Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 583
Filtrar
1.
Diabetologia ; 51(12): 2263-70, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18795252

RESUMO

AIMS/HYPOTHESIS: The glucose-lowering effect of glucagon-like peptide-1 (GLP-1) is based not only upon its potent insulinotropic actions but also on its ability to restrain glucagon secretion. Surprisingly, the closely related glucose-dependent insulinotropic peptide (GIP) stimulates glucagon release. We examined whether the islet hormone somatostatin, which strongly inhibits glucagon secretion, is involved in this divergent behaviour. METHODS: At 1.5 mmol/l glucose and therefore minimal insulin secretion, the glucagon, insulin and somatostatin responses to 20 mmol/l glucose, GLP-1, GIP and somatostatin were studied in the presence of a high-affinity monoclonal somatostatin antibody and of a highly specific somatostatin receptor subtype 2 (SSTR2) antagonist (PRL-2903) in the isolated perfused rat pancreas. RESULTS: In control experiments, GLP-1 at 1 and 10 nmol/l reduced glucagon secretion significantly to 59.0 +/- 6.3% (p < 0.004; n = 5; SSTR2 series; each vs pre-infusion level) and to 48.0 +/- 2.6% (p < 0.001; n = 6; somatostatin antibody series) respectively. During somatostatin antibody administration, GLP-1 still inhibited glucagon secretion significantly, but the effect was less pronounced than in control experiments (p < 0.018). Co-infusion of the SSTR2 antagonist completely abolished the GLP-1-induced suppression of glucagon secretion. In contrast, neither the GIP-induced stimulation of glucagon release nor its inhibition by 20 mmol/l glucose was altered by somatostatin antibody or SSTR2 antagonist administration. CONCLUSIONS/INTERPRETATION: We conclude that GLP-1 is capable of inhibiting glucagon secretion even in the absence of secretory products from the beta cell. It is highly likely that this is mediated via somatostatin interacting with SSTR2 on rat alpha cells. In contrast, GIP and glucose seem to influence the alpha cell independently of somatostatin secretion.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/farmacologia , Glucagon/metabolismo , Pâncreas/metabolismo , Receptores de Somatostatina/metabolismo , Animais , Anticorpos/imunologia , Feminino , Polipeptídeo Inibidor Gástrico/farmacologia , Insulina/metabolismo , Secreção de Insulina , Pâncreas/efeitos dos fármacos , Pâncreas/imunologia , Perfusão , Ratos , Ratos Wistar , Receptores de Somatostatina/antagonistas & inibidores , Somatostatina/imunologia
2.
Neurogastroenterol Motil ; 18(3): 217-25, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16487413

RESUMO

This study demonstrates the expression of functional somatostatin receptor (sstr) subtypes in human circular and longitudinal colonic smooth muscle cells (SMC). Native somatostatin (SS) and sstr subtype-specific analogues were used to characterize the sstr subtypes present in both cell types by contraction/relaxation studies. Qualitative and quantitative mRNA analysis and immunohistochemistry of sstr subtypes were also carried out. sstr subtype 2 mRNA was expressed in circular SMC, and various levels of subtypes 1, 2 and 3 mRNA were expressed in longitudinal colonic SMC. Native SS and each subtype-specific analogue exerted a modest, but significant, contraction, although inhibition of carbachol-induced contraction (relaxation) was the main effect on SMC from both layers. CH-288, a sstr subtype 1-specific analogue, and octreotide, a sstr subtype 2-specific analogue, were the most effective relaxant analogues on longitudinal and circular SMC, respectively. sstr subtypes display a distinct expression pattern on human colonic SMC; on circular SMC, subtype 2 is the only sstr, whereas sstr subtypes 1, 2 and 3 are expressed on human SMC isolated from the longitudinal layer. The contractile effects of SS are mediated through sstr subtype 2 and sstr subtype 1 on circular and longitudinal human colonic SMC, respectively.


Assuntos
Colo/fisiologia , Contração Muscular/fisiologia , Miócitos de Músculo Liso/metabolismo , Receptores de Somatostatina/biossíntese , Células Cultivadas , Colo/efeitos dos fármacos , Fármacos Gastrointestinais/farmacologia , Humanos , Imuno-Histoquímica , Contração Muscular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Octreotida/farmacologia , RNA Mensageiro/análise , Receptores de Somatostatina/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Somatostatina/análogos & derivados , Somatostatina/farmacologia
3.
Peptides ; 23(12): 2259-64, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12535707

RESUMO

Urotensin II is the latest of a growing list of peptides exhibiting potent cardiovascular effects. It is an extremely potent vasoconstrictor in primates; its excretion is elevated in hypertensive patients thus suggesting therapeutic potential for urotensin II analogues, particularly receptor antagonists. In the present study, a number of interesting structural features pertaining to the N-terminus of urotensin II have been evaluated for binding to cloned human and rat urotensin II receptors and functional effects on rat upper thoracic aorta smooth muscle preparations. Shortened octapeptides retained full binding affinities and functional activities, did not require a free N-terminal amino group, and could tolerate an amidated C-terminus. The N-terminal Asp residue present in the octapeptides did not require a negatively charged side chain, merely one which contained a hydrogen bond acceptor CO group which could be present at a variety of positions on the side chain. The side chain could be constrained into a trans-olefinic configuration with full retention of potency, but potency was lost in the cis configuration. N-terminal aromatic amino substituted with polar groups such as OH and NO(2) also resulted in high affinity analogues. Overall, the correlation between binding affinities for the human and rat receptors was quite good. These findings could be of value in the development of more potent urotensin II receptor antagonists based on the previously identified somatostatin antagonist octapeptides which we have recently found, function as relatively weak urotensin II antagonists.


Assuntos
Oligopeptídeos/química , Receptores Acoplados a Proteínas G , Urotensinas/química , Animais , Aorta Torácica/metabolismo , Asparagina/metabolismo , Humanos , Masculino , Oligopeptídeos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Superfície Celular/metabolismo , Relação Estrutura-Atividade , Urotensinas/metabolismo
4.
Peptides ; 22(9): 1427-34, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11514024

RESUMO

Responses to human calcitonin gene-related peptide (hCGRP) and human adrenomedullin (hADM) hAmylin were investigated in isolated mesenteric resistance arteries from the rat. The results of the present investigation show that hCGRP, hAmylin, and hADM induce dose-related vasodilator responses in isolated resistance arteries from the rat mesenteric vascular bed. Vasodilator responses to hCGRP and hAmylin were not altered after denuding the vascular endothelium, after administration of the nitric oxide synthase inhibitor L-NA, or after administration of the soluble guanylate cyclase inhibitor ODQ, suggesting that vasodilator responses to hCGRP and hAmylin are not mediated by the release of nitric oxide from the vascular endothelium and the subsequent increase in cGMP. Vasodilator responses to hCGRP, hAmylin, and hADM were not altered by the vascular selective K+(ATP) channel antagonist U-37883A. The role of the CGRP1 receptor was investigated and responses to hCGRP and hAmylin, but not hADM, were significantly reduced following administration of hCGRP-(8-37). Moreover, vasodilator responses to hCGRP and hAmylin, but not hADM, were significantly reduced by hAmylin-(8-37), suggesting that an hAmylin-(8-37)-sensitive receptor mediates responses to hCGRP and hAmylin in the rat mesenteric artery. These data suggest that hCGRP and hAmylin have direct vasodilator effects in the isolated mesenteric resistance artery that are mediated by hAmylin-(8-37)- and hCGRP-(8-37)-sensitive receptors.


Assuntos
Adamantano/análogos & derivados , Amiloide/farmacologia , Arginina/análogos & derivados , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Artérias Mesentéricas/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Peptídeos/farmacologia , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/fisiologia , Receptores de Peptídeos/fisiologia , Acetilcolina/farmacologia , Adamantano/farmacologia , Adrenomedulina , Animais , Arginina/farmacologia , Cromakalim/metabolismo , Relação Dose-Resposta a Droga , Endotélio Vascular/fisiologia , Inibidores Enzimáticos/farmacologia , Humanos , Técnicas In Vitro , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Artérias Mesentéricas/fisiologia , Morfolinas/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Oxidiazóis/farmacologia , Perfusão , Bloqueadores dos Canais de Potássio , Quinoxalinas/farmacologia , Ratos , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/efeitos dos fármacos , Receptores de Polipeptídeo Amiloide de Ilhotas Pancreáticas , Receptores de Peptídeos/efeitos dos fármacos , Resistência Vascular/efeitos dos fármacos , Vasodilatadores/farmacologia
5.
J Biol Chem ; 276(39): 36652-63, 2001 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-11463790

RESUMO

Few gastrointestinal hormones/neurotransmitters have high affinity peptide receptor antagonists, and little is known about the molecular basis of their selectivity or affinity. The receptor mediating the action of the mammalian bombesin (Bn) peptide, gastrin-releasing peptide receptor (GRPR), is an exception, because numerous classes of peptide antagonists are described. To investigate the molecular basis for their high affinity for the GRPR, two classes of peptide antagonists, a statine analogue, JMV594 ([d-Phe(6),Stat(13)]Bn(6-14)), and a pseudopeptide analogue, JMV641 (d-Phe-Gln-Trp-Ala-Val-Gly-His-Leupsi(CHOH-CH(2))-(CH(2))(2)-CH(3)), were studied. Each had high affinity for the GRPR and >3,000-fold selectivity for GRPR over the closely related neuromedin B receptor (NMBR). To investigate the basis for this, we used a chimeric receptor approach to make both GRPR loss of affinity and NMBR gain of affinity chimeras and a site-directed mutagenesis approach. Chimeric or mutated receptors were transiently expressed in Balb/c 3T3. Only substitution of the fourth extracellular (EC) domain of the GRPR by the comparable NMBR domain markedly decreased the affinity for both antagonists. Substituting the fourth EC domain of NMBR into the GRPR resulted in a 300-fold gain in affinity for JMV594 and an 11-fold gain for JMV641. Each of the 11 amino acid differences between the GRPR and NMBR in this domain were exchanged. The substitutions of Thr(297) in GRPR by Pro from the comparable position in NMBR, Phe(302) by Met, and Ser(305) by Thr decreased the affinity of each antagonist. Simultaneous replacement of Thr(297), Phe(302), and Ser(305) in GRPR by the three comparable NMBR amino acids caused a 500-fold decrease in affinity for both antagonists. Replacing the comparable three amino acids in NMBR by those from GRPR caused a gain in affinity for each antagonist. Receptor modeling showed that each of these three amino acids faced inward and was within 5 A of the putative binding pocket. These results demonstrate that differences in the fourth EC domain of the mammalian Bn receptors are responsible for the selectivity of these two peptide antagonists. They demonstrate that Thr(297), Phe(302), and Ser(305) of the fourth EC domain of GRPR are the critical residues for determining GRPR selectivity and suggest that both receptor-ligand cation-pi interactions and hydrogen bonding are important for their high affinity interaction.


