Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Pharmacol Res Perspect ; 8(6): e00693, 2020 12.
Article in English | MEDLINE | ID: mdl-33280279

ABSTRACT

Melanoma is responsible for most skin cancer deaths, and its incidence continues to rise year after year. Different treatment options have been developed for melanoma depending on the stage of the disease. Despite recent advances in immuno- and targeted therapies, advanced melanoma remains incurable and thus an urgent need persists for safe and more effective melanoma therapeutics. In this study, we demonstrate that a novel compound MM902 (3-(3-(bromomethyl)-5-(4-(tert-butyl) phenyl)-1H-1,2,4-triazol-1-yl) phenol) exhibited potent efficacies in inhibiting the growth of different cancer cells, and suppressed tumor growth in a mouse xenograft model of malignant melanoma. Beginning with MM902 instead of specific targets, computational similarity- and docking-based approaches were conducted to search for known anticancer drugs whose structural features match MM902 and whose pharmacological target would accommodate an irreversible inhibitor. Peroxisome proliferator-activated receptor (PPAR) was computationally identified as one of the pharmacological targets and confirmed by in vitro biochemical assays. MM902 was shown to bind to PPARγ in an irreversible mode of action and to function as a selective antagonist for PPARγ over PPARα and PPARδ. It is hoped that MM902 will serve as a valuable research probe to study the functions of PPARγ in tumorigenesis and other pathological processes.


Subject(s)
Antineoplastic Agents/therapeutic use , Melanoma/drug therapy , Molecular Docking Simulation/methods , PPAR gamma/antagonists & inhibitors , Skin Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Computational Biology/methods , Dose-Response Relationship, Drug , Female , Humans , Melanoma/pathology , Mice , Mice, SCID , PPAR gamma/chemistry , Skin Neoplasms/pathology , Xenograft Model Antitumor Assays/methods , Melanoma, Cutaneous Malignant
2.
J Pharmacol Exp Ther ; 342(2): 273-87, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22537770

ABSTRACT

It has been demonstrated previously that immune cell activation and proliferation were sensitive to the effects of naltrindole, a nonpeptidic δ-opioid receptor-selective antagonist; therefore, we hypothesized that human multiple myeloma (MM) would be a valuable model for studying potential antineoplastic properties of naltrindole. [(3)H]naltrindole exhibited saturable, low-affinity binding to intact human MM cells; however, the pharmacological profile of the binding site differed considerably from the properties of δ-, κ-, and µ-opioid receptors, and opioid receptor mRNA was not detected in MM cells by reverse transcriptase-polymerase chain reaction. Naltrindole inhibited the proliferation of cultured human U266 MM cells in a time- and dose-dependent manner with an EC(50) of 16 µM. The naltrindole-induced inhibition of U266 cell proliferation was not blocked by a 10-fold molar excess of naltrexone, a nonselective opioid antagonist. Additive inhibition of MM cell proliferation was observed when using a combination of naltrindole with the histone deacetylase inhibitor sodium valproate, the proteasome inhibitor bortezomib, the glucocorticoid receptor agonist dexamethasone, and the HMG CoA reductase inhibitor simvastatin. Treatment of U266 cells with naltrindole significantly decreased the level of the active, phosphorylated form of the kinases, extracellular signal-regulated kinase and Akt, which may be related to its antiproliferative activity. The antiproliferative activity of naltrindole toward MM cells was maintained in cocultures of MM and bone marrow-derived stromal cells, mimicking the bone marrow microenvironment. In vivo, naltrindole significantly decreased tumor cell volumes in human MM cell xenografts in severe combined immunodeficient mice. We hypothesize that naltrindole inhibits the proliferation of MM cells through a nonopioid receptor-dependent mechanism.


Subject(s)
Antineoplastic Agents/pharmacology , Multiple Myeloma/drug therapy , Naltrexone/analogs & derivatives , Animals , Binding Sites , Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Mice , Mice, SCID , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Naltrexone/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Opioid/metabolism , Xenograft Model Antitumor Assays
3.
Bioorg Med Chem ; 17(17): 6442-50, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19646882

ABSTRACT

A novel family of 1,3,5-trisubstituted 1,2,4-triazoles was discovered as potent and selective ligands for the delta opioid receptor by rational design. Compound 5b exhibited low-nanomolar in vitro binding affinity (IC(50)=5.8 nM), excellent selectivity for the delta opioid receptor over the alternative mu and kappa opioid receptors, full agonist efficacy in receptor down-regulation and MAP kinase activation assays, and low-efficacy partial agonist activity in stimulation of GTPgammaS binding. The apparent discrepancy observed in these functional assays may stem from different signaling pathways involved in each case, as found previously for other G-protein coupled receptors. More biological studies are underway to better understand the differential stimulation of signaling pathways by these novel compounds.


Subject(s)
Receptors, Opioid, delta/agonists , Triazoles/chemistry , Catalytic Domain , Cell Differentiation , Cell Line , Computer Simulation , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Molecular Conformation , Receptors, Opioid, delta/metabolism , Signal Transduction , Triazoles/chemical synthesis , Triazoles/pharmacology
4.
Brain Res ; 1230: 13-26, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18656460

ABSTRACT

A clonal human embryonic kidney (HEK) 293 cell line was established that stably expressed the rat kappa-opioid receptor (rKOR) with a FLAG epitope at the amino terminus. The Kd for [3H]diprenorphine was 1.1+/-0.2 nM, and the Bmax was 2.6+/-0.4 pmol/mg. Dynorphin A (1-13), U69,593 and naloxone competitively inhibited [3H]diprenorphine binding with Ki values of 2.0, 18 and 18 nM, respectively, in good agreement with previously reported affinities for the unmodified receptor. U69,593 stimulated [35S]GTPgammaS binding in a concentration-dependent manner and caused phosphorylation of mitogen-activated protein (MAP) kinase, indicating that the activated epitope-tagged receptor triggered appropriate signaling pathways. Immunoblot analysis demonstrated that two immunoreactive receptor species with apparent molecular masses of 42 and 52 kDa were expressed. Previous studies indicated that the 42 kDa protein was localized intracellularly and was a precursor of the 52 kDa receptor, which was present at the cell surface. rKOR was extracted from transfected HEK 293 cell membranes with n-dodecyl-beta-D-maltopyranoside. Sequential use of wheat germ agglutinin chromatography, Sephacryl S300 gel filtration chromatography, anti-FLAG immunoaffinity chromatography and SDS/PAGE permitted purification of the 52 kDa receptor. MALDI-TOF mass spectrometry was used to identify peptides derived from rKOR following sequential in-gel digestion with trypsin and cyanogen bromide. Eighteen rKOR peptides were detected, corresponding to 27.1% coverage of the receptor. Precursor-selective MS/MS confirmed the identity of most of these peptides. In addition, we have identified heat shock protein 70 (HSP70) as a rKOR-interacting protein.


Subject(s)
Receptors, Opioid, kappa/isolation & purification , Amino Acid Sequence , Blotting, Western , Cell Line , Chromatography, Affinity , Chromatography, Agarose , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , HSP70 Heat-Shock Proteins/analysis , Humans , Immunochemistry , Immunoprecipitation , Mass Spectrometry , Membranes/chemistry , Membranes/metabolism , Protein Hydrolysates/chemistry , Radioligand Assay , Receptors, Opioid, kappa/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Wheat Germ Agglutinins/chemistry
5.
Life Sci ; 81(23-24): 1593-601, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17964607

ABSTRACT

Based on the promising opioid pharmacological profile of the peptide, Tyr-Pro-Trp-Gly-NH(2) (Tyr-W-MIF), Zadina et al. [Zadina, J.E., Hackler, L., Ge, L.-J., Kastin, A.J., 1997. A potent and selective endogenous agonist for the mu-opiate receptor. Nature 386, 499-5502] synthesized and screened other Gly(4)-substituted peptides, culminating in the synthesis of Tyr-Pro-Trp-Phe-NH(2) (endomorphin-1), which displayed high affinity and selectivity for the mu-opioid receptor. The amidated peptide was then isolated from bovine brain frontal cortex, as was a related peptide, Tyr-Pro-Phe-Phe-NH(2) (endomorphin-2), that displayed similar high affinity and selectivity for the mu-opioid receptor. The biosynthesis of the endomorphins in the brain remains obscure, since the putative precursor proteins for the peptides have not been identified. With the completion of the human genome sequencing project, we hypothesized that we should uncover the biological precursors of the peptides using a bioinformatic approach to search the current human proteome for proteins that contained the endomorphin peptide sequences followed by Gly-Lys/Arg, the consensus sequence for peptide alpha-amidation and precursor cleavage. Twelve proteins were identified that contained the endomorphin-1 Tyr-Pro-Trp-Phe sequence, however none contained the Tyr-Pro-Trp-Phe-Gly sequence necessary for alpha-amidation. Twenty-two distinct proteins contained the endomorphin-2 tetrapeptide sequence, and two of those contained the sequence, Tyr-Pro-Phe-Phe-Gly, however, none contained the requisite peptide-Gly-Lys/Arg sequence. Western blot analysis using an endomorphin-2 antibody detected 4 prominent proteins in mouse brain, necessitating reinterpretation of previous immunocytolocalization studies in the brain. Screening of the current human proteome yielded no evidence for endomorphin precursor proteins based on accepted biochemical criteria.


Subject(s)
Oligopeptides/genetics , Protein Precursors/genetics , Proteome , Animals , Blotting, Western , Brain Chemistry/physiology , Cell Line , Computational Biology , Databases, Genetic , Glycine/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Oligopeptides/biosynthesis , Protein Precursors/biosynthesis
6.
J Pharmacol Exp Ther ; 323(2): 614-25, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17720886

ABSTRACT

Ligand-induced regulation of the rat kappa-opioid receptor (rKOR) was investigated in human embryonic kidney 293 cells stably expressing the FLAG-tagged rKOR. Incubation of rKOR cells with naltrexone for 24 h increased the B(max) >3-fold, with no change in the affinity of [(3)H]diprenorphine. Two immunoreactive receptor species were present in cell lysates: naltrexone treatment caused a >3-fold increase in the 52-kDa species while decreasing the level of the 42-kDa species. Dynorphin(1-13), U69,593 [(5alpha,7alpha,8beta)-(+)-N-methyl-N-(7-[1-pyrrolidinyl]-1-oxaspiro[4,5]dec-8-yl)benzeneacetamide], or salvinorin A [2S,4aR,6aR,7R,9S,10aS, 10bR)-9-(acetyloxy)-2-(3-furanyl)dodecahydro-6a,10b-dimethyl-4,10-dioxo-2H-naphtho[2,1c]pyran-7-carboxylic acid methyl ester] treatment did not alter the level of immunoreactive rKOR protein, whereas etorphine, cyclazocine, naloxone, and naloxone methiodide increased the 52-kDa and decreased the 42-kDa rKOR bands. Receptor up-regulation was associated with an increase in the number of cell surface receptors and a 2-fold increase in the E(max) for guanosine 5'-O-(3-[(35)S]thio)triphosphate binding. Glycosidase digestion indicated that the 52- and 42-kDa receptors contained complex and high-mannose N-glycans, respectively, Pulse-chase analysis and glycosidase digestion sensitivities suggested that the 42-kDa rKOR species was a precursor of the 52-kDa species. Naltrexone did not alter rKOR mRNA levels or translational efficiency, and rKOR up-regulation was not inhibited by cycloheximide. Brefeldin A caused accumulation of intracellular rKOR intermediates, and coincubation with naltrexone increased the levels of the brefeldin-induced species significantly. These results suggest that select opioid ligands up-regulate rKOR by enhancing the rate of receptor folding and maturation and by protecting the receptor from degradation, resulting in an increase in the number of rKOR binding sites, immunoreactive protein, and functional receptors.


Subject(s)
Receptors, Opioid, kappa/genetics , Animals , Benzeneacetamides/pharmacokinetics , Brefeldin A/pharmacology , Cells, Cultured , Cycloheximide/pharmacology , Diprenorphine/pharmacology , Humans , Ligands , Naloxone/pharmacokinetics , Naloxone/pharmacology , Naltrexone/pharmacology , Pyrrolidines/pharmacokinetics , RNA, Messenger/analysis , Rats , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/metabolism , Transfection , Up-Regulation
7.
J Pharmacol Exp Ther ; 320(3): 1186-94, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17159161

ABSTRACT

This study was designed to test the hypothesis that inhibition of agonist-induced delta-receptor down-regulation would block the development of opioid tolerance in a cell-based model. A human embryonic kidney 293 cell line was established that expressed an epitope-tagged delta-opioid receptor (DOR). Treatment of DOR cells with Tyr-d-Ala-Gly-Phe-d-Leu-enkephalin (DADL) resulted in a time-dependent decrease in the B(max) of delta-opioid receptor binding sites and immunoreactive receptor protein. When cells were coincubated with the proteasome inhibitor N-benzyloxycarbonyl-l-leucyl-l-leucyl-l-leucinal (ZLLL) and DADL, the magnitude of the agonist-induced decrease in B(max) and immunoreactive receptor protein was reduced compared with DADL treatment alone. Acute treatment of DOR cells with DADL caused a 3-fold increase in the level of phosphorylated mitogen-activated protein (MAP) kinase. Prior exposure of DOR cells to DADL completely abrogated the agonist-induced activation of MAP kinase. When DOR cells were coincubated with DADL and ZLLL, the proteasome inhibitor prevented the loss of agonist activation of MAP kinase. Acute treatment of DOR cell membranes with DADL stimulated [(35)S]guanosine 5'-3-O-(thio-)triphosphate (GTPgammaS) binding. When DOR cells were preincubated with DADL, the agonist-induced increase in [(35)S]GTPgammaS binding was attenuated. Coincubation of ZLLL and agonist partially prevented the decreased responsiveness to agonist stimulation. The results of this study demonstrated that inhibition of agonist-induced down regulation with a proteasome inhibitor attenuated opioid tolerance in a cellular model, and suggest that coadministration of a proteasome inhibitor with chronic opioid agonist treatment may be useful for limiting opioid tolerance in vivo.


Subject(s)
Cell Membrane/drug effects , Enkephalin, Leucine-2-Alanine/pharmacology , Leupeptins/pharmacology , Proteasome Inhibitors , Receptors, Opioid, delta , Binding Sites , Cell Line , Cell Membrane/metabolism , Down-Regulation , Electrophoresis, Polyacrylamide Gel , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Kidney/cytology , Kidney/embryology , Ligands , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , Protein Binding , Radioligand Assay , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, delta/genetics , Transfection
8.
J Med Chem ; 49(14): 4044-7, 2006 Jul 13.
Article in English | MEDLINE | ID: mdl-16821764

ABSTRACT

We report the computer-aided design, chemical synthesis, and biological evaluation of a novel family of delta opioid receptor (DOR) antagonists containing a 1,2,4-triazole core structure that are structurally distinct from other known opioid receptor active ligands. Among those delta antagonists sharing this core structure, 8 exhibited strong binding affinity (K(i) = 50 nM) for the DOR and appreciable selectivity for delta over mu and kappa opioid receptors (delta/mu = 80; delta/kappa > 200).


Subject(s)
Receptors, Opioid, delta/antagonists & inhibitors , Triazoles/chemical synthesis , Cell Line , Humans , Ligands , Radioligand Assay , Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship , Triazoles/chemistry , Triazoles/pharmacology , Up-Regulation
9.
Brain Res Mol Brain Res ; 136(1-2): 54-64, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15893587

ABSTRACT

A mouse delta opioid receptor was engineered to contain a FLAG epitope at the amino-terminus and a hexahistidine tag at the carboxyl terminus to facilitate purification. Selection of transfected human embryonic kidney (HEK) 293 cells yielded a cell line that expressed the receptor with a B(max) of 10.5 pmol/mg protein. [3H]Bremazocine exhibited high affinity binding to the epitope-tagged delta opioid receptor with a K(D) of 1.4 nM. The agonists DADL, morphine, and DAMGO competitively inhibited bremazocine binding to the tagged delta receptor with K(I)'s of 0.9, 370, and 620 nM, respectively. Chronic treatment of cells expressing the epitope-tagged delta receptor with DADL resulted in downregulation of the receptor, indicating that the tagged receptor retained the capacity to mediate signal transduction. The delta receptor was solubilized from HEK 293 cell membranes with n-dodecyl-beta-d-maltoside in an active form that maintained high affinity bremazocine binding. Sequential use of Sephacryl S300 gel filtration chromatography, wheat germ agglutinin (WGA)-agarose chromatography, immobilized metal affinity chromatography, immunoaffinity chromatography, and SDS/PAGE permitted purification of the receptor. The purified delta opioid receptor was a glycoprotein that migrated on SDS/PAGE with an apparent molecular mass of 65 kDa. MALDI-TOF mass spectrometry was used to identify and characterize peptides derived from the delta opioid receptor following in-gel digestion with trypsin, and precursor-derived ms/ms confirmed the identity of peptides derived from enzymatic digestion of the delta opioid receptor.


Subject(s)
Mass Spectrometry , Receptors, Opioid, delta/analysis , Analgesics/pharmacokinetics , Benzomorphans/pharmacokinetics , Blotting, Western/methods , Cell Line , Chromatography, Affinity , Chromatography, Gel/methods , Humans , Models, Molecular , Molecular Weight , Radioligand Assay/methods , Receptors, Opioid, delta/chemistry , Receptors, Opioid, delta/drug effects , Receptors, Opioid, delta/isolation & purification , Solubility , Transfection/methods , Tritium/pharmacokinetics , Trypsin/pharmacology
10.
Brain Res Mol Brain Res ; 118(1-2): 119-31, 2003 Oct 21.
Article in English | MEDLINE | ID: mdl-14559361

ABSTRACT

A mouse mu opioid receptor was engineered to contain a FLAG epitope at the amino-terminus and a hexahistidine tag at the carboxyl-terminus to facilitate purification. Selection of transfected human embryonic kidney (HEK) 293 cells yielded a cell line that expressed the receptor with a B(max) of 10 pmol/mg protein. 3[H]Bremazocine exhibited high affinity binding to the epitope-tagged mu opioid receptor with a KD of 1.0 nM. The agonists [D-Ala(2),N-Me-Phe(4),Gly(5)-ol]enkephalin (DAMGO), morphine and [D-Ala(2),D-Leu(5)]enkephalin (DADL) competitively inhibited bremazocine binding to the tagged mu receptor with KI's of 3.5, 17 and 70 nM, respectively. Chronic treatment of cells expressing the epitope-tagged mu receptor with DAMGO resulted in down-regulation of the receptor, indicating that the tagged receptor retained the capacity to mediate signal transduction. The mu receptor was solubilized from HEK 293 cell membranes with n-dodecyl-beta-D-maltoside in an active form that maintained high affinity bremazocine binding. Sequential use of wheat germ agglutinin (WGA)-agarose chromatography, Sephacryl S300 gel filtration chromatography, immobilized metal affinity chromatography, immunoaffinity chromatography, and sodium dodecyl sulfate/polyacrylamide gel electrophoresis (SDS/PAGE) permitted purification of the receptor. The purified mu opioid receptor was a glycoprotein that migrated on SDS/PAGE with an apparent molecular mass of 80 kDa. Matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry was used to identify and characterize peptides derived from the mu opioid receptor following in-gel digestion with trypsin or chymotrypsin, and precursor-derived tandem mass spectrometry (ms/ms) confirmed the identity of several peptides derived from enzymatic digestion of the mu opioid receptor.


Subject(s)
Cell Membrane/metabolism , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/isolation & purification , Analgesics/metabolism , Analgesics/pharmacology , Benzomorphans/metabolism , Benzomorphans/pharmacology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Cell Line , Down-Regulation/drug effects , Down-Regulation/physiology , Epitopes , Humans , Ligands , Mass Spectrometry , Narcotics/metabolism , Narcotics/pharmacology , Peptides/chemistry , Peptides/isolation & purification , Receptors, Opioid, mu/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Solubility
11.
Regul Pept ; 105(1): 9-14, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11853866

ABSTRACT

We reported recently that the ubiquitin-proteasome pathway is involved in agonist-induced down regulation of mu and delta opioid receptors [J. Biol. Chem. 276 (2001) 12345]. While evaluating the effects of various protease inhibitors on agonist-induced opioid receptor down regulation, we observed that while the peptide aldehyde, leupeptin (acetyl-L-Leucyl-L-Leucyl-L-Arginal), did not affect agonist-induced down regulation, leupeptin at submillimolar concentrations directly inhibited radioligand binding to opioid receptors. In this study, the inhibitory activity of leupeptin on radioligand binding was characterized utilizing human embryonic kidney (HEK) 293 cell lines expressing transfected mu, delta, or kappa opioid receptors. The rank order of potency for leupeptin inhibition of [3H]bremazocine binding to opioid receptors was mu > delta > kappa. In contrast to the effect of leupeptin, the peptide aldehyde proteasome inhibitor, MG 132 (carbobenzoxy-L-Leucyl-L-Leucyl-L-Leucinal), had significantly less effect on bremazocine binding to mu, delta, or kappa opioid receptors. We propose that leupeptin inhibits ligand binding by reacting reversibly with essential sulfhydryl groups that are necessary for high-affinity ligand/receptor interactions.


Subject(s)
Cysteine Proteinase Inhibitors/pharmacology , Leupeptins/pharmacology , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/metabolism , Benzomorphans/antagonists & inhibitors , Benzomorphans/metabolism , Cell Line , Cell Membrane/metabolism , Humans , Ligands , Protein Binding/drug effects , Receptors, Opioid, kappa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...