Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Med Chem ; 64(8): 4841-4856, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33826325

ABSTRACT

Proteins adopt unique folded secondary and tertiary structures that are responsible for their remarkable biological properties. This structural complexity is key in designing efficacious peptides that can mimic the three-dimensional structure needed for biological function. In this study, we employ different chemical strategies to induce and stabilize a ß-hairpin fold of peptides targeting cholecystokinin-2 receptors for theranostic application (combination of a targeted therapeutic and a diagnostic companion). The newly developed peptides exhibited enhanced folding capacity as demonstrated by circular dichroism (CD) spectroscopy, ion-mobility spectrometry-mass spectrometry, and two-dimensional (2D) NMR experiments. Enhanced folding characteristics of the peptides led to increased biological potency, affording four optimal Ga-68 labeled radiotracers ([68Ga]Ga-4b, [68Ga]Ga-11b-13b) targeting CCK-2R. In particular, [68Ga]Ga-12b and [68Ga]Ga-13b presented improved metabolic stability, enhanced cell internalization, and up to 6 fold increase in tumor uptake. These peptides hold great promise as next-generation theranostic radiopharmaceuticals.


Subject(s)
Neoplasms/diagnosis , Peptides/chemistry , Radiopharmaceuticals/chemistry , Receptor, Cholecystokinin B/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Gallium Radioisotopes/chemistry , Humans , Mice , Mice, Nude , Neoplasms/pathology , Peptides/chemical synthesis , Peptides/metabolism , Positron Emission Tomography Computed Tomography , Precision Medicine , Protein Binding , Protein Structure, Tertiary , Radiopharmaceuticals/metabolism , Receptor, Cholecystokinin B/chemistry , Tissue Distribution , Transplantation, Heterologous
2.
Nanoscale ; 11(29): 13714-13719, 2019 Aug 07.
Article in English | MEDLINE | ID: mdl-31314031

ABSTRACT

The selective formation of nanomedicines around cancer cells is very important for cancer therapy because it increases the inhibitory capacity and decreases the systemic toxicity. However, successful examples are rare. Taking advantage of the overexpression of both the enzyme alkaline phosphatase (ALP) and the cell membrane receptor (CCK2R), we demonstrated in this study the selective formation of supramolecular nanofibers and hydrogels in the pericellular space of two cancer cell lines (HeLa and HepG2 cells). Both cell lines showed high expression levels of extracellular ALP and membrane-bound CCK2R. ALP efficiently converted Comp. 1 to a self-assembling molecule (Comp. 2). Comp. 2 interacted with CCK2R, thereby facilitating the self-assembly and formation of hydrogels around the cancer cells. The selective pericellular hydrogelations efficiently inhibited cancer cells. Pericellular hydrogelation around cancer cells is a promising strategy to control the formation of nanomedicines spatiotemporally in cellular microenvironments for cancer therapy and diagnostics.


Subject(s)
Alkaline Phosphatase/metabolism , Hydrogels/chemistry , Receptor, Cholecystokinin B/metabolism , Alkaline Phosphatase/chemistry , Alkaline Phosphatase/genetics , Cell Line, Tumor , Cell Survival/drug effects , Humans , Microscopy, Confocal , Peptides/chemistry , Peptides/pharmacology , Receptor, Cholecystokinin B/chemistry , Receptor, Cholecystokinin B/genetics
3.
Daru ; 27(1): 21-34, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30607886

ABSTRACT

BACKGROUND: As a membrane G protein coupled receptors (GPCRs) family, gastrin/cholecystokinin-2 receptor (CCK2R) plays a key role in the initiation and development of gastric cancer. OBJECTIVES: Targeting CCK2R by immunotherapeutics such as single-chain variable fragments (scFvs) may provide an effective treatment modality against gastric cancer. Thus, the main objective of this study was to isolate scFvs specific to CCK2R. METHODS: To isolate scFvs specific to the CCK2R, we capitalized on a semi-synthetic diverse phage antibody library (PAL) and a solution-phase biopanning process. The library was panned against a biotinylated peptide of the second extracellular loop (ECL2) of CCK2R. After four rounds of biopanning, the selected soluble scFv clones were screened by enzyme-linked immunosorbent assay (ELISA) and examined for specific binding to the peptide. The selected scFvs were purified using immobilized metal affinity chromatography (IMAC). The binding affinity and specificity of the scFvs were examined by the surface plasmon resonance (SPR), immunoblotting and flow cytometry assays and molecular docking using ZDOCK v3.0.2. RESULTS: Ten different scFvs were isolated, which displayed binding affinity ranging from 0.68 to 8.0 (nM). Immunoblotting and molecular docking analysis revealed that eight scFvs were able to detect the denatured form of CCK2R protein. Of the isolated scFvs, two scFvs showed high-binding affinity to the human gastric adenocarcinoma AGS cells. CONCLUSIONS: Based on our findings, a couple of the selected scFvs showed markedly high-binding affinity to immobilized CCK2R peptide and CCK2R-overexpressing AGS cells. Therefore, these scFvs are proposed to serve as targeting and/or treatment agents in the diagnosis and immunotherapy of CCK2R-positive tumors. Graphical abstract ᅟ.


