Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Endocr Soc ; 2(7): 779-793, 2018 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-30151433

RESUMO

Acromegaly is a neuroendocrine disorder caused by excess secretion of GH by somatotroph tumor cells. It is often treated with somatostatin receptor (SSTR) 2 agonists, which suppress GH secretion. SOM230 is a somatostatin analogue that targets multiple SSTRs and was recently approved for patients with treatment-resistant acromegaly. Previous reports indicate that SOM230 may function as a biased agonist, suggesting that its ability to selectively activate SSTR-dependent signaling events may contribute to its therapeutic efficacy. To better understand how SOM230 modulates Sstr2A function, which is the most commonly expressed SSTR in somatotrophs, we used real-time assays to study SOM230-dependent signaling in rat pituitary tumor cells. We observed that SOM230 suppressed cAMP production in a Gαi-dependent manner, similar to conventional Sstr2A agonists. However, it did not cause receptor internalization as would be expected for an Sstr2A agonist. Surprisingly, SOM230 did not cause membrane hyperpolarization, which is an important mechanism by which Sstr2a activation suppresses intracellular calcium (Ca2+) accumulation and GH secretion. In fact, SOM230 inhibited the ability of conventional somatostatin analogues to control membrane potential. However, SOM230 still inhibited intracellular Ca2+ accumulation in a novel, Gßγ-dependent manner. These studies show that SOM230 exhibits strong agonist bias in regulating signaling pathways downstream of Sstr2A that control GH secretion.

2.
ACS Med Chem Lett ; 8(7): 720-725, 2017 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-28740605

RESUMO

Fluorescently labeled imaging agents can identify surgical margins in real-time to help achieve complete resections and minimize the likelihood of local recurrence. However, photon attenuation limits fluorescence-based imaging to superficial lesions or lesions that are a few millimeters beneath the tissue surface. Contrast agents that are dual-labeled with a radionuclide and fluorescent dye can overcome this limitation and combine quantitative, whole-body nuclear imaging with intraoperative fluorescence imaging. Using a multimodality chelation (MMC) scaffold, IRDye 800CW was conjugated to the clinically used somatostatin analog, 68Ga-DOTA-TOC, to produce the dual-labeled analog, 68Ga-MMC(IRDye 800CW)-TOC, with high yield and specific activity. In vitro pharmacological assays demonstrated retention of receptor-targeting properties for the dual-labeled compound with robust internalization that was somatostatin receptor (SSTR) 2-mediated. Biodistribution studies in mice identified the kidneys as the primary excretion route for 68Ga-MMC(IRDye 800CW)-TOC, along with clearance via the reticuloendothelial system. Higher uptake was observed in most tissues compared to 68Ga-DOTA-TOC but decreased as a function of time. The combination of excellent specificity for SSTR2-expressing cells and suitable biodistribution indicate potential application of 68Ga-MMC(IRDye 800CW)-TOC for intraoperative detection of SSTR2-expressing tumors.

3.
J Nucl Med ; 58(11): 1858-1864, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28572490

RESUMO

Fluorescence-guided surgery is an emerging imaging technique that can enhance the ability of surgeons to detect tumors when compared with visual observation. To facilitate characterization, fluorescently labeled probes have been dual-labeled with a radionuclide to enable cross-validation with nuclear imaging. In this study, we selected the somatostatin receptor imaging agent DOTATOC as the foundation for developing a dual-labeled analog. We hypothesized that a customized dual-labeling approach with a multimodality chelation (MMC) scaffold would minimize steric effects of dye conjugation and retain agonist properties. Methods: An MMC conjugate (MMC-TOC) was synthesized on solid-phase and compared with an analog prepared using conventional methods (DA-TOC). Both analogs were conjugated to IRDye 800 using copper-free click chemistry. The resulting compounds, MMC(IR800)-TOC and DA(IR800)-TOC, were labeled with Cu and 64Cu and tested in vitro in somatostatin receptor subtype 2-overexpressing HEK-293 cells to assess agonist properties, and in AR42J rat pancreatic cancer cells to determine receptor binding characteristics. Multimodality imaging was performed in AR42J xenografts. Results: Cu-MMC(IR800)-TOC demonstrated higher potency for cyclic adenosine monophosphate inhibition (half maximal effective concentration [EC50]: 0.21 ± 0.18 vs. 1.38 ± 0.54 nM) and receptor internalization (EC50: 41.9 ± 29.8 vs. 455 ± 299 nM) than Cu-DA(IR800)-TOC. Radioactive uptake studies showed that blocking with octreotide caused a dose-dependent reduction in 64Cu-MMC(IR800)-TOC uptake whereas 64Cu-DA(IR800)-TOC was not affected. In vivo studies revealed higher tumor uptake for 64Cu-MMC(IR800)-TOC than 64Cu-DA(IR800)-TOC (5.2 ± 0.2 vs. 3.6 ± 0.4 percentage injected dose per gram). In vivo blocking studies with octreotide reduced tumor uptake of 64Cu-MMC(IR800)-TOC by 66%. Excretion of 64Cu-MMC(IR800)-TOC was primarily through the liver and spleen whereas 64Cu-DA(IR800)-TOC was cleared through the kidneys. Ex vivo analysis at 24 h confirmed PET/CT data by showing near-infrared fluorescence signal in tumors and a tumor-to-muscle ratio of 5.3 ± 0.8 as determined by γ-counting. Conclusion: The findings demonstrate that drug design affected receptor pharmacology and suggest that the MMC scaffold is a useful tool for the development of dual-labeled imaging agents.


Assuntos
Quelantes/química , Compostos Radiofarmacêuticos/síntese química , Receptores de Somatostatina/agonistas , Animais , Quelantes/farmacologia , Radioisótopos de Cobre , Relação Dose-Resposta a Droga , Desenho de Fármacos , Feminino , Células HEK293 , Humanos , Marcação por Isótopo , Camundongos , Camundongos Nus , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/metabolismo , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/metabolismo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética , Receptores de Somatostatina/química , Distribuição Tecidual
6.
Endocr Relat Cancer ; 21(5): 691-704, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25012983

RESUMO

Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) raise difficult therapeutic problems despite the emergence of targeted therapies. Somatostatin analogs (SSA) remain pivotal therapeutic drugs. However, the tachyphylaxis and the limited antitumoral effects observed with the classical somatostatin 2 (sst2) agonists (octreotide and lanreotide) led to the development of new SSA, such as the pan sst receptor agonist pasireotide. Our aim was to compare the effects of pasireotide and octreotide on cell survival, chromogranin A (CgA) secretion, and sst2 phosphorylation/trafficking in pancreatic NET (pNET) primary cells from 15 tumors. We established and characterized the primary cultures of human pancreatic tumors (pNETs) as powerful preclinical models for understanding the biological effects of SSA. At clinically relevant concentrations (1-10 nM), pasireotide was at least as efficient as octreotide in inhibiting CgA secretion and cell viability through caspase-dependent apoptosis during short treatments, irrespective of the expression levels of the different sst receptors or the WHO grade of the parental tumor. Interestingly, unlike octreotide, which induces a rapid and persistent partial internalization of sst2 associated with its phosphorylation on Ser341/343, pasireotide did not phosphorylate sst2 and induced a rapid and transient internalization of the receptor followed by a persistent recycling at the cell surface. These results provide the first evidence, to our knowledge, of striking differences in the dynamics of sst2 trafficking in pNET cells treated with the two SSAs, but with similar efficiency in the control of CgA secretion and cell viability.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias Intestinais/metabolismo , Tumores Neuroendócrinos/metabolismo , Octreotida/farmacologia , Neoplasias Pancreáticas/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Neoplasias Gástricas/metabolismo , Adulto , Idoso , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 7/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cromogranina A/metabolismo , AMP Cíclico/metabolismo , Fragmentação do DNA , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Somatostatina/farmacologia , Células Tumorais Cultivadas
7.
Trends Pharmacol Sci ; 34(12): 676-88, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24183675

RESUMO

Somatostatin analogs for the diagnosis and therapy of neuroendocrine tumors (NETs) have been used in clinical applications for more than two decades. Five somatostatin receptor subtypes have been identified and molecular mechanisms of somatostatin receptor signaling and regulation have been elucidated. These advances increased understanding of the biological role of each somatostatin receptor subtype, their distribution in NETs, as well as agonist-specific regulation of receptor signaling, internalization, and phosphorylation, particularly for the sst2 receptor subtype, which is the primary target of current somatostatin analog therapy for NETs. Various hypotheses exist to explain differences in patient responsiveness to somatostatin analog inhibition of tumor secretion and growth as well as differences in the development of tumor resistance to therapy. In addition, we now have a better understanding of the action of both first generation (octreotide, lanreotide, Octreoscan) and second generation (pasireotide) FDA-approved somatostatin analogs, including the biased agonistic character of some agonists. The increased understanding of somatostatin receptor pharmacology provides new opportunities to design more sophisticated assays to aid the future development of somatostatin analogs with increased efficacy.


Assuntos
Tumores Neuroendócrinos/tratamento farmacológico , Tumores Neuroendócrinos/metabolismo , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Animais , Humanos , Fosforilação , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/metabolismo , Transdução de Sinais , Somatostatina/uso terapêutico
8.
Am J Pathol ; 180(5): 1942-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22538189

RESUMO

Somatostatin analogues, which are used to treat neuroendocrine tumors, target the high levels of somatostatin receptor subtype 2 (SSTR1; alias sst2) expressed in these cancers. However, some tumors are resistant to somatostatin analogues, and it is unknown whether the defect lies in sst2 activation or downstream signaling events. Because sst2 phosphorylation occurs rapidly after receptor activation, we examined whether sst2 is phosphorylated in neuroendocrine tumors. The sst2 receptor phosphorylation was evaluated by IHC and Western blot analysis with the new Ra-1124 antibody specific for the sst2 receptor phosphorylated at Ser341/343 in receptor-positive neuroendocrine tumors obtained from 10 octreotide-treated and 7 octreotide-naïve patients. The specificity, time course, and subcellular localization of sst2 receptor phosphorylation were examined in human embryo kinase-sst2 cell cultures by immunofluorescence and confocal microscopy. All seven octreotide-naïve tumors displayed exclusively nonphosphorylated cell surface sst2 expression. In contrast, 9 of the 10 octreotide-treated tumors contained phosphorylated sst2 that was predominantly internalized. Western blot analysis confirmed the IHC data. Octreotide treatment of human embryo kinase-sst2 cells in culture demonstrated that phosphorylated sst2 was localized at the plasma membrane after 10 seconds of stimulation and was subsequently internalized into endocytic vesicles. These data show, for the first time to our knowledge, that phosphorylated sst2 is present in most gastrointestinal neuroendocrine tumors from patients treated with octreotide but that a striking variability exists in the subcellular distribution of phosphorylated receptors among such tumors.


Assuntos
Antineoplásicos Hormonais/farmacologia , Carcinoma Neuroendócrino/metabolismo , Octreotida/farmacologia , Receptores de Somatostatina/metabolismo , Antineoplásicos Hormonais/uso terapêutico , Carcinoma Neuroendócrino/tratamento farmacológico , Carcinoma Neuroendócrino/cirurgia , Quimioterapia Adjuvante , Humanos , Microscopia Confocal , Microscopia de Fluorescência , Proteínas de Neoplasias/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Octreotida/uso terapêutico , Fosforilação/efeitos dos fármacos , Receptores de Somatostatina/efeitos dos fármacos
9.
Am J Surg Pathol ; 36(2): 242-52, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22251942

RESUMO

High overexpression of somatostatin receptors in neuroendocrine tumors allows imaging and radiotherapy with radiolabeled somatostatin analogues. To ascertain whether a tumor is suitable for in vivo somatostatin receptor targeting, its somatostatin receptor expression has to be determined. There are specific indications for use of immunohistochemistry for the somatostatin receptor subtype 2A, but this has up to now been limited by the lack of an adequate reliable antibody. The aim of this study was to correlate immunohistochemistry using the new monoclonal anti-somatostatin receptor subtype 2A antibody UMB-1 with the gold standard in vitro method quantifying somatostatin receptor levels in tumor tissues. A UMB-1 immunohistochemistry protocol was developed, and tumoral UMB-1 staining levels were compared with somatostatin receptor binding site levels quantified with in vitro I-[Tyr]-octreotide autoradiography in 89 tumors. This allowed defining an immunohistochemical staining threshold permitting to distinguish tumors with somatostatin receptor levels high enough for clinical applications from those with low receptor expression. The presence of >10% positive tumor cells correctly predicted high receptor levels in 95% of cases. In contrast, absence of UMB-1 staining truly reflected low or undetectable somatostatin receptor expression in 96% of tumors. If 1% to 10% of tumor cells were stained, a weak staining intensity was suggestive of low somatostatin receptor levels. This study allows for the first time a reliable recommendation for eligibility of an individual patient for in vivo somatostatin receptor targeting based on somatostatin receptor immunohistochemistry. Under optimal methodological conditions, UMB-1 immunohistochemistry may be equivalent to in vitro receptor autoradiography.


Assuntos
Neoplasias/química , Receptores de Somatostatina/análise , Anticorpos Monoclonais , Autorradiografia , Humanos , Imuno-Histoquímica , Neoplasias/patologia , Receptores de Somatostatina/imunologia , Estudos Retrospectivos
10.
Mol Endocrinol ; 25(6): 1040-54, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21493671

RESUMO

The somatostatin receptor subtype 2A (sst2A) mediates many of somatostatin's neuroendocrine actions and is the primary therapeutic target for the stable somatostatin analogs used to inhibit hormone secretion by pituitary and gastroenteropancreatic tumors. Two new multireceptor targeting somatostatin analogs currently under clinical investigation, the multisomatostatin receptor agonist cyclo-[diaminoethylcarbamoyl-HydroxyPro-Phenylglycine-D-Trp-Lys-(4-O-benzyl)Tyr-Phe] (SOM230) (Pasireotide) and pan-somatostatin receptor agonist Tyr-cyclo-[D-diaminobutyric acid-Arg-Phe-Phe-D-Trp-Lys-Thr-Phe] (KE108), behave as functionally selective ligands at the sst2A receptor, mimicking some of somatostatin's actions but antagonizing others. Further, SOM230 and KE108 are less able to induce receptor internalization than somatostatin, indicating that they exhibit functional selectivity for receptor regulation as well as signaling. Here, we identify agonist-specific differences in the molecular events regulating sst2A receptor endocytosis. SOM230 and KE108 were less potent and less effective than somatostatin at stimulating sst2A receptor phosphorylation at two pairs of residues, Ser341/343 and Thr353/354. Only the pattern of Thr353/354 phosphorylation correlated with receptor internalization, consistent with the known importance of Thr phosphorylation for sst2A receptor endocytosis. As expected, arrestin recruitment to membrane receptors was reduced with SOM230 and KE108. In addition, both receptor dephosphorylation and receptor recycling occurred more rapidly with SOM230 and KE108 than with somatostatin. Surprisingly, however, SOM230 and KE108 also altered sst2A internalization in a phosphorylation-independent manner, because these analogs were less effective than somatostatin at stimulating the endocytosis of a phosphorylation-negative receptor mutant. These results show that the decreased receptor internalization produced by SOM230 and KE108 compared with somatostatin result from phosphorylation-independent effects as well as reduced site-specific receptor phosphorylation and receptor-arrestin association.


Assuntos
Peptídeos Cíclicos/farmacologia , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Animais , Arrestinas/metabolismo , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Endocitose/efeitos dos fármacos , Pâncreas Exócrino/citologia , Pâncreas Exócrino/efeitos dos fármacos , Pâncreas Exócrino/metabolismo , Fosforilação , Transporte Proteico , Ratos , Receptores de Somatostatina/agonistas , Somatostatina/farmacologia , beta-Arrestinas
11.
J Biol Chem ; 286(15): 13561-73, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21343287

RESUMO

The G(i)-coupled somatostatin 2A receptor (sst2A) mediates many of the neuromodulatory and neuroendocrine actions of somatostatin (SS) and is targeted by the SS analogs used to treat neuroendocrine tumors. As for other G protein-coupled receptors, agonists stimulate sst2A receptor phosphorylation on multiple residues, and phosphorylation at different sites has distinct effects on receptor internalization and uncoupling. To elucidate the spatial and temporal regulation of sst2A receptor phosphorylation, we examined agonist-stimulated phosphorylation of multiple receptor GPCR kinase sites using phospho-site-specific antibodies. SS increased receptor phosphorylation sequentially, first on Ser-341/343 and then on Thr-353/354, followed by receptor internalization. Reversal of receptor phosphorylation was determined by the duration of prior agonist exposure. In acutely stimulated cells, in which most receptors remained on the cell surface, dephosphorylation occurred only on Thr-353/354. In contrast, both Ser-341/343 and Thr-353/354 were rapidly dephosphorylated when cells were stimulated long enough to allow receptor internalization before agonist removal. Consistent with these observations, dephosphorylation of Thr-353/354 was not affected by either hypertonic sucrose or dynasore, which prevent receptor internalization, whereas dephosphorylation of Ser-341/343 was completely blocked. An okadaic acid- and fostriecin-sensitive phosphatase catalyzed the dephosphorylation of Thr-353/354 both intracellularly and at the cell surface. In contrast, dephosphorylation of Ser-341/343 was insensitive to these inhibitors. Our results show that the phosphorylation and dephosphorylation of neighboring GPCR kinase sites in the sst2A receptor are subject to differential spatial and temporal regulation. Thus, the pattern of receptor phosphorylation is determined by the duration of agonist stimulation and compartment-specific enzymatic activity.


Assuntos
Quinases de Receptores Acoplados a Proteína G/metabolismo , Receptores de Somatostatina/metabolismo , Substituição de Aminoácidos , Animais , Células CHO , Cricetinae , Cricetulus , Inibidores Enzimáticos/farmacologia , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/genética , Humanos , Mutação de Sentido Incorreto , Ácido Okadáico/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Polienos/farmacologia , Pironas/farmacologia , Receptores de Somatostatina/genética
12.
Mol Endocrinol ; 24(1): 240-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19910453

RESUMO

Somatostatin analogs that activate the somatostatin subtype 2A (sst2A) receptor are used to treat neuroendocrine cancers because they inhibit tumor secretion and growth. Recently, new analogs capable of activating multiple somatostatin receptor subtypes have been developed to increase tumor responsiveness. We tested two such multi-somatostatin analogs for functional selectivity at the sst2A receptor: SOM230, which activates sst1, sst2, sst3, and sst5 receptors, and KE108, which activates all sst receptor subtypes. Both compounds are reported to act as full agonists at their target sst receptors. In sst2A-expressing HEK293 cells, somatostatin inhibited cAMP production, stimulated intracellular calcium accumulation, and increased ERK phosphorylation. SOM230 and KE108 were also potent inhibitors of cAMP accumulation, as expected. However, they antagonized somatostatin stimulation of intracellular calcium and behaved as partial agonists/antagonists for ERK phosphorylation. In pancreatic AR42J cells, which express sst2A receptors endogenously, SOM230 and KE108 were both full agonists for cAMP inhibition. However, although somatostatin increased intracellular calcium and ERK phosphorylation, SOM230 and KE108 again antagonized these effects. Distinct mechanisms were involved in sst2A receptor signaling in AR42J cells; pertussis toxin pretreatment blocked somatostatin inhibition of cAMP accumulation but not the stimulation of intracellular calcium and ERK phosphorylation. Our results demonstrate that SOM230 and KE108 behave as agonists for inhibition of adenylyl cyclase but antagonize somatostatin's actions on intracellular calcium and ERK phosphorylation. Thus, SOM230 and KE108 are not somatostatin mimics, and their functional selectivity at sst2A receptors must be considered in clinical applications where it may have important consequences for therapy.


Assuntos
Inibidores Enzimáticos/farmacologia , Antagonistas de Hormônios/farmacologia , Peptídeos Cíclicos/farmacologia , Receptores de Somatostatina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Somatostatina/análogos & derivados , Inibidores de Adenilil Ciclases , Adenilil Ciclases/metabolismo , Animais , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas de Ligação ao GTP/antagonistas & inibidores , Proteínas de Ligação ao GTP/metabolismo , Humanos , Fosforilação/efeitos dos fármacos , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/metabolismo , Ratos , Receptores de Somatostatina/genética , Somatostatina/agonistas , Somatostatina/antagonistas & inibidores , Somatostatina/farmacologia , Células Secretoras de Somatostatina/efeitos dos fármacos , Células Secretoras de Somatostatina/metabolismo , Fatores de Tempo
13.
Mol Pharmacol ; 76(1): 68-80, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19389921

RESUMO

Somatostatin receptor subtype 2A (sst2A) mediates many of the endocrine and neuronal actions of somatostatin and is the target of somatostatin analogs in cancer therapy. As with many G-protein-coupled receptors, agonist stimulation causes sst2A receptor desensitization and internalization, events that require receptor phosphorylation. Furthermore, heterologous receptor activation of protein kinase C (PKC) also increases sst2A receptor phosphorylation and internalization. Here we analyzed a series of sst2A receptor mutants biochemically to identify residues in the receptor carboxyl terminus that were phosphorylated upon agonist stimulation, and we then generated four phosphorylation-sensitive antibodies to those residues. Once the selectivity of each antibody for its phosphorylated and nonphosphorylated target sequence was determined, the phospho-site-specific antibodies were used to demonstrate that somatostatin treatment of Chinese hamster ovary (CHO) cells expressing the wild type sst2A receptor increased phosphorylation on five residues in the receptor C terminus: Ser341, Ser343, Ser348, Thr353, and Thr354. Phorbol 12-myristate 13-acetate (PMA) increased receptor phosphorylation only on Ser343. Inhibition of PKC blocked PMA but not somatostatin stimulation, showing that different kinases catalyzed Ser343 phosphorylation. In contrast, somatostatin-stimulated sst2A receptor phosphorylation was inhibited by knockdown of G-protein coupled receptor kinase-2 with siRNA. Somatostatin increased sst2A receptor phosphorylation on the same five residues in GH4C1 pituitary cells as in CHO cells. However, PMA stimulated sst2A receptor phosphorylation on both Ser343 and Ser348 in GH4C1 cells. These results characterize the complex pattern of sst2A receptor phosphorylation by agonist and second messenger-activated kinases for the first time and indicate that cell type-specific regulation of sst2A receptor phosphorylation occurs.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Proteína Quinase C/fisiologia , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/metabolismo , Animais , Células CHO , Colforsina/farmacologia , Cricetinae , Cricetulus , Ensaio de Imunoadsorção Enzimática , Quinase 2 de Receptor Acoplado a Proteína G/fisiologia , Humanos , Fosforilação , Receptores de Somatostatina/química , Relação Estrutura-Atividade , Acetato de Tetradecanoilforbol/farmacologia
14.
Mol Cell Endocrinol ; 286(1-2): 35-9, 2008 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-18006219

RESUMO

The five somatostatin receptor subtypes, named sst1-sst5, activate both distinct and common signaling pathways and exhibit different patterns of receptor regulation. Until recently it was believed that once a particular somatostatin receptor was activated by an agonist, all the down-stream signaling and regulatory effects characteristic of that receptor subtype in that cellular environment would be triggered. Thus, differences in the actions of somatostatin analogs between tissues were attributed to variability in the nature and concentration of the sst receptor subtypes and effectors expressed in different targets. However, agonists have recently been shown to exhibit functional selectivity at individual sst receptors such that they can elicit a subset of that receptor's potential effects, a property known as biased agonism. This review will summarize the evidence for functionally selective somatostatin receptor agonists and discuss the implications and promise of these new findings.


Assuntos
Receptores de Somatostatina/agonistas , Receptores de Somatostatina/fisiologia , Somatostatina/metabolismo , Animais , Humanos , Transdução de Sinais
15.
J Nucl Med ; 47(3): 502-11, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16513620

RESUMO

UNLABELLED: The uptake of radiolabeled somatostatin analogs by tumor cells through receptor-mediated internalization is a critical process for the in vivo targeting of tumoral somatostatin receptors. In the present study, the somatostatin receptor internalization induced by a variety of somatostatin analogs was measured with new immunocytochemical methods that allow characterization of trafficking of the somatostatin receptor subtype 2 (sst2), somatostatin receptor subtype 3 (sst3), and somatostatin receptor subtype 5 (sst5) in vitro at the protein level. METHODS: Human embryonic kidney 293 (HEK293) cells expressing the sst2, sst3, or the sst5 were used in a morphologic immunocytochemical internalization assay using specific sst2, sst3 and sst5 antibodies to qualitatively and quantitatively determine the capability of somatostatin agonists or antagonists to induce somatostatin receptor internalization. In addition, the internalization properties of a selection of these agonists have been compared and quantified in sst2-expressing CHO-K1 cells using an ELISA. RESULTS: Agonists with a high sst2-binding affinity were able to induce sst2 internalization in the HEK293 and CHO-K1 cell lines. New sst2 agonists, such as Y-DOTA-TATE, Y-DOTA-NOC, Lu-DOTA-BOC-ATE (where DOTA is 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid; TATE is [Tyr3, Thr8]-octreotide; NOC is [1-NaI3]-octreotide; and BOC-ATE is [BzThi3, Thr8]-octreotide), iodinated sugar-containing octreotide analogs, or BIM-23244 were considerably more potent in internalizing sst2 than was DTPA-octreotide (where DTPA is diethylenetriaminepentaacetic acid). Similarly, compounds with high sst3 affinity such as KE108 were able to induce sst3 internalization. In sst2- or sst3-expressing cell lines, agonist-induced receptor internalization was efficiently abolished by sst2- or sst3-selective antagonists, respectively. Antagonists alone had no effect on sst2 or sst3 internalization. We also showed that somatostatin-28 and somatostatin-14 can induce sst5 internalization. Unexpectedly, however, potent sst5 agonists such as KE108, BIM-23244, and L-817,818 were not able to induce sst5 internalization under the same conditions. CONCLUSION: Using sensitive and reproducible immunocytochemical methods, the ability of various somatostatin analogs to induce sst2, sst3, and sst5 internalization has been qualitatively and quantitatively determined. Whereas all agonists triggered sst2 and sst3 internalization, sst5 internalization was induced by natural somatostatin peptides but not by synthetic high-affinity sst5 agonists. Such assays will be of considerable help for the future characterization of ligands foreseen for nuclear medicine applications.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Radioisótopos do Iodo/farmacocinética , Rim/diagnóstico por imagem , Receptores de Somatostatina/metabolismo , Somatostatina/agonistas , Somatostatina/antagonistas & inibidores , Animais , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Humanos , Rim/metabolismo , Cintilografia , Compostos Radiofarmacêuticos/farmacocinética , Somatostatina/análogos & derivados
16.
Am J Surg Pathol ; 29(12): 1642-51, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16327437

RESUMO

The somatostatin receptor sst2A is highly expressed at the cellular surface of neuroendocrine and other human tumor cells, allowing somatostatin receptor-targeted scintigraphic tumor imaging and tumor therapy. In vitro information on tumoral somatostatin receptor expression is provided mainly by receptor autoradiography, based on binding of radiolabeled somatostatin analogs to tumoral somatostatin receptors. Recent availability of anti-sst2A antibodies opens the possibility of sst2A assessment in human tumors with immunohistochemistry. The aim of the present study was to evaluate the usefulness of several commercial anti-sst2A antibodies for this purpose in comparison with the extensively characterized antibody R2-88 and with receptor autoradiography. sst2A immunohistochemistry was performed in 64 formalin-fixed tumors known to frequently express sst2A (pancreatic, gastrointestinal, pulmonary neuroendocrine tumors, breast carcinomas, meningiomas, pituitary adenomas, neuroblastomas, medulloblastomas, pheochromocytomas, and paragangliomas); receptor autoradiography could be performed simultaneously in 37 of these cases. The commercial antibody SS-800 clearly identified sst2A and correlated excellently with R2-88: compared with R2-88, SS-800 immunohistochemistry generally yielded the same tissular and subcellular staining distribution. Results of R2-88 and SS-800 immunohistochemistry correlated excellently with those obtained with receptor autoradiography; compared with receptor autoradiography, immunohistochemistry with both R2-88 and SS-800 resulted in a slightly lower tumoral sst2A incidence, but a higher resolution, with frequent identification of heterogeneous sst2A distribution at high magnification. Finally, not only membranous, but also cytoplasmic, sst2A immunostaining was simultaneously observed with both antibodies in some tumors. In conclusion, the commercially available SS-800 antibody promises to be useful for the routine immunohistochemical assessment of sst2A in formalin-fixed human tumors.


Assuntos
Anticorpos/imunologia , Imuno-Histoquímica , Neoplasias/metabolismo , Receptores de Somatostatina/metabolismo , Aminoácidos/química , Animais , Anticorpos/química , Autorradiografia , Estudos de Avaliação como Assunto , Humanos , Neoplasias/classificação , Peptídeos/química , Coelhos
17.
Mol Pharmacol ; 68(1): 90-101, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15855408

RESUMO

Upon hormone stimulation, the sst2 somatostatin receptor couples to adenylyl cyclase through G(i/o) proteins and undergoes rapid endocytosis via clathrin-coated pits. In this study, we determined the relationship between the ability of ligands to induce sst2 receptor internalization and inhibit adenylyl cyclase. Immunocytochemical studies demonstrated that peptide agonists [such as somatostatin-14, cortistatin-17, octreotide, vapreotide, KE108 (Tyr0-cyclo[d-diaminobutyric acid-Arg-Phe-Phe-d-Trp-Lys-Thr-Phe]), and SOM230 (cyclo[diaminoethylcarbamoyl-hydroxyproline-phenylglycine-d-Trp-Lys-(4-O-benzyl)-l-Tyr-Phe])] and nonpeptide agonists (such as L-779,976), stimulated the rapid endocytosis of sst2 receptors in human embryonic kidney 293 and CHO-K1 cells. In contrast, two antagonists did not induce receptor endocytosis by themselves and completely blocked agonist stimulation. Using a quantitative enzyme-linked immunosorbent assay to measure sst2 receptor sequestration, we found that peptide agonists varied by more than 100-fold in their potencies but exhibited the same efficacy as somatostatin14. In contrast, L-779,976 did not induce maximal receptor internalization. It is interesting that although betaarrestin-2 was recruited to cell surface sst2 receptors after stimulation with either somatostatin14 or L-779,976, the betaarrestin-receptor complex dissociated earlier in the endocytic pathway with the nonpeptide ligand. Although all agonists, including L-779,976, produced the same maximal inhibition of cyclic AMP, the potency ratio for inhibition of cyclic AMP and stimulation of receptor endocytosis varied by 15-fold. In general, native peptides showed similar potencies for cyclic AMP inhibition and receptor endocytosis, whereas short therapeutic analogs were substantially more potent at inhibiting cyclic AMP synthesis. These results demonstrate that the activity of somatostatin analogs to regulate receptor endocytosis and signaling are not tightly linked and provide compelling evidence for the induction of agonist specific states of the sst2 receptor.


Assuntos
Endocitose/efeitos dos fármacos , Receptores de Somatostatina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Animais , Células CHO , Cricetinae , Relação Dose-Resposta a Droga , Endocitose/fisiologia , Humanos , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/antagonistas & inibidores
18.
J Biol Chem ; 279(9): 8029-37, 2004 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-14660576

RESUMO

Amphiphysins are SH3 domain-containing proteins thought to function in clathrin-mediated endocytosis. To investigate the potential role of amphiphysin II in cellular trafficking of G protein-coupled somatostatin (SRIF) receptors, we generated an AtT-20 cell line stably overexpressing amphiphysin IIb, a splice variant that does not bind clathrin. Endocytosis of (125)I-[d-Trp(8)]SRIF was not affected by amphiphysin IIb overexpression. However, the maximal binding capacity (B(max)) of the ligand on intact cells was significantly lower in amphiphysin IIb overexpressing than in non-transfected cells. This difference was no longer apparent when the experiments were performed on crude cell homogenates, suggesting that amphiphysin IIb overexpression interferes with SRIF receptor targeting to the cell surface and not with receptor synthesis. Accordingly, immunofluorescence experiments demonstrated that, in amphiphysin overexpressing cells, sst(2A) and sst(5) receptors were segregated in a juxtanuclear compartment identified as the trans-Golgi network. Amphiphysin IIb overexpression had no effect on corticotrophin-releasing factor 41-stimulated adrenocorticotropic hormone secretion, suggesting that it is not involved in the regulated secretory pathway. Taken together, these results suggest that amphiphysin II is not necessary for SRIF receptor endocytosis but is critical for its constitutive targeting to the plasma membrane. Therefore, amphiphysin IIb may be an important component of the constitutive secretory pathway.


Assuntos
Proteínas do Tecido Nervoso/fisiologia , Sistemas Neurossecretores/metabolismo , Receptores de Somatostatina/metabolismo , Hormônio Adrenocorticotrópico/metabolismo , Processamento Alternativo , Animais , Linhagem Celular , Membrana Celular/metabolismo , Clatrina/metabolismo , Hormônio Liberador da Corticotropina/farmacologia , Imunofluorescência , Expressão Gênica , Camundongos , Microscopia Confocal , Proteínas do Tecido Nervoso/genética , Proteínas Recombinantes , Somatostatina/metabolismo , Somatostatina/farmacologia , Transfecção , Células Tumorais Cultivadas
19.
J Clin Endocrinol Metab ; 88(12): 6073-9, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14671213

RESUMO

Hormone-stimulated receptor internalization and desensitization occur widely in the G protein-coupled receptor (GPCR) family. A critical first step in both these processes is thought to be receptor phosphorylation, a reaction which has been extensively characterized in cell culture. However, little is known about GPCR phosphorylation in vivo. The somatostatin (SS) receptor subtype (sst)2A is widely distributed in human neuroendocrine tumors, and SS analogs are commonly used to target this receptor for both therapy and diagnosis. In cultured pituitary cells sst2A is rapidly phosphorylated and internalized after hormone binding. The aim of the present study was to go one crucial step further and characterize the phosphorylation state of this receptor in human neuroendocrine tumors using a newly developed gel-shift assay. The receptor from a somatostatinoma was completely phosphorylated. In contrast, only unphosphorylated sst2A was present in human tumors that were not exposed to autocrine stimulation. Both in vivo and in cultured cells, the phosphorylation state of the sst2A receptor was correlated with its subcellular localization: phosphorylated receptor was mostly intracellular, whereas unphosphorylated receptor was localized at the cell surface. These results are the first to demonstrate ligand-stimulated GPCR phosphorylation in human tissue in situ, providing a crucial step toward a better understanding of receptor regulation in vivo. Analysis of sst2A phosphorylation promises to provide a sensitive indicator of the effectiveness of SS analogs in diagnostic and therapeutic situations in tumor patients.


Assuntos
Tumores Neuroendócrinos/metabolismo , Receptores de Somatostatina/metabolismo , Neoplasias das Glândulas Suprarrenais/metabolismo , Animais , Transporte Biológico , Células CHO , Tumor Carcinoide/metabolismo , Cricetinae , Humanos , Neoplasias do Íleo/metabolismo , Neoplasias Pancreáticas/metabolismo , Feocromocitoma/metabolismo , Fosforilação , Receptores de Somatostatina/agonistas , Somatostatinoma/metabolismo , Frações Subcelulares/metabolismo , Distribuição Tecidual
20.
Mol Endocrinol ; 16(11): 2502-14, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12403839

RESUMO

We previously demonstrated that phosphorylation of somatostatin receptor 2A (sst2A) is rapidly increased in transfected cells both by agonist and by the protein kinase C (PKC) activator phorbol myristate acetate (PMA). Here, we investigate whether PKC-mediated receptor phosphorylation is involved in the homologous or heterologous regulation of endogenous sst2 receptors in AR42J pancreatic acinar cells upon stimulation by agonist or by cholecystokinin (CCK) or bombesin (BBS). Somatostatin, PMA, CCK, and BBS all increased sst2A receptor phosphorylation 5- to 10-fold within minutes. Somatostatin binding also caused rapid internalization of the ligand-receptor complex, and PMA, CCK, and BBS all stimulated this internalization further. Additionally, sst2 receptor-mediated inhibition of adenylyl cyclase was desensitized by all treatments. Somatostatin, as well as peptidic (SMS201-995) and nonpeptidic (L-779,976) sst2 receptor agonists increased the EC(50) for somatostatin inhibition 20-fold. In contrast, pretreatment with BBS, CCK, or PMA caused a modest 2-fold increase in the EC(50) for cyclase inhibition. Whereas the PKC inhibitor GF109203X abolished sst2A receptor phosphorylation by CCK, BBS, and PMA, it did not alter the effect of somatostatin, demonstrating that these reactions were catalyzed by different kinases. Consistent with a functional role for PKC-mediated receptor phosphorylation, GF109203X prevented PMA stimulation of sst2 receptor internalization. Surprisingly, however, GF109203X did not inhibit BBS and CCK stimulation of sst2A receptor endocytosis. These results demonstrate that homologous and heterologous hormones induce sst2A receptor phosphorylation by PKC-independent and -dependent mechanisms, respectively, and produce distinct effects on receptor signaling and internalization. In addition, the heterologous hormones also modulate sst2 receptor internalization by a novel mechanism that is independent of receptor phosphorylation.


Assuntos
Receptores de Somatostatina/genética , Inibidores de Adenilil Ciclases , Amidas/farmacologia , Animais , Linhagem Celular , Indóis/farmacologia , Octreotida/farmacologia , Pâncreas , Fosforilação , Proteína Quinase C/metabolismo , Ratos , Receptores de Somatostatina/efeitos dos fármacos , Receptores de Somatostatina/metabolismo , Somatostatina/farmacologia , Acetato de Tetradecanoilforbol/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...