Assuntos
Peptídeos/química , Receptores da Bombesina/antagonistas & inibidores , Receptores da Bombesina/química , Células 3T3 , Sequência de Aminoácidos , Aminoácidos/química , Animais , Sítios de Ligação , DNA Complementar/metabolismo , Concentração Inibidora 50 , Cinética , Metionina/química , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fenilalanina/química , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Homologia de Sequência de Aminoácidos , Serina/química , Transfecção
6.
Peptides ; 22(7): 1119-28, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11445243

RESUMO

Bombesin-like peptides have been implicated as growth factors in various human cancers. Human adenocarcinoma cell lines (Capan-1, Capan-2, MiaPaCa-2 and HPAF) were tested to determine whether they express the gastrin-releasing peptide-preferring bombesin receptor (GRPR) and neuromedin B-preferring bombesin receptor (NMBR). Using RT-PCR the highest level of GRP receptor mRNA was found in HPAF cells. NMB receptor mRNA expression moderate in all cell lines investigated. We therefore selected the HPAF cell line to investigate whether bombesin treatment affects intracellular Ca(2+) ([Ca(2+)](i)), cAMP level, DNA synthesis as a measure of cell proliferation, and expression of three transcription factors: c-fos, c-myc and high mobility group protein IY (HMG-I(Y)).Bombesin administration led to an immediate increase in free intracellular Ca(2+) concentration ([Ca(2+)](i)) but did not change cAMP levels. The peptide also enhanced [(3)H]thymidine incorporation in HPAF cells (but not in the other cell lines), an effect that was concentration dependent, reaching 36 +/- 5% stimulation over control values at 24 h with an EC(50) of 2.27 x 10(-12) M. Furthermore, bombesin stimulated c-fos, c-myc and HMG-I(Y) expression in a time-dependent manner: the c-fos mRNA level increased dramatically in the first 30 min of exposure, then returned to basal level within 2 h, while the c-myc and HMG-I(Y) mRNA levels peaked at 2 h and 4h, respectively. All actions of bombesin were blocked by BME (D-Phe(6)-bombesin-(6-13)-methylester), a selective GRP receptor antagonist, but not by the NMB receptor antagonist BIM-23127 (D-Nal-cyclo[Cys-Tyr-D-Trp-Orn-Val-Cys]-Nal-NH(2)). We conclude that HPAF cells express mRNA for GRP receptors and that functional receptors are present in the cell membrane. The occupation of these receptors leads to a sequence of intracellular events involving rapid mobilization of intracellular Ca(2+), expression of c-fos, c-myc and HMG-I(Y) mRNA, and stimulation of cell proliferation. Conversely, although NMB receptor mRNA can be detected, its actual translation to functional receptors does not reach a detectable level.


Assuntos
Adenocarcinoma/metabolismo , DNA/biossíntese , Neoplasias Pancreáticas/metabolismo , Receptores da Bombesina/metabolismo , Transdução de Sinais , Northern Blotting , Bombesina/farmacologia , Cálcio/metabolismo , Membrana Celular/metabolismo , Clonagem Molecular , Colforsina/farmacologia , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Humanos , Processamento de Imagem Assistida por Computador , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrometria de Fluorescência , Fatores de Tempo , Células Tumorais Cultivadas
7.
Peptides ; 22(7): 1139-51, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11445245

RESUMO

Growth hormone (GH) is used or is being evaluated for efficacy in treatment of short stature, aspects of aging, cardiac disorders, Crohn's disease, and short bowel syndrome. Therefore, we synthesized several stable growth hormone-releasing factor (GRF) analogues that could be therapeutically useful. One potent analog, [D-Ala(2),Aib(8, 18,)Ala(9, 15, 16, 22, 24-26,)Gab(27)]hGRF(1-27)NH(2) (GRF-6), with prolonged infusion caused severe diarrhea in monkeys; however, it had no side-effects in rats. Because GRF has similarity to VIP/PACAP and VIPomas cause diarrhea, this study investigated the ability of this and other GRF analogues to interact with the VIP/PACAP receptors. Rat VPAC(1)-R (rVPAC(1)-R), human VPAC(1)-R (hVPAC(1)-R), rVPAC(2)-R and hVPAC(2)-R stably transfected CHO and PANC 1 cells were made and T47D breast cancer cells containing native human VPAC(1)-R and AR4-2J cells containing PAC(1)-R were used. hGRF(1-29)NH(2) had low affinity for both rVPAC(1)-R and rVPAC(2)-R while VIP had a high affinity for both receptors. GRF-6 had a low affinity for both rVPAC(1)-R and rVPAC(2)-R and very low affinity for the rPAC(1)-R. VIP had a high affinity, whereas hGRF(1-29)NH(2) had a low affinity for both hVPAC(1)-R and hVPAC(2)-R. In contrast GRF-6, while having a low affinity for hVPAC(2)-R, had relatively higher affinity for the hVPAC(1)-R. In guinea pig pancreatic acini, all GRF analogues were full agonists at the VPAC(1)-R causing enzyme secretion. These results demonstrate that in contrast to native hGRF(1-29)NH(2,) GRF-6 has a relatively high affinity for the human VPAC(1)-R but not for the human VPAC(2)-R, rat VPAC(1)-R, rat VPAC(2)-R or rat PAC(1)-R. These results suggest that the substituted GRF analog, GRF-6, likely causes the diarrheal side-effects in monkeys by interacting with the VPAC(1)-R. Furthermore, they demonstrate significant species differences can exist for possible therapeutic peptide agonists of the VIP/PACAP/GRF receptor family and that it is essential that receptor affinity assessments be performed in human cells or from a closely related species.


Assuntos
Hormônio Liberador de Hormônio do Crescimento/análogos & derivados , Hormônio do Crescimento/farmacologia , Receptores de Peptídeo Intestinal Vasoativo/agonistas , Sequência de Aminoácidos , Amilases/metabolismo , Animais , Células CHO , Células Cultivadas , Cricetinae , Relação Dose-Resposta a Droga , Cobaias , Haplorrinos , Humanos , Dados de Sequência Molecular , Pâncreas/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Hipófise/citologia , Hipófise/metabolismo , Ligação Proteica , Ratos , Homologia de Sequência de Aminoácidos , Transfecção , Células Tumorais Cultivadas
8.
J Endocrinol ; 170(1): 251-7, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11431158

RESUMO

Adrenomedullin is a 52-amino acid peptide first described in a human phaeochromocytoma but since been found to be present in many tissues, including the vascular system and bone. Because of its structural similarity to amylin and calcitonin gene-related peptide, both of which have actions on bone cells, we have previously assessed the effects of adrenomedullin on the skeleton, and found that it increases osteoblast proliferation in vitro and bone formation following local injection in vivo. The present study carries this work forward by assessing the effects on bone of the systemic administration of a fragment of this peptide lacking the structural requirements for vasodilator activity. Two groups of 20 adult male mice received 20 injections of human adrenomedullin(27-52) 8.1 microg or vehicle over a 4-week period and bone histomorphometry and strength were assessed. In the tibia, adrenomedullin(27-52) produced increases in the indices of osteoblast activity, osteoid perimeter and osteoblast perimeter (P<0.05 for both using Student's t-test). Osteoclast perimeter was not affected. There was a 21% increase in cortical width and a 45% increase in trabecular bone volume in animals treated with adrenomedullin(27-52) (P<0.002 for both). Assessment of bone strength by three-point bending of the humerus showed both the maximal force and the displacement to the point of failure were increased in the animals treated with adrenomedullin(27-52) (P<0.03 for both). There was also a significant increase in the thickness of the epiphyseal growth plate. No adverse effects of the treatment were noted. It is concluded that adrenomedullin(27-52) acts as an anabolic agent on bone. These findings may be relevant to the normal regulation of bone mass and to the design of agents for the treatment of osteoporosis.


Assuntos
Osso e Ossos/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Adrenomedulina , Animais , Fenômenos Biomecânicos , Composição Corporal/efeitos dos fármacos , Osso e Ossos/anatomia & histologia , Osso e Ossos/fisiologia , Úmero/efeitos dos fármacos , Úmero/fisiologia , Masculino , Camundongos , Osteoblastos/fisiologia , Estatísticas não Paramétricas , Tíbia/anatomia & histologia , Tíbia/efeitos dos fármacos , Tíbia/fisiologia
9.
Gut ; 49(1): 23-8, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11413106

RESUMO

BACKGROUND AND AIMS: The main goal of our study was to characterise the activity of BIM26226 as a peripheral gastrin releasing peptide (GRP) receptor antagonist in healthy human subjects and to determine if endogenous GRP is a physiological regulator of gastric acid secretion and gastrin release. METHODS: Our study consisted of three parts. In part I, subjects received saline or BIM26226 followed by graded doses of intravenous human GRP in a four period crossover design. In part II, subjects received BIM26226 or saline during oral meal ingestion or modified sham feeding. In part III, subjects received an acidified meal in the presence and absence of BIM26226 in a two period crossover design. In addition, gastrin and somatostatin mRNA were measured in biopsy specimens during saline and BIM26226 infusion. RESULTS: BIM26226 dose dependently inhibited GRP induced acid output. Acid secretion after oral liquid meal intake and sham feeding was significantly inhibited by BIM26226 (p<0.01) whereas plasma gastrin release remained unchanged. Gastrin and somatostatin mRNAs were not significantly different after saline or BIM26226. CONCLUSIONS: BIM26226 is a potent GRP antagonist in humans. Endogenous GRP may be a physiological regulator of gastric acid secretion. Gastrin release does not seem to be under the control of GRP.


Assuntos
Bombesina/análogos & derivados , Bombesina/farmacologia , Ácido Gástrico/metabolismo , Peptídeo Liberador de Gastrina/fisiologia , Fragmentos de Peptídeos/farmacologia , Adulto , Análise de Variância , Northern Blotting , Estudos Cross-Over , Relação Dose-Resposta a Droga , Método Duplo-Cego , Ingestão de Alimentos/fisiologia , Determinação da Acidez Gástrica , Peptídeo Liberador de Gastrina/antagonistas & inibidores , Gastrinas/análise , Humanos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/análise , Somatostatina/análise , Estatísticas não Paramétricas
10.
J Surg Res ; 97(2): 131-7, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11341788

RESUMO

INTRODUCTION: Expression of somatostatin receptor subtype 2 (sst 2) in angiogenic tumor vessels appears to be homogeneous, while tumor cell expression of this receptor is often heterogeneous. We have developed a novel in vitro three-dimensional tumor angiogenesis model to study the antitumor and the antiangiogenic effects of radiolabeled somatostatin analogs. We hypothesized that targeted in situ radiation with an Auger electron-emitting radiolabeled somatostatin analog would produce receptor-specific cytotoxicity in sst 2-expressing cells. MATERIALS AND METHODS: IMR-32 human neuroblastoma (sst 2-positive) and MDA MB-231 human breast cancer (sst 2-negative) xenografts were created in nude mice from monolayer cell cultures. Fragments of these tumors were embedded in three-dimensional fibrin gels supplemented with endothelial growth media and incubated for a period of 14 days. Tumor fragments were treated with 50 microCi/ml of (111)In-JIC 2DL, a sst 2-preferring somatostatin analog, or medium on Day 1. Initial angiogenic activity was determined at 48 h and the mean angiogenic score and tumoricidal responses were assessed on Day 14. RESULTS AND CONCLUSION: Tumoricidal effects of (111)In-JIC 2DL were seen only in sst 2-positive IMR-32 tumors. However, the angiogenic response was inhibited in both IMR-32 and MDA MB-231 tumors independent of the tumor cells' sst 2 status. Somatostatin receptor-mediated in situ radiation therapy has profound cytotoxic effects on angiogenic blood vessels and sst 2-expressing tumor cells.


Assuntos
Meios de Contraste/farmacologia , Radioisótopos de Índio/farmacologia , Neovascularização Patológica/radioterapia , Ácido Pentético/farmacologia , Receptores de Somatostatina/metabolismo , Adenocarcinoma , Sequência de Aminoácidos , Animais , Neoplasias da Mama , Feminino , Humanos , Técnicas In Vitro , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Transplante de Neoplasias , Neuroblastoma , Octreotida/química , Octreotida/farmacologia , Ácido Pentético/análogos & derivados , Células Tumorais Cultivadas
11.
J Med Chem ; 44(8): 1305-11, 2001 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-11312929

RESUMO

The search for synthetic peptide analogues of somatostatin (SRIF) which exhibit selective affinities for the five known receptor subtypes (sst1-5) has generated a large number of potent agonists. Some of these agonists display good subtype selectivities and affinities for the subtypes 1, 2, 3, and 5, including analogues created by N-methyl amino acid substitutions in a standard octapeptide analogue format. We have now extended this peptide backbone N-methylation approach to a potent somatostatin receptor antagonist series using the antagonist Cpa-cyclo(DCys-Pal-DTrp-Lys-Thr-Cys)-Nal-NH2 9 reported from this laboratory as the lead structure. Synthetic analogues were tested for their ability to inhibit somatostatin-stimulated GH release from rat pituitary cells in culture and to displace 125I-labeled somatostatin from CHO cells transfected with the five known human somatostatin receptors. Several interesting observations resulted from the study. N-Methylation at the Lys(9) residue (5) increased the rat GH release inhibitory potency nearly 4-fold to 0.73 nM but resulted in little change in the binding affinity for human type 2 receptor. This analogue also had a high affinity of 5.98 nM for sst5 receptor (compared to 1.4 nM for somatostatin itself) and is the first antagonist analogue to be reported with high affinity for sst5. It also had high potency on in vitro inhibition of sst5 mediated intracellular calcium mobilization. These results were considered surprising, since the Lys(9) residue has long been considered to constitute the active center of somatostatin, important both for receptor binding and activation, and suggests important conformational differences between D-Cys(9) somatostatin antagonists and normal agonist structures. More modifications were carried out on this analogue with the aim of improving antagonist potency and/or specificity. Tyr(7) substitution of 5 resulted in an analogue, which had the highest affinity in the series for hsst2 (K(I) 5.51 nM) and an extraordinarily low IC50 of 0.53 nM in the rat pituitary cell assay. However, this analogue lost considerable affinity for sst5 relative to analogue 5. Analogue 16 with DTrp(12) at C-terminus had the highest affinity for hsst2, however, the IC50 in the rat GH release assay was only 11.6 nM. Replacement of Lys(9) in 9 with Dab(9) gave 11 which displayed high binding affinity for sst3, and it was also quite selective for that receptor. Both the sst3 and sst5 antagonists should be of value in assigning the physiological roles to type 3 and 5 receptor, respectively.


Assuntos
Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Somatostatina/síntese química , Animais , Ligação Competitiva , Células CHO , Cálcio/metabolismo , Células Cultivadas , Cricetinae , Hormônio do Crescimento/metabolismo , Humanos , Técnicas In Vitro , Líquido Intracelular/metabolismo , Masculino , Metilação , Modelos Moleculares , Adeno-Hipófise/citologia , Adeno-Hipófise/metabolismo , Ensaio Radioligante , Ratos , Receptores de Somatostatina/antagonistas & inibidores , Receptores de Somatostatina/genética , Somatostatina/química , Somatostatina/farmacologia , Transfecção
12.
J Med Chem ; 44(9): 1416-21, 2001 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-11311064

RESUMO

The search for synthetic analogues of somatostatin which exhibit selective affinities for the five receptor subtypes is of considerable basic and therapeutic interest and has generated a large number of potent agonist analogues with a wide spectrum of binding profiles. In the past, conformational restriction of side chain groups and the peptide backbone has yielded the most interesting results. Under the latter category and as part of the present study, we were interested in the potential effects of N-methylation of peptide bond NH groups on binding affinity since this approach had not been systematically examined with these peptides. This was aided by new chemistries for introducing an N-Me group during regular solid-phase peptide synthesis using Boc protection. A number of interesting effects were noted on relative binding affinities of the two series of agonist sequences chosen, DPhe(5)(or Tyr(5))-c[Cys(6)-Phe(7)-DTrp(8)-Lys(9)-Thr(10)-Cys(11)]Thr(12)-NH(2) (SRIF numbering), at the five known human somatostatin receptors transfected into and stably expressed by CHO cells. N-Methylation of residues 7 (Phe), 10 (Thr), 11 (Cys), and 12 (Thr) largely destroyed affinities for all five receptors. N-Methylation of DTrp in the DPhe series gave an analogue with extraordinarily high affinity for the type 5 receptor for which it was also quite selective. N-Methylation of Lys in both series resulted in retention of type 2 affinity despite this residue constituting the "active center" of somatostatin peptides. N-Methylation of either the N-terminal Tyr residue or of Cys(6) in the Tyr series resulted in analogues with extraordinarily high affinity for the type 3 receptor, also with a degree of specificity. N-Methylation of the peptide bond constrains the conformational space of the amino acid and eliminates the possibility of donor hydrogen bond formation from the amide linkage. The beta-bend conformation of the agonists around DTrp-Lys is stabilized by a transannular intramolecular hydrogen bond(s) between Phe(7) and Thr(10) so methylation of these residues eliminates this source of stabilization. It is expected that several of these analogues will provide additional tools for determining some of the physiological roles played by type 3 and 5 somatostatin receptors which are still far from being fully elucidated.


Assuntos
Fragmentos de Peptídeos/química , Receptores de Somatostatina/agonistas , Somatostatina/química , Animais , Células CHO , Cricetinae , Hormônio do Crescimento/metabolismo , Humanos , Técnicas In Vitro , Masculino , Metilação , Modelos Moleculares , Fragmentos de Peptídeos/metabolismo , Adeno-Hipófise/metabolismo , Ensaio Radioligante , Ratos , Receptores de Somatostatina/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Relação Estrutura-Atividade , Transfecção
13.
Eur J Pharmacol ; 412(1): 13-20, 2001 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-11166731

RESUMO

A synthetic peptide, (D-Phe(6), beta-Ala(11), Phe(13), Nle(14))bombesin-(6-14) was used to investigate the signal transduction mechanisms of bombesin receptor subtype-3. Using NCI-1299#5 human lung cancer cells stably transfected with bombesin receptor subtype-3, 100 nM (D-Phe(6), beta-Ala(11), Phe(13), Nle(14))bombesin-(6-14) elevated the cytosolic Ca2+ from 150 to 250 nM within 10 s. Addition of (D-Phe(6), beta-Ala(11), Phe(13), Nle(14))bombesin-(6-14) caused phosphorylation of mitogen activated protein kinase in a time- and concentration-dependent manner. The mitogen activated protein kinase phosphorylation caused by (D-Phe(6), beta-Ala(11), Phe(13), Nle(14))bombesin-(6-14) was inhibited by 2'-amino-3'-methyoxyflavone (PD98059), a mitogen activated protein kinase kinase (MEK-1) inhibitor. Using a luciferase reporter gene construct, (D-Phe(6), beta-Ala(11), Phe(13), Nle(14))bombesin-(6-14) caused Elk-1 activation after 10 min and the increase in Elk-1 activation caused by (D-Phe(6), beta-Ala(11), Phe(13), Nle(14))bombesin-(6-14) was inhibited by PD98059 as well as a dominant-negative MEK-1. (D-Phe(6), beta-Ala(11), Phe(13), Nle(14))bombesin-(6-14) caused increased c-fos as well as c-jun mRNAs 1 h after addition to NCI-H1299#5 cells. The 47-fold increase in c-fos mRNA caused by 100 nM (D-Phe(6), beta-Ala(11), Phe(13), Nle(14))bombesin-(6-14) was inhibited by PD98059, a dominant-negative MEK-1 and a substance P antagonist but not (3-phenylpropanoyl-D-Ala(24), Pro(26), Psi(26,27), Phe(27))GRP-(20-27) (BW2258U89), a GRP receptor antagonist. These results indicate that (D-Phe(6), beta-Ala(11), Phe(13), Nle(14))bombesin-(6-14) caused increased nuclear oncogene expression and upstream events include mitogen activated protein kinase phosphorylation and Elk-1 activation.


Assuntos
Bombesina/análogos & derivados , Proteínas de Ligação a DNA , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Genes fos/efeitos dos fármacos , Neoplasias Pulmonares/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Proto-Oncogênicas/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , Receptores da Bombesina/efeitos dos fármacos , Fatores de Transcrição , Bombesina/farmacologia , Cálcio/metabolismo , Genes fos/fisiologia , Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Oncogenes/efeitos dos fármacos , Oncogenes/fisiologia , Fragmentos de Peptídeos/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/metabolismo , Receptores da Bombesina/metabolismo , Células Tumorais Cultivadas , Proteínas Elk-1 do Domínio ets
14.
J Biol Chem ; 276(12): 9219-29, 2001 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-11112777

RESUMO

The orphan receptor, bombesin (Bn) receptor subtype 3 (BRS-3), shares high homology with bombesin receptors (neuromedin B receptor (NMB-R) and gastrin-releasing peptide receptor (GRP-R)). This receptor is widely distributed in the central nervous system and gastrointestinal tract; target disruption leads to obesity, diabetes, and hypertension, however, its role in physiological and pathological processes remain unknown due to lack of selective ligands or identification of its natural ligand. We have recently discovered (Mantey, S. A., Weber, H. C., Sainz, E., Akeson, M., Ryan, R. R. Pradhan, T. K., Searles, R. P., Spindel, E. R., Battey, J. F., Coy, D. H., and Jensen, R. T. (1997) J. Biol. Chem. 272, 26062-26071) that [d-Tyr(6),beta-Ala(11),Phe(13),Nle(14)]Bn-(6-14) has high affinity for BRS-3 and using this ligand showed BRS-3 has a unique pharmacology with high affinity for no known natural Bn peptides. However, use of this ligand is limited because it has high affinity for all known Bn receptors. In the present study we have attempted to identify BRS-3 selective ligands using a strategy of rational peptide design with the substitution of conformationally restricted amino acids into the prototype ligand [d-Tyr(6),beta-Ala(11),Phe(13),Nle(14)]Bn-(6-14) or its d-Phe(6) analogue. Each of the 22 peptides synthesized had binding affinities determined for hBRS-3, hGRPR, and hNMBR, and hBRS-3 selective ligands were tested for their ability to activate phospholipase C and increase inositol phosphates ([(3)H]inositol phosphate). Using this approach we have identified a number of BRS-3 selective ligands. These ligands functioned as receptor agonists and their binding affinities were reflected in their potencies for altering [(3)H]inositol phosphate. Two peptides with an (R)- or (S)-amino-3-phenylpropionic acid substitution for beta-Ala(11) in the prototype ligand had the highest selectivity for the hBRS-3 over the mammalian Bn receptors and did not interact with receptors for other gastrointestinal hormones/neurotransmitters. Molecular modeling demonstrated these two selective BRS-3 ligands had a unique conformation of the position 11 beta-amino acid. This selectivity was of sufficient magnitude that these should be useful in explaining the role of hBRS-3 activation in obesity, glucose homeostasis, hypertension, and other physiological or pathological processes.


Assuntos
Peptídeos/farmacologia , Receptores da Bombesina/agonistas , Células 3T3 , Sequência de Aminoácidos , Animais , Desenho de Fármacos , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Peptídeos/química , Peptídeos/metabolismo , Receptores da Bombesina/metabolismo
15.
J Biol Chem ; 276(1): 495-504, 2001 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-11013243

RESUMO

Peptoid antagonists are increasingly being described for G protein-coupled receptors; however, little is known about the molecular basis of their binding. Recently, the peptoid PD168368 was found to be a potent selective neuromedin B receptor (NMBR) antagonist. To investigate the molecular basis for its selectivity for the NMBR over the closely related receptor for gastrin-releasing peptide (GRPR), we used a chimeric receptor approach and a site-directed mutagenesis approach. Mutated receptors were transiently expressed in Balb 3T3. The extracellular domains of the NMBR were not important for the selectivity of PD168368. However, substitution of the 5th upper transmembrane domain (uTM5) of the NMBR by the comparable GRPR domains decreased the affinity 16-fold. When the reverse study was performed by substituting the uTM5 of NMBR into the GRPR, a 9-fold increase in affinity occurred. Each of the 4 amino acids that differed between NMBR and GRPR in the uTM5 region were exchanged, but only the substitution of Phe(220) for Tyr in the NMBR caused a decrease in affinity. When the reverse study was performed to attempt to demonstrate a gain of affinity in the GRPR, the substitution of Tyr(219) for Phe caused an increase in affinity. These results suggest that the hydroxyl group of Tyr(220) in uTM5 of NMBR plays a critical role for high selectivity of PD168368 for NMBR over GRPR. Receptor and ligand modeling suggests that the hydroxyl of the Tyr(220) interacts with nitrophenyl group of PD168368 likely primarily by hydrogen bonding. This result shows the selectivity of the peptoid PD168368, similar to that reported for numerous non-peptide analogues with other G protein-coupled receptors, is primarily dependent on interaction with transmembrane amino acids.


Assuntos
Indóis/farmacologia , Peptídeos/farmacologia , Piridinas/farmacologia , Receptores da Bombesina/antagonistas & inibidores , Receptores da Bombesina/metabolismo , Tirosina/metabolismo , Células 3T3 , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Animais , Sítios de Ligação , Indóis/química , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida/genética , Neurocinina B/análogos & derivados , Neurocinina B/antagonistas & inibidores , Neurocinina B/química , Neurocinina B/metabolismo , Peptídeos/química , Peptoides , Mutação Puntual/genética , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Piridinas/química , Receptores da Bombesina/química , Receptores da Bombesina/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Especificidade por Substrato , Transfecção , Tirosina/genética
16.
J Cardiovasc Pharmacol ; 36(4): 526-32, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11026656

RESUMO

Biologic actions attributed to adrenomedullin include reduction of arterial pressure and suppression of aldosterone secretion. To assess possible in vivo antiangiotensin II actions of adrenomedullin, we examined hemodynamic and adrenal responses to stepped angiotensin II infusions with or without co-infusions of adrenomedullin (33 ng/kg/min) in conscious sheep under controlled conditions of a low sodium intake. Plasma adrenomedullin levels rose during peptide infusions (p < 0.001) to plateau at approximately 15-18 pM. The dose-dependent pressor response (15-20 mm Hg) of angiotensin II was both delayed and markedly attenuated (p = 0.017) by adrenomedullin, which also stimulated heart rate (p < 0.001) and cardiac output (p < 0.001). Adrenomedullin prevented the angiotensin II-induced increase in peripheral resistance (p < 0.001). Plasma aldosterone responses to angiotensin II were variable and were not significantly altered by concomitant adrenomedullin infusion. In conclusion, low-dose infusion of adrenomedullin administered to conscious sheep on a low-salt diet clearly antagonized the vasopressor actions of administered angiotensin II while stimulating cardiac output and heart rate. The data suggest a possible role for adrenomedullin in cardiovascular homeostasis in part through antagonism of the vasopressor action of angiotensin II.


Assuntos
Angiotensina II/antagonistas & inibidores , Pressão Sanguínea/efeitos dos fármacos , Peptídeos/farmacologia , Vasoconstritores/antagonistas & inibidores , Adrenomedulina , Aldosterona/sangue , Angiotensina II/sangue , Angiotensina II/farmacologia , Animais , Débito Cardíaco/efeitos dos fármacos , Dieta Hipossódica , Feminino , Frequência Cardíaca/efeitos dos fármacos , Humanos , Ovinos , Resistência Vascular/efeitos dos fármacos , Vasoconstritores/sangue , Vasoconstritores/farmacologia
17.
Am J Physiol Endocrinol Metab ; 279(4): E730-5, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11001752

RESUMO

Amylin increases bone mass when administered systemically to mice. However, because of its size, the full peptide is not an ideal candidate for the therapy of osteoporosis. The fragment, amylin-(1---8), stimulates osteoblast proliferation in vitro, although it is without effect on carbohydrate metabolism. The present study assessed the effects of daily administration of this peptide on sexually mature male mice for 4 wk. Amylin-(1---8) almost doubled histomorphometric indices of osteoblast activity but did not change measures of bone resorption. Trabecular bone volume increased by 36% as a result of increases in both trabecular number and trabecular thickness, and tibial cortical width increased by 8%. On three-point bending tests of bone strength, displacement to fracture was increased by amylin-(1---8), from 0.302 +/- 0.013 to 0.351 +/- 0. 017 mm (P = 0.02). In a separate experiment using dynamic histomorphometry with bone-seeking fluorochrome labels, amylin-(1---8) was administered by local injection over the calvariae of female mice. Amylin-(1---8) (40 nM) increased the double-labeled surface threefold. The effect was dose dependent from 0.4 to 40 nM and was greater than that of an equimolar dose of human parathyroid hormone-(1---34) [hPTH-(1---34)]. Mineral apposition rate was increased by 40 nM amylin-(1---8) but not by hPTH-(1---34). Amylin-(1---8) thus has significant anabolic effects in vivo, suggesting that this peptide or analogs of it should be further evaluated as potential therapies for osteoporosis.


Assuntos
Amiloide/administração & dosagem , Densidade Óssea/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Amiloide/química , Animais , Peso Corporal/efeitos dos fármacos , Osso e Ossos/anatomia & histologia , Osso e Ossos/metabolismo , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Corantes Fluorescentes , Humanos , Injeções Subcutâneas , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Masculino , Camundongos , Osteoblastos/efeitos dos fármacos , Fragmentos de Peptídeos/química , Ratos , Crânio/efeitos dos fármacos , Teriparatida/farmacologia , Tíbia/efeitos dos fármacos
18.
Nitric Oxide ; 4(2): 94-102, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10835289

RESUMO

The purpose of this study was to investigate the in vivo effects of intracavernosal injections of galanin and galantide (a specific galanin receptor antagonist) on penile erection in the anesthetized cat. Erectile responses to galanin and galantide were compared with responses to a standard triple drug combination [1.65 mg papaverine, 25 microg phentolamine, and 0.5 microg prostaglandin E(1) (PGE(1))]. Intracavernosal injections of galanin (3-100 nmol) and galantide (0. 1-3 nmol) induced penile erection in a dose-dependent manner. In terms of relative potency, galantide was approximately 100-fold more potent than galanin at increasing cavernosal pressure. The maximal increases in intracavernosal pressure in response to galanin and galantide were 83 and 95%, respectively, of the control triple drug combination. The total durations of erectile response caused by these peptides were significantly shorter (P<0.05) than those by the triple drug combination. The nitric oxide synthase inhibitor L-NAME (20 mg) significantly decreased the erectile response in the cat to galantide but not to galanin, while the K(+)(ATP) channel antagonist U-37883A (3 mg) had no effect on the erectile response to galanin nor galantide. The results of the present study demonstrate that galantide, a putative antagonist for the galanin receptor, has more potent agonist activity than galanin in increasing intracavernosal pressure in the cat. Moreover, these data suggest that galantide, but not galanin, causes penile erection by an NO/cGMP-dependent mechanism. This is the first study to demonstrate that galanin may play a role in the physiology of penile erection.


Assuntos
Galanina/análogos & derivados , Galanina/farmacologia , Óxido Nítrico/fisiologia , Ereção Peniana/efeitos dos fármacos , Substância P/análogos & derivados , Animais , Gatos , Masculino , Músculo Liso/efeitos dos fármacos , Músculo Liso/metabolismo , Óxido Nítrico/metabolismo , Ereção Peniana/fisiologia , Canais de Potássio/metabolismo , Substância P/farmacologia
19.
Am J Physiol Gastrointest Liver Physiol ; 278(1): G64-74, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10644563

RESUMO

Pancreatic acini from most species possess vasoactive intestinal peptide (VIP) receptors. Recently, two subtypes of VIP receptors, VIP(1)-R and VIP(2)-R, were cloned. Which subtype exists on pancreatic acini or mediates secretion is unclear. To address this, we examined pancreatic acini from both rat and guinea pig. VIP(1)-R and VIP(2)-R mRNA were identified in dispersed acini from both species by Northern blot analysis and in rat by Southern blot analysis. With the use of the VIP(2)-R-selective ligand Ro-25-1553 in both species, inhibition of binding of (125)I-labeled VIP to acini showed a biphasic pattern with a high-affinity component (10%) and a second representing 90%. The VIP(1)-R-selective ligand, [Lys(15),Arg(16),Leu(27)]VIP-(1-7)-GRF-(8-27), gave a monophasic pattern. Binding of Ro-25-1553 was better fit by a two-site model. In both rat and guinea pig acini, the dose-response curve of Ro-25-1553 for stimulation of enzyme secretion was biphasic, with a high-affinity component of 10-15% of the maximal secretion and a low-affinity component accounting for 85-90%. At low concentrations (10 nM) of Ro-25-1553 and [Lys(15),Arg(16), Leu(27)]VIP-(1-7)-GRF(8-27), which only occupy VIP receptors, a 4-fold and a 56-fold increase in cAMP occurred, respectively. These results show that both VIP(1)-R and VIP(2)-R subtypes exist on pancreatic acini of rat and guinea pig, their activation stimulates enzyme secretion by a cAMP-mediated mechanism, and the effects of VIP are mediated 90% by activation of VIP(1)-R and 10% by VIP(2)-R. Because VIP has a high affinity for both VIP-R subtypes, its effect on pancreatic acini is mediated by two receptor subtypes, which will need to be considered in future studies of the action of VIP in the pancreas.


Assuntos
Pâncreas/metabolismo , Receptores de Peptídeo Intestinal Vasoativo/metabolismo , Animais , AMP Cíclico/metabolismo , Enzimas/metabolismo , Cobaias , Ligantes , Masculino , Pâncreas/enzimologia , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Peptídeos Cíclicos/metabolismo , Peptídeos Cíclicos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Tipo II de Peptídeo Intestinal Vasoativo , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo , Peptídeo Intestinal Vasoativo/análogos & derivados , Peptídeo Intestinal Vasoativo/metabolismo , Peptídeo Intestinal Vasoativo/farmacologia
20.
Life Sci ; 65(10): PL115-20, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10499878

RESUMO

Urethane increases the release of somatostatin (SRIF) which inhibits gastric acid secretion. The SRIF monoclonal antibody, CURE.S6 and the novel sst2 antagonist, PRL-2903 injected intravenously at maximal effective doses increased gastric acid secretion by 2 and 10 fold respectively from basal values within 30 min in urethane-anesthetized rats. Plasma gastrin levels were elevated 2.5 fold within 15 min by PRL-2903 (1.3 micromol/kg, i.v.). These data indicate that the low gastrin and acid secretion levels induced by urethane result from endogenous SRIF acting on sst2 and that PRL-2903 is a valuable SRIF antagonist to assess sst2 mediated events.


Assuntos
Anestésicos Intravenosos/administração & dosagem , Ácido Gástrico/metabolismo , Mucosa Gástrica/efeitos dos fármacos , Peptídeos Cíclicos/farmacologia , Receptores de Somatostatina/antagonistas & inibidores , Somatostatina/metabolismo , Uretana/administração & dosagem , Animais , Anticorpos Monoclonais , Mucosa Gástrica/metabolismo , Gastrinas/sangue , Masculino , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA