Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Mol Pharmacol ; 98(4): 462-474, 2020 10.
Article in English | MEDLINE | ID: mdl-32958572

ABSTRACT

Opioid receptors (ORs) convert extracellular messages to signaling events by coupling to the heterotrimeric G proteins, Gα•ßγ Classic pharmacological methods, such as [35S]GTPγS binding and inhibition of cyclic AMP production, allow for general opioid characterization, but they are subject to the varying endogenous Gα proteins in a given cell type. Bioluminescence resonance energy transfer (BRET) technology offers new insight by allowing the direct observation of Gα subunit-specific effects on opioid pharmacology. Using a Venus-tagged Gßγ and nanoluciferase-tagged truncated G protein receptor kinase 3, an increase in BRET signal correlated with OR activation mediated by a specific Gα protein. The magnitude of the BRET signal was normalized to the maximum response obtained with 10 µM 2-(3,4-dichlorophenyl)-N-methyl-N-[(1R,2R)-2-pyrrolidin-1-ylcyclohexyl]acetamide (U50,488) for the kappa OR (KOR). Opioids reached equilibrium with the KOR, and concentration-response curves were generated. Although the full agonists U50,488, salvinorin A, nalfurafine, and dynorphin peptides were equally efficacious regardless of the Gα subunit present, the concentration-response curves were leftward shifted when the KOR was signaling through Gαz compared with other Gαi/o subunits. In contrast, the Gα subunit distinctly affected both the efficacy and potency of partial kappa agonists, such as the benzomorphans, and the classic mu opioid antagonists, naloxone, naltrexone, and nalmefene. For example, (-)pentazocine had EC50 values of 7.3 and 110 nM and maximal stimulation values of 79% and 35% when the KOR signaled through Gαz and Gαi1, respectively. Together, these observations suggest KOR pharmacology varies based on the specific Gα subunit coupled to the KOR. SIGNIFICANCE STATEMENT: Opioid receptors couple to various heterotrimeric Gαßγ proteins to convert extracellular cues to precise intracellular events. This paper focuses on how the various inhibitory Gα subunits influence the pharmacology of full and partial agonists at the kappa opioid receptor. Using a bioluminescent assay, the efficacy and potency of kappa opioids was determined. Opioid signaling was more potent through Gαz compared with other Gα proteins. These observations suggest that Gαz may impact opioid pharmacology and cellular physiology more than previously thought.


Subject(s)
Analgesics, Opioid/pharmacology , Bioluminescence Resonance Energy Transfer Techniques/methods , GTP-Binding Protein alpha Subunits/metabolism , Receptors, Opioid, kappa/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Diterpenes, Clerodane/pharmacology , Dynorphins/pharmacology , HEK293 Cells , Humans , Morphinans/pharmacology , Signal Transduction/drug effects , Spiro Compounds/pharmacology
2.
J Pharmacol Exp Ther ; 367(2): 267-281, 2018 11.
Article in English | MEDLINE | ID: mdl-30108159

ABSTRACT

A combination of buprenorphine (BUP) and samidorphan (SAM) at a 1:1 (mg/mg) fixed-ratio dose is being investigated as an adjunctive treatment of major depressive disorder (BUP/SAM, ALKS 5461). Both [3H]BUP and [3H]SAM bound to the µ-, κ-, and δ-opioid receptors (MOR, KOR, and DOR, respectively) with Kd values of 3 nM or less. [3H]BUP dissociated from the MOR more slowly than [3H]SAM did. In the [35S]GTPγS assay, BUP was a partial agonist at the MOR, KOR, and DOR. SAM was an antagonist at the MOR and a partial agonist at the KOR and DOR. The pharmacology of the combination of SAM and BUP was characterized at ratios like the molar ratios of both compounds at steady state in humans. In all assessments, SAM reduced the efficacy of BUP at the MOR without altering its potency. At the KOR, SAM had no significant effect on the activity of BUP. In bioluminescent resonance energy transfer assays, SAM, naltrexone, and naloxone were partial agonists when the MOR was coupled to the Gα oB and Gα z, and were antagonists when coupled to Gα i At the KOR, SAM was a partial agonist activating Gα oA and Gα oB and a full agonist in stimulating Gα z SAM inhibited BUP's recruitment of ß-arrestin to the MOR, suggesting an attenuation of BUP's efficacy in activating G proteins correlated with an inhibition of ß-arrestin recruitment. The collective data suggest that SAM attenuates the efficacy of BUP under all conditions tested at the MOR and DOR but had little effect on BUP activity at the KOR.


Subject(s)
Buprenorphine/pharmacology , Depressive Disorder, Major/drug therapy , Naltrexone/analogs & derivatives , Animals , CHO Cells , Cell Line , Cricetulus , Drug Combinations , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Naloxone/pharmacology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , beta-Arrestins/metabolism
3.
Brain Behav Immun ; 67: 152-162, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28844812

ABSTRACT

Patients receiving the cytokine immunotherapy, interferon-alpha (IFN-α) frequently present with neuropsychiatric consequences and cognitive impairments. Patients (25-80%) report symptoms of depression, including, anhedonia, irritability, fatigue and impaired motivation. Our lab has previously demonstrated treatment (170,000IU/kg sc, 3 times per week for 4weeks) of the pro-inflammatory cytokine, IFN-α, induced a depressive phenotype in rats in the forced swim test (FST). Here, we examine the biological mechanisms underlying behavioral changes induced by IFN-α, which may be reflective of mechanisms underlying inflammation associated depression. We also investigate the potential of 3-carboxamido seco-nalmefene (3CS-nalmefene), a novel opioid modulator (antagonist at mu and partial agonist at kappa and delta opioid receptors in vitro), to reverse IFN-α induced changes. In vitro radioligand receptor binding assays and the [35S] GTPγS were performed to determine the affinity of 3CS-nalmefene for the mu, kappa and delta opioid receptors. IFN-α treatment increased circulating and central markers of inflammation and hypothalamic-pituitaryadrenal (HPA) axis activity (IL-6, IL-1ß and corticosterone) while increasing immobility in the FST, impairing of object displacement learning in the object exploration task (OET), and decreasing neuronal proliferation and brain-derived neurotrophic factor (BDNF) in the hippocampus. Treatment with 3CS-nalmefene (0.3mg/kg/sc twice per day, 3 times per week for 4weeks) prevented IFN-α-induced immobility in the FST and impaired object displacement learning. In addition, 3CS-nalmefene prevented IFN-α-induced increases in inflammation and hyperactivity of the HPA-axis, the IFN-α-induced reduction in both neuronal proliferation and BDNF expression in the hippocampus. Overall, these preclinical data would support the hypothesis that opioid receptor modulation is a relevant target for treatment of depression.


Subject(s)
Antidepressive Agents/administration & dosage , Depressive Disorder/drug therapy , Naltrexone/analogs & derivatives , Narcotic Antagonists/administration & dosage , Receptors, Opioid/agonists , Animals , Anxiety/chemically induced , Anxiety/drug therapy , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation/drug effects , Depression/chemically induced , Depression/drug therapy , Depressive Disorder/chemically induced , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Interferon-alpha/administration & dosage , Male , Naltrexone/administration & dosage , Neurons/drug effects , Neurons/metabolism , Rats, Wistar
4.
Bioorg Med Chem Lett ; 23(7): 2128-33, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23434225

ABSTRACT

Derivatives of the lead compound N-BPE-8-CAC (1) where each CH of the biphenyl group was individually replaced by N were prepared in hopes of identifying high affinity ligands with improved aqueous solubility. Compared to 1, binding affinities of the five possible pyridinyl derivatives for the µ opioid receptor were between threefold lower to fivefold higher with the Ki of the most potent compound being 0.064 nM. Docking of 8-CAC (2) into the unliganded binding site of the mouse µ opioid receptor (pdb: 4DKL) revealed that 8-CAC and ß-FNA (from 4DKL) make nearly identical interactions with the receptor. However, for 1 and the new pyridinyl derivatives 4-8, binding is not tolerated in the 8-CAC binding mode due to the steric constraints of the large N-substituents. Either an alternative binding mode or rearrangement of the protein to accommodate these modifications may account for their high binding affinity.


Subject(s)
Biphenyl Compounds/pharmacology , Cyclopropanes/pharmacology , Pyridines/pharmacology , Receptors, Opioid/chemistry , Animals , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/chemistry , Cyclopropanes/chemical synthesis , Cyclopropanes/chemistry , Dose-Response Relationship, Drug , Humans , Ligands , Mice , Models, Molecular , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Receptors, Opioid/metabolism , Structure-Activity Relationship
5.
J Pharmacol Exp Ther ; 342(3): 642-53, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22637724

ABSTRACT

The Kv1.3 channel is a recognized target for pharmaceutical development to treat autoimmune diseases and organ rejection. ShK-186, a specific peptide inhibitor of Kv1.3, has shown promise in animal models of multiple sclerosis and rheumatoid arthritis. Here, we describe the pharmacokinetic-pharmacodynamic relationship for ShK-186 in rats and monkeys. The pharmacokinetic profile of ShK-186 was evaluated with a validated high-performance liquid chromatography-tandem mass spectrometry method to measure the peptide's concentration in plasma. These results were compared with single-photon emission computed tomography/computed tomography data collected with an ¹¹¹In-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid-conjugate of ShK-186 to assess whole-blood pharmacokinetic parameters as well as the peptide's absorption, distribution, and excretion. Analysis of these data support a model wherein ShK-186 is absorbed slowly from the injection site, resulting in blood concentrations above the Kv1.3 channel-blocking IC50 value for up to 7 days in monkeys. Pharmacodynamic studies on human peripheral blood mononuclear cells showed that brief exposure to ShK-186 resulted in sustained suppression of cytokine responses and may contribute to prolonged drug effects. In delayed-type hypersensitivity, chronic relapsing-remitting experimental autoimmune encephalomyelitis, and pristane-induced arthritis rat models, a single dose of ShK-186 every 2 to 5 days was as effective as daily administration. ShK-186's slow distribution from the injection site and its long residence time on the Kv1.3 channel contribute to the prolonged therapeutic effect of ShK-186 in animal models of autoimmune disease.


Subject(s)
Autoimmune Diseases/drug therapy , Kv1.3 Potassium Channel/antagonists & inhibitors , Proteins/pharmacology , T-Lymphocytes/drug effects , Absorption/drug effects , Absorption/immunology , Animals , Arthritis/drug therapy , Arthritis/immunology , Arthritis/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Humans , Inhibitory Concentration 50 , Kv1.3 Potassium Channel/immunology , Kv1.3 Potassium Channel/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Macaca fascicularis , Potassium Channel Blockers/immunology , Potassium Channel Blockers/pharmacokinetics , Potassium Channel Blockers/pharmacology , Proteins/pharmacokinetics , Rats , Rats, Sprague-Dawley , Saimiri , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tissue Distribution/drug effects , Tissue Distribution/immunology
6.
Neuropharmacology ; 202: 108858, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34715121

ABSTRACT

Because of increased opioid misuse, there is a need to identify new targets for minimizing opioid tolerance, and physical and psychological dependence. Previous studies showed that fibroblast growth factor 21 (FGF21) decreased alcohol and sweet preference in mice. In this study, FGF21-transgenic (FGF21-Tg) mice, expressing high FGF21 serum levels, and wildtype (WT) C57BL/6J littermates were treated with morphine and saline to determine if differences exist in their physiological and behavioral responses to opioids. FGF21-Tg mice displayed reduced preference for morphine in the conditioned place preference assay compared to WT littermates. Similarly, FGF21-Tg mice had an attenuation of the magnitude and rate of acute morphine antinociceptive tolerance development, and acute and chronic morphine physical dependence, but exhibited no change in chronic morphine antinociceptive tolerance. The ED50 values for morphine-induced antinociception in the 55 °C hot plate and the 55 °C warm-water tail withdrawal assays were similar in both strains of mice. Likewise, FGF21-Tg and WT littermates had comparable responses to morphine-induced respiratory depression. Overall, FGF21-Tg mice had a decrease in the development of acute analgesic tolerance, and the development of physical dependence, and morphine preference. FGF21 and its receptor have therapeutic potential for reducing opioid withdrawal symptoms and craving, and augmenting opioid therapeutics for acute pain patients to minimize tolerance development.


Subject(s)
Drug Tolerance/genetics , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/physiology , Morphine Dependence/genetics , Morphine/adverse effects , Nociception/drug effects , Animals , Behavior, Animal/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/genetics , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/therapy
7.
Bioorg Med Chem Lett ; 20(5): 1507-9, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20144870

ABSTRACT

A series of bivalent hydroxy ether butorphan ligands were prepared and their binding affinities at the opioid receptors determined. Addition of a hydroxy group to a hydrocarbon chain can potentiate binding affinity up to 27- and 86-fold at the mu and kappa opioid receptors, respectively. Two bivalent ligands with sub-nanomolar binding affinity at the mu and kappa opioid receptors were discovered.


Subject(s)
Analgesics/chemistry , Ligands , Morphinans/chemistry , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Analgesics/chemical synthesis , Analgesics/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Morphinans/chemical synthesis , Morphinans/pharmacology , Protein Binding , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/antagonists & inhibitors
8.
Bioorg Med Chem Lett ; 18(16): 4474-6, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18674902

ABSTRACT

We synthesized several hydrophobic esters and ethers of butorphanol and assessed their affinities at opioid receptors in CHO cell membranes. Tested compounds displayed moderate to high affinities to the mu and kappa receptors. The findings accord with previous evidence of a lipophilic binding pocket in the opioid receptors that can be accessed to afford good binding affinity without the need for a phenolic hydrogen-bond donor group. The most potent (K(i)=61 pM at mu and 48 pM at kappa) novel agent was (-)-N-cyclobutylmethylmorphinan-3-yl-14-ol phenoxyacetate (4d).


Subject(s)
Butorphanol/chemistry , Butorphanol/chemical synthesis , Chemistry, Pharmaceutical/methods , Narcotic Antagonists , Receptors, Opioid/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Design , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Chemical , Phenol , Protein Binding , Temperature
9.
J Med Chem ; 50(9): 2254-8, 2007 May 03.
Article in English | MEDLINE | ID: mdl-17407276

ABSTRACT

Our investigation of bivalent ligands at mu, delta, and kappa opioid receptors is focused on the preparation of ligands containing kappa agonist and mu agonist/antagonist pharmacophores at one end joined by a chain containing the mu antagonist pharmacophores (naltrexone, naloxone, or nalbuphine) at the other end. These ligands were evaluated in vitro by their binding affinity at mu, delta, and kappa opioid receptors and their relative efficacy in the [35S]GTPgammaS assay.


Subject(s)
Morphinans/chemical synthesis , Nalbuphine/analogs & derivatives , Naloxone/analogs & derivatives , Naltrexone/analogs & derivatives , Receptors, Opioid, delta/drug effects , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, mu/drug effects , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Humans , Ligands , Morphinans/chemistry , Morphinans/pharmacology , Nalbuphine/chemical synthesis , Nalbuphine/pharmacology , Naloxone/chemical synthesis , Naloxone/pharmacology , Naltrexone/chemical synthesis , Naltrexone/pharmacology , Radioligand Assay , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Structure-Activity Relationship
10.
J Med Chem ; 50(11): 2747-51, 2007 May 31.
Article in English | MEDLINE | ID: mdl-17488103

ABSTRACT

To further extend the structure-activity relationships of levorphanol, two series of novel morphinans were prepared by incorporation of an indole or aminothiazole fragment to the hexyl ring (ring C) in levorphanol. Such morphinans differed from previously reported ligands in that such indole- or aminothiazole-containing morphinans displayed enhanced binding affinity to the delta opioid receptor, while the affinity to kappa and micro receptors was slightly reduced.


Subject(s)
Analgesics, Opioid/chemical synthesis , Indoles/chemical synthesis , Levorphanol/analogs & derivatives , Levorphanol/chemical synthesis , Thiazoles/chemical synthesis , Analgesics, Opioid/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Indoles/pharmacology , Levorphanol/pharmacology , Radioligand Assay , Structure-Activity Relationship , Thiazoles/pharmacology
11.
Mol Cancer Ther ; 5(7): 1817-26, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16891468

ABSTRACT

Dysregulation of enzymes involved in prostaglandin biosynthesis plays a critical role in influencing the biological behavior and clinical outcome of several tumors. In human gliomas, overexpression of cyclooxygenase-2 has been linked to increased aggressiveness and poor prognosis. In contrast, the role of prostaglandin E synthase in influencing the biological behavior of human gliomas has not been established. We report that constitutive expression of the microsomal prostaglandin E synthase-1 (mPGES-1) is associated with increased prostaglandin E(2) (PGE(2)) production and stimulation of growth in the human astroglioma cell line U87-MG compared with human primary astrocytes. Consistently, pharmacologic and genetic inhibition of mPGES-1 activity and expression blocked the release of PGE(2) from U87-MG cells and decreased their proliferation. Conversely, exogenous PGE(2) partially overcame the antiproliferative effects of mPGES-1 inhibition and stimulated U87-MG cell proliferation in the absence of mPGES-1 inhibitors. The EP2/EP4 subtype PGE(2) receptors, which are linked to stimulation of adenylate cyclase, were expressed in U87-MG cells to a greater extent than in human astrocytes. PGE(2) increased cyclic AMP levels and stimulated protein kinase A (PKA) activity in U87-MG cells. Treatment with a selective type II PKA inhibitor decreased PGE(2)-induced U87-MG cell proliferation, whereas a selective type I PKA inhibitor had no effect. Taken together, these results are consistent with the hypothesis that mPGES-1 plays a critical role in promoting astroglioma cell growth via PGE(2)-dependent activation of type II PKA.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/metabolism , Glioma/enzymology , Intramolecular Oxidoreductases/metabolism , Astrocytes/drug effects , Astrocytes/enzymology , Cell Proliferation/drug effects , Cyclic AMP-Dependent Protein Kinase Type II , Dinoprostone/pharmacology , Glioma/pathology , Humans , Intramolecular Oxidoreductases/genetics , Microsomes/enzymology , Prostaglandin-E Synthases , Tumor Cells, Cultured
12.
J Med Chem ; 49(1): 256-62, 2006 Jan 12.
Article in English | MEDLINE | ID: mdl-16392810

ABSTRACT

A series of homo- and heterodimeric ligands containing kappa agonist and mu agonist/antagonist pharmacophores joined by a linker chain of varying lengths was synthesized and evaluated in vitro by their binding affinity at mu, delta, and kappa opioid receptors. The functional activities of these compounds were measured in the [(35)S]GTPgammaS binding assay. The data suggest that the stereochemistry of the pharmacophores, the N-substituents of the pharmacophore, ester linkages, and the spacer length were crucial factors for optimum interactions of such ligands at opioid receptor binding sites. These novel ligands as well as their pharmacological properties will serve as the basis for our continuing investigation of such bivalent ligands as probes of the opioid receptor oligomerization phenomena and for in vivo studies as analgesics.


Subject(s)
Morphinans/chemical synthesis , Morphinans/pharmacology , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, kappa , Receptors, Opioid, mu , Animals , CHO Cells , Cricetinae , Drug Evaluation, Preclinical , Humans , In Vitro Techniques , Ligands , Molecular Conformation , Morphinans/chemistry , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Stereoisomerism , Structure-Activity Relationship
13.
J Med Chem ; 49(18): 5640-3, 2006 Sep 07.
Article in English | MEDLINE | ID: mdl-16942040

ABSTRACT

Here, we report the synthesis of a designed multi-pharmacophore ligand derived from the linkage of a delta selective peptide antagonist (Dmt-Tic) and a mu/kappa morphinan agonist butorphan (MCL 101) through a two methylene spacer. The new compound MCL 450 maintains the same characteristics as those the two reference compounds. MCL 450 represents a useful starting point for the synthesis of other multiple opioid ligands endowed with analgesic properties with low tolerance and dependence.


Subject(s)
Dipeptides/chemical synthesis , Morphinans/chemical synthesis , Narcotic Antagonists , Receptors, Opioid/agonists , Tetrahydroisoquinolines/chemical synthesis , Animals , CHO Cells , Cricetinae , Cricetulus , Dipeptides/chemistry , Dipeptides/pharmacology , Ligands , Morphinans/chemistry , Morphinans/pharmacology , Radioligand Assay , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists , Structure-Activity Relationship , Tetrahydroisoquinolines/chemistry , Tetrahydroisoquinolines/pharmacology
14.
Mol Cancer Ther ; 2(1): 1-7, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12533667

ABSTRACT

Angiogenesis plays a crucial role in tumor development and growth. The present investigation was undertaken to test the potential involvement of the cyclooxygenase-2 (COX-2) pathway in the regulation of angiogenesis and growth in pancreatic cancer. We compared the angiogenic characteristics of a COX-2-positive human pancreatic tumor cell line, BxPC-3, with those of a COX-2-negative pancreatic tumor cell line, AsPC-1. Cultured BxPC-3 cells promoted a marked increase of endothelial cell migration in comparison with migration that occurred in the absence of cancer cells. Furthermore, BxPC-3 cell culture supernatants induced endothelial cell capillary morphogenesis in vitro and neovascularization in vivo. In contrast, cultured AsPC-1 cells elicited a modest effect on endothelial cell migration and neovascularization in vivo. Pretreatment of BxPC-3 cells with the selective COX-2 inhibitor NS-398 (50 micro M) dramatically decreased angiogenic responses of endothelial cells. NS-398 (25-100 micro M) caused inhibition of BxPC-3 cell proliferation but had no effect on AsPC-1 cell growth. SC-560, a selective COX-1 inhibitor, had no effect on growth of either cell lines. These results suggest an involvement of COX-2 in the control of tumor-dependent angiogenesis and growth in certain pancreatic cancers and provide the rational for inhibition of the COX pathway as an effective therapeutic approach for pancreatic tumors.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Isoenzymes/physiology , Neovascularization, Pathologic/enzymology , Nitrobenzenes/pharmacology , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Prostaglandin-Endoperoxide Synthases/physiology , Sulfonamides/pharmacology , Capillaries/drug effects , Capillaries/pathology , Cell Division/drug effects , Cell Division/physiology , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Dinoprostone/metabolism , Endothelial Growth Factors/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Gene Expression Regulation, Enzymologic/drug effects , Homeostasis , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Lymphokines/metabolism , Membrane Proteins , Neovascularization, Pathologic/prevention & control , Pancreatic Neoplasms/enzymology , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
15.
J Med Chem ; 58(15): 5728-41, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26125201

ABSTRACT

Glycopeptides related to ß-endorphin penetrate the blood-brain barrier (BBB) of mice to produce antinociception. Two series of glycopeptides were assessed for opioid receptor binding affinity. Attempts to alter the mu-selectivity of [D-Ala(2),N-MePhe(4),Gly-ol(5)]enkephalin (DAMGO)-related glycopeptides by altering the charged residues of the amphipathic helical address were unsuccessful. A series of pan-agonists was evaluated for antinociceptive activity (55 °C tail flick) in mice. A flexible linker was required to maintain antinociceptive activity. Circular dichroism (CD) in H2O, trifluoroethanol (TFE), and SDS micelles confirmed the importance of the amphipathic helices (11s → 11sG → 11) for antinociception. The glycosylated analogues showed only nascent helices and random coil conformations in H2O. Chemical shift indices (CSI) and nuclear Overhauser effects (NOE) with 600 MHz NMR and CD confirmed helical structures in micelles, which were rationalized by molecular dynamics calculations. Antinociceptive studies with mice confirm that these glycosylated endorphin analogues are potential drug candidates that penetrate the BBB to produce potent central effects.


Subject(s)
Central Nervous System/drug effects , Glycopeptides/pharmacology , Opioid Peptides/pharmacology , Amino Acid Sequence , Animals , Circular Dichroism , Glycopeptides/chemistry , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Molecular Sequence Data , Opioid Peptides/chemistry , Protein Conformation , Receptors, Opioid, mu/drug effects
16.
J Med Chem ; 47(1): 165-74, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14695830

ABSTRACT

A series of 10-ketomorphinan analogues were synthesized, and their binding affinity at all three opioid receptors was investigated. In most cases, high affinity at micro and kappa receptors, and lower affinity at delta receptor was observed, resulting in good selectivity for micro and kappa receptors. A wide range of substituents can be accommodated on the nitrogen position. The N-(S)-tetrahydrofurfuryl analogue 11 displayed the highest affinity at all three receptors. The N-cyclobutylmethyl analogue 13 gave both high affinity and selectivity at kappa receptor, and N-2-phenylethyl analogue 18 exhibited good affinity and selectivity at micro receptor. Further modifications of the 3-substituent indicated that one H-bond donor was an essential requirement for good affinity at micro and kappa receptors. Similar modifications were investigated at the 3-OH group of morphinans: levorphanol (2a), cyclorphan (2b), and MCL-101 (2c) lacking the 10-keto group. The 3-amino bioisosteric analogues (40 and 41) displayed reasonably good affinity at micro and kappa receptors. The 3-carboxamido replacement (compounds 46-48) in the morphinan subseries resulted in similar affinities comparable to their corresponding 3-OH congeners. The high affinity of these carboxamido analogues, along with their greater lipophilicity and metabolic stability, make them promising candidates for further pharmacological investigation.


Subject(s)
Morphinans/chemical synthesis , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Animals , Brain/metabolism , CHO Cells , Cricetinae , Guinea Pigs , Hydrogen Bonding , In Vitro Techniques , Ligands , Models, Molecular , Molecular Conformation , Morphinans/chemistry , Morphinans/pharmacology , Radioligand Assay , Structure-Activity Relationship
17.
J Med Chem ; 46(24): 5162-70, 2003 Nov 20.
Article in English | MEDLINE | ID: mdl-14613319

ABSTRACT

A novel series of morphinans were synthesized, and their binding affinity at and functional selectivity for micro, delta, and kappa opioid receptors were evaluated. These dimeric ligands can be viewed as dimeric morphinans, which were formed by coupling two identical morphinan pharmacophores (cyclorphan (1) or MCL 101 (2)) with varying connecting spacers. Ligands 6 and 7 with alkyl spacers on the nitrogen position and ligands 8 and 9 in which the two morphinan pharmacophores were coupled by ether moieties at the 3-hydroxyl positions showed significant decrease in affinity at all three opioid receptors. An improvement in the affinity was achieved by introducing an ester moiety as the spacer in the dimeric morphinans. It was observed that the affinity of these ligands was sensitive to the character and length of the spacer. Compound 13 (MCL-139) with a 4-carbon ester spacer, compound 17 (MCL-144) containing a 10-carbon spacer, and compound 19 (MCL-145) with the conformationally constrained fumaryl spacer were the most potent ligands in this series, displaying excellent affinities at micro and kappa receptors (K(i) = 0.09-0.2 nM at micro and K(i) = 0.078-0.049 nM at kappa), which were comparable to the parent compound 2. Ligand 12, a compound containing only one morphinan pharmacophore and a long-chain ester group, had affinity at both micro and kappa receptors almost identical to that of the parent ligand 2. In the [(35)S]GTPgammaS binding assay, ligands 13, 17, and 19 and their parent morphinans 1 and 2 stimulated [(35)S]GTPgammaS binding mediated by the micro and kappa receptors. Compounds 13 and 17 were full kappa agonists and partial micro agonists, while compound 19 was a partial agonist at both micro and kappa receptors. These novel ligands, as well as their interesting pharmacological properties, will serve as the basis for our continuing investigation of the dimeric ligands as potential probes for the pharmacotherapy of cocaine abuse and may also open new avenues for the characterization of opioid receptors.


Subject(s)
Morphinans/chemical synthesis , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Animals , CHO Cells , Cricetinae , Dimerization , Drug Design , Humans , Ligands , Morphinans/chemistry , Morphinans/pharmacology , Radioligand Assay , Structure-Activity Relationship
19.
J Med Chem ; 57(6): 2237-46, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24576160

ABSTRACT

Glycosylated ß-endorphin analogues of various amphipathicity were studied in vitro and in vivo in mice. Opioid binding affinities of the O-linked glycopeptides (mono- or disaccharides) and unglycosylated peptide controls were measured in human receptors expressed in CHO cells. All were pan-agonists, binding to µ-, δ-, or κ-opioid receptors in the low nanomolar range (2.2-35 nM K(i)'s). The glycoside moiety was required for intravenous (i.v.) but not for intracerebroventricular (i.c.v.) activity. Circular dichroism and NMR indicated the degree of helicity in H2O, aqueous trifluoroethanol, or micelles. Glycosylation was essential for activity after i.v. administration. It was possible to manipulate the degree of helicity by the alteration of only two amino acid residues in the helical address region of the ß-endorphin analogues without destroying µ-, δ-, or κ-agonism, but the antinociceptive activity after i.v. administration could not be directly correlated to the degree of helicity in micelles.


Subject(s)
Analgesics/chemical synthesis , Analgesics/pharmacology , Glycopeptides/chemistry , Glycopeptides/pharmacology , beta-Endorphin/analogs & derivatives , beta-Endorphin/pharmacology , Animals , CHO Cells , Circular Dichroism , Cricetinae , Cricetulus , Drug Design , Glycopeptides/chemical synthesis , Humans , Injections, Intravenous , Injections, Intraventricular , Magnetic Resonance Spectroscopy , Male , Mice , Micelles , Models, Molecular , Protein Conformation , Protein Structure, Secondary , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
20.
ACS Chem Neurosci ; 5(2): 93-9, 2014 Feb 19.
Article in English | MEDLINE | ID: mdl-24393077

ABSTRACT

A series of levo- and dextromorphinan pairs have been synthesized and evaluated for their affinities to the mu, kappa, and delta opioid receptors, the N-methyl-D-aspartate (NMDA) channel, and sigma 1 and 2 receptors. It was found that levo isomers tended to have higher affinities at the opioid receptors and moderate to high affinities to the NMDA and sigma receptors, while dextro isomers tended to have lower affinities to the opioid receptors but comparatively higher affinities to the NMDA and sigma receptors. This series of compounds have interesting and complex pharmacological profiles, and merit further investigation as potential therapies for drug abuse treatment.


Subject(s)
Morphinans/pharmacology , N-Methylaspartate/drug effects , Receptors, Opioid/drug effects , Receptors, sigma/drug effects , Animals , Humans , Male , Morphinans/chemical synthesis , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL