Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 341
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Biol ; 22(7): e3002673, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39083706

ABSTRACT

Development of optimal therapeutics for disease states that can be associated with increased membrane cholesterol requires better molecular understanding of lipid modulation of the drug target. Type 1 cholecystokinin receptor (CCK1R) agonist actions are affected by increased membrane cholesterol, enhancing ligand binding and reducing calcium signaling, while agonist actions of the closely related CCK2R are not. In this work, we identified a set of chimeric human CCK1R/CCK2R mutations that exchange the cholesterol sensitivity of these 2 receptors, providing powerful tools when expressed in CHO and HEK-293 model cell lines to explore mechanisms. Static, low energy, high-resolution structures of the mutant CCK1R constructs, stabilized in complex with G protein, were not substantially different, suggesting that alterations to receptor dynamics were key to altered function. We reveal that cholesterol-dependent dynamic changes in the conformation of the helical bundle of CCK receptors affects both ligand binding at the extracellular surface and G protein coupling at the cytosolic surface, as well as their interrelationships involved in stimulus-response coupling. This provides an ideal setting for potential allosteric modulators to correct the negative impact of membrane cholesterol on CCK1R.


Subject(s)
Cholesterol , GTP-Binding Proteins , Protein Binding , Receptor, Cholecystokinin A , Receptor, Cholecystokinin B , Animals , Humans , CHO Cells , Cholesterol/metabolism , Cricetulus , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/genetics , HEK293 Cells , Ligands , Mutation , Protein Conformation , Receptor, Cholecystokinin A/metabolism , Receptor, Cholecystokinin A/genetics , Receptor, Cholecystokinin B/metabolism , Receptor, Cholecystokinin B/genetics
2.
Am J Physiol Gastrointest Liver Physiol ; 326(3): G291-G309, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38252699

ABSTRACT

Hepatocellular carcinoma (HCC) is the fastest-growing cause of cancer-related deaths worldwide. Chronic inflammation and fibrosis are the greatest risk factors for the development of HCC. Although the cell of origin for HCC is uncertain, many theories believe this cancer may arise from liver progenitor cells or stem cells. Here, we describe the activation of hepatic stem cells that overexpress the cholecystokinin-B receptor (CCK-BR) after liver injury with either a DDC diet (0.1% 3, 5-diethoxy-carbonyl 1,4-dihydrocollidine) or a NASH-inducing CDE diet (choline-deficient ethionine) in murine models. Pharmacologic blockade of the CCK-BR with a receptor antagonist proglumide or knockout of the CCK-BR in genetically engineered mice during the injury diet reduces the expression of hepatic stem cells and prevents the formation of three-dimensional tumorspheres in culture. RNA sequencing of livers from DDC-fed mice treated with proglumide or DDC-fed CCK-BR knockout mice showed downregulation of differentially expressed genes involved in cell proliferation and oncogenesis and upregulation of tumor suppressor genes compared with controls. Inhibition of the CCK-BR decreases hepatic transaminases, fibrosis, cytokine expression, and alters the hepatic immune cell signature rendering the liver microenvironment less oncogenic. Furthermore, proglumide hastened recovery after liver injury by reversing fibrosis and improving markers of synthetic function. Proglumide is an older drug that is orally bioavailable and being repurposed for liver conditions. These findings support a promising therapeutic intervention applicable to patients to prevent the development of HCC and decrease hepatic fibrosis.NEW & NOTEWORTHY This investigation identified a novel pathway involving the activation of hepatic stem cells and liver oncogenesis. Receptor blockade or genetic disruption of the cholecystokinin-B receptor (CCK-BR) signaling pathway decreased the activation and proliferation of hepatic stem cells after liver injury without eliminating the regenerative capacity of healthy hepatocytes.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Mice , Animals , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism , Carcinoma, Hepatocellular/pathology , Proglumide/pharmacology , Liver Neoplasms/metabolism , Liver/metabolism , Fibrosis , Stem Cells/metabolism , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/metabolism , Cholecystokinin/metabolism , Tumor Microenvironment
3.
Mol Pharm ; 21(8): 3848-3865, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38959127

ABSTRACT

The cholecystokinin type 2 receptor (CCK2-R) represents an ideal target for cancer therapy since it is overexpressed in several tumors and is associated with poor prognosis. Nastorazepide (Z-360), a selective CCK2-R antagonist, has been widely investigated as a CCK2-R ligand for targeted therapy; however, its high hydrophobicity may represent a limit to cell selectivity and optimal in vivo biodistribution. Here, we present three new fluorescent Z-360 derivatives (IP-002G-Rho, IP-002L-Rho, and IP-002M-Rho) in which nastorazepide was linked, through spacers bearing different saccharides (glucose (G), lactose (L), and maltotriose (M)), to sulforhodamine B. A fourth compound (IP-002H-Rho) with no pendant sugar was also synthesized as a control. Through two-dimensional (2D) and three-dimensional (3D) in vitro studies, we evaluated the compound association with and selectivity for CCK2-R-overexpressing cells (A431-CCK2-R+) vs CCK2-R-underexpressing cells (A431 WT). 2D in vitro studies highlighted a progressive increase of IP-002x-Rho association with A431-CCK2-R+ cells according to the linker hydrophilicity, that is, maltotriose > lactose > glucose > hydrogen, with IP-002M-Rho showing a 2.4- and a 1.36-fold higher uptake than IP-002G-Rho and IP-002L-Rho, respectively. Unexpectedly, IP-002H-Rho showed a similar cell association to that of IP-002L-Rho but with no difference between the two tested cell lines. On the contrary, association with A431-CCK2-R+ cells as compared to the A431 WT was found to be 1.08-, 1.14-, and 1.37-fold higher for IP-002G-Rho, IP-002L-Rho, and IP-002M-Rho, respectively, proving IP-002M-Rho to be the best-performing compound, as also confirmed by competition studies. Trafficking studies on A431-CCK2-R+ cells incubated with IP-002M-Rho suggested the coexistence of receptor-mediated endocytosis and simple diffusion. On the contrary, a high and selective uptake of IP-002M-Rho by A431-CCK2-R+ cells only was observed on 3D scaffolds embedded with cells, underlining the importance of 3D models in in vitro preliminary evaluation.


Subject(s)
Receptor, Cholecystokinin B , Humans , Receptor, Cholecystokinin B/antagonists & inhibitors , Receptor, Cholecystokinin B/metabolism , Cell Line, Tumor , Trisaccharides/chemistry , Lactose/analogs & derivatives , Lactose/chemistry , Glucose/metabolism
4.
J Pept Sci ; 30(10): e3607, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38710638

ABSTRACT

There is an expanding body of evidence showing that synthetic peptides in combination with radioactive isotopes can be utilized for medical purposes. This area is of particular interest in oncology where applications in diagnosis and therapy are at different stages of development. We review the contributions in this area by the group originally founded by Carlo Pedone in Naples many years ago. We highlight the work of this group in the context of other developments in this area, focusing on three biologically relevant receptor systems: somatostatin, gastrin-releasing peptide, and cholecystokinin-2/gastrin receptors. We focus on key milestones, state of the art, and challenges in this area of research as well as the current and future outlook for expanding clinical applications.


Subject(s)
Neoplasms , Peptides , Radiopharmaceuticals , Humans , Neoplasms/drug therapy , Neoplasms/diagnostic imaging , Peptides/chemistry , Peptides/therapeutic use , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/therapeutic use , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Gastrin-Releasing Peptide/chemistry , Gastrin-Releasing Peptide/metabolism , Gastrin-Releasing Peptide/antagonists & inhibitors , Receptor, Cholecystokinin B/metabolism , Receptor, Cholecystokinin B/antagonists & inhibitors , Radioisotopes/chemistry , Radioisotopes/therapeutic use , Animals , Molecular Targeted Therapy/methods
5.
Fish Physiol Biochem ; 50(3): 941-954, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38381278

ABSTRACT

Gastrin is an important intragastrointestinal hormone, but reports on its regulation of feeding behavior in fish are still scarce. This study aimed to determine the feeding regulatory function of gastrin in sturgeon. In this study, a gastrin/cholecystokinin-like peptide was identified in the genomes of sturgeon and proved to be gastrin by evolutionary tree analysis. Tissue distribution of gastrin and its receptor, cholecystokinin receptor B (CCKRB), showed that both had high mRNA abundance in the hypothalamus and gastrointestinal tract. In the duodenum, gastrin and CCKRB mRNAs were reduced at 1 h of fasting, and both were also observed in the stomach and hypothalamus in response to changes in feeding status. Sulfated gastrin 17 is the major form of gastrin in vivo. Therefore, we investigated the effect of sulfated gastrin 17 on feeding by intraperitoneal injection into Siberian sturgeon using sulfated gastrin 17. The results showed that gastrin 17 significantly reduced the cumulative feeding of Siberian sturgeon in the short term (1, 3 and 6 h) and long term (1, 2, 3, 4, 5 and 7 days). Finally, we explored the potential mechanism of feeding inhibition after intraperitoneal injection of gastrin 17 for 7 consecutive days. The results showed that gastrin 17 treatment significantly increased the mRNA levels of anorexigenic peptides (cart, cck and pyy), while it had no significant effect on the mRNA abundance of orexigenic peptides (npy and agrp). In addition, gastrin 17 treatment significantly affected the expression of appetite signaling pathways in the hypothalamus, such that the mRNA expression of ampkα1 was significantly reduced, whereas the mRNA abundance of stat3, mtor and s6k was significantly increased. In conclusion, the present study confirmed the anorectic effect of gastrin on Siberian sturgeon.


Subject(s)
Fishes , Gastrins , Receptor, Cholecystokinin B , Animals , Gastrins/metabolism , Fishes/physiology , Fishes/metabolism , Receptor, Cholecystokinin B/metabolism , Receptor, Cholecystokinin B/genetics , Feeding Behavior/drug effects , RNA, Messenger/metabolism , RNA, Messenger/genetics , Hypothalamus/metabolism
6.
Eur J Nucl Med Mol Imaging ; 50(3): 892-907, 2023 02.
Article in English | MEDLINE | ID: mdl-36334104

ABSTRACT

INTRODUCTION: Medullary thyroid cancer (MTC) is a rare malignant tumour of the parafollicular C-cells with an unpredictable clinical course and currently suboptimal diagnostic and therapeutic options, in particular in advanced disease. Overexpression of cholecystokinin-2 receptors (CCK2R) represents a promising avenue to diagnostic imaging and targeted therapy, ideally through a theranostic approach. MATERIALS AND METHODS: A translational study (GRAN-T-MTC) conducted through a Phase I multicentre clinical trial of the indium-111 labelled CP04 ([111In]In-CP04), a CCK2R-seeking ligand was initiated with the goal of developing a theranostic compound. Patients with proven advanced/metastatic MTC or short calcitonin doubling time were enrolled. A two-step concept was developed through the use of low- and high-peptide mass (10 and 50 µg, respectively) for safety assessment, with the higher peptide mass considered appropriate for therapeutic application. Gelofusine was co-infused in a randomized fashion in the second step for the evaluation of potential reduction of the absorbed dose to the kidneys. Imaging for the purpose of biodistribution, dosimetry evaluation, and diagnostic assessment were performed as well as pre-, peri-, and postprocedural clinical and biochemical assessment. RESULTS: Sixteen patients were enrolled. No serious adverse events after application of the compound at both peptide amounts were witnessed; transient tachycardia and flushing were observed in two patients. No changes in biochemistry and clinical status were observed on follow-up. Preliminary dosimetry assessment revealed the highest dose to urinary bladder, followed by the kidneys and stomach wall. The effective dose for 200 MBq of [111In]In-CP04 was estimated at 7±3 mSv and 7±1 mSv for 10 µg and 50 µg CP04, respectively. Administration of Gelofusine reduced the dose to the kidneys by 53%, resulting in the organ absorbed dose of 0.044±0.019 mSv/MBq. Projected absorbed dose to the kidneys with the use of [177Lu]Lu-CP04 was estimated at 0.9±0.4 Gy/7.4 GBq. [111In]In-CP04 scintigraphy was positive in 13 patients (detection rate of 81%) with superior diagnostic performance over conventional imaging. CONCLUSION: In the present study, [111In]In-CP04 was shown to be a safe and effective radiopharmaceutical with promising theranostic characteristics for patients with advanced MTC.


Subject(s)
Receptor, Cholecystokinin B , Thyroid Neoplasms , Humans , Receptor, Cholecystokinin B/metabolism , Receptor, Cholecystokinin B/therapeutic use , Precision Medicine , Polygeline/therapeutic use , Ligands , Tissue Distribution , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/drug therapy , Peptides
7.
Int J Mol Sci ; 24(23)2023 Nov 28.
Article in English | MEDLINE | ID: mdl-38069171

ABSTRACT

Micro-environmental factors, including stromal and immune cells, cytokines, and circulating hormones are well recognized to determine cancer progression. Melanoma cell growth was recently shown to be suppressed by cholecystokinin/gastrin (CCK) receptor antagonists, and our preliminary data suggested that melanoma patients with Helicobacter gastritis (which is associated with elevated serum gastrin) might have an increased risk of cancer progression. Therefore, in the present study, we examined how gastrin may act on melanoma cells. In 89 melanoma patients, we found a statistically significant association between circulating gastrin concentrations and melanoma thickness and metastasis, which are known risk factors of melanoma progression and prognosis. Immunocytochemistry using a validated antibody confirmed weak to moderate CCK2R expression in both primary malignant melanoma cells and the melanoma cell lines SK-MEL-2 and G361. Furthermore, among the 219 tumors in the Skin Cutaneous Melanoma TCGA Pan-Cancer dataset showing gastrin receptor (CCKBR) expression, significantly higher CCKBR mRNA levels were linked to stage III-IV than stage I-II melanomas. In both cell lines, gastrin increased intracellular calcium levels and stimulated cell migration and invasion through mechanisms inhibited by a CCK2 receptor antagonist. Proteomic studies identified increased MMP-2 and reduced TIMP-3 levels in response to gastrin that were likely to contribute to the increased migration of both cell lines. However, the effects of gastrin on tumor cell invasion were relatively weak in the presence of the extracellular matrix. Nevertheless, dermal fibroblasts/myofibroblasts, known also to express CCK2R, increased gastrin-induced cancer cell invasion. Our data suggest that in a subset of melanoma patients, an elevated serum gastrin concentration is a risk factor for melanoma tumor progression, and that gastrin may act on both melanoma and adjacent stromal cells through CCK2 receptors to promote mechanisms of tumor migration and invasion.


Subject(s)
Melanoma , Skin Neoplasms , Humans , Melanoma/metabolism , Gastrins/pharmacology , Gastrins/metabolism , Proteomics , Receptors, Cholecystokinin , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism
8.
Biochem Biophys Res Commun ; 588: 23-28, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34942530

ABSTRACT

Morphine is the pain releasing and abusing drug. Morphine leads to addiction by activating dopaminergic rewarding system consisted of the ventral tegmental area (VTA) and nucleus accumbens (NAc). Cholecystokinin (CCK) is a gut-brain neuropeptide and involved in morphine dependence. Brain-derived neurotrophic factor (BDNF) is a neurotrophin and plays roles in regulating addiction. Geranylgeranylacetone (GGA) is a medicine of protecting gastric mucosal injury and protecting neurons. Our previous study showed that GGA blocked morphine-induced withdrawal and relapse through inducing thioredoxin 1(Trx1). In this study, we investigated that whether cholecystokinin-B receptor (CCKB receptor) and BDNF were related to GGA inhibition on morphine addiction. At first, we made conditioned place preference (CPP) model and confirmed again that GGA blocked the expression of morphine-CPP in present study. Then, our results showed that morphine increased the expressions of dopamine D1 receptor, tyrosine hydroxylase (TH), CCKB receptor and BDNF in the VTA and NAc in mice, which was inhibited by GGA. These results suggest that CCK and BDNF in dopaminergic systems are associated with the role of GGA blocking morphine-CPP.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Diterpenes/pharmacology , Morphine/adverse effects , Receptor, Cholecystokinin B/metabolism , Receptors, Dopamine D1/metabolism , Animals , Conditioning, Classical , Male , Mice, Inbred C57BL , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
9.
J Pharmacol Sci ; 148(4): 364-368, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35300811

ABSTRACT

We examined whether galantamine (GAL), a cholinesterase inhibitor and allosteric potentiating ligand for α7 nicotinic acetylcholine receptor (nAChR), had an impact on emotional abnormalities in forebrain-specific cholecystokinin receptor-2 overexpressed transgenic mice. Treatment with GAL (1 mg/kg, s.c.) attenuated the decrease of social interaction time, but failed to attenuate anxiety-like behavior in the elevated plus-maze test. The effect of GAL was blocked by an α7 nAChR antagonist, methyllycaconitine (3 mg/kg, i.p.). These results suggest that GAL improved social interaction impairments via α7 nAChR and could be useful to treat sociability-related emotional abnormalities.


Subject(s)
Cholinesterase Inhibitors , Galantamine , Receptor, Cholecystokinin B , Social Behavior Disorders , Social Interaction , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , Animals , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/therapeutic use , Galantamine/pharmacology , Galantamine/therapeutic use , Mice , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism , Social Behavior Disorders/drug therapy , Social Interaction/drug effects
10.
Proc Natl Acad Sci U S A ; 116(13): 6397-6406, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30850520

ABSTRACT

Memory is stored in neural networks via changes in synaptic strength mediated in part by NMDA receptor (NMDAR)-dependent long-term potentiation (LTP). Here we show that a cholecystokinin (CCK)-B receptor (CCKBR) antagonist blocks high-frequency stimulation-induced neocortical LTP, whereas local infusion of CCK induces LTP. CCK-/- mice lacked neocortical LTP and showed deficits in a cue-cue associative learning paradigm; and administration of CCK rescued associative learning deficits. High-frequency stimulation-induced neocortical LTP was completely blocked by either the NMDAR antagonist or the CCKBR antagonist, while application of either NMDA or CCK induced LTP after low-frequency stimulation. In the presence of CCK, LTP was still induced even after blockade of NMDARs. Local application of NMDA induced the release of CCK in the neocortex. These findings suggest that NMDARs control the release of CCK, which enables neocortical LTP and the formation of cue-cue associative memory.


Subject(s)
Cholecystokinin/metabolism , Long-Term Potentiation/physiology , Memory/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Auditory Cortex/metabolism , Behavior, Animal , Cholecystokinin/genetics , Electric Stimulation , Entorhinal Cortex/metabolism , Female , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Methylaspartate/metabolism , Neocortex/metabolism , Neurons/metabolism , Rats, Sprague-Dawley , Receptor, Cholecystokinin B/drug effects , Receptor, Cholecystokinin B/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synapses/metabolism
11.
Molecules ; 27(6)2022 Mar 21.
Article in English | MEDLINE | ID: mdl-35335396

ABSTRACT

The new minigastrin analog DOTA-MGS8 targeting the cholecystokinin-2 receptor (CCK2R) used in this study displays the combination of two site-specific modifications within the C-terminal receptor binding sequence together with an additional N-terminal amino acid substitution preventing fast metabolic degradation. Within this study, the preparation of 68Ga-labeled DOTA-MGS8 was validated using an automated synthesis module, describing the specifications and analytical methods for quality control for possible clinical use. In addition, preclinical studies were carried out to characterize the targeting potential. [68Ga]Ga-DOTA-MGS8 showed a high receptor-specific cell internalization into AR42J rat pancreatic cells (~40%) with physiological expression of rat CCK2R as well as A431-CCK2R cells transfected to stably express human CCK2R (~47%). A favorable biodistribution profile was observed in BALB/c nude mice xenografted with A431-CCK2R cells and mock-transfected A431 cells as control. The high tumor uptake of ~27% IA/g together with low background activity and limited uptake in non-target tissue confirms the potential for high-sensitivity positron emission tomography of stabilized MG analogs in patients with MTC and other CCK2R-related malignancies.


Subject(s)
Gallium Radioisotopes , Receptor, Cholecystokinin B , Animals , Cell Line, Tumor , Heterocyclic Compounds, 1-Ring , Humans , Mice , Mice, Nude , Rats , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism , Tissue Distribution
12.
Biol Pharm Bull ; 44(5): 701-706, 2021.
Article in English | MEDLINE | ID: mdl-33952826

ABSTRACT

We previously reported that exposure of human colon adenocarcinoma (Caco-2) cells to the bitter substance phenylthiocarbamide (PTC) rapidly enhanced the transport function of P-glycoprotein (P-gp). In this study, we investigated the short-term effect of etoposide, another bitter-tasting P-gp substrate, on P-gp transport function in the same cell line. We found that etoposide exposure significantly increased both the P-gp protein level in the plasma membrane fraction and the efflux rate of rhodamine123 (Rho123) in Caco-2 cells within 10 min. The efflux ratio (ratio of the apparent permeability coefficient in the basal-to-apical direction to that in the apical-to-basal direction) of Rho123 in etoposide-treated cells was also significantly increased compared with the control. These results indicated that etoposide rapidly enhances P-gp function in Caco-2 cells. In contrast, P-gp expression in whole cells at both the mRNA and protein level was unchanged by etoposide exposure, compared with the levels in non-treated cells. Furthermore, etoposide increased the level of phosphorylated ezrin, radixin and moesin (P-ERM) proteins in the plasma membrane fraction of Caco-2 cells within 10 min. P-gp functional changes were blocked by YM022, an inhibitor of cholecystokinin (CCK) receptor. These results suggest that etoposide induces release of CCK, causing activation of the CCK receptor followed by phosphorylation of ERM proteins, which recruit intracellular P-gp for trafficking to the gastrointestinal membrane, thereby increasing the functional activity of P-gp.


Subject(s)
Etoposide/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , Benzodiazepines/pharmacology , Caco-2 Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Cholecystokinin/metabolism , Cytoskeletal Proteins/metabolism , Humans , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Phosphorylation/drug effects , Receptor, Cholecystokinin B/antagonists & inhibitors , Receptor, Cholecystokinin B/metabolism
13.
Molecules ; 26(4)2021 Feb 09.
Article in English | MEDLINE | ID: mdl-33572353

ABSTRACT

The cholecystokinin-2 receptor (CCK-2R) is overexpressed in several human cancers but displays limited expression in normal tissues. For this reason, it is a suitable target for developing specific radiotracers. In this study, a nastorazepide-based ligand functionalized with a 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) chelator (IP-001) was synthesized and labelled with indium-111. The radiolabeling process yielded >95% with a molar activity of 10 MBq/nmol and a radiochemical purity of >98%. Stability studies have shown a remarkable resistance to degradation (>93%) within 120 h of incubation in human blood. The in vitro uptake of [111In]In-IP-001 was assessed for up to 24 h on a high CCK-2R-expressing tumor cell line (A549) showing maximal accumulation after 4 h of incubation. Biodistribution and single photon emission tomography (SPECT)/CT imaging were evaluated on BALB/c nude mice bearing A549 xenograft tumors. Implanted tumors could be clearly visualized after only 4 h post injection (2.36 ± 0.26% ID/cc), although a high amount of radiotracer was also found in the liver, kidneys, and spleen (8.25 ± 2.21%, 6.99 ± 0.97%, and 3.88 ± 0.36% ID/cc, respectively). Clearance was slow by both hepatobiliary and renal excretion. Tumor retention persisted for up to 24 h, with the tumor to organs ratio increasing over-time and ending with a tumor uptake (1.52 ± 0.71% ID/cc) comparable to liver and kidneys.


Subject(s)
Benzodiazepines/chemistry , Indium Radioisotopes/pharmacokinetics , Lung Neoplasms/metabolism , Radiopharmaceuticals/pharmacokinetics , Receptor, Cholecystokinin B/antagonists & inhibitors , Animals , Apoptosis , Cell Proliferation , Female , Humans , Indium Radioisotopes/administration & dosage , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Mice , Mice, Inbred BALB C , Mice, Nude , Radiopharmaceuticals/administration & dosage , Receptor, Cholecystokinin B/metabolism , Tissue Distribution , Tomography, Emission-Computed, Single-Photon , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Dig Dis Sci ; 65(1): 189-203, 2020 01.
Article in English | MEDLINE | ID: mdl-31297627

ABSTRACT

BACKGROUND AND AIMS: Nonalcoholic steatohepatitis (NASH) is a common inflammatory liver condition that may lead to cirrhosis and hepatocellular carcinoma (HCC). Risk factors for NASH include a saturated fat diet, altered lipid metabolism, and genetic and epigenetic factors, including microRNAs. Serum levels of cholecystokinin (CCK) are elevated in mice and humans that consume a high-saturated fat diet. CCK receptors (CCK-Rs) have been reported on fibroblasts which when activated can induce fibrosis; however, their role in hepatic fibrosis remains unknown. We hypothesized that elevated levels of CCK acting on the CCK-Rs play a role in the development of NASH and in NASH-associated HCC. METHODS: We performed a NASH Prevention study and Reversal study in mice fed a saturated fat 75% choline-deficient-ethionine-supplemented (CDE) diet for 12 or 18 weeks. In each study, half of the mice received untreated drinking water, while the other half received water supplemented with the CCK-R antagonist proglumide. CCK-R expression was evaluated in mouse liver and murine HCC cells. RESULTS: CCK receptor antagonist treatment not only prevented NASH but also reversed hepatic inflammation, fibrosis, and steatosis and normalized hepatic transaminases after NASH was established. Thirty-five percent of the mice on the CDE diet developed HCC compared with none in the proglumide-treated group. We found that CCK-BR expression was markedly upregulated in mouse CDE liver and HCC cells compared with normal hepatic parenchymal cells, and this expression was epigenetically regulated by microRNA-148a. CONCLUSION: These results support the novel role of CCK receptors in the pathogenesis of NASH and HCC.


Subject(s)
Carcinoma, Hepatocellular/prevention & control , Hormone Antagonists/pharmacology , Liver Neoplasms/prevention & control , Liver/drug effects , Non-alcoholic Fatty Liver Disease/prevention & control , Proglumide/pharmacology , Receptor, Cholecystokinin B/antagonists & inhibitors , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Choline Deficiency/complications , Disease Models, Animal , Epigenesis, Genetic , Ethionine , Female , Gene Expression Regulation, Neoplastic , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism , Signal Transduction
15.
Can J Physiol Pharmacol ; 98(5): 282-295, 2020 May.
Article in English | MEDLINE | ID: mdl-31821012

ABSTRACT

Apelin is a peptide that plays a role in physiological processes such as angiogenesis, apoptosis, and proliferation. The aim of this study was to investigate the role of capsaicin-sensitive afferent neurons and vagus in the effect of apelin against ischemia/reperfusion (I/R) injury. The experimental groups were (1) control, (2) I/R, (3) apelin + I/R, (4) vagotomy + I/R, (5) vagotomy + apelin + I/R, (6) capsaicin + I/R, (7) capsaicin + apelin + I/R, (8) lorglumide + I/R, and (9) lorglumide + apelin + I/R. To test the potential gastroprotective effect of apelin-13, apelin-13 (2 mg/kg i.v.) was administered just before both ischemia and reperfusion. A vagotomy was performed 1 week before I/R in the vagotomized groups; capsaicin (125 mg/kg s.c.) was administrated 2 weeks before I/R in the capsaicin-treated groups and lorglumide (5 mg/kg i.p.) was administered 30 min before I/R in the lorglumide-treated groups. After I/R, a variety parameters in gastric tissue were analyzed. cfos expression was determined in brainstem samples. In the I/R group, the lesion index, myeloperoxidase activity, lipid peroxidation, nitric oxide, and tumor necrosis factor-α increased, and mucosal blood flow, prostaglandin-E2, and calcitonin gene related peptide decreased. Apelin prevented the damaging effects of I/R and increased cfos expression in brainstem areas. Vagotomy, capsaicin, and lorglumide largely eliminated the gastroprotective effects of apelin-13. This study showed that sensory nerves and the vagus play regulatory roles in apelin-induced gastroprotection. Cholecystokinin may play a role in the effect of apelin through sensory neurons.


Subject(s)
Intercellular Signaling Peptides and Proteins/pharmacology , Reperfusion Injury/drug therapy , Reperfusion Injury/physiopathology , Sensory Receptor Cells/drug effects , Stomach/drug effects , Stomach/innervation , Vagus Nerve/drug effects , Animals , Cytoprotection/drug effects , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Male , Nitric Oxide/metabolism , Peroxidase/metabolism , Rats , Rats, Wistar , Receptor, Cholecystokinin B/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Sensory Receptor Cells/pathology , Vagus Nerve/physiopathology
16.
Molecules ; 25(19)2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33049999

ABSTRACT

Targeting of cholecystokinin-2 receptor (CCK2R) expressing tumors using radiolabeled minigastrin (MG) analogs is hampered by rapid digestion of the linear peptide in vivo. In this study, a new MG analog stabilized against enzymatic degradation was investigated in preclinical studies to characterize the metabolites formed in vivo. The new MG analog DOTA-DGlu-Pro-Tyr-Gly-Trp-(N-Me)Nle-Asp-1Nal-NH2 comprising site-specific amino acid substitutions in position 2, 6 and 8 and different possible metabolites thereof were synthesized. The receptor interaction of the peptide and selected metabolites was evaluated in a CCK2R-expressing cell line. The enzymatic stability of the 177Lu-labeled peptide analog was evaluated in vitro in different media as well as in BALB/c mice up to 1 h after injection and the metabolites were identified based on radio-HPLC analysis. The new radiopeptide showed a highly increased stability in vivo with >56% intact radiopeptide in the blood of BALB/c mice 1 h after injection. High CCK2R affinity and cell uptake was confirmed only for the intact peptide, whereas enzymatic cleavage within the receptor specific C-terminal amino acid sequence resulted in complete loss of affinity and cell uptake. A favorable biodistribution profile was observed in BALB/c mice with low background activity, preferential renal excretion and prolonged uptake in CCK2R-expressing tissues. The novel stabilized MG analog shows high potential for diagnostic and therapeutic use. The radiometabolites characterized give new insights into the enzymatic degradation in vivo.


Subject(s)
Lutetium/metabolism , Peptides/metabolism , Radioisotopes/metabolism , Receptor, Cholecystokinin B/metabolism , Amino Acid Sequence , Amino Acid Substitution/physiology , Animals , Cell Line, Tumor , Female , Gastrins/metabolism , Humans , Mice , Mice, Inbred BALB C , Tissue Distribution/physiology
17.
Am J Physiol Regul Integr Comp Physiol ; 316(5): R628-R639, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30892908

ABSTRACT

We investigated expression of cholecystokinin (CCK) in humans and mice, and the bitter taste receptor TAS2R14 in the human placenta. Because CCK and gastrin activate the CCKBR receptor, we also explored placental gastrin expression. Finally, we investigated calcium signaling by CCK and TAS2R14. By RT-PCR, we found CCK/Cck and GAST/Gast mRNA expression in both normal human and mouse placentas, as well as in human trophoblast cell lines (TCL). Although both Cckar and -br mRNA were expressed in the mouse placenta, only CCKBR mRNA was detected in the human placenta and TCL. mRNA expression for TAS2R14 was also observed in the human placenta and TCL. Using immunohistochemistry, CCK protein was localized to the syncytiotrophoblast (ST) and extravillous trophoblast (EVT) in the human term placenta, and to trophoblast glycogen cells in mouse and human placentas. Gastrin and TAS2R14 proteins were also observed in ST and EVT of the human placenta. Both sulfated and nonsulfated CCK elicited a comparable rise in intracellular calcium in TCL, consistent with CCKBR expression. Three TAS2R14 agonists, flufenamic acid, chlorhexidine, and diphenhydramine, also evoked rises in intracellular calcium in TCL. These results establish CCK, gastrin, and their receptor(s) in both human and mouse placentas, and TAS2R14 in the human placenta. Both CCK and TAS2R14 agonists increased intracellular calcium in human TCL. Although the roles of these ligands and receptors, and their potential cross talk in normal and pathological placentas, are currently unknown, this study opens new avenues for placental research.


Subject(s)
Cholecystokinin/metabolism , Gastrins/metabolism , Receptor, Cholecystokinin B/metabolism , Receptors, Cholecystokinin/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Trophoblasts/metabolism , Animals , Calcium Signaling , Cell Line , Cholecystokinin/genetics , Cholecystokinin/pharmacology , Female , Gastrins/genetics , Gene Expression Regulation, Developmental , Humans , Ligands , Mice , Mice, Inbred C57BL , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Cholecystokinin B/genetics , Receptors, Cholecystokinin/agonists , Receptors, Cholecystokinin/genetics , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Trophoblasts/drug effects
18.
Bioconjug Chem ; 30(3): 657-666, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30608664

ABSTRACT

Derivatized minigastrin analogues make up a promising class of candidates for targeting cholecystokinin receptor subtype 2 (CCK2R), which is overexpressed on cancer cells of various neuroendocrine tumors. The pentaglutamic acid sequence of minigastrin influences its biological properties. In particular, it plays a crucial role in the kidney reuptake mechanism. However, the importance of the binding affinity and interaction of this region with the receptor on a molecular level remains unclear. To elucidate its structure-activity relationship with CCK2R, we replaced this sequence with various linkers differing in their amount of anionic charge, structural characteristics, and flexibility. Specifically, a flexible aliphatic linker, a linker with only three d-Glu residues, and a structured linker with four adjacent ß3-glutamic acid residues were evaluated and compared to the lead compound PP-F11N (DOTA-[d-Glu1-6,Nle11]gastrin-13). 1,4,7,10-Tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) was conjugated to the minigastrin derivatives, which allowed radiolabeling with Lutetium-177. The levels of In vitro internalization into MZ-CRC1 cells and in vivo tumor uptake as well as human blood plasma stability increased in the following order: aliphatic linker < three d-Glu < (ß3-Glu)4 < (d-Glu)6. The in vitro and in vivo behavior was therefore significantly improved with anionic charges. Computational modeling of a CCK2 receptor-ligand complex revealed ionic interactions between cationic residues (Arg and His) of the receptor and anionic residues of the ligand in the linker.


Subject(s)
Gastrins/chemistry , Gastrins/pharmacology , Receptor, Cholecystokinin B/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Drug Stability , Gastrins/pharmacokinetics , Humans , Mice , Molecular Docking Simulation , Structure-Activity Relationship , Tissue Distribution
20.
Cell Commun Signal ; 17(1): 68, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31215470

ABSTRACT

BACKGROUND: Cholecystokinin (CCK) is implicated in the regulation of nociceptive sensitivity of primary afferent neurons. Nevertheless, the underlying cellular and molecular mechanisms remain unknown. METHODS: Using patch clamp recording, western blot analysis, immunofluorescent labelling, enzyme-linked immunosorbent assays, adenovirus-mediated shRNA knockdown and animal behaviour tests, we studied the effects of CCK-8 on the sensory neuronal excitability and peripheral pain sensitivity mediated by A-type K+ channels. RESULTS: CCK-8 reversibly and concentration-dependently decreased A-type K+ channel (IA) in small-sized dorsal root ganglion (DRG) neurons through the activation of CCK type B receptor (CCK-BR), while the sustained delayed rectifier K+ current was unaffected. The intracellular subunit of CCK-BR coimmunoprecipitated with Gαo. Blocking G-protein signaling with pertussis toxin or by the intracellular application of anti-Gß antibody reversed the inhibitory effects of CCK-8. Antagonism of phosphatidylinositol 3-kinase (PI3K) but not of its common downstream target Akts abolished the CCK-BR-mediated IA response. CCK-8 application significantly activated JNK mitogen-activated protein kinase. Antagonism of either JNK or c-Src prevented the CCK-BR-mediated IA decrease, whereas c-Src inhibition attenuated the CCK-8-induced p-JNK activation. Application of CCK-8 enhanced the action potential firing rate of DRG neurons and elicited mechanical and thermal pain hypersensitivity in mice. These effects were mediated by CCK-BR and were occluded by IA blockade. CONCLUSION: Our findings indicate that CCK-8 attenuated IA through CCK-BR that is coupled to the Gßγ-dependent PI3K and c-Src-mediated JNK pathways, thereby enhancing the sensory neuronal excitability in DRG neurons and peripheral pain sensitivity in mice.


Subject(s)
CSK Tyrosine-Protein Kinase/metabolism , MAP Kinase Signaling System/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Potassium Channel Blockers/pharmacology , Receptor, Cholecystokinin B/metabolism , Sensory Receptor Cells/cytology , Sensory Receptor Cells/drug effects , Animals , Ganglia, Spinal/cytology , Male , Mice , Mice, Inbred ICR , Nociception/drug effects , Pain/pathology , Pain/physiopathology , Sincalide/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL