Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Molecules ; 27(19)2022 Oct 05.
Article in English | MEDLINE | ID: mdl-36235124

ABSTRACT

COX-2 can be considered as a clinically relevant molecular target for adjuvant, in particular radiosensitizing treatments. In this regard, using selective COX-2 inhibitors, e.g., in combination with radiotherapy or endoradiotherapy, represents an interesting treatment option. Based on our own findings that nitric oxide (NO)-releasing and celecoxib-derived COX-2 inhibitors (COXIBs) showed promising radiosensitizing effects in vitro, we herein present the development of a series of eight novel NO-COXIBs differing in the peripheral substitution pattern and their chemical and in vitro characterization. COX-1 and COX-2 inhibition potency was found to be comparable to the lead NO-COXIBs, and NO-releasing properties were demonstrated to be mainly influenced by the substituent in 4-position of the pyrazole (Cl vs. H). Introduction of the N-propionamide at the sulfamoyl residue as a potential prodrug strategy lowered lipophilicity markedly and abolished COX inhibition while NO-releasing properties were not markedly influenced. NO-COXIBs were tested in vitro for a combination with single-dose external X-ray irradiation as well as [177Lu]LuCl3 treatment in HIF2α-positive mouse pheochromocytoma (MPC-HIF2a) tumor spheroids. When applied directly before X-ray irradiation or 177Lu treatment, NO-COXIBs showed radioprotective effects, as did celecoxib, which was used as a control. Radiosensitizing effects were observed when applied shortly after X-ray irradiation. Overall, the NO-COXIBs were found to be more radioprotective compared with celecoxib, which does not warrant further preclinical studies with the NO-COXIBs for the treatment of pheochromocytoma. However, evaluation as radioprotective agents for healthy tissues could be considered for the NO-COXIBs developed here, especially when used directly before irradiation.


Subject(s)
Adrenal Gland Neoplasms , Pheochromocytoma , Prodrugs , Radiation-Protective Agents , Radiation-Sensitizing Agents , Adrenal Gland Neoplasms/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Celecoxib/pharmacology , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors/chemistry , Mice , Nitric Oxide , Pheochromocytoma/drug therapy , Prodrugs/pharmacology , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Radiation-Sensitizing Agents/pharmacology
2.
Cells ; 11(8)2022 04 14.
Article in English | MEDLINE | ID: mdl-35456021

ABSTRACT

Transglutaminase 2 (TG2) is a protein expressed in many tissues that exerts numerous, sometimes contradictory, intra- and extracellular functions, under both physiological and pathophysiological conditions. In the context of tumor progression, it has been found to be involved in cell adhesion, DNA repair mechanisms, induction of apoptosis, and mesenchymal transdifferentiation, among others. Here, we hypothesized that TG2 also contributes to the radioresistance of two human melanoma cell lines, A375 and MeWo, which can be seen to differ in their basal TG2 biosynthesis by examining their proliferation and clonal expansion after irradiation. For this purpose, cellular TG2 biosynthesis and TG2 activity were modulated by transfection-induced overexpression or TG2 knock-out and application of TG2-selective inhibitors. Proliferation and clonal expansion of TG2-overexpressing cells was not enhanced over wildtype cells, suggesting that increased TG2 biosynthesis does not further enhance the radioresistance of melanoma cells. Conversely, TG2 knock-out in A375 cells reduced their proliferation, as well as clonal and spheroidal expansion after irradiation, which indicates a contribution of TG2 to the radioresistance of melanoma cells. Since TG1, TG3, and partly also, TG6 biosynthesis was detectable in A375 and MeWo cells, it can be assumed that these other members of the TG family may exert a partially compensatory effect.


Subject(s)
Melanoma , Radiation Tolerance , Cell Adhesion , Cell Line, Tumor/radiation effects , Humans , Melanoma/genetics , Melanoma/radiotherapy , Protein Glutamine gamma Glutamyltransferase 2
3.
Cells ; 11(4)2022 02 21.
Article in English | MEDLINE | ID: mdl-35203404

ABSTRACT

The inducible isoenzyme cyclooxygenase-2 (COX-2) is an important hub in cellular signaling, which contributes to tumor progression by modulating and enhancing a pro-inflammatory tumor microenvironment, tumor growth, apoptosis resistance, angiogenesis and metastasis. In order to understand the role of COX-2 expression in melanoma, we investigated the functional knockout effect of COX-2 in A2058 human melanoma cells. COX-2 knockout was validated by Western blot and flow cytometry analysis. When comparing COX-2 knockout cells to controls, we observed significantly reduced invasion, colony and spheroid formation potential in cell monolayers and three-dimensional models in vitro, and significantly reduced tumor development in xenograft mouse models in vivo. Moreover, COX-2 knockout alters the metabolic activity of cells under normoxia and experimental hypoxia as demonstrated by using the radiotracers [18F]FDG and [18F]FMISO. Finally, a pilot protein array analysis in COX-2 knockout cells verified significantly altered downstream signaling pathways that can be linked to cellular and molecular mechanisms of cancer metastasis closely related to the enzyme. Given the complexity of the signaling pathways and the multifaceted role of COX-2, targeted suppression of COX-2 in melanoma cells, in combination with modulation of related signaling pathways, appears to be a promising therapeutic approach.


Subject(s)
CRISPR-Cas Systems , Cyclooxygenase 2 , Melanoma , Neoplasm Invasiveness , Animals , Cell Line, Tumor , Cyclooxygenase 2/genetics , Gene Knockdown Techniques , Humans , Melanoma/pathology , Mice , Tumor Microenvironment
4.
RSC Adv ; 10(63): 38601-38611, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-35517533

ABSTRACT

The inducible isoenzyme cyclooxygenase-2 (COX-2) is closely associated with chemo-/radioresistance and poor prognosis of solid tumors. Therefore, COX-2 represents an attractive target for functional characterization of tumors by positron emission tomography (PET). In this study, the celecoxib derivative 3-([18F]fluoromethyl)-1-[4-(methylsulfonyl)phenyl]-5-(p-tolyl)-1H-pyrazole ([18F]5a) was chosen as a lead compound having a reported high COX-2 inhibitory potency and a potentially low carbonic anhydrase binding tendency. The respective deuterated analog [D2,18F]5a and the fluoroethyl-substituted derivative [18F]5b were selected to study the influence of these modifications with respect to COX inhibition potency in vitro and metabolic stability of the radiolabeled tracers in vivo. COX-2 inhibitory potency was found to be influenced by elongation of the side chain but, as expected, not by deuteration. An automated radiosynthesis comprising 18F-fluorination and purification under comparable conditions provided the radiotracers [18F]5a,b and [D2,18F]5a in good radiochemical yields (RCY) and high radiochemical purity (RCP). Biodistribution and PET studies comparing all three compounds revealed bone accumulation of 18F-activity to be lowest for the ethyl derivative [18F]5b. However, the deuterated analog [D2,18F]5a turned out to be the most stable compound of the three derivatives studied here. Time-dependent degradation of [18F]5a,b and [D2,18F]5a after incubation in murine liver microsomes was in accordance with the data on metabolism in vivo. Furthermore, metabolites were identified based on UPLC-MS/MS.

5.
J Labelled Comp Radiopharm ; 62(8): 495-509, 2019 06 30.
Article in English | MEDLINE | ID: mdl-30912594

ABSTRACT

Radiolabeled α-melanocyte-stimulating hormone (α-MSH) derivatives have a high potential for diagnosis and treatment of melanoma, because of high specificity and binding affinity to the melanocortin-1 receptor (MC1R). Hence, the α-MSH-derived peptide NAP-NS1 with a ß-Ala linker (ε-Ahx-ß-Ala-Nle-Asp-His-D-Phe-Arg-Trp-Gly-NH2 ) was conjugated to different chelators: either to NOTA (p-SCN-Bn-1,4,7-triazacyclononane-1,4,7-triacetic acid), to a hexadentate bispidine carbonate derivative (dimethyl-9-(((4-nitrophenoxy)carbonyl)oxy)-2,4-di(pyridin-2-yl)-3,7-bis(pyridin-2-ylmethyl)-3,7-diazabicyclo[3.3.1]nonane-1,5-dicarboxylate), or to DMPTACN (p-SCN-Ph-bis(2-pyridyl-methyl)-1,4,7-triaza-cyclononane), labeled with 64 Cu, and investigated in terms of radiochemical and radiopharmacological properties. For the three 64 Cu-labeled conjugates negligible transchelation, suitable buffer and serum stability, as well as appropriate water solubility, was determined. The three conjugates exhibited high binding affinity (low nanomolar range) in murine B16F10, human MeWo, and human TXM13 cells. The Bmax values of [64 Cu]Cu-bispidine-NAP-NS1 ([64 Cu]Cu-2) and [64 Cu]Cu-DMPTACN-NAP-NS1 ([64 Cu]Cu-3) were higher than those of [64 Cu]Cu-NOTA-NAP-NS1 ([64 Cu]Cu-1), implying that different charged chelate units might have an impact on binding capacity. Preliminary in vivo biodistribution studies suggested the main excretion pathway of [64 Cu]Cu-1 and [64 Cu]Cu-3 to be renal, while that of [64 Cu]Cu-2 seemed to be both renal and hepatobiliary. An initial moderate uptake in the kidney decreased clearly after 60 minutes. All three 64 Cu-labeled conjugates should be considered for further in vivo investigations using a suitable xenograft mouse model.


Subject(s)
Chelating Agents/chemistry , Copper Radioisotopes/chemistry , alpha-MSH/chemistry , Animals , Cell Line , Drug Stability , Humans , Isotope Labeling , Radiochemistry , Rats , Tissue Distribution , alpha-MSH/metabolism , alpha-MSH/pharmacokinetics
6.
Methods Mol Biol ; 1929: 461-485, 2019.
Article in English | MEDLINE | ID: mdl-30710291

ABSTRACT

The interaction of S100 proteins (S100s), a multigenic family of Ca2+-binding and Ca2+-modulated proteins, with pattern recognition receptors, e.g., Toll-like receptors (TLRs), the receptor for advanced glycation end products (RAGE), or scavenger receptors (SR), is hypothesized to be of high relevance in the pathogenesis of various diseases. This includes chronic inflammatory conditions, atherosclerosis, cardiomyopathies, neurodegeneration, and progression of cancers. However, data concerning the role of circulating S100s in these pathologies are scarce. One reason for this is the shortage of suitable radiolabeling methods for direct assessment of the metabolic fate of circulating S100s in vivo. We report a radiotracer approach using radiolabeling of recombinant human S100s with the positron emitter fluorine-18 (18F) by conjugation with N-succinimidyl 4-[18F]fluorobenzoate ([18F]SFB). The methodological radiochemical part focuses on an optimized and automated synthesis of [18F]SFB comprising HPLC purification to achieve higher chemical purity. The respective radioligands, [18F]fluorobenzoylated S100s ([18F]FB-S100s), were obtained with appropriate radiochemical purities, yields, and effective molar activities. Biological applications comprise cell and tissue binding experiments in vitro, biodistribution and metabolite studies in rodents in vivo/ex vivo, and dynamic positron emission tomography studies using dedicated small animal PET systems. Radiolabeling of S100s with 18F and, particularly, the use of small animal PET provide novel probes to delineate both their metabolic fate and the functional expression of their specific receptors under normal and pathophysiological conditions in rodent models of disease.


Subject(s)
Benzoates/chemistry , Radiopharmaceuticals/chemical synthesis , S100 Proteins/chemical synthesis , Succinimides/chemistry , Animals , Chromatography, High Pressure Liquid , Humans , Isotope Labeling/methods , Positron-Emission Tomography/methods , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Rats , S100 Proteins/chemistry , S100 Proteins/pharmacokinetics , Tissue Distribution
7.
Amino Acids ; 51(2): 219-244, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30264172

ABSTRACT

The cell surface receptor claudin-4 (Cld-4) is upregulated in various tumours and represents an important emerging target for both diagnosis and treatment of solid tumours of epithelial origin. The C-terminal fragment of the Clostridium perfringens enterotoxin cCPE290-319 appears as a suitable ligand for targeting Cld-4. The synthesis of this 30mer peptide was attempted via several approaches, which has revealed sequential SPPS using three pseudoproline dipeptide building blocks to be the most efficient one. Labelling with fluorine-18 was achieved on solid phase using N-succinimidyl 4-[18F]fluorobenzoate ([18F]SFB) and 4-[18F]fluorobenzoyl chloride as 18F-acylating agents, which was the most advantageous when [18F]SFB was reacted with the resin-bound 30mer containing an N-terminal 6-aminohexanoic spacer. Binding to Cld-4 was demonstrated via surface plasmon resonance using a protein construct containing both extracellular loops of Cld-4. In addition, cell binding experiments were performed for 18F-labelled cCPE290-319 with the Cld-4 expressing tumour cell lines HT-29 and A431 that were complemented by fluorescence microscopy studies using the corresponding fluorescein isothiocyanate-conjugated peptide. The 30mer peptide proved to be sufficiently stable in blood plasma. Studying the in vivo behaviour of 18F-labelled cCPE290-319 in healthy mice and rats by dynamic PET imaging and radiometabolite analyses has revealed that the peptide is subject to substantial liver uptake and rapid metabolic degradation in vivo, which limits its suitability as imaging probe for tumour-associated Cld-4.


Subject(s)
Claudin-4/antagonists & inhibitors , Enterotoxins/chemical synthesis , Enterotoxins/pharmacokinetics , Animals , Claudin-4/chemistry , Claudin-4/metabolism , Enterotoxins/chemistry , Enterotoxins/pharmacology , Fluorine Radioisotopes/chemistry , HT29 Cells , Humans , Isotope Labeling , Ligands , Male , Mice , Mice, Nude , Molecular Imaging , Molecular Mimicry/physiology , Molecular Targeted Therapy , Neoplasms/drug therapy , Positron-Emission Tomography , Rats , Rats, Wistar , Solid-Phase Synthesis Techniques
8.
J Cell Mol Med ; 20(5): 825-35, 2016 May.
Article in English | MEDLINE | ID: mdl-26928771

ABSTRACT

S100A4, a member of the S100 protein family of EF-hand calcium-binding proteins, is overexpressed in various tumour entities, including melanoma, and plays an important role in tumour progression. Several studies in epithelial and mesenchymal tumours revealed a correlation between extracellular S100A4 and metastasis. However, exact mechanisms how S100A4 stimulates metastasis in melanoma are still unknown. From a pilot experiment on baseline synthesis and secretion of S100A4 in human melanoma cell lines, which are in broad laboratory use, A375 wild-type cells and, additionally, newly generated A375 cell lines stably transfected with human S100A4 (A375-hS100A4) or human receptor for advanced glycation endproducts (A375-hRAGE), were selected to investigate the influence of extracellular S100A4 on cell motility, adhesion, migration and invasion in more detail. We demonstrated that A375 cells actively secrete S100A4 in the extracellular space via an endoplasmic reticulum-Golgi-dependent pathway. S100A4 overexpression and secretion resulted in prometastatic activation of A375 cells. Moreover, we determined the influence of S100A4-RAGE interaction and its blockade on A375, A375-hS100A4, A375-hRAGE cells, and showed that interaction of RAGE with extracellular S100A4 contributes to the observed activation of A375 cells. This investigation reveals additional molecular targets for therapeutic approaches aiming at blockade of ligand binding to RAGE or RAGE signalling to inhibit melanoma metastasis.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Receptor for Advanced Glycation End Products/metabolism , S100 Calcium-Binding Protein A4/metabolism , Skin Neoplasms/metabolism , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Endoplasmic Reticulum/metabolism , Female , Golgi Apparatus/metabolism , Humans , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Protein Binding , Protein Transport , Receptor for Advanced Glycation End Products/genetics , S100 Calcium-Binding Protein A4/genetics , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Xenograft Model Antitumor Assays
9.
Amino Acids ; 48(3): 833-847, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26643502

ABSTRACT

The melanocortin-1 receptor (MC1R) plays an important role in melanoma growth, angiogenesis and metastasis, and is overexpressed in melanoma cells. α-Melanocyte stimulating hormone (α-MSH) and derivatives are known to bind with high affinity at this receptor that provides the potential for selective targeting of melanoma. In this study, one linear α-MSH-derived peptide Nle-Asp-His-D-Phe-Arg-Trp-Gly-NH2 (NAP-NS1) without linker and with εAhx-ß-Ala linker, and a cyclic α-MSH derivative, [Lys-Glu-His-D-Phe-Arg-Trp-Glu]-Arg-Pro-Val-NH2 (NAP-NS2) with εAhx-ß-Ala linker were conjugated with p-SCN-Bn-NOTA and labeled with (64)Cu. Radiochemical and radiopharmacological investigations were performed with regard to transchelation, stability, lipophilicity and in vitro binding assays as well as biodistribution in healthy rats. No transchelation reactions, but high metabolic stability and water solubility were demonstrated. The linear derivatives showed higher affinity than the cyclic one. [(64)Cu]Cu-NOTA-εAhx-ß-Ala-NAP-NS1 ([(64)Cu]Cu-2) displayed rapid cellular association and dissociation in murine B16F10 cell homogenate. All [(64)Cu]Cu-labeled conjugates exhibited affinities in the low nanomolar range in B16F10. [(64)Cu]Cu-2 showed also high affinity in human MeWo and TXM13 cell homogenate. In vivo studies suggested that [(64)Cu]Cu-2 was stable, with about 85 % of intact peptide in rat plasma at 2 h p.i. Biodistribution confirmed the renal pathway as the major elimination route. The uptake of [(64)Cu]Cu-2 in the kidney was 5.9 % ID/g at 5 min p.i. and decreased to 2.0 % ID/g at 60 min p.i. Due to the prospective radiochemical and radiopharmacological properties of the linear α-MSH derivative [(64)Cu]Cu-2, this conjugate is a promising candidate for tracer development in human melanoma imaging.


Subject(s)
Copper Radioisotopes/chemistry , Diagnostic Imaging/instrumentation , Melanoma/diagnosis , Radiopharmaceuticals/chemistry , alpha-MSH/analogs & derivatives , Animals , Copper Radioisotopes/administration & dosage , Copper Radioisotopes/pharmacokinetics , Drug Stability , Humans , Male , Melanoma/diagnostic imaging , Melanoma/metabolism , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Wistar , Skin Neoplasms , Tissue Distribution , alpha-MSH/administration & dosage , alpha-MSH/pharmacokinetics , Melanoma, Cutaneous Malignant
10.
Clin Hemorheol Microcirc ; 61(3): 523-39, 2015.
Article in English | MEDLINE | ID: mdl-26444589

ABSTRACT

Gold surfaces functionalized with nickel-nitrilotriacetic acid (Ni²âº-NTA) as self-assembled monolayers (SAM) to immobilize histidine (His)-tagged biomolecules are broadly reported in the literature. However, the increasing demand of using microfluidic systems and biosensors takes more and more advantage on silicon technology which provides dedicated glass surfaces and substantially allows cost and resource savings. Here we present a novel method for the controlled oriented immobilization of His-tagged proteins on glass surfaces functionalized with a Ni²âº-NTA layer. Exemplarily, the protein pattern morphology after immobilization on the Ni²âº-NTA layer of His6-tagged soluble receptor for advanced glycation endproducts (sRAGE) was investigated and compared to non-oriented immobilization of sRAGE on amino SAM by using scanning electron microscopy (SEM). Moreover, we demonstrated interaction of immobilized sRAGE with three structurally different ligands, S100A12, S100A4, and glycated low density lipoproteins (glycLDL), by means of peak-force tapping atomic force microscopy (PF-AFM). We showed a clear discrimination of different protein-ligand orientations by differential height measurements.


Subject(s)
Histidine/genetics , Nitrilotriacetic Acid/analogs & derivatives , Oligopeptides/genetics , Organometallic Compounds/metabolism , Biosensing Techniques , Glass , Ligands , Nitrilotriacetic Acid/metabolism , S100 Proteins
11.
Biochem Biophys Res Commun ; 446(1): 143-8, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24613382

ABSTRACT

S100A4, synthesized and secreted from both tumor and stroma cells, modulates an aggressive tumor phenotype in various cancers by intracellular and extracellular interactions which are not completely understood. Because of the high content of tumor-associated macrophages in melanoma, here, a syngeneic model (coculture of mouse B16-F10 melanoma cells (Mel) and RAW264.7 macrophages (Mϕ); administration (i.v.) of Mel and Mϕ/Mel in NMRI nu/nu mice) was used to investigate synthesis and secretion of (a) S100A4, (b) S100A4-mediated signaling and activation of NFκB, and (c) S100A4-mediated modulation of Mel invasiveness in vitro (transwell assay, transwell matrigel assay) and in vivo (metastatic lung colonization), respectively. In this model substantial S100A4 synthesis and secretion is demonstrated in Mϕ. Macrophage-derived S100A4 promotes Mel invasiveness in a paracrine manner in vitro, which is further substantiated in control experiments using recombinant human S100A4 and Mel stably transfected with mouse S100A4. Moreover, the participation of S100A4-mediated signaling, e.g., via the receptor for advanced glycation endproducts (RAGE), resulting in activation of NFκB was demonstrated in all experimental settings. Finally, we demonstrated that interaction of macrophage-derived S100A4 with Mel results in increased metastatic lung colonization in vivo.


Subject(s)
Macrophages/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/secondary , S100 Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Coculture Techniques , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Mice , Mice, Nude , NF-kappa B/metabolism , Neoplasm Invasiveness , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , S100 Calcium-Binding Protein A4 , S100 Proteins/genetics , Signal Transduction
12.
Biochem Biophys Res Commun ; 413(3): 494-8, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21924240

ABSTRACT

The aim of this study was to investigate the response to and the physiological consequences of copper-mediated cross-linking of S100A2 and S100A4, two members of the S100 family of EF-hand calcium-binding proteins. As demonstrated by electrophoresis and mass spectrometry techniques S100A2 and S100A4 show formation of cross-links due to copper-mediated oxidation of cysteine residues. For S100A4, but not for S100A2, this results in both increased activation of NFκB and secretion of TNF-α in human A375 and, to a higher extent, in RAGE-transfected melanoma cells. The data suggest that a prooxidative tumor microenvironment enhances proinflammatory and prometastatic action of S100A4.


Subject(s)
Chemotactic Factors/metabolism , Copper/metabolism , Inflammation/metabolism , Melanoma/metabolism , S100 Proteins/metabolism , Tumor Microenvironment , Amino Acid Sequence , Cell Line, Tumor , Chemotactic Factors/chemistry , Copper/chemistry , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/pharmacology , Cysteine/chemistry , Cysteine/metabolism , Glycation End Products, Advanced/metabolism , Humans , Molecular Sequence Data , NF-kappa B/metabolism , Oxidation-Reduction , Protein Structure, Secondary , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , S100 Calcium-Binding Protein A4 , S100 Proteins/chemistry , Tumor Necrosis Factor-alpha/metabolism
13.
Amino Acids ; 41(4): 849-61, 2011 Oct.
Article in English | MEDLINE | ID: mdl-20521072

ABSTRACT

Owing to the exceptional intracellular distribution and the heterogeneous expression pattern during transformation and metastasis in various tumors, the EF-hand calcium-binding protein S100A2 attracts increasing attention. Unlike the majority of S100 proteins, S100A2 expression is downregulated in many cancers and the loss in nuclear expression has been associated with poor prognosis. On the other hand, S100A2 is upregulated in some cancers. This mini review highlights the general characteristics of S100A2 and discusses recent findings on its putative functional implication as a suppressor or promoter in cancerogenesis.


Subject(s)
Chemotactic Factors/chemistry , Chemotactic Factors/genetics , Chemotactic Factors/metabolism , Neoplasms/metabolism , S100 Proteins/chemistry , S100 Proteins/genetics , S100 Proteins/metabolism , Amino Acid Sequence , Animals , Humans , Mice , Molecular Sequence Data , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Up-Regulation
14.
Amino Acids ; 41(4): 809-20, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21153848

ABSTRACT

Data concerning the pathophysiological role of extracellular S100A4, a member of the multigenic family of Ca(2+)-modulated S100 proteins, and its interaction with the receptor for advanced glycation endproducts (RAGE) or other putative receptors in tumorigenesis, metastasis, and inflammatory processes in vivo are scarce. One reason is the shortage of suitable radiotracer methods. We report a novel methodology using recombinant human S100A4 as potential probe for molecular imaging and functional characterization of this interaction. Therefore, human S100A4 was cloned as GST fusion protein in the bacterial expression vector pGEX-6P-1 and expressed in E. coli strain BL21. Purified recombinant human S100A4 was radiolabeled with the positron emitter fluorine-18 ((18)F) by conjugation with N-succinimidyl-4-[(18)F]fluorobenzoate ([(18)F]SFB). The radioligand [(18)F]fluorobenzoyl-S100A4 ((18)F-S100A4) was used in cell binding experiments in RAGE-bearing human melanoma cells and endothelial cells in vitro, and in both biodistribution experiments and small animal positron emission tomography (PET) studies in normal rats in vivo. The cellular association and tissue-specific distribution of (18)F-S100A4 in vitro and in vivo correlated well with the protein expression and anatomical localization of RAGE, e.g., in the vascular system and in lung. Compared to other S100 RAGE radioligands, the overall findings of this study indicate that extracellular S100A4 in vivo shows only a moderate interaction with RAGE and, furthermore, exhibits a substantially faster metabolic degradation. On the other hand, the approach allows the use of quantitative small animal PET and provides a novel probe to both delineate functional expression and differentiate multiligand interaction of RAGE under normal and pathophysiological conditions in rodent models of disease.


Subject(s)
Receptors, Immunologic/analysis , Receptors, Immunologic/metabolism , S100 Proteins/isolation & purification , S100 Proteins/metabolism , Animals , Benzoates/chemistry , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Fluorine Radioisotopes/chemistry , Glycation End Products, Advanced , Humans , Male , Melanoma/metabolism , Positron-Emission Tomography/methods , Rats , Rats, Wistar , Receptor for Advanced Glycation End Products , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , S100 Calcium-Binding Protein A4 , S100 Proteins/genetics , Succinimides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...