Subject(s)
Adenocarcinoma/metabolism , Receptor, Cholecystokinin B/chemistry , Receptor, Cholecystokinin B/immunology , Single-Chain Antibodies/pharmacology , Stomach Neoplasms/metabolism , Adenocarcinoma/drug therapy , Cell Line, Tumor , Drug Screening Assays, Antitumor , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunotherapy , Models, Molecular , Molecular Docking Simulation , Peptide Library , Protein Structure, Tertiary , Single-Chain Antibodies/chemistry , Stomach Neoplasms/drug therapy , Surface Plasmon Resonance , Up-Regulation/drug effects
4.
Q J Nucl Med Mol Imaging ; 59(3): 287-302, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26158215

ABSTRACT

Cholecystokinin subtype 2 receptors (CCK2R) are overexpressed in several human cancers, including medullary thyroid carcinoma. Gastrin and cholecystokinin (CCK) peptides that bind with high affinity and specificity to CCK2R can be used as carriers of radioactivity to CCK2R-expressing tumor sites. Several gastrin and CCK related peptides have been proposed for diagnostic imaging and radionuclide therapy of primary and metastatic CCK2R-positive human tumors. Their clinical application has been restricted to a great extent by their fast in vivo degradation that eventually compromises tumor uptake. This problem has been addressed by structural modifications of gastrin and CCK motifs, which, however, often lead to suboptimal pharmacokinetic profiles. A major enzyme implicated in the catabolism of gastrin and CCK based peptides is neutral endopeptidase (NEP), which is widely distributed in the body. Coinjection of the NEP inhibitor phosphoramidon (PA) with radiolabeled gastrin and other peptide analogs has been recently proposed as a new promising strategy to increase bioavailability and tumor-localization of radiopeptides in tumor sites. Specifically, co-administration of PA with the truncated gastrin analog [(111)In-DOTA]MG11 ([((111)In-DOTA)DGlu(10)]gastrin(10-17)) impressively enhanced the levels of intact radiopeptide in mouse circulation and has led to an 8-fold increase of CCK2R-positive tumor uptake in SCID mice. This increased tumor uptake, visualized also by SPECT/CT imaging, is expected to eventually translate into higher diagnostic sensitivity and improved therapeutic efficacy of radiolabeled gastrin analogs in CCK2R-expressing cancer patients.


Subject(s)
Gastrins/chemistry , Neoplasms/diagnostic imaging , Neoplasms/diagnosis , Radiopharmaceuticals/chemistry , Receptor, Cholecystokinin B/chemistry , Animals , Carcinoma, Neuroendocrine/diagnostic imaging , Cholecystokinin/chemistry , Gene Expression Regulation, Neoplastic , Glycopeptides/chemistry , Humans , Kidney/diagnostic imaging , Kidney Neoplasms/diagnostic imaging , Ligands , Mice , Mice, SCID , Models, Chemical , Neoplasm Transplantation , Neprilysin/chemistry , Peptides/chemistry , Thyroid Neoplasms/diagnostic imaging , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed
5.
Bioorg Med Chem ; 21(17): 5029-38, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23890524

ABSTRACT

Probes for use in time-resolved fluorescence competitive binding assays at melanocortin receptors based on the parental ligands MSH(4), MSH(7), and NDP-α-MSH were prepared by solid phase synthesis methods, purified, and characterized. The saturation binding of these probes was studied using HEK-293 cells engineered to overexpress the human melanocortin 4 receptor (hMC4R) as well as the human cholecystokinin 2 receptor (hCCK2R). The ratios of non-specific binding to total binding approached unity at high concentrations for each probe. At low probe concentrations, receptor-mediated binding and uptake was discernable, and so probe concentrations were kept as low as possible in determining Kd values. The Eu-DTPA-PEGO-MSH(4) probe exhibited low specific binding relative to non-specific binding, even at low nanomolar concentrations, and was deemed unsuitable for use in competition binding assays. The Eu-DTPA-PEGO probes based on MSH(7) and NDP-α-MSH exhibited Kd values of 27±3.9nM and 4.2±0.48nM, respectively, for binding with hMC4R. These probes were employed in competitive binding assays to characterize the interactions of hMC4R with monovalent and divalent MSH(4), MSH(7), and NDP-α-MSH constructs derived from squalene. Results from assays with both probes reflected only statistical enhancements, suggesting improper ligand spacing on the squalene scaffold for the divalent constructs. The Ki values from competitive binding assays that employed the MSH(7)-based probe were generally lower than the Ki values obtained when the probe based on NDP-α-MSH was employed, which is consistent with the greater potency of the latter probe. The probe based on MSH(7) was also competed with monovalent, divalent, and trivalent MSH(4) constructs that previously demonstrated multivalent binding in competitive binding assays against a variant of the probe based on NDP-α-MSH. Results from these assays confirm multivalent binding, but suggest a more modest increase in avidity for these MSH(4) constructs than was previously reported.


Subject(s)
Fluorescent Dyes/chemical synthesis , Receptors, Melanocortin/metabolism , Binding, Competitive , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , HEK293 Cells , Humans , Pentetic Acid/chemistry , Peptides/chemical synthesis , Peptides/chemistry , Peptides/metabolism , Protein Binding , Receptor, Cholecystokinin B/chemistry , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism , Receptor, Melanocortin, Type 4/chemistry , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Receptors, Melanocortin/chemistry , Solid-Phase Synthesis Techniques
6.
J Biol Chem ; 288(29): 21082-21095, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23754289

ABSTRACT

Understanding the molecular basis of drug action can facilitate development of more potent and selective drugs. Here, we explore the molecular basis for action of a unique small molecule ligand that is a type 1 cholecystokinin (CCK) receptor agonist and type 2 CCK receptor antagonist, GI181771X. We characterize its binding utilizing structurally related radioiodinated ligands selective for CCK receptor subtypes that utilize the same allosteric ligand-binding pocket, using wild-type receptors and chimeric constructs exchanging the distinct residues lining this pocket. Intracellular calcium assays were performed to determine biological activity. Molecular models for docking small molecule agonists to the type 1 CCK receptor were developed using a ligand-guided refinement approach. The optimal model was distinct from the previous antagonist model for the same receptor and was mechanistically consistent with the current mutagenesis data. This study revealed a key role for Leu(7.39) that was predicted to interact with the isopropyl group in the N1 position of the benzodiazepine that acts as a "trigger" for biological activity. The molecular model was predictive of binding of other small molecule agonists, effectively distinguishing these from 1065 approved drug decoys with an area under curve value of 99%. The model also selectively enriched for agonist compounds, with 130 agonists identified by ROC analysis when seeded in 2175 non-agonist ligands of the type 1 CCK receptor (area under curve 78%). Benzodiazepine agonists in this series docked in consistent pose within this pocket, with a key role played by Leu(7.39), whereas the role of this residue was less clear for chemically distinct agonists.


Subject(s)
Benzodiazepines/pharmacology , Receptor, Cholecystokinin A/agonists , Amino Acid Sequence , Animals , Benzodiazepines/chemistry , CHO Cells , Cricetinae , Cricetulus , Models, Molecular , Molecular Sequence Data , Mutant Proteins/agonists , Mutant Proteins/chemistry , Mutant Proteins/metabolism , ROC Curve , Receptor, Cholecystokinin A/chemistry , Receptor, Cholecystokinin A/metabolism , Receptor, Cholecystokinin B/chemistry , Receptor, Cholecystokinin B/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment
7.
J Am Chem Soc ; 135(7): 2560-73, 2013 Feb 20.
Article in English | MEDLINE | ID: mdl-23323542

ABSTRACT

Seven-transmembrane receptors (7TMRs), also termed G protein-coupled receptors (GPCRs), form the largest class of cell surface membrane receptors, involving several hundred members in the human genome. Nearly 30% of marketed pharmacological agents target 7TMRs. 7TMRs adopt multiple conformations upon agonist binding. Biased agonists, in contrast to non-biased agonists, are believed to stabilize conformations preferentially activating either G-protein- or ß-arrestin-dependent signaling pathways. However, proof that cognate conformations of receptors display structural differences within their binding site where biased agonism initiates, are still lacking. Here, we show that a non-biased agonist, cholecystokinin (CCK) induces conformational states of the CCK2R activating Gq-protein-dependent pathway (CCK2R(G)) or recruiting ß-arrestin2 (CCK2R(ß)) that are pharmacologically and structurally distinct. Two structurally unrelated antagonists competitively inhibited both pathways. A third ligand (GV150013X) acted as a high affinity competitive antagonist on CCK2R(G) but was nearly inefficient as inhibitor of CCK2R(ß). Several structural elements on both GV150013X and in CCK2R binding cavity, which hinder binding of GV150013X only to the CCK2R(ß) were identified. At last, proximity between two conserved amino acids from transmembrane helices 3 and 7 interacting through sulfur-aromatic interaction was shown to be crucial for selective stabilization of the CCK2R(ß) state. These data establish structural evidence for distinct conformations of a 7TMR associated with ß-arrestin-2 recruitment or G-protein coupling and validate relevance of the design of biased ligands able to selectively target each functional conformation of 7TMRs.


Subject(s)
Arrestins/chemistry , Receptor, Cholecystokinin B/chemistry , Type C Phospholipases/metabolism , Adamantane/analogs & derivatives , Adamantane/pharmacology , Binding Sites , Enzyme Activation/drug effects , Humans , Microscopy, Confocal , Models, Molecular , Molecular Structure , Mutation , Phenylurea Compounds/pharmacology , Protein Conformation , Receptor, Cholecystokinin B/antagonists & inhibitors , Receptor, Cholecystokinin B/genetics , Signal Transduction , Type C Phospholipases/chemistry , Up-Regulation , beta-Arrestin 2 , beta-Arrestins
8.
J Chem Inf Model ; 53(1): 176-87, 2013 Jan 28.
Article in English | MEDLINE | ID: mdl-23240656

ABSTRACT

The present study revisited the three-dimensional (3D) homology model of CCK-2R using human A(2a) adenosine receptor and the resolved NMR based structure of the third extracellular loop of the CCK-2R as templates. Further in order to identify novel antiulcer agents, rational designing have been performed utilizing the substructure of a well-known CCK-2R antagonist benzotript as a lead molecule and submitted to the combined docking and simulation studies. This led to the understanding of the essential structure requirement as well as variation of binding mode among conformational isomers of small molecule CCK-2R antagonists. In the next step, preparation of each configurational isomer of these molecules was carried out and submitted for their in vitro activity followed by in vivo screening into antiulcer rat model. The biological screening of these compounds has not only validated the developed homology model of CCK-2R but also led to the identification of highly potent CCK-2R antagonist 6a as an orally active and safe candidate molecule having better antiulcer properties than the well-known drug benzotript.


Subject(s)
Amino Acids/pharmacology , Drug Design , Molecular Docking Simulation , Receptor, Cholecystokinin B/antagonists & inhibitors , Receptor, Cholecystokinin B/metabolism , Sequence Homology, Amino Acid , Stomach Ulcer/drug therapy , Amino Acid Sequence , Amino Acids/chemical synthesis , Amino Acids/metabolism , Amino Acids/therapeutic use , Animals , Cattle , Chemistry Techniques, Synthetic , Humans , Molecular Sequence Data , Protein Conformation , Rats , Receptor, Cholecystokinin B/chemistry
9.
Mol Cell Endocrinol ; 349(2): 170-9, 2012 Feb 26.
Article in English | MEDLINE | ID: mdl-22040601

ABSTRACT

The cholecystokinin-2 receptor (CCK2R), is expressed in cancers where it contributes to tumor progression. The CCK2R is over-expressed in a sub-set of tumors, allowing its use in tumor targeting with a radiolabel ligand. Since discrepancies between mRNA levels and CCK2R binding sites were noticed, we searched for abnormally spliced variants in tumors from various origins having been previously reported to frequently express cholecystokinin receptors, such as medullary thyroid carcinomas, gastrointestinal stromal tumors, leiomyomas and leiomyosarcomas, and gastroenteropancreatic tumors. A variant of the CCK2R coding for a putative five-transmembrane domains receptor has been cloned. This variant represented as much as 6% of CCK2R levels. Ectopic expression in COS-7 cells revealed that this variant lacks biological activity due to its sequestration in endoplasmic reticulum. When co-expressed with the CCK2R, this variant diminished membrane density of the CCK2R and CCK2R-mediated activity (phospholipase-C and ERK activation). In conclusion, a novel splice variant acting as a dominant negative on membrane density of the CCK2R may be of importance for the pathophysiology of certain tumors and for their in vivo CCK2R-targeting.


Subject(s)
Alternative Splicing , Receptor, Cholecystokinin B/genetics , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Carcinoma, Medullary/metabolism , Chlorocebus aethiops , Cholecystokinin/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gastrins/metabolism , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Stromal Tumors/metabolism , Humans , Insulinoma/metabolism , Leiomyoma/metabolism , Leiomyosarcoma/metabolism , Molecular Sequence Data , Pancreatic Neoplasms/metabolism , Protein Structure, Tertiary , RNA, Messenger/biosynthesis , Receptor, Cholecystokinin B/chemistry , Receptor, Cholecystokinin B/metabolism , Signal Transduction , Thyroid Neoplasms/metabolism , Type C Phospholipases/metabolism
10.
Amino Acids ; 41(5): 1049-58, 2011 Nov.
Article in English | MEDLINE | ID: mdl-20198494

ABSTRACT

Cholecystokinin (CCK) receptors are overexpressed in numerous human cancers, like medullary thyroid carcinomas, small cell lung cancers and stromal ovarian cancers. The specific receptor-binding property of the endogenous ligands for these receptors can be exploited by labeling peptides with a radionuclide and using these as carriers to guide the radioactivity to the tissues that express the receptors. In this way, tumors can be visualized using positron emission tomography and single photon emission computed tomography imaging. A variety of radiolabeled CCK/gastrin-related peptides has been synthesized and characterized for imaging. All peptides have the C-terminal CCK receptor-binding tetrapeptide sequence Trp-Met-Asp-Phe-NH(2) in common or derivatives thereof. This review focuses on the development and application of radiolabeled CCK/gastrin peptides for radionuclide imaging and radionuclide therapy of tumors expressing CCK receptors. We discuss both preclinical studies as well as clinical studies with CCK and gastrin peptides.


Subject(s)
Cholecystokinin , Gastrins , Neoplasms/diagnostic imaging , Neoplasms/radiotherapy , Receptor, Cholecystokinin B/genetics , Animals , Cholecystokinin/chemistry , Cholecystokinin/genetics , Gastrins/chemistry , Gastrins/genetics , Humans , Isotope Labeling , Neoplasms/genetics , Neoplasms/metabolism , Radioisotopes/chemistry , Radioisotopes/therapeutic use , Radionuclide Imaging , Receptor, Cholecystokinin B/chemistry , Receptor, Cholecystokinin B/metabolism
11.
Bioorg Med Chem ; 18(14): 5400-12, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20542702

ABSTRACT

To improve the targeting to tumors expressing the cholecystokinin receptor subtype 2 (CCK2R) with limited kidney uptake, we synthesized a novel cholecystokinin C-terminal tetrapeptide (CCK4)-based derivative conjugated to an original bipyridine-chelator (BPCA), 111In-BPCA-(Ahx)2-CCK4. To our knowledge this is the first CCK4-based radioligand that presents a high affinity for the CCK2R, a high and specific tumor uptake, a low renal accumulation and a very good visualization of tumors in vivo compared with an internal control, 111Indium-trans-cyclohexyldiethylenetriaminepenta-acetic acid-cholecystokinin octapeptide (111In-CHX-A''-DTPA-CCK8). These properties make 111In-BPCA-(Ahx)2-CCK4, a promising candidate for imaging and peptide receptor radionuclide therapy of CCK2R positive tumors.


Subject(s)
Neoplasms/diagnostic imaging , Oligopeptides , Radionuclide Imaging/methods , Radiopharmaceuticals , Receptor, Cholecystokinin B/metabolism , Animals , COS Cells , Chlorocebus aethiops , Humans , Mice , Mice, Nude , Models, Molecular , NIH 3T3 Cells , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Oligopeptides/pharmacology , Pentetic Acid/analogs & derivatives , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/pharmacology , Receptor, Cholecystokinin B/chemistry
12.
ACS Nano ; 4(3): 1279-87, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20180585

ABSTRACT

The early diagnosis of cancer is the critical element in successful treatment and long-term favorable patient prognoses. The high rate of mortality is mainly attributed to the tendency for late diagnoses as symptoms may not occur until the disease has metastasized, as well as the lack of effective systemic therapies. Late diagnosis is often associated with the lack of timely sensitive imaging modalities. The promise of nanotechnology is presently limited by the inability to simultaneously seek, treat, and image cancerous lesions. This study describes the design and synthesis of fluorescent calcium phosphosilicate nanocomposite particles (CPNPs) that can be systemically targeted to breast and pancreatic cancer lesions. The CPNPs are a approximately 20 nm diameter composite composed of an amorphous calcium phosphate matrix doped with silicate in which a near-infrared imaging agent, indocyanine green (ICG), is embedded. In the present studies, we describe and validate CPNP bioconjugation of human holotransferrin, anti-CD71 antibody, and short gastrin peptides via an avidin-biotin or a novel PEG-maleimide coupling strategy. The conjugation of biotinylated human holotransferrin (diferric transferrin) and biotinylated anti-CD71 antibody (anti-transferrin receptor antibody) to avidin-conjugated CPNPs (Avidin-CPNPs) permits targeting of transferrin receptors, which are highly expressed on breast cancer cells. Similarly, the conjugation of biotinylated pentagastrin to Avidin-CPNPs and decagastrin (gastrin-10) to PEG-CPNPs via PEG-maleimide coupling permits targeting of gastrin receptors, which are overexpressed in pancreatic cancer lesions. These bioconjugated CPNPs have the potential to perform as a theranostic modality, simultaneously enhancing drug delivery, targeting, and imaging of breast and pancreatic cancer tumors.


Subject(s)
Breast Neoplasms/metabolism , Nanoparticles/chemistry , Pancreatic Neoplasms/metabolism , Silicates/chemistry , Animals , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic , Drug Design , Humans , Mice , Organ Specificity , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Receptor, Cholecystokinin B/chemistry , Receptor, Cholecystokinin B/metabolism
13.
PLoS One ; 5(12): e15999, 2010 Dec 30.
Article in English | MEDLINE | ID: mdl-21209861

ABSTRACT

Fear, an emotional response of animals to environmental stress/threats, plays an important role in initiating and driving adaptive response, by which the homeostasis in the body is maintained. Overwhelming/uncontrollable fear, however, represents a core symptom of anxiety disorders, and may disturb the homeostasis. Because to recall or imagine certain cue(s) of stress/threats is a compulsory inducer for the expression of anxiety, it is generally believed that the pathogenesis of anxiety is associated with higher attention (acquisition) selectively to stress or mal-enhanced fear memory, despite that the actual relationship between fear memory and anxiety is not yet really established. In this study, inducible forebrain-specific cholecystokinin receptor-2 transgenic (IF-CCKR-2 tg) mice, different stress paradigms, batteries of behavioral tests, and biochemical assays were used to evaluate how different CCKergic activities drive fear behavior and hormonal reaction in response to stresses with different intensities. We found that in IF-CCKR-2 tg mice, contextual fear was impaired following 1 trial of footshock, while overall fear behavior was enhanced following 36 trials of footshock, compared to their littermate controls. In contrast to a standard Yerkes-Dodson (inverted-U shaped) stress-fear relationship in control mice, a linearized stress-fear curve was observed in CCKR-2 tg mice following gradient stresses. Moreover, compared to 1 trial, 36 trials of footshock in these transgenic mice enhanced anxiety-like behavior in other behavioral tests, impaired spatial and recognition memories, and prolonged the activation of adrenocorticotropic hormone (ACTH) and glucocorticoids (CORT) following new acute stress. Taken together, these results indicate that stress may trigger two distinctive neurobehavioral systems, depending on both of the intensity of stress and the CCKergic tone in the brain. A "threshold theory" for this two-behavior system has been suggested.


Subject(s)
Anxiety/metabolism , Fear , Memory , Receptor, Cholecystokinin B/chemistry , Adrenocorticotropic Hormone/metabolism , Amygdala/metabolism , Animals , Glucocorticoids/metabolism , Hippocampus/metabolism , Hypothalamo-Hypophyseal System , Maze Learning , Mice , Mice, Transgenic , Prosencephalon/metabolism , Stress, Psychological/metabolism
14.
Anal Biochem ; 398(1): 15-23, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-19852924

ABSTRACT

Lanthanide-based luminescent ligand binding assays are superior to traditional radiolabel assays due to improving sensitivity and affordability in high-throughput screening while eliminating the use of radioactivity. Despite significant progress using lanthanide(III)-coordinated chelators such as diethylenetriaminepentaacetic acid (DTPA) derivatives, dissociation-enhanced lanthanide fluoroimmunoassays (DELFIAs) have not yet been successfully used with more stable chelators (e.g., tetraazacyclododecyltetraacetic acid [DOTA] derivatives) due to the incomplete release of lanthanide(III) ions from the complex. Here a modified and optimized DELFIA procedure incorporating an acid treatment protocol is introduced for use with Eu(III)-DOTA-labeled peptides. Complete release of Eu(III) ions from DOTA-labeled ligands was observed using hydrochloric acid (2.0M) prior to the luminescent enhancement step. [Nle(4),d-Phe(7)]-alpha-melanocyte-stimulating hormone (NDP-alpha-MSH) labeled with Eu(III)-DOTA was synthesized, and the binding affinity to cells overexpressing the human melanocortin-4 (hMC4) receptor was evaluated using the modified protocol. Binding data indicate that the Eu(III)-DOTA-linked peptide bound to these cells with an affinity similar to its DTPA analogue. The modified DELFIA procedure was further used to monitor the binding of an Eu(III)-DOTA-labeled heterobivalent peptide to the cells expressing both hMC4 and cholecystokinin-2 (CCK-2) receptors. The modified assay provides superior results and is appropriate for high-throughput screening of ligand libraries.


Subject(s)
Europium/chemistry , Fluorescent Dyes/chemistry , Fluoroimmunoassay/methods , Heterocyclic Compounds, 1-Ring/chemistry , Ligands , Receptors, Cell Surface/analysis , Amino Acid Sequence , Cell Line , High-Throughput Screening Assays , Humans , Lanthanoid Series Elements/chemistry , Melanocyte-Stimulating Hormones/chemistry , Melanocyte-Stimulating Hormones/metabolism , Receptor, Cholecystokinin B/chemistry , Receptor, Cholecystokinin B/metabolism , Receptor, Melanocortin, Type 4/chemistry , Receptor, Melanocortin, Type 4/metabolism , Staining and Labeling , Time Factors
15.
Mol Pharmacol ; 75(3): 502-13, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19064631

ABSTRACT

Signaling of G protein-coupled receptors (GPCRs) is regulated by different mechanisms. One of these involves regulators of G protein signaling (RGS), which are diverse and multifunctional proteins that bind to active Galpha subunits of G proteins and act as GTPase-activating proteins. Little is known about the molecular mechanisms that govern the selective use of RGS proteins in living cells. We first demonstrated that CCK2R-mediated inositol phosphate production, known to be G(q)-dependent, is more sensitive to RGS2 than to RGS4 and is insensitive to RGS8. Both basal and agonist-stimulated activities of the CCK2R are regulated by RGS2. By combining biochemical, functional, and in silico structural approaches, we demonstrate that a direct and functional interaction occurs between RGS2 and agonist-stimulated cholecystokinin receptor-2 (CCK2R) and identified the precise residues involved: phosphorylated Ser434 and Thr439 located in the C-terminal tail of CCK2R and Lys62, Lys63, and Gln67, located in the N-terminal domain of RGS2. These findings confirm previous reports that RGS proteins can interact with GPCRs to modulate their signaling and provide a molecular basis for RGS2 recognition by the CCK2R.


Subject(s)
Peptide Fragments/physiology , RGS Proteins/physiology , Receptor, Cholecystokinin B/physiology , Signal Transduction/physiology , Amino Acid Motifs/physiology , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Humans , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation/physiology , Protein Binding/physiology , RGS Proteins/chemistry , RGS Proteins/metabolism , Receptor, Cholecystokinin B/chemistry , Receptor, Cholecystokinin B/metabolism
16.
J Biol Chem ; 283(51): 35860-8, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-18936102

ABSTRACT

Given the importance of G-protein-coupled receptors as pharmacological targets in medicine, efforts directed at the understanding the molecular mechanism by which pharmacological compounds regulate their activity is of paramount importance. Here, we investigated at an atomic level the mechanism of inverse agonism and partial agonism of two high affinity, high selectivity very similar non-peptide ligands of the cholecystokinin-2 receptor (CCK2R) which differ by the absence or presence of a methyl group on their indole moiety. Using in silico, site-directed mutagenesis and pharmacological experiments, we demonstrated that these functionally different activities are due to differing anchoring modes of the two compounds to a residue of helix II (Thr-2.61) in the inactive state of the CCK2R. The binding mode of the inverse agonist allows the ligand to interact through its phenyl moiety with a key amino acid for CCK2R activation (Trp-6.48), preventing rotation of helix VI and, thus, CCK2R activation, whereas the partial agonist binds deeper into the binding pocket and closer to helix V, so that CCK2R activation is favored. This study on the molecular mechanism of ligand action opens the possibility of target-based optimization of G protein-coupled receptor non-peptide ligands.


Subject(s)
Ligands , Models, Molecular , Receptor, Cholecystokinin B/agonists , Receptor, Cholecystokinin B/chemistry , Binding Sites/physiology , Humans , Protein Structure, Secondary/physiology
17.
J Med Chem ; 51(3): 565-73, 2008 Feb 14.
Article in English | MEDLINE | ID: mdl-18201065

ABSTRACT

Cholecystokinin 2 receptor antagonists encompass a wide range of structures. This makes them unsuitable candidates for existing 3D-QSAR methods and has led us to develop an alternative approach to account for their observed biological activities. A diverse set of 21 antagonists was subjected to a novel molecular field-based similarity analysis. The hypothesis is that compounds with similar field patterns will bind at the same target site regardless of their underlying structure. This initial report demonstrates a linear correlation between ligand similarity and biological activity for this challenging data set. A model generated with three molecules was used to predict the activity of 18 test compounds, with different chemotypes, with a root-mean-square error of 0.68 pKB units. The ability to automatically derive a molecular alignment without knowledge of the protein structure represents an improvement over existing pharmacophore methods and makes the method particularly suitable for scaffold-hopping.


Subject(s)
Quantitative Structure-Activity Relationship , Receptor, Cholecystokinin B/antagonists & inhibitors , Receptor, Cholecystokinin B/chemistry , Animals , Benzoates/chemistry , Benzoates/pharmacology , Binding Sites , Binding, Competitive , Bridged-Ring Compounds/chemistry , Bridged-Ring Compounds/pharmacology , Cyclohexanes/chemistry , Cyclohexanes/pharmacology , Gastric Mucosa/metabolism , Guinea Pigs , Heterocyclic Compounds, 2-Ring/chemistry , Heterocyclic Compounds, 2-Ring/pharmacology , Hydrophobic and Hydrophilic Interactions , In Vitro Techniques , Least-Squares Analysis , Ligands , Linear Models , Models, Molecular , Pancreas/cytology , Phthalic Acids/chemistry , Phthalic Acids/pharmacology , Radioligand Assay , Rats , Receptor, Cholecystokinin A/antagonists & inhibitors
18.
Regul Pept ; 145(1-3): 17-23, 2008 Jan 10.
Article in English | MEDLINE | ID: mdl-17961734

ABSTRACT

CCK receptors represent potential targets in a number of diseases. Knowledge of CCK receptor binding sites is a prerequisite for the understanding of the molecular basis for their ligand recognition, partial agonism, ligand-induced trafficking of signalling. In the current paper, we report studies from our laboratory and others which have provided new data on the molecular basis of the pharmacology and functioning of CCK1 and CCK2 receptors. It has been shown that: 1) homologous regions of the two receptors are involved in the binding site of CCK, however, positioning of CCK slightly differs in agreement with distinct pharmacophores of CCK toward the two receptors and receptor sequence variations; 2) Binding sites of most of non-peptide agonists/ antagonist are buried in the pocket formed by transmembrane helices and overlap that of CCK; Aromatic amino acids within and near the binding site, especially in helix VI, are involved in receptor activation; 4) Like for other members of family A of G-protein coupled receptors, residues of the binding sites as well as of conserved motifs such as E/DRY, NPXXY are crucial for receptor activation.


Subject(s)
Cholecystokinin/metabolism , Gastrins/metabolism , Receptor, Cholecystokinin B/metabolism , Receptors, Cholecystokinin/metabolism , Animals , Binding Sites , Humans , Receptor, Cholecystokinin B/chemistry , Receptors, Cholecystokinin/chemistry
19.
Peptides ; 28(11): 2211-22, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17950490

ABSTRACT

A comparison of the conformational characteristics of the related hormones [Nle(15)] gastrin-17 and [Tyr(9)-SO(3)] cholecystokinin-15, in membrane-mimetic solutions of dodecylphosphocholine micelles and water, was undertaken using NMR spectroscopy to investigate the possibility of a structural motif responsible for the two hormones common ability to stimulate the CCK(2) receptor. Distance geometry calculations and NOE-restrained molecular dynamics simulations in biphasic solvent boxes of decane and water pointed to the two peptides adopting near identical helical C-terminal configurations, which extended one residue further than their shared pentapeptide sequence of Gly-Trp-Met-Asp-Phe-NH(2). The C-terminal conformation of [Nle(15)] gastrin-17 contained a short alpha-helix spanning the Ala(11)-Trp(14) sequence and an inverse gamma-turn centered on Nle(15) while that of [Tyr(9)-SO(3)] cholecystokinin-15 contained a short 3(10) helix spanning its Met(10) to Met(13) sequence and an inverse gamma-turn centered on Asp(14). Significantly, both the C-terminal helices were found to terminate in type I beta-turns spanning the homologous Gly-Trp-Met-Asp sequences. This finding supports the hypothesis that this structural motif is a necessary condition for CCK(2) receptor activation given that both gastrin and cholecystokinin have been established to follow a membrane-associated pathway to receptor recognition and activation. Comparison of the conformations for the non-homologous C-terminal tyrosyl residues of [Nle(15)] gastrin-17 and [Tyr(9)-SO(3)] cholecystokinin-15 found that they lie on opposite faces of the conserved C-terminal helices. The positioning of this tyrosyl residue is known to be essential for CCK(1) activity and non-essential for CCK(2) activity, pointing to it as a possible differentiator in CCK(1)/CCK(2) receptor selection. The different tyrosyl orientations were retained in molecular models for the [Nle(15)] gastrin-17/CCK(2) receptor and [Tyr(9)-SO(3)] cholecystokinin-15/CCK(1) receptor complexes, highlighting the role of this residue as a likely CCK(1)/CCK(2) receptor differentiator.


Subject(s)
Amino Acid Motifs , Gastrins/chemistry , Receptor, Cholecystokinin B/chemistry , Amino Acid Sequence , Cholecystokinin/chemistry , Cholecystokinin/metabolism , Gastrins/metabolism , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/chemistry , Phosphorylcholine/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Receptor, Cholecystokinin B/metabolism
20.
J Biol Chem ; 282(39): 28779-28790, 2007 Sep 28.
Article in English | MEDLINE | ID: mdl-17599907

ABSTRACT

G protein-coupled receptors (GPCRs) represent a major focus in functional genomics programs and drug development research, but their important potential as drug targets contrasts with the still limited data available concerning their activation mechanism. Here, we investigated the activation mechanism of the cholecystokinin-2 receptor (CCK2R). The three-dimensional structure of inactive CCK2R was homology-modeled on the basis of crystal coordinates of inactive rhodopsin. Starting from the inactive CCK2R modeled structure, active CCK2R (namely cholecystokinin-occupied CCK2R) was modeled by means of steered molecular dynamics in a lipid bilayer and by using available data from other GPCRs, including rhodopsin. By comparing the modeled structures of the inactive and active CCK2R, we identified changes in the relative position of helices and networks of interacting residues, which were expected to stabilize either the active or inactive states of CCK2R. Using targeted molecular dynamics simulations capable of converting CCK2R from the inactive to the active state, we delineated structural changes at the atomic level. The activation mechanism involved significant movements of helices VI and V, a slight movement of helices IV and VII, and changes in the position of critical residues within or near the binding site. The mutation of key amino acids yielded inactive or constitutively active CCK2R mutants, supporting this proposed mechanism. Such progress in the refinement of the CCK2R binding site structure and in knowledge of CCK2R activation mechanisms will enable target-based optimization of nonpeptide ligands.


Subject(s)
Computer Simulation , Models, Molecular , Receptor, Cholecystokinin B/metabolism , Amino Acid Substitution , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Cholecystokinin , Drug Design , Genomics , Humans , Lipid Bilayers , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Receptor, Cholecystokinin B/chemistry , Receptor, Cholecystokinin B/genetics , Rhodopsin , Structural Homology, Protein , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...