890 resultados para GLP-1 receptor agonists


Relevância:

100.00% 100.00%

Publicador:

Resumo:

The midbrain dorsal periaqueductal gray (dPAG) has an important role in orchestrating anxiety-and panic-related responses. Given the cellular and behavioral evidence suggesting opposite functions for cannabinoid type 1 receptor (CB1) and transient receptor potential vanilloid type-1 channel (TRPV1), we hypothesized that they could differentially influence panic-like reactions induced by electrical stimulation of the dPAG. Drugs were injected locally and the expression of CB1 and TRPV1 in this structure was assessed by immunofluorescence and confocal microscopy. The CB1-selective agonist, ACEA (0.01, 0.05 and 0.5 pmol) increased the threshold for the induction of panic-like responses solely at the intermediary dose, an effect prevented by the CB1-selective antagonist, AM251 (75 pmol). Panicolytic-like effects of ACEA at the higher dose were unmasked by pre-treatment with the TRPV1 antagonist capsazepine (0.1 nmol). Similarly to ACEA, capsazepine (1 and 10 nmol) raised the threshold for triggering panic-like reactions, an effect mimicked by another TRPV1 antagonist, SB366791 (1 nmol). Remarkably, the effects of both capsazepine and SB366791 were prevented by AM251 (75 pmol). These pharmacological data suggest that a common endogenous agonist may have opposite functions at a given synapse. Supporting this view, we observed that several neurons in the dPAG co-expressed CB1 and TRPV1. Thus, the present work provides evidence that an endogenous substance, possibly anandamide, may exert both panicolytic and panicogenic effects via its actions at CB1 receptors and TRPV1 channels, respectively. This tripartite set-point system might be exploited for the pharmacotherapy of panic attacks and anxiety-related disorders. Neuropsychopharmacology (2012) 37, 478-486; doi:10.1038/npp.2011.207; published online 21 September 2011

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Bradykinin is not only important for inflammation and blood pressure regulation, but also involved in neuromodulation and neuroprotection. Here we describe novel functions for bradykinin and the kinin-B2 receptor (B2BkR) in differentiation of neural stem cells. In the presence of the B2BkR antagonist HOE-140 during rat neurosphere differentiation, neuron-specific beta 3-tubulin and enolase expression was reduced together with an increase in glial protein expression, indicating that bradykinin- induced receptor activity contributes to neurogenesis. In agreement, HOE-140 affected in the same way expression levels of neural markers during neural differentiation of murine P19 and human iPS cells. Kinin-B1 receptor agonists and antagonists did not affect expression levels of neural markers, suggesting that bradykinin-mediated effects are exclusively mediated via B2BkR. Neurogenesis was augmented by bradykinin in the middle and late stages of the differentiation process. Chronic treatment with HOE-140 diminished eNOS and nNOS as well as M1-M4 muscarinic receptor expression and also affected purinergic receptor expression and activity. Neurogenesis, gliogenesis, and neural migration were altered during differentiation of neurospheres isolated from B2BkR knock-out mice. Whole mount in situ hybridization revealed the presence of B2BkR mRNA throughout the nervous system in mouse embryos, and less beta 3-tubulin and more glial proteins were expressed in developing and adult B2BkR knock-out mice brains. As a underlying transcriptional mechanism for neural fate determination, HOE-140 induced up-regulation of Notch1 and Stat3 gene expression. Because pharmacological treatments did not affect cell viability and proliferation, we conclude that bradykinin-induced signaling provides a switch for neural fate determination and specification of neurotransmitter receptor expression.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Arthritic pain is a serious health problem that affects a large number of patients. Toll-like receptors (TLRs) activation within the joints has been implicated in pathophysiology of arthritis. However, their role in the genesis of arthritic pain needs to be demonstrated. In the present study, it was addressed the participation of TLR2 and TLR4 and their adaptor molecule MyD88 in the genesis of joint hypernociception (a decrease in the nociceptive threshold) during zymosan-induced arthritis. Zymosan injected in the tibio-tarsal joint induced mechanical hypernociception in C57BL/6 wild type mice that was reduced in TLR2 and MyD88 null mice. On the other hand, zymosan-induced hypernociception was similar in C3H/HePas and C3H/Hej mice (TLR4 mutant mice). Zymosan-induced joint hypernociception was also reduced in TNFR1 null mice and in mice treated with IL-1 receptor antagonist or with an antagonist of CXCR1/2. Moreover, the joint production of TNF-alpha, IL-1 beta and CXCL1/KC by zymosan was dependent on TLR2/MyD88 signaling. Investigating the mechanisms by which TNF-alpha, IL-1 beta and CXCL1/KC mediate joint hypernociception, joint administration of these cytokines produced mechanical hypernociception, and they act in an interdependent manner. In last instance, their hypernociceptive effects were dependent on the production of hypernociceptive mediators, prostaglandins and sympathetic amines. These results indicate that in zymosan-induced experimental arthritis, TLR2/MyD88 is involved in the cascade of events of joint hypernociception through a mechanism dependent on cytokines and chemokines production. Thus, TLR2/MyD88 signaling might be a target for the development of novel drugs to control pain in arthritis. (C) 2011 Elsevier B.V. All rights reserved.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Hyperthyroidism promotes cardiac hypertrophy and the Angiotensin type 1 receptor (AT1R) has been demonstrated to mediate part of this response. Recent studies have uncovered a potentially important role for the microRNAs (miRNAs) in the control of diverse aspects of cardiac function. Then, the objective of the present study was to investigate the action promoted by hyperthyroidism on β-MHC/miR-208b expression and on α-MHC/miR-208a expression, as well as the possible contribution of the AT1R in this event. The findings of this study confirmed that AT1R is a key mediator of the cardiac hypertrophy induced by hyperthyroidism. Additionally, we demonstrated that like β-MHC, miR-208b was down-regulated in the hyperthyroid group. Similarly, like the expression of its host gene, α-MHC, miR-208a expression was up-regulated in response to hyperthyroidism. Finally, our data suggest for the first time that AT1R mediates the hyperthyroidism-induced increase on cardiac miRNA-208a/α-MHC levels, while does not influence on the reduction of miRNA-208b/β-MHC levels.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

The submitted work concentrated on the study of mRNA expression of two distinct GABA transporters, GAT-1 and GAT-3, in the rat brain. For the detection and quantification of the chosen mRNAs, appropriate methods had to be established. Two methods, ribonuclease protection assay (RPA) and competitive RT-PCR were emloyed in the present study. Competitive RT-PCR worked out to be 20 times more sensitive as RPA. Unlike the sensitivity, the fidelity of both techniques was comparable with respect to their intra- and inter-assay variability.The basal mRNA levels of GAT-1 and GAT-3 were measured in various brain regions. Messenger RNAs for both transporters were detected in all tested brain regions. Depending on the region, the observed mRNA level for GAT-1 was 100-300 higher than for GAT-3. The GAT-1 mRNA levels were similar in all tested regions. The distribution of GAT-3 mRNA seemed to be more region specific. The strongest GAT-3 mRNA expression was detected in striatum, medulla oblongata and thalamus. The lowest levels of GAT-3 were in cortex frontalis and cerebellum.Furthermore, the mRNA expression for GAT-1 and GAT-3 was analysed under altered physiological conditions; in kindling model of epilepsy and also after long-term treatment drugs modulating GABAergic transmission. In kindling model of epilepsy, altered GABA transporter function was hypothesised by During and coworkers (During et al., 1995) after observed decrease in binding of nipecotic acid, a GAT ligand, in hippocampus of kindled animals. In the present work, the mRNA levels were measured in hippocampus and whole brain samples. Neither GAT-1 nor GAT-3 showed altered transcription in any tested region of kindled animals compared to controls. This leads to conclusion that an altered functionality of GABA transporters is involved in epilepsy rather than a change in their expression.The levels of GAT-1 and GAT-3 mRNAs were also measured in the brain of rats chronically treated with diazepam or zolpidem, GABAA receptor agonists. Prior to the molecular biology tests, behavioural analysis was carried out with chronically and acutely treated animals. In two tests, open field and elevated plus-maze, the basal activity exploration and anxiety-like behaviour were analysed. Zolpidem treatment increased exploratory activity. There were observed no differencies between chronically and acutely treated animals. Diazepam increased exploratory activity and decresed anxiety-like behaviour when applied acutely. This effect disappeard after chronic administration of diazepam. The loss of effect suggested a development of tolerance to effects of diazepam following long-term administration. Double treatment, acute injection of diazepam after chronic diazepam treatment, confirmed development of a tolerance to effects of diazepam. Also, the mRNAs for GAT-1 and GAT-3 were analysed in cortex frontalis, hippocampus, cerebellum and whole brain samples of chronically treated animals. The mRNA levels for any of tested GABA transporters did not show significant changes in any of tested region neither after diazepam nor zolpidem treatment. Therefore, changes in GAT-1 and GAT-3 transcription are probably not involved in adaptation of GABAergic system to long-term benzodiazepine administration and so in development of tolerance to benzodiazepines.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

In den letzten Jahren gewann die Erforschung des Sphingolipidstoffwechsels in den verschiedensten Zellsystemen immer mehr an Bedeutung, da es sich zeigte, dass einige Sphingolipidspezies, vor allem Ceramid und Sphingosin-1-Phosphat, als wichtige intra- und extrazelluläre Botenstoffe wirken und bei einer Vielzahl unterschiedlicher zellulärer Antworten, wie Apoptose, Proliferation und Migration, eine wichtige Rolle spielen. Während Ceramid eher pro-apoptotisch und wachstumshemmend wirkt, begünstigt Sphingosin-1-Phosphat als „Gegenspieler“ eher die Proliferation und das Zellwachstum. Ceramid kann relativ schnell in Sphingosin-1-Phosphat umgewandelt werden durch die Wirkung zweier Enzymklassen, den Ceramidasen und den Sphingosinkinasen. Konsequenterweise ist die Regulation dieser Enzyme von entscheidender Bedeutung für das zelluläre Gleichgewicht zwischen Ceramid und Sphingosin-1-Phosphat. Im Rahmen dieser Dissertation wurde die Wirkung von extrazellulären Nukleotiden, die ebenfalls als Regulatoren zahlreicher zellulärer Antworten, wie z.B. Proliferation und Migration, bekannt sind und über entsprechende Oberflächenrezeptoren, die Purinrezeptoren, wirken, auf die Aktivität besonders der Sphingosinkinasen 1 und 2 näher untersucht. Es sollte geklärt werden, ob die Sphingosinkinasen an einigen durch extrazelluläre Nukleotide induzierbaren zellulären Antworten, in diesem Falle der Migration und der Proliferation von Zellen, beteiligt sind. Als Zellsystem wurden Nierenmesangiumzellen verwendet, da diese Zellen bei verschiedenen entzündlichen Nierenerkrankungen (Glomerulonephritiden) eine wichtige Rolle spielen. Es konnte in dieser Arbeit gezeigt werden, dass extrazelluläre Nukleotide die Aktivität der Sphingosinkinase 1 in den Mesangiumzellen stimulieren können. Zu beobachten ist dabei eine biphasische Aktivitätssteigerung der Sphingosinkinase 1. Die erste Aktivitätssteigerung nach einer Kurzzeitstimulation ist dabei auf eine Phosphorylierung des Enzyms zurückzuführen, während die zweite Aktivitätssteigerung mit einer Aktivierung des Sphingosinkinase 1-Promotors, einer verstärkten mRNA-Expression und einer de novo Proteinsynthese zu erklären ist. Diese Induktion kann durch die Verwendung von Hemmstoffen des PKC- und MAPK-Signalweges, sowie durch Verwendung eines Transkriptions- (Actinomycin D) oder eines Translationsinhibitors (Cycloheximid) blockiert werden. Die Halbwertszeit der mRNA der Sphingosinkinase 1 in den Mesangiumzellen konnte auf ca. 20 Minuten bestimmt werden. Im Gegensatz dazu ist die Sphingosinkinase 2 nicht durch ATP aktivierbar, wohl aber durch diverse Abbauprodukte von ATP, wie AMP und Adenosin, sowie durch UTP und seine Abbauprodukten UDP und UMP. Die neutrale Ceramidase kann nicht durch ATP und UTP aktiviert werden, wohl aber durch P2X7-Rezeptoragonisten (Bz-ATP, αβ-Me-ATP, γS-ATP) und TPA. In einem zweiten Schritt wurde die Rolle der Sphingosinkinasen und der neutralen Ceramidase bei der durch extrazelluläre Nukleotide induzierten Migration und Proliferation untersucht. Es zeigte sich mit Hilfe von genspezifischer siRNA zur Depletion der Sphingosinkinasen und der neutralen Ceramidase, sowie durch Verwendung von Kinase-Hemmstoffen und damit einhergehend der Inhibierung der Signalwege und mit Hilfe von verschiedenen Zelllinien isoliert aus Wildtyp-, SPHK 1-überexprimierenden und mSPHK1-defizienten Mäusen, dass die Aktivierung der Sphingosinkinase 1 durch extrazelluläre Nukleotide von entscheidender Bedeutung für die Migrationsfähigkeit der Zellen ist, jedoch keinen signifikanten Einfluss auf die Proliferationsrate der Mesangiumzellen hat. Auch die Aktivität der neutralen Ceramidase ist von entscheidender Bedeutung für die Migrationsfähigkeit der Zellen. Durch Depletion der neutralen Ceramidase scheint Ceramid in den Zellen zu akkumulieren, was die Proliferationsrate reduziert. Für die Proliferation der Mesangiumzellen könnte die Sphingosinkinase 2 als negativer Regulator fungieren, wie die Experimente mit der genspezifischen siRNA unter UTP-Stimulation gezeigt haben. Für die Migration der Mesangiumzellen gilt darüber hinaus, dass auch das Produkt der Sphingosinkinase 1, Sphingosin-1-Phosphat, in der Lage ist, die Migration zu stimulieren. Im Gegensatz dazu spielt Sphingosin-1-Phosphat für die Induktion der Proliferation der hier verwendeten Zellen keine wesentliche Rolle. Zusammenfassend zeigen die Daten, dass die Sphingosinkinase 1 und vorgeschaltet auch die neutrale Ceramidase bei der Migration von Mesangiumzellen eine zentrale Rolle spielen und damit als therapeutische Angriffspunkte bei der Behandlung von Krankheiten, die durch eine vermehrte Migration gekennzeichnet sind, in Frage kommen.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Sphingosine-1-phosphate (S1P) acts as high affinity agonist at specific G-protein-coupled receptors, S1P(1-5), that play important roles e.g. in the cardiovascular and immune systems. A S1P receptor modulating drug, FTY720 (fingolimod), has been effective in phase III clinical trials for multiple sclerosis. FTY720 is a sphingosine analogue and prodrug of FTY720-phosphate, which activates all S1P receptors except S1P(2) and disrupts lymphocyte trafficking by internalizing the S1P(1) receptor. Cis-4-methylsphingosine (cis-4M-Sph) is another synthetic sphingosine analogue that is readily taken up by cells and phosphorylated to cis-4-methylsphingosine-1-phosphate (cis-4M-S1P). Therefore, we analysed whether cis-4M-Sph interacted with S1P receptors through its metabolite cis-4M-S1P in a manner similar to FTY720. Indeed, cis-4M-Sph caused an internalization of S1P receptors, but differed from FTY720 as it acted on S1P(2) and S1P(3) and only weakly on S1P(1), while FTY720 internalized S1P(1) and S1P(3) but not S1P(2). Consequently, pre-incubation with cis-4M-Sph specifically desensitized S1P-induced [Ca(2+)](i) increases, which are mediated by S1P(2) and S1P(3), in a time- and concentration-dependent manner. This effect was not shared by sphingosine or FTY720, indicating that metabolic stability and targeting of S1P(2) receptors were important. The desensitization of S1P-induced [Ca(2+)](i) increases was dependent on the expression of SphKs, predominantly of SphK2, and thus mediated by cis-4M-S1P. In agreement, cis-4M-S1P was detected in the supernatants of cells exposed to cis-4M-Sph. It is concluded that cis-4M-Sph, through its metabolite cis-4M-S1P, acts as a S1P receptor modulator and causes S1P receptor internalization and desensitization. The data furthermore help to define requirements for sphingosine kinase substrates as S1P receptor modulating prodrugs.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Peptide hormones of the glucagon-like peptide (GLP) family play an increasing clinical role, as reported for GLP-1 in diabetes therapy and insulinoma diagnostics. GLP-2, despite its known trophic and anti-inflammatory intestinal actions translated into preliminary clinical studies using the GLP-2 analogue teduglutide for treatment of short bowel syndrome and Crohn's disease, remains poorly characterized in terms of expression of its receptor in tissues of interest. Therefore, the GLP-2 receptor expression was assessed in 237 tumor and 148 non-neoplastic tissue samples with in vitro receptor autoradiography. A GLP-2 receptor expression was present in 68% of gastrointestinal stromal tumors (GIST). Furthermore, GLP-2 receptors were identified in the intestinal myenteric plexus, with significant up-regulation in active Crohn's disease. The GLP-2 receptors in GIST may be used for clinical applications like in vivo targeting with radiolabelled GLP-2 analogues for imaging and therapy. Moreover, the over-expressed GLP-2 receptor in the myenteric plexus may represent the morphological correlate of the clinical target of teduglutide in Crohn's disease.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

The recent identification of a cellular balance between ceramide and sphingosine 1-phosphate (S1P) as a critical regulator of cell growth and death has stimulated increasing research effort to clarify the role of ceramide and S1P in various diseases associated with dysregulated cell proliferation and apoptosis. S1P acts mainly, but not exclusively, by binding to and activating specific cell surface receptors, the so-called S1P receptors. These receptors belong to the class of G protein-coupled receptors that constitute five subtypes, denoted as S1P(1)-S1P(5), and represent attractive pharmacological targets to interfere with S1P action. Whereas classical receptor antagonists will directly block S1P action, S1P receptor agonists have also proven useful, as recently shown for the sphingolipid-like immunomodulatory substance FTY720. When phosphorylated by sphingosine kinase to yield FTY720 phosphate, it acutely acts as an agonist at S1P receptors, but upon prolonged presence, it displays antagonistic activity by specifically desensitizing the S1P(1) receptor subtype. This commentary will cover the most recent developments in the field of S1P receptor pharmacology and highlights the potential therapeutic benefit that can be expected from these novel drug targets in the future.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

PURPOSE: G protein-coupled receptor agonists are being used as radiolabeled vectors for in vivo localization and therapy of tumors. Recently, somatostatin-based antagonists were shown to be superior to agonists. Here, we compare the new [111In/68Ga]-labeled bombesin-based antagonist RM1 with the agonist [111In]-AMBA for targeting the gastrin-releasing peptide receptor (GRPR). EXPERIMENTAL DESIGN: IC50, Kd values, and antagonist potency were determined using PC-3 and HEK-GRPR cells. Biodistribution and imaging studies were done in nude mice transplanted with the PC-3 tumor. The antagonist potency was assessed by evaluating the effects on calcium release and on receptor internalization monitored by immunofluorescence microscopy. RESULTS: The IC50 value of [(nat)In]-RM1 was 14 +/- 3.4 nmol/L. [(nat/111)In]-RM1 was found to bind to the GRPR with a Kd of 8.5 +/- 2.7 nmol/L compared with a Kd of 0.6 +/- 0.3 nmol/L of [111In]-AMBA. A higher maximum number of binding site value was observed for [111In]-RM1 (2.4 +/- 0.2 nmol/L) compared with [111In]-AMBA (0.7 +/- 0.1 nmol/L). [(nat)Lu]-AMBA is a potent agonist in the immunofluorescence-based internalization assay, whereas [(nat)In]-RM1 is inactive alone but efficiently antagonizes the bombesin effect. These data are confirmed by the calcium release assay. The pharmacokinetics showed a superiority of the radioantagonist with regard to the high tumor uptake (13.4 +/- 0.8% IA/g versus 3.69 +/- 0.75% IA/g at 4 hours after injection. as well as to all tumor-to-normal tissue ratios. CONCLUSION: Despite their relatively low GRPR affinity, the antagonists [111In/68Ga]-RM1 showed superior targeting properties compared with [111In]-AMBA. As found for somatostatin receptor-targeting radiopeptides, GRP-based radioantagonists seem to be superior to radioagonists for in vivo imaging and potentially also for targeted radiotherapy of GRPR-positive tumors.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Radiolabeled antagonists of specific peptide receptors identify a higher number of receptor binding sites than agonists and may thus be preferable for in vivo tumor targeting. In this study, two novel radioiodinated 1,4-benzodiazepines, (S)-1-(3-iodophenyl)-3-(1-methyl-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diazepin-3-yl)urea (9) and (R)-1-(3-iodophenyl)-3-(1-methyl-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diazepin-3-yl)urea (7), were developed. They were characterized in vitro as high affinity selective antagonists at cholecystokinin types 1 and 2 (CCK(1) and CCK(2)) receptors using receptor binding, calcium mobilization, and internalization studies. Their binding to human tumor tissues was assessed with in vitro receptor autoradiography and compared with an established peptidic CCK agonist radioligand. The (125)I-labeled CCK(1) receptor-selective compound 9 often revealed a substantially higher amount of CCK(1) receptor binding sites in tumors than the agonist (125)I-CCK. Conversely, the radioiodinated CCK(2) receptor-selective compound 7 showed generally weaker tumor binding than (125)I-CCK. In conclusion, compound 9 is an excellent radioiodinated nonpeptidic antagonist ligand for direct and selective labeling of CCK(1) receptors in vitro. Moreover, it represents a suitable candidate to test antagonist binding to CCK(1) receptor-expressing tumors in vivo.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

The pharmacological characterization of ligands depends upon the ability to accurately measure their binding properties. Fluorescence provides an alternative to more traditional approaches such as radioligand binding. Here we describe the binding and spectroscopic properties of eight fluorescent 5-HT3 receptor ligands. These were tested on purified receptors, expressed receptors on live cells, or in vivo. All compounds had nanomolar affinities with fluorescent properties extending from blue to near infra-red emission. A fluorescein-derivative had the highest affinity as measured by fluorescence polarization (FP; 1.14 nM), flow cytometry (FC; 3.23 nM) and radioligand binding (RB; 1.90 nM). Competition binding with unlabeled 5-HT3 receptor agonists (5-HT, mCPBG, quipazine) and antagonists (granisetron, palonosetron, tropisetron) yielded similar affinities in all three assays. When cysteine substitutions were introduced into the 5-HT3 receptor binding site the same changes in binding affinity were seen for both granisetron and the fluorescein-derivative, suggesting that they both adopt orientations that are consistent with co-crystal structures of granisetron with a homologous protein (5HTBP). As expected, in vivo live imaging in anaesthetized mice revealed staining in the abdominal cavity in intestines, but also in salivary glands. The unexpected presence of 5-HT3 receptors in mouse salivary glands was confirmed by Western blots. Overall, these results demonstrate the wide utility of our new high-affinity fluorescently-labeled 5-HT3 receptor probes, ranging from in vitro receptor pharmacology, including FC and FP ligand competition, to live imaging of 5-HT3 expressing tissues.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

BACKGROUND Neuroendocrine tumors are well vascularized and express specific cell surface markers, such as somatostatin receptors and the glucagon-like peptide-1 receptor (GLP-1R). Using the Rip1Tag2 transgenic mouse model of pancreatic neuroendocrine tumors (pNET), we have investigated the potential benefit of a combination of anti-angiogenic treatment with targeted internal radiotherapy. METHODS [Lys40(Ahx-DTPA-111In)NH2]-exendin-4, a radiopeptide that selectively binds to GLP-1R expressed on insulinoma and other neuroendocrine tumor cells, was co-administered with oral vatalanib (an inhibitor of vascular endothelial growth factor receptors (VEGFR)) or imatinib (a c-kit/PDGFR inhibitor). The control groups included single-agent kinase inhibitor treatments and [Lys40(Ahx-DTPA-natIn)NH2]-exendin-4 monotherapy. For biodistribution, Rip1Tag2 mice were pre-treated with oral vatalanib or imatinib for 0, 3, 5, or 7 days at a dose of 100 mg/kg. Subsequently, [Lys40(Ahx-DTPA-111In)NH2]-exendin-4 was administered i.v., and the biodistribution was assessed after 4 h. For therapy, the mice were injected with 1.1 MBq [Lys40(Ahx-DTPA-111In)NH2]-exendin-4 and treated with vatalanib or imatinib 100 mg/kg orally for another 7 days. Tumor volume, tumor cell apoptosis and proliferation, and microvessel density were quantified. RESULTS Combination of [Lys40(Ahx-DTPA-111In)NH2]-exendin-4 and vatalanib was significantly more effective than single treatments (p < 0.05) and reduced the tumor volume by 97% in the absence of organ damage. The pre-treatment of mice with vatalanib led to a reduction in the tumor uptake of [Lys40(Ahx-DTPA-111In)NH2]-exendin-4, indicating that concomitant administration of vatalanib and the radiopeptide was the best approach. Imatinib did not show a synergistic effect with [Lys40(Ahx-DTPA-111In)NH2]-exendin-4. CONCLUSION The combination of 1.1 MBq of [Lys40(Ahx-DTPA-111In)NH2]-exendin-4 with 100 mg/kg vatalanib had the same effect on a neuroendocrine tumor as the injection of 28 MBq of the radiopeptide alone but without any apparent side effects, such as radiation damage of the kidneys.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

UNLABELLED A high proportion of gut and bronchial neuroendocrine tumors (NETs) overexpresses somatostatin receptors, especially the sst2 subtype. It has also recently been observed that incretin receptors, namely glucagonlike peptide 1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP) receptors, can be overexpressed in gut and bronchial NETs. However, because not all tumors can express these receptors in sufficient amounts, in vivo imaging with a single radioligand may not always be successful. We therefore evaluated with in vitro methods whether a cocktail of radioligands targeting these 3 receptors would improve tumor labeling. METHODS In vitro receptor autoradiography was performed on 55 NETs, comparing in each successive section of tumor the binding with a single radioligand, either (125)I-Tyr(3)-octreotide, (125)I-GLP-1(7-36)amide, or (125)I-GIP(1-30), with the binding using a cocktail of all 3 radioligands, given concomitantly under identical experimental conditions. RESULTS Using the cocktail of radioligands, all tumors without exception showed moderate to very high binding, with a receptor density corresponding to 1,000-10,000 dpm/mg of tissue; conversely, single-ligand binding, although identifying most tumors as receptor-positive, failed to detect receptors or measured only a low density of receptors below 1,000 dpm/mg in a significant number of tumors. In addition, the cocktail of radioligands always provided a homogeneous labeling of the whole tumor, whereas single radioligands occasionally showed heterogeneous labeling. CONCLUSION The study suggests that the use of a cocktail of 3 radioligands binding to somatostatin receptors, GLP-1 receptors, and GIP receptors would allow detecting virtually all NETs and labeling them homogeneously in vivo, representing a significant improvement for imaging and therapy in NETs.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Cytokines interact with hematopoietin superfamily receptors and stimulate receptor dimerization. We demonstrate that chemoattractant cytokines (chemokines) also trigger biological responses through receptor dimerization. Functional responses are induced after pairwise crosslinking of chemokine receptors by bivalent agonistic antichemokine receptor mAb, but not by their Fab fragments. Monocyte chemoattractant protein (MCP)-1-triggered receptor dimerization was studied in human embryonic kidney (HEK)-293 cells cotransfected with genes coding for the CCR2b receptor tagged with YSK or Myc sequences. After MCP-1 stimulation, immunoprecipitation with Myc-specific antibodies revealed YSK-tagged receptors in immunoblotting. Receptor dimerization also was validated by chemical crosslinking in both HEK-293 cells and the human monocytic cell line Mono Mac 1. Finally, we constructed a loss-of-function CCR2bY139F mutant that acted as a dominant negative, blocking signaling through the CCR2 wild-type receptor. This study provides functional support for a model in which the MCP-1 receptor is activated by ligand-induced homodimerization, allowing discussion of the similarities between bacterial and leukocyte chemotaxis.