29(3): 719-726.e5, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30449683

RESUMO

Studies on isolated pancreatic islets suggest that neuromedin U (NMU), a brain and gastrointestinal peptide, acts as a decretin hormone, inhibiting glucose-stimulated insulin secretion. We investigated whether this effect could be reproduced in vivo and in isolated perfused rat pancreas. Unlike the incretin hormone, glucagon-like peptide 1 (GLP-1), intravenous NMU administration had no effects on blood glucose and plasma insulin and glucagon in vivo. Moreover, NMU neither changed insulin, glucagon, or somatostatin secretion from isolated perfused rat pancreas, nor affected GLP-1-stimulated insulin and somatostatin secretion. For NMU to act as a decretin hormone, its secretion should increase following glucose ingestion; however, glucose did not affect NMU secretion from isolated perfused rat small intestine, which contained extractable NMU. Furthermore, the two NMU receptors were not detected in endocrine rat or human pancreas. We conclude that NMU does not act as a decretin hormone in rats.


Assuntos
Glucagon/metabolismo , Insulina/metabolismo , Intestino Delgado/metabolismo , Ilhotas Pancreáticas/metabolismo , Neuropeptídeos , Pâncreas/metabolismo , Somatostatina/metabolismo , Animais , Células COS , Chlorocebus aethiops , Humanos , Masculino , Neuropeptídeos/farmacologia , Neuropeptídeos/fisiologia , Ratos , Ratos Wistar , Receptores de Neurotransmissores/metabolismo
8.
Cell Rep ; 25(5): 1127-1134.e2, 2018 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-30380405

RESUMO

The intra-islet theory states that glucagon secretion is suppressed when insulin secretion is stimulated, but glucagon's role in intra-islet paracrine regulation is controversial. This study investigated intra-islet functions of glucagon in mice. We examined glucagon-induced insulin secretion using isolated perfused pancreata from wild-type, GLP-1 receptor (GLP-1R) knockout, diphtheria toxin-induced proglucagon knockdown, ß cell-specific glucagon receptor (Gcgr) knockout, and global Gcgr knockout (Gcgr-/-) mice. We found that glucagon stimulates insulin secretion through both Gcgr and GLP-1R. Moreover, loss of either Gcgr or GLP-1R does not change insulin responses, whereas combined blockage of both receptors significantly reduces insulin secretion. Active GLP-1 is identified in pancreatic perfusate from Gcgr-/- but not wild-type mice, suggesting that ß cell GLP-1R activation results predominantly from glucagon action. Our results suggest that combined activity of glucagon and GLP-1 receptors is essential for ß cell secretory responses, emphasizing a role for paracrine intra-islet glucagon actions to maintain appropriate insulin secretion.


Assuntos
Glucagon/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Transdução de Sinais , Animais , Células COS , Chlorocebus aethiops , AMP Cíclico/metabolismo , Exenatida/farmacologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/antagonistas & inibidores , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Camundongos Endogâmicos C57BL , Perfusão
9.
Am J Physiol Gastrointest Liver Physiol ; 315(1): G53-G65, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29494208

RESUMO

The colonic epithelium harbors a large number of endocrine cells, but little is known about the endocrine functions of the colon. However, the high density of glucagon like peptide-1 (GLP-1)- and peptide-YY (PYY)-secreting L cells is of great interest because of the potential antidiabetic and antiobesity effects of GLP-1 and PYY. Short-chain fatty acids (SCFAs) produced by local bacterial fermentation are suggested to activate the colonic free fatty acid receptors FFAR2 (GPR43) and FFAR3 (GPR41), stimulating the colonic L cells. We used the isolated perfused rat colon as a model of colonic endocrine secretion and studied the effects of the predominant SCFAs formed: acetate, propionate, and butyrate. We show that luminal and especially vascular infusion of acetate and butyrate significantly increases colonic GLP-1 secretion, and to a minor extent also PYY secretion, but only after enhancement of intracellular cAMP. Propionate neither affected GLP-1 nor PYY secretion whether administered luminally or vascularly. A FFAR2- and FFAR3-specific agonist [( S)-2-(4-chlorophenyl)-3,3-dimethyl- N-(5-phenylthiazol-2-yl)butamide (CFMB)/ AR420626 ] had no effect on colonic GLP-1 output, and a FFAR3 antagonist ( AR399519 ) did not decrease the SCFA-induced GLP-1 response. However, the voltage-gated Ca2+-channel blocker nifedipine, the KATP-channel opener diazoxide, and the ATP synthesis inhibitor 2,4-dinitrophenol completely abolished the responses. FFAR2 receptor studies confirmed low-potent partial agonism of acetate, propionate, and butyrate, compared with CFMB, which is a full agonist with ~750-fold higher potency than the SCFAs. In conclusion, SCFAs may increase colonic GLP-1/PYY secretion, but FFAR2/FFAR3 do not seem to be involved. Rather, SCFAs are metabolized and appear to function as a colonocyte energy source. NEW & NOTEWORTHY By the use of in situ isolated perfused rat colon we show that short-chain fatty acids (SCFAs) primarily are used as a colonocyte energy source in the rat, subsequently triggering glucagon like peptide-1 (GLP-1) secretion independent of the free fatty acid receptors FFAR2 and FFAR3. Opposite many previous studies on SCFAs and FFAR2/FFAR3 and GLP-1 secretion, this experimental model allows investigation of the physiological interactions between luminal nutrients and secretion from cells whose function depend critically on their blood supply as well as nerve and paracrine interactions.


Assuntos
Colo , Ácidos Graxos Voláteis/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo YY/metabolismo , Animais , Colo/irrigação sanguínea , Colo/inervação , Colo/metabolismo , Ácidos Graxos Voláteis/classificação , Hormônios Gastrointestinais/metabolismo , Mucosa Intestinal/metabolismo , Modelos Teóricos , Comunicação Parácrina/fisiologia , Ratos , Receptores Acoplados a Proteínas G/classificação , Receptores Acoplados a Proteínas G/metabolismo
10.
Peptides ; 100: 173-181, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29412817

RESUMO

Glucose-dependent insulinotropic polypeptide (GIP) is an intestinal hormone with a broad range of physiological actions. In the postprandial state, the hormone stimulates insulin secretion and during eu- and hypoglycemia, it stimulates glucagon secretion. In addition, GIP increases triacylglycerol (TAG) uptake in adipose tissue and decreases bone resorption. However, the importance of these actions in humans are not clearly understood as a specific GIP receptor (GIPR) antagonist - an essential tool to study GIP physiology - has been missing. Several different GIPR antagonists have been identified comprising both peptides, vaccines against GIP, GIP antibodies or antibodies against the GIPR. However, most of these have only been tested in rodents. In vitro, N- and C-terminally truncated GIP variants are potent and efficacious GIPR antagonists. Recently, GIP(3-30)NH2, a naturally occurring peptide, was shown to block the GIPR in humans and decrease GIP-induced insulin secretion as well as adipose tissue blood flow and TAG uptake. So far, there are no studies with a GIPR antagonist in patients with type 2 diabetes (T2D), but because the elevations in fasting plasma glucagon and paradoxical postprandial glucagon excursions, seen in patients with T2D, are aggravated by GIP, a GIPR antagonist could partly alleviate this and possibly improve the fasting and postprandial glycemia. Since the majority of patients with T2D are overweight, inhibition of GIP-induced fat deposition may be beneficial as well. Here we summarize the studies of GIPR antagonists and discuss the therapeutic potential of the GIP system in humans.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Insulina/metabolismo , Receptores dos Hormônios Gastrointestinais/genética , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Glicemia/efeitos dos fármacos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Polipeptídeo Inibidor Gástrico/metabolismo , Glucagon/metabolismo , Glucose/metabolismo , Humanos , Hipoglicemiantes/química , Receptores dos Hormônios Gastrointestinais/antagonistas & inibidores , Receptores dos Hormônios Gastrointestinais/uso terapêutico
11.
Peptides ; 100: 48-53, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29412831

RESUMO

Oxyntomodulin is a product of the glucagon precursor, proglucagon, produced and released from the endocrine L-cells of the gut after enzymatic processing by the precursor prohormone convertase 1/3. It corresponds to the proglucagon sequence 33-69 and thus contains the entire glucagon sequence plus a C-terminal octapeptide, comprising in total 37 amino acids. As might have been expected, it has glucagon-like bioactivity, but also and more surprisingly also activates the receptor for GLP-1. This has given the molecule an interesting status as a glucagon-GLP-1 co-agonist, which is currently attracting considerable interest for its potential in the treatment of diabetes and obesity. Here, we provide an update on oxyntomodulin with a focus on its potential role in metabolic diseases.


Assuntos
Diabetes Mellitus/tratamento farmacológico , Glucagon/metabolismo , Obesidade/tratamento farmacológico , Oxintomodulina/metabolismo , Aminoácidos/genética , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Glucagon/química , Glucagon/uso terapêutico , Humanos , Obesidade/genética , Obesidade/metabolismo , Oxintomodulina/química , Oxintomodulina/uso terapêutico , Proglucagon/química , Proglucagon/metabolismo , Proglucagon/uso terapêutico , Pró-Proteína Convertases/química , Pró-Proteína Convertases/metabolismo
12.
Biochem Pharmacol ; 150: 97-107, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29378179

RESUMO

GIP(3-30)NH2 is a high affinity antagonist of the GIP receptor (GIPR) in humans inhibiting insulin secretion via G protein-dependent pathways. However, its ability to inhibit G protein-independent signaling is unknown. Here we determine its action on arrestin-recruitment and receptor internalization in recombinant cells. As GIP is adipogenic, we evaluate the inhibitory actions of GIP(3-30)NH2 in human adipocytes. Finally, we determine the receptor selectivity of GIP(3-30)NH2 among other human and animal GPCRs. cAMP accumulation and ß-arrestin 1 and 2 recruitment were studied in transiently transfected HEK293 cells and real-time internalization in transiently transfected HEK293A and in HEK293A ß-arrestin 1 and 2 knockout cells. Furthermore, human subcutaneous adipocytes were assessed for cAMP accumulation following ligand stimulation. Competition binding was examined in transiently transfected COS-7 cells using human 125I-GIP(3-30)NH2. The selectivity of human GIP(3-30)NH2 was examined by testing for agonistic and antagonistic properties on 62 human GPCRs. Human GIP(3-30)NH2 inhibited GIP(1-42)-induced cAMP and ß-arrestin 1 and 2 recruitment on the human GIPR and Schild plot analysis showed competitive antagonism with a pA2 and Hill slope of 16.8 nM and 1.11 ±â€¯0.02 in cAMP, 10.6 nM and 1.15 ±â€¯0.05 in ß-arrestin 1 recruitment, and 10.2 nM and 1.06 ±â€¯0.05 in ß-arrestin 2 recruitment. Efficient internalization of the GIPR was dependent on the presence of either ß-arrestin 1 or 2. Moreover, GIP(3-30)NH2 inhibited GIP(1-42)-induced internalization in a concentration-dependent manner and notably also inhibited GIP-mediated signaling in human subcutaneous adipocytes. Finally, the antagonist was established as GIPR selective among 62 human GPCRs being species-specific with high affinity binding to the human and non-human primate (Macaca fascicularis) GIPRs, and low affinity binding to the rat and mouse GIPRs (Kd values of 2.0, 2.5, 31.6 and 100 nM, respectively). In conclusion, human GIP(3-30)NH2 is a selective and species-specific GIPR antagonist with broad inhibition of signaling and internalization in transfected cells as well as in human adipocytes.


Assuntos
Polipeptídeo Inibidor Gástrico/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Receptores dos Hormônios Gastrointestinais/antagonistas & inibidores , Receptores dos Hormônios Gastrointestinais/metabolismo , Transdução de Sinais/fisiologia , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Polipeptídeo Inibidor Gástrico/farmacologia , Células HEK293 , Humanos , Macaca fascicularis , Camundongos , Fragmentos de Peptídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA