Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
J Biol Chem ; 289(32): 22078-89, 2014 Aug 08.
Article En | MEDLINE | ID: mdl-24962584

Quercetin, a flavonoid abundantly present in plants, is widely used as a phytotherapy in prostatitis and prostate cancer. Although quercetin has been reported to have a number of therapeutic effects, the cellular target(s) responsible for its anti-cancer action has not yet been clearly elucidated. Here, employing affinity chromatography and mass spectrometry, we identified heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) as a direct target of quercetin. A specific interaction between quercetin and hnRNPA1 was validated by immunoblotting and in vitro binding experiments. We found that quercetin bound the C-terminal region of hnRNPA1, impairing the ability of hnRNPA1 to shuttle between the nucleus and cytoplasm and ultimately resulting in its cytoplasmic retention. In addition, hnRNPA1 was recruited to stress granules after treatment of cells with quercetin for up to 48 h, and the levels of cIAP1 (cellular inhibitor of apoptosis), an internal ribosome entry site translation-dependent protein, were reduced by hnRNPA1 regulation. This is the first report that anti-cancer effects of quercetin are mediated, in part, by impairing functions of hnRNPA1, insights that were obtained using a chemical proteomics strategy.


Antineoplastic Agents, Phytogenic/pharmacology , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Quercetin/pharmacology , Amino Acid Sequence , Antineoplastic Agents, Phytogenic/pharmacokinetics , Apoptosis/drug effects , Binding Sites , Biological Transport, Active , Cell Line, Tumor , Cell Survival/drug effects , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Humans , Inhibitor of Apoptosis Proteins/metabolism , Male , Models, Biological , Molecular Sequence Data , Mutant Proteins/genetics , Mutant Proteins/metabolism , Phytotherapy , Prostatic Neoplasms/genetics , Protein Binding , Proteomics , Quercetin/pharmacokinetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , beta Karyopherins/metabolism
2.
Arch Biochem Biophys ; 506(1): 66-72, 2011 Feb 01.
Article En | MEDLINE | ID: mdl-21056540

[(3)H]8-OH-DPAT is a selective ligand for labeling 5-HT(1A) receptor sites. In competition binding experiments, we found that classic biogenic amine transporter inhibitors displaced [(3)H]8-OH-DPAT binding at its high-affinity binding sites in HeLaS3 cells. [(125)I]RTI-55 and [(3)H]paroxetine are known to specifically label amine transporter sites, and this was observed in our cells. Displacement studies showed that 8-OH-DPAT displayed affinity in a dose-dependent manner for the labeled amine transporter sites. These data suggest that [(3)H]8-OH-DPAT binds to amine uptake sites in HeLaS3 cells. A variety of drugs targeting different classes of receptors did not significantly affect [(3)H]8-OH-DPAT binding. Moreover, we determined the specific binding effects of various serotonergic ligands (i.e. [(125)I]cyanopindolol, [(3)H]ketanserin/[(3)H]mesulergine, [(3)H]GR-65630, [(3)H]GR-113808 and [(3)H]LSD) that specifically labeled 5-HT(1), 5-HT(2), 5-HT(3), 5-HT(4) and 5-HT(5-7) receptors, respectively. It is suggested that HeLaS3 cells contain distinct types of the related to 5-HT receptor recognition binding sites. These observations could help elucidate the relevant characteristics of different types of 5-HT receptors and 5-HT membrane transporters in tumor cells and their role in tumorigenesis.


8-Hydroxy-2-(di-n-propylamino)tetralin/metabolism , Receptors, Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Binding, Competitive , Biogenic Amines/metabolism , Cocaine/analogs & derivatives , Cocaine/metabolism , Fluoxetine/metabolism , HeLa Cells , Humans , Kinetics , Ligands , Paroxetine/metabolism , Radioligand Assay , Selective Serotonin Reuptake Inhibitors/metabolism , Vesicular Biogenic Amine Transport Proteins/antagonists & inhibitors
3.
Bioorg Med Chem ; 18(2): 597-604, 2010 Jan 15.
Article En | MEDLINE | ID: mdl-20056549

In the present study we have discovered compound 1, a benzo[1.3.2]dithiazolium ylide-based compound, as a new prototype dual inhibitor of cyclooxygenase (COX) and 5-lipoxygenase (5-LOX). Compound 1 was initially discovered as a COX-2 inhibitor, resulting indirectly from the COX-2 structure-based virtual screening that identified compound 2 as a virtual hit. Compounds 1 and 2 inhibited COX-1 and COX-2 in mouse macrophages with IC(50) in the range of 1.5-18.1microM. Both compounds 1 and 2 were also found to be potent inhibitors of human 5-LOX (IC(50)=1.22 and 0.47microM, respectively). Interestingly, compound 1 also had an inhibitory effect on tumor necrosis factor-alpha (TNF-alpha) production (IC(50)=0.44microM), which was not observed with compound 2. Docking studies suggested the (S)-enantiomer of 1 as the biologically active isomer that binds to COX-2. Being a cytokine-suppressive dual COX/5-LOX inhibitor, compound 1 may represent a useful lead structure for the development of advantageous new anti-inflammatory agents.


Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Lipoxygenase Inhibitors , Lipoxygenase Inhibitors/pharmacology , Nitro Compounds/pharmacology , Thiazoles/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Benzothiazoles , Cell Line , Crystallography, X-Ray , Cyclic S-Oxides , Cyclooxygenase Inhibitors/chemical synthesis , Cyclooxygenase Inhibitors/chemistry , Drug Discovery , Humans , Lipoxygenase Inhibitors/chemical synthesis , Lipoxygenase Inhibitors/chemistry , Mice , Models, Molecular , Molecular Structure , Nitro Compounds/chemical synthesis , Nitro Compounds/chemistry , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry , Tumor Necrosis Factor-alpha/biosynthesis
4.
Arch Biochem Biophys ; 495(1): 14-20, 2010 Mar 01.
Article En | MEDLINE | ID: mdl-20018167

Some G protein-coupled receptors (GPCRs) have functional links to cancer biology, yet the manifestation of GPCRs in tumor types is little studied to date. Using a battery of radioligand binding assays, we sought to characterize GPCR recognition binding sites on HeLaS3 tumor cells. High levels of binding of the selective serotonin 5-HT(1A) receptor agonist [3H]8-OH-DPAT were observed in these cells. Saturation and homologous competition experiments indicated that [3H]8-OH-DPAT bound different populations of high- and low-affinity sites. In competition experiments, several serotonergic compounds displaced [3H]8-OH-DPAT binding with low potency from its high-affinity binding sites, suggesting that low-affinity binding is the predominant mode of binding. A variety of drugs targeting different classes of receptors did not affect [3H]8-OH-DPAT binding. These observations may help elucidate the pathophysiological and functional relevance of 5-HT receptors in tumor cells and link GPCRs and tumorigenic mechanisms to pharmacological and chemotherapeutic paradigms.


8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Radioligand Assay , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin Receptor Agonists/pharmacology , Binding, Competitive , Dimethyl Sulfoxide/metabolism , HeLa Cells , Humans , Receptors, G-Protein-Coupled/metabolism
5.
Phytother Res ; 24(1): 43-8, 2010 Jan.
Article En | MEDLINE | ID: mdl-19517465

A Chlorella powder was tested in a total of 129 in vitro receptor binding assay systems. The results showed a potent inhibition of this powder on cysteinyl leukotriene CysLT2, and glutamate AMPA in a dose-concentration manner with IC(50) mean +/- SEM values of 20 +/- 4.5 microg/mL and 44 +/- 14 microg/mL, respectively. Other moderate and weak activities reflected in competitive binding experiments were seen versus adenosine transporter; calcium channel L-type, benzothiazepine; gabapentin; kainate, NMDA-glycine; inositol trisphosphate IP(3); cysteinyl CysLT(1), LTB(4); purinergic P(2Y); tachykinin NK(2); serotonin 5-HT(2B) and prostanoid, thromboxane A(2). Together, the results suggest that the various inhibitory effects of Chlorella powder in these receptor binding assays could reflect its actions in modulating Ca(2+)-dependent signal related targets and might be relevant to the mechanisms of its biological effects. These results reveal important potential biochemical activities that might be exploited for the prevention or treatment of several pathologies. From these results, the possible therapeutic usage of the product is discussed.


Chlorella/chemistry , Ion Channels/metabolism , Nucleoside Transport Proteins/metabolism , Receptors, Leukotriene/metabolism , Receptors, Neurotransmitter/metabolism , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Guinea Pigs , Humans , Inhibitory Concentration 50 , Male , Protein Binding , Rats , Rats, Wistar
6.
Int J Food Sci Nutr ; 60 Suppl 1: 89-98, 2009.
Article En | MEDLINE | ID: mdl-19107625

A Chlorella powder was tested in 118 in vitro enzyme assay systems. The powder showed potent inhibitions of peptidase cathepsin S, thromboxane A(2) synthase and cyclooxygenase-2 in a dose-concentration manner with IC(50)+/-standard error of the mean values of 3.46+/-0.93 microg/ml, 3.23+/-0.69 microg/ml, and 44.26+/-9.98 microg/ml, respectively. Other activities observed were inhibitions of tumor necrosis factor-alpha converting enzyme, protein tyrosine phosphatase (SHP-2), calpain, protein kinases and protein tyrosine phosphatases. Chlorella powder had no significant effect on cyclooxygenase-1. These actions to inhibit cyclooxygenase-2 and thromboxane synthase could contribute to the purported anti-inflammatory and anti-thrombotic effects of Chlorella. These results reveal important potential biochemical activities to be developed that, if confirmed by in vivo studies, might be exploited for the prevention or treatment of several serious pathologies, including inflammatory diseases, immune and cancer.


Anti-Inflammatory Agents/pharmacology , Chlorella , Enzyme Inhibitors/pharmacology , Plant Preparations/pharmacology , ADAM Proteins/antagonists & inhibitors , ADAM17 Protein , Calpain/antagonists & inhibitors , Cathepsins/antagonists & inhibitors , Cyclooxygenase 2/metabolism , Group II Phospholipases A2/antagonists & inhibitors , Peptide Hydrolases/metabolism , Phosphotransferases/antagonists & inhibitors , Platelet Aggregation Inhibitors/pharmacology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Thromboxane-A Synthase/antagonists & inhibitors
7.
Mol Cancer Ther ; 6(1): 147-53, 2007 Jan.
Article En | MEDLINE | ID: mdl-17237275

Cell cycle G(2) checkpoint abrogation is an attractive strategy for sensitizing cancer cells to DNA-damaging anticancer agent without increasing adverse effects on normal cells. However, there is no single proven molecular target for this therapeutic approach. High-throughput screening for molecules inhibiting CHK1, a kinase that is essential for the G(2) checkpoint, has not yet yielded therapeutic G(2) checkpoint inhibitors, and the tumor suppressor phenotypes of ATM and CHK2 suggest they may not be ideal targets. Here, we optimized two G(2) checkpoint-abrogating peptides, TAT-S216 and TAT-S216A, based on their ability to reduce G(2) phase accumulation of DNA-damaged cells without affecting M phase accumulation of cells treated with a microtubule-disrupting compound. This approach yielded a peptide CBP501, which has a unique, focused activity against molecules that phosphorylate Ser(216) of CDC25C, including MAPKAP-K2, C-Tak1, and CHK1. CBP501 is >100-fold more potent than TAT-S216A and retains its selectivity for cancer cells. CBP501 is unusually stable, enters cells rapidly, and increases the cytotoxicity of DNA-damaging anticancer drugs against cancer cells without increasing adverse effects. These findings highlight the potency of CBP501 as a G(2)-abrogating drug candidate. This report also shows the usefulness of the cell cycle phenotype-based protocol for identifying G(2) checkpoint-abrogating compounds as well as the potential of peptide-based compounds as focused multitarget inhibitors.


Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , G2 Phase/drug effects , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Peptides/pharmacology , cdc25 Phosphatases/metabolism , cdc25 Phosphatases/pharmacology , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Bleomycin/adverse effects , Bleomycin/pharmacology , Cell Cycle Proteins/metabolism , Cell Death/drug effects , Cisplatin/adverse effects , Cisplatin/pharmacology , HCT116 Cells , Humans , Jurkat Cells , Male , Mice , Mice, SCID , Models, Molecular , Molecular Sequence Data , Neoplasms/pathology , Peptide Fragments/chemistry , Peptides/chemistry , Phenotype , Phosphorylation/drug effects , Xenograft Model Antitumor Assays , cdc25 Phosphatases/chemistry
8.
Biochem Biophys Res Commun ; 352(3): 656-61, 2007 Jan 19.
Article En | MEDLINE | ID: mdl-17141198

The pro-inflammatory cytokines TNF-alpha and IL-1beta are two of the important mediators involved in the several chronic inflammatory diseases. We used the release of TNF-alpha and IL-1beta from lipopolysaccharide-stimulated human PBMC as inflammatory indexes to discover the potential anti-inflammatory candidates. Among near 500 chemical compounds, MT4 had the suppressive action on the release of TNF-alpha and IL-1beta in PBMC with IC50 values of 22 and 44 nM, respectively. After verified the MT4 inhibitory mechanism, the results revealed that p38alpha and p38beta MAPK activity was inhibited by MT4 with an IC50 value of 0.13 and 0.55 microM, respectively. Further characterization of enzyme kinetics showed the binding mode of MT4 was competitive with the ATP substrate-binding site of p38alpha MAPK.


Interleukin-1beta/metabolism , Leukocytes, Mononuclear/metabolism , Matrix Metalloproteinase 17/administration & dosage , Matrix Metalloproteinase 17/chemistry , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/chemistry , Tumor Necrosis Factor-alpha/metabolism , Biological Assay/methods , Cells, Cultured , Computer Simulation , Dose-Response Relationship, Drug , Drug Design , Humans , Leukocytes, Mononuclear/drug effects , Lipopolysaccharides/administration & dosage , Mitogen-Activated Protein Kinase 14/metabolism , Models, Chemical , Models, Molecular , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry
9.
Org Lett ; 8(20): 4397-400, 2006 Sep 28.
Article En | MEDLINE | ID: mdl-16986909

The design, synthesis, and evaluation of a series of catechol-based non-peptide peptidomimetics of the peptide hormone somatostatin have been achieved. These ligands comprise the simplest known non-peptide mimetics of the i + 1 and i + 2 positions of the somatostatin beta-turn. Incorporation of an additional side chain to include the i position of the beta-turn induces a selective 9-fold affinity enhancement at the sst2 receptor.


Catechols/chemistry , Molecular Mimicry , Somatostatin/chemistry , Models, Molecular
10.
Anticancer Res ; 26(4B): 3057-63, 2006.
Article En | MEDLINE | ID: mdl-16886634

BACKGROUND: Essiac is a herbal compound that has been in common use with cancer patients in North America for over 80 years. Despite its relatively widespread use, there are no peer-reviewed published reports of in vivo studies regarding the use of this compound. MATERIALS AND METHODS: Essiac was administered orally to test animals prior to all experiments. Standard assays to test protection from ethanol-induced gastric ulceration and carbon tetrachloride-induced hepatic injury were performed on Wistar rats. Assays of postglucose-load serum glucose and cellular and humoral immune modulation were conducted on ICR and BALB/C mice, respectively. RESULTS: Essiac demonstrated a modest gastric protective effect via reduction of ethanol-induced gastric ulceration. However, Essiac did not demonstrate significant hepatoprotective, hypoglycemic or immunomodulatory properties. CONCLUSION: Essiac, administered in established in vivo experimental models, did not significantly demonstrate its purported physiological modifying effects.


Plant Extracts/pharmacology , Animals , Blood Glucose/drug effects , Carbon Tetrachloride Poisoning/prevention & control , Chemical and Drug Induced Liver Injury , Liver Diseases/prevention & control , Male , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Rats , Rats, Wistar , Stomach Ulcer/chemically induced , Stomach Ulcer/prevention & control
11.
Org Lett ; 8(9): 1799-802, 2006 Apr 27.
Article En | MEDLINE | ID: mdl-16623554

[structure: see text] Cyclic hexapeptides, incorporating a dipeptide unit in place of the disulfide bond found in urotensin, were prepared and screened at the human urotensin receptor. The bridging dipeptide unit was found to influence dramatically the affinity for the urotensin receptor. Alanyl-N-methylalanyl and alanylprolyl dipeptide bridges failed to afford active ligands, while the alanyl-alanyl unit yielded a ligand with submicromolar affinity for the urotensin receptor. Further development led to a hexapeptide agonist with nanomolar affinity (2.8 nM).


Dipeptides/chemistry , Peptides, Cyclic/chemistry , Peptides, Cyclic/chemical synthesis , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , Urotensins/agonists , Amino Acid Sequence , Binding Sites , Cystine/chemistry , Humans , Models, Molecular , Molecular Structure , Urotensins/chemistry
12.
J Med Chem ; 48(12): 4025-30, 2005 Jun 16.
Article En | MEDLINE | ID: mdl-15943475

An alanine scan performed in the 1970s suggested that Phe(6) and Phe(11) are required for the binding of somatostatin (SRIF-14). Molecular modeling studies and replacement of Phe(6) and Phe(11) with a cystine bridge affording ligands with the retention of high biological activity, however, led to the alternate conclusion that Phe(6) and Phe(11) stabilize the bioactive conformation of SRIF-14. Subsequent studies revealed that Phe(11) shields Phe(6) in a "herringbone" arrangement. More recently, a report from this laboratory demonstrated that Spartan 3-21G MO calculations can be invaluable in explaining SARs in medicinal chemistry. For example, the ability of benzene and indole rings to bind the Trp(8) binding pocket for SRIF-14 and the inability of pyrazine to do so was explained through differences in electrostatic potentials. To investigate the role of Phe(6) and Phe(11) more fully, we report here the synthesis of two analogues of D-Trp(8)-SRIF in which Phe(6) and Phe(11) were replaced by the pryazinylalanine congeners, respectively. The NMR spectra in D(2)O and the K(i)s fully support the proposition that Phe(11) stabilizes the bioactive conformation through pi-bonding or aromatic edge-to-face interaction and that pyrazinylalanine(6) can be shielded by Phe(11). The data also show unexpectedly that Phe(6), via the pi-bond, interacts with the receptor, consistent with the original interpretation of the alanine scan. Heretofore it had only been known that Lys(9) interacts with an aspartate anion of the receptor. These conclusions are supported by recent studies of Lewis et al. on the effects on K(i)s of Ala(6)-SRIF-14-amide at the five receptor subtargets. We also found that pyrazinylalanine(7)-D-Trp(8)-SRIF-14 does not bind, suggesting a repulsive interaction with the receptor. Taken together, our results not only validate predictions based on Spartan 3-21G MO analysis but also provide valuable information about the nature of the receptor interaction at the molecular level. Finally, the chirality of Trp(8) was unexpectedly found to have a striking effect on NMR spectra in methanol, especially at lower temperatures.


Alanine/analogs & derivatives , Alanine/chemistry , Membrane Proteins/chemistry , Phenylalanine/chemistry , Pyrazines/chemistry , Receptors, Somatostatin/chemistry , Somatostatin/analogs & derivatives , Somatostatin/chemical synthesis , Binding Sites , Magnetic Resonance Spectroscopy , Methanol , Models, Molecular , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry , Solvents , Somatostatin/chemistry , Stereoisomerism , Structure-Activity Relationship
13.
Thromb Haemost ; 93(5): 940-8, 2005 May.
Article En | MEDLINE | ID: mdl-15886813

To evaluate the anti-sepsis potential of YC-1, we have examined the effect of YC-1 on the regulation of cytokine production in human leukocytes and endotoxemic mice. The data demonstrated that YC-1 showed a preferential inhibition on proinflammatory cytokine production without inhibition of cell growth or induction of cytotoxicity in human leukocytes. On the other hand, in the septic mouse model, treatment with an intraperitoneal application of LPS caused a cumulative death within 27 hours. The post-treatment administration of YC-1 significantly increased the survival rate in endotoxemic mice. Furthermore, several mediators were detected and the data showed that YC-1 profoundly blocked LPS-induced NO as well as TNF-alpha production, and prevented lung damage by histological examination. Samples from the animal model showed that LPS-induced NF-kappaB/DNA binding activity and consequent up-regulation of iNOS expression in tissues were abolished by post-administration of YC-1. Furthermore, YC-1, by itself, did not modify cGMP content while significantly inhibit LPS-induced cGMP formation, suggesting that YC-1-mediated effect was not through a cGMP-elevating pathway. Taken together, it is evident that the post-treatment administration of YC-1 after LPS application significantly inhibits NF-kappaB activation, iNOS expression, NO over-production, and cytokine release reaction resulting in an improved survival rate in endotoxemic mice. It is suggested that YC-1 may be a potential agent for the therapeutic treatment of sepsis.


Cytokines/metabolism , Indazoles/pharmacology , Leukocytes, Mononuclear/cytology , Platelet Aggregation Inhibitors/pharmacology , Animals , Blotting, Western , Cyclic GMP/metabolism , Cytokines/biosynthesis , DNA/metabolism , Disease Models, Animal , Endotoxemia/blood , Humans , Immunohistochemistry , Leukocytes/metabolism , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Lung/pathology , Male , Mice , Mice, Inbred ICR , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Rats , Rats, Wistar , Shock, Septic , Time Factors , Up-Regulation
14.
Am J Chin Med ; 33(1): 61-71, 2005.
Article En | MEDLINE | ID: mdl-15844834

BNG-1, a novel mixture of traditional Chinese medicines with a long history in the treatment of stroke, exhibited acute neuroprotection effect on rats with middle cerebral artery occlusion (MCAO). Anti-ischemic effects were seen in both animals receiving BNG-1 before the ischemic insult as well as in animals receiving the drug formulation after surgical occlusion of the artery. Anti-thrombic activity was seen in vitro to inhibit arachidonic acid-induced platelet aggregation and in vivo to prolong bleeding time in mice. BNG-1 was also found to inhibit several phosphodiesterase (PDE) isoforms with potency order of the following rank: PDE 1 > PDE 3 > PDE 6 > PDE 2 > PDE 4 > PDE 5. Other pre-clinical results and emerging clinical data coupled with the present findings suggest that BNG-1 may be a safe and effective therapy for both the prevention and treatment of cerebral stroke. Moreover, the fundamental cellular mechanism underlying its therapeutic effects may result from phosphodiesterase inhibition.


Drugs, Chinese Herbal/pharmacology , Neuroprotective Agents/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Stroke/drug therapy , Animals , Female , Guinea Pigs , Male , Mice , Mice, Inbred ICR , Panax , Platelet Aggregation Inhibitors/pharmacology , Rabbits , Rats , Rats, Sprague-Dawley
15.
J Pharmacol Exp Ther ; 314(1): 35-42, 2005 Jul.
Article En | MEDLINE | ID: mdl-15784655

Angiogenesis is a process that involves endothelial cell proliferation, migration, invasion, and tube formation, and inhibition of these processes has implications for angiogenesis-mediated disorders. The purpose of this study was to evaluate the antiangiogenic efficacy of YC-1 [3-(5'-hydroxymethyl-2'-furyl)-1-benzyl indazole] in well characterized in vitro and in vivo systems. YC-1 inhibited the ability of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) in a dose-dependent manner to induce proliferation, migration, and tube formation in human umbilical vascular endothelial cells; these outcomes were evaluated using [3H]thymidine incorporation, transwell chamber, and Matrigel-coated slide assays, respectively. YC-1 inhibited VEGF- and bFGF-induced p42/p44 mitogen-activated protein kinase and Akt phosphorylation as well as protein kinase C alpha translocation using Western blot analysis. The effect of YC-1 on angiogenesis in vivo was evaluated using the mouse Matrigel implant model. YC-1 administered orally in doses of 1 to 100 mg/kg/day inhibited VEGF- and bFGF-induced neovascularization in a dose-dependent manner over 7 days. These results indicate that YC-1 has antiangiogenic activity at very low doses. Moreover, in transplantable murine tumor models, YC-1 administered orally displayed a high degree of antitumor activity (treatment-to-control life span ratio > 175%) without cytotoxicity. YC-1 may be useful for treating angiogenesis-dependent human diseases such as cancer.


Angiogenesis Inducing Agents/pharmacology , Angiogenesis Inhibitors/pharmacology , Endothelial Cells/drug effects , Enzyme Activators/pharmacology , Indazoles/pharmacology , Neovascularization, Pathologic/prevention & control , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 2/antagonists & inhibitors , Guanylate Cyclase/metabolism , Humans , Longevity , Microtubules/drug effects , Neoplasms/blood supply , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Signal Transduction/drug effects , Tetrazolium Salts , Thiazoles , Thymidine/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors
16.
Org Lett ; 7(3): 399-402, 2005 Feb 03.
Article En | MEDLINE | ID: mdl-15673249

[structure: see text] Tetrapyrrolinone somatostatin (SRIF) mimetics (cf. 1), based on a heterochiral (D,L-mixed) pyrrolinone scaffold, were designed, synthesized, and evaluated for biological activity. The iterative synthetic sequence, incorporating the requisite functionalized coded and noncoded amino acid side chains, comprised a longest linear synthetic sequence of 23 steps. Binding affinities at two somatostatin receptor subtypes (hsst 4 and 5) reveal micromolar activity, demonstrating that the d,l-mixed pyrrolinone scaffold can be employed to generate functional mimetics of peptide beta-turns.


Hormone Antagonists/chemistry , Pyrroles/chemical synthesis , Pyrroles/metabolism , Somatostatin/chemistry , Somatostatin/metabolism , Amino Acids/chemistry , Drug Design , Hormone Antagonists/metabolism , Humans , Indicators and Reagents , Ligands , Models, Molecular , Molecular Mimicry , Protein Conformation , Pyrroles/pharmacology , Receptors, Somatostatin/metabolism
17.
J Med Food ; 7(2): 146-52, 2004.
Article En | MEDLINE | ID: mdl-15298760

A Chlorella powder was screened using 52 in vitro assay systems for enzyme activity, receptor binding, cellular cytokine release, and B and T cell proliferation. The screening revealed a very potent inhibition of human protein tyrosine phosphatase (PTP) activity of CD45 and PTP1C with 50% inhibitory concentration (IC(50)) values of 0.678 and 1.56 microg/mL, respectively. It also showed a moderate inhibition of other PTPs, including PTP1B (IC(50) = 65.3 microg/mL) and T-cell-PTP (114 microg/mL). Other inhibitory activities and their IC(50) values included inhibition of the human matrix metalloproteinases (MMPs) MMP-1 (127 microg/mL), MMP-3 (185 microg/mL), MMP-7 (18.1 microg/mL), and MMP-9 (237 microg/mL) and the human peptidase caspases caspase 1 (300 microg/mL), caspase 3 (203 microg/mL), caspase 6 (301 microg/mL), caspase 7 (291 microg/mL), and caspase 8 (261 microg/mL), as well as release of the cytokines interleukin (IL)-1 (44.9 microg/mL), IL-2 (14.8 microg/mL), IL-4 (49.2 microg/mL), IL-6 (34.7 microg/mL), interferon-gamma (31.6 microg/mL), and tumor necrosis factor-alpha (11 microg/mL) from human peripheral blood mononuclear cells. Chlorella also inhibited B cell proliferation (16.6 microg/mL) in mouse splenocytes and T cell proliferation (54.2 microg/mL) in mouse thymocytes. The binding of a phorbol ester, phorbol 12,13-dibutyrate, to its receptors was also inhibited by Chlorella with an IC(50) of 152 microg/mL. These results reveal potential pharmacological activities that, if confirmed by in vivo studies, might be exploited for the prevention or treatment of several serious pathologies, including inflammatory disease and cancer.


Caspase Inhibitors , Chlorella/chemistry , Cytokines/biosynthesis , Lymphocyte Activation , Matrix Metalloproteinase Inhibitors , Protein Tyrosine Phosphatases/antagonists & inhibitors , Animals , B-Lymphocytes/physiology , Concanavalin A/pharmacology , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Humans , Leukocyte Common Antigens , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Protein Kinase C/metabolism , Receptors, Drug/metabolism , T-Lymphocytes/physiology
18.
J Pharmacol Sci ; 95(3): 311-9, 2004 Jul.
Article En | MEDLINE | ID: mdl-15272206

A radioligand binding assay for the HERG (human ether-a-go-go-related gene) K(+) channel was developed to identify compounds which may have inhibitory activity and potential cardiotoxicity. Pharmacological characterization of the [(3)H]astemizole binding assay for HERG K(+) channels was performed using HERG-expressing HEK293 cells. The assay conditions employed yielded 90% specific binding using 10 microg/well of membrane protein with 1.5 nM of [(3)H]astemizole at 25 degrees C. The K(d) and B(max) values were 5.91 +/- 0.81 nM and 6.36 +/- 0.26 pmol/mg, respectively. The intraassay and interassay variations were 11.4% and 14.9%, respectively. Binding affinities for 32 reference compounds (including dofetilide, cisapride, and terfenadine) with diverse structures demonstrated a similar potency rank order for HERG inhibition to that reported in the literature. Moreover, the [(3)H]astemizole binding data demonstrated a rank order of affinity that was highly correlated to that of inhibitory potency in the electrophysiological studies for HERG in HEK293 (r(SP) = 0.91, P<0.05). In conclusion, the [(3)H]astemizole binding assay is rapid and capable of detecting HERG inhibitors.


Astemizole/pharmacology , Calcium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/metabolism , Astemizole/metabolism , Binding Sites , Binding, Competitive , Calcium Channel Blockers/metabolism , Cell Line , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Ligands , Patch-Clamp Techniques , Radioligand Assay , Time Factors , Transfection , Tritium
...