375 results on '"Philip S Portoghese"'
Search Results
2. A bivalent compound targeting CCR5 and the mu opioid receptor treats inflammatory arthritis pain in mice without inducing pharmacologic tolerance
- Author
-
Raini Dutta, Mary M. Lunzer, Jennifer L. Auger, Eyup Akgün, Philip S. Portoghese, and Bryce A. Binstadt
- Subjects
Analgesia ,CCR5 ,Chemokine receptor ,Heteromer ,Inflammation ,Opioid receptor ,Diseases of the musculoskeletal system ,RC925-935 - Abstract
Abstract Background Pain accompanies rheumatoid arthritis and other chronic inflammatory conditions and is difficult to manage. Although opioids provide potent analgesia, chronic opioid use can cause tolerance and addiction. Recent studies have demonstrated functional interactions between chemokine and opioid receptor signaling pathways. Reported heterodimerization of chemokine and opioid receptors led our group to develop bivalent compounds that bind both types of receptors, with the goal of targeting opioids to sites of inflammation. MCC22 is a novel bivalent compound containing a CCR5 antagonist and mu opioid receptor (MOR) agonist pharmacophores linked through a 22-atom spacer. We evaluated the efficacy of MCC22 in the K/B.g7 T-cell receptor transgenic mouse model of spontaneous inflammatory arthritis. Methods MCC22 or morphine was administered intraperitoneally at varying doses to arthritic K/B.g7 mice or nonarthritic control mice. Mechanical pain hypersensitivity was measured each day before and after drug administration, using the electronic von Frey test. The potency of MCC22 relative to that of morphine was calculated. Functional readouts of pain included grip strength and nesting behavior. A separate dosing regimen was used to determine whether the drugs induced pharmacologic tolerance. Results MCC22 provided ~ 3000-fold more potent analgesia than morphine in this model. Daily treatment with MCC22 also led to a cumulative analgesic effect, reducing the daily baseline pain level. MCC22 produced no observable analgesic effect in nonarthritic control mice. Importantly, repeated administration of MCC22 did not induce pharmacologic tolerance, whereas a similar regimen of morphine did. Both grip strength and nesting behaviors improved among arthritic mice treated with MCC22. Ankle thickness and arthritis scores were not affected by MCC22. The analgesic effect of MCC22 was abolished in K/B.g7 mice genetically lacking CCR5, demonstrating the receptor specificity of the antagonist pharmacophore. Conclusions MCC22 is a novel bivalent ligand that targets CCR5 and MOR. Our findings demonstrate that MCC22 provides highly potent analgesia and improved functional outcomes in a model of inflammatory arthritis, without inducing typical opioid tolerance. These findings suggest that MCC22 or similar compounds could be used to treat the pain associated with inflammatory arthritis and related conditions, while minimizing the risks typically associated with chronic opioid use.
- Published
- 2018
- Full Text
- View/download PDF
3. Opioid receptors in GtoPdb v.2023.1
- Author
-
Andreas Zimmer, Nurulain Zaveri, Yung H. Wong, Hiroshi Ueda, John R. Traynor, Lawrence Toll, Eric J. Simon, Toni S. Shippenberg, Stefan Schulz, Philip S. Portoghese, Jean-Claude Meunier, Dominique Massot, Davide Malfacini, Lee-Yuan Liu-Chen, Mary-Jeanne Kreek, Ian Kitchen, Brigitte Kieffer, Eamonn Kelly, Stephen Husbands, Graeme Henderson, Volker Höllt, Christopher Evans, Lakshmi A. Devi, Brian M. Cox, Mark Connor, Olivier Civelli, MacDonald J. Christie, Charles Chavkin, Girolamo Caló, Michael Bruchas, and Anna Borsodi
- Subjects
General Medicine ,General Chemistry - Abstract
Opioid and opioid-like receptors are activated by a variety of endogenous peptides including [Met]enkephalin (met), [Leu]enkephalin (leu), β-endorphin (β-end), α-neodynorphin, dynorphin A (dynA), dynorphin B (dynB), big dynorphin (Big dyn), nociceptin/orphanin FQ (N/OFQ); endomorphin-1 and endomorphin-2 are also potential endogenous peptides. The Greek letter nomenclature for the opioid receptors, μ, δ and κ, is well established, and NC-IUPHAR considers this nomenclature appropriate, along with the symbols spelled out (mu, delta, and kappa), and the acronyms, MOP, DOP, and KOP [124, 101, 92]. However the acronyms MOR, DOR and KOR are still widely used in the literature. The human N/OFQ receptor, NOP, is considered 'opioid-related' rather than opioid because, while it exhibits a high degree of structural homology with the conventional opioid receptors [304], it displays a distinct pharmacology. Currently there are numerous clinically used drugs, such as morphine and many other opioid analgesics, as well as antagonists such as naloxone. The majority of clinically used opiates are relatively selective μ agonists or partial agonists, though there are some μ/κ compounds, such as butorphanol, in clinical use. κ opioid agonists, such as the alkaloid nalfurafine and the peripherally acting peptide difelikefalin, are in clinical use for itch.
- Published
- 2023
- Full Text
- View/download PDF
4. FBNTI, a DOR-Selective Antagonist That Allosterically Activates MOR within a MOR–DOR Heteromer
- Author
-
Philip S. Portoghese, George L. Wilcox, Daniel J. Bruce, Michael Ansonoff, Defeng Tian, Eyup Akgün, Mary M. Lunzer, John E. Pintar, and Jon E. Hawkinson
- Subjects
Male ,Agonist ,medicine.drug_class ,Allosteric regulation ,Receptors, Opioid, mu ,Heteromer ,Pharmacology ,Biochemistry ,Mice ,Receptors, Opioid, delta ,mental disorders ,polycyclic compounds ,medicine ,Animals ,Humans ,Receptor ,Injections, Spinal ,Mice, Knockout ,Mice, Inbred ICR ,Molecular Structure ,Chemistry ,HEK 293 cells ,Antagonist ,Analgesics, Opioid ,HEK293 Cells ,nervous system ,Opioid ,Knockout mouse ,Calcium ,human activities ,medicine.drug - Abstract
This report describes the unique pharmacological profile of FBNTI, a potent DOR antagonist that acts as a MOR agonist via an allosteric mechanism. Binding of FBNTI to opioid receptors expressed in HEK 293 cells revealed a 190-fold greater affinity for DOR (Ki = 0.84 nM) over MOR (Ki = 160 nM). In mice, intrathecal FBNTI produced potent antinociception (ED50 = 46.9 pmol/mouse), which was antagonized by selective MOR antagonists (CTOP, β-FNA). Autoantagonism of the MOR agonism by FBNTI was observed above the ED75 dose, suggesting antagonism of activated MOR. That FBNTI is devoid of agonism in DOR knockout mice is consistent with allosteric activation of the MOR protomer via FBNTI bound to within a MOR-DOR heteromer. This proposed mechanism is supported by calcium mobilization assays, which indicate that FBNTI selectively activates the MOR-DOR heteromer and functionally antagonizes the MOR protomer at >ED75. The unprecedented mode of MOR activation by FBNTI may be responsible for the lack of tolerance after intrathecal (i.t.) administration. FBNTI was highly effective upon topical administration to the ipsolateral hind paw in the Hargreaves assay (EC50 = 0.17 ± 0.08 μM) and without significant contralateral activity, suggesting a lack of systemic exposure.
- Published
- 2020
- Full Text
- View/download PDF
5. Opioid receptors in GtoPdb v.2021.3
- Author
-
Eric J. Simon, Nurulain T. Zaveri, Dominique Massot, Brian M. Cox, Lawrence Toll, Lakshmi A. Devi, Toni S. Shippenberg, Eamonn Kelly, John R. Traynor, Philip S. Portoghese, Mary Jeanne Kreek, Brigitte L. Kieffer, Hiroshi Ueda, Andreas Zimmer, Volker Höllt, Graeme Henderson, Charles Chavkin, Stephen M. Husbands, MacDonald J. Christie, Michael R. Bruchas, Mark Connor, Ian Kitchen, Girolamo Calò, Anna Borsodi, Stefan Schulz, Olivier Civelli, Jean-Claude Meunier, Lee-Yuan Liu-Chen, Yung Hou Wong, and Christopher J. Evans
- Subjects
chemistry.chemical_compound ,Nociceptin receptor ,chemistry ,Enkephalin ,Opioid ,Dynorphin B ,medicine ,Dynorphin A ,(+)-Naloxone ,Pharmacology ,Receptor ,Big dynorphin ,medicine.drug - Abstract
Opioid and opioid-like receptors are activated by a variety of endogenous peptides including [Met]enkephalin (met), [Leu]enkephalin (leu), β-endorphin (β-end), α-neodynorphin, dynorphin A (dynA), dynorphin B (dynB), big dynorphin (Big dyn), nociceptin/orphanin FQ (N/OFQ); endomorphin-1 and endomorphin-2 are also potential endogenous peptides. The Greek letter nomenclature for the opioid receptors, μ, δ and κ, is well established, and NC-IUPHAR considers this nomenclature appropriate, along with the symbols spelled out (mu, delta, and kappa), and the acronyms, MOP, DOP, and KOP. [121, 100, 91]. The human N/OFQ receptor, NOP, is considered 'opioid-related' rather than opioid because, while it exhibits a high degree of structural homology with the conventional opioid receptors [294], it displays a distinct pharmacology. Currently there are numerous clinically used drugs, such as morphine and many other opioid analgesics, as well as antagonists such as naloxone, however only for the μ receptor.
- Published
- 2021
- Full Text
- View/download PDF
6. An Analysis of the Conserved Residues between Halobacterial Retinal Proteins and G-Protein Coupled Receptors: Implications for GPCR Modeling.
- Author
-
Thomas G. Metzger, M. Germana Paterlini, Philip S. Portoghese, and David M. Ferguson
- Published
- 1996
- Full Text
- View/download PDF
7. Combined Glia Inhibition and Opioid Receptor Agonism Afford Highly Potent Analgesics without Tolerance
- Author
-
Eyup Akgün, Mary M. Lunzer, and Philip S. Portoghese
- Subjects
Male ,Physiology ,medicine.drug_class ,Cognitive Neuroscience ,Pain ,Pharmacology ,Biochemistry ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Piperidines ,Opioid receptor ,medicine ,Animals ,Receptor ,Pain Measurement ,030304 developmental biology ,Mice, Inbred ICR ,0303 health sciences ,Microglia ,Chemistry ,Antagonist ,Drug Tolerance ,Cell Biology ,General Medicine ,Isoquinolines ,Analgesics, Opioid ,medicine.anatomical_structure ,Receptors, Opioid ,Hyperalgesia ,NMDA receptor ,medicine.symptom ,μ-opioid receptor ,Neuroglia ,030217 neurology & neurosurgery ,Astrocyte - Abstract
Commonly prescribed opioid analgesics produce tolerance upon chronic use due in part to induction of hyperalgesia. Given that two reported bivalent ligands (MMG22 and MCC22) produce potent antinociception without tolerance only in inflamed mice, we have investigated the possible cellular and receptor targets of these ligands. The selective microglia inhibitors, minocycline and SB290157, antagonized intrathecal (i.t.) MCC22 antinociception orders of magnitude more potently than MMG22, suggesting that MCC22 selectively targets activated microglia. The astrocyte toxin, l-α-aminoadipic acid antagonized MMG22 antinociception 126-fold without reducing the potency of MCC22, indicating that activated astrocytes are targets of MMG22. MK-801 and Ro25-6981 antagonism of MMG22 antinociception, but not MCC22, is consistent with selective inhibition of activated NMDAR in astrocytes. The antinociception produced by i.t. MMG22 or MCC22 were both antagonized by the selective mu opioid receptor antagonist, β-FNA, implicating interaction of these ligands with MOR in spinal afferent neurons. MCC22 antinociception was potently blocked by kainate or AMPA ion channel antagonists (LY382884; NBQX), in contrast to MMG22. It is concluded that i.t. MMG22 and MCC22 produce exceptional antinociception via potent inhibition of activated spinal glia, thereby leading to desensitization of spinal neurons and enhanced activation of neuronal MOR. Thus, the present study suggests a new approach to treatment of chronic inflammatory pain without tolerance through a single molecular entity that simultaneously inhibits activated glia and stimulates MOR in spinal neurons.
- Published
- 2018
- Full Text
- View/download PDF
8. Bivalent ligand that activates mu opioid receptor and antagonizes mGluR5 receptor reduces neuropathic pain in mice
- Author
-
Maureen S. Riedl, Eyup Akgün, Mary M. Lunzer, Carolyn A. Fairbanks, George L. Wilcox, Lucy Vulchanova, Cristina D. Peterson, Philip S. Portoghese, and Kelley F. Kitto
- Subjects
Male ,0301 basic medicine ,Agonist ,medicine.drug_class ,Narcotic Antagonists ,Receptor, Metabotropic Glutamate 5 ,Receptors, Opioid, mu ,Pharmacology ,Ligands ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,mental disorders ,medicine ,Enzyme-linked receptor ,Animals ,Injections, Spinal ,Analgesics ,Chemistry ,Metabotropic glutamate receptor 5 ,030104 developmental biology ,Anesthesiology and Pain Medicine ,nervous system ,Neurology ,Opioid ,Hyperalgesia ,Oxymorphone ,Anesthesia ,Neuropathic pain ,Neuralgia ,Neurology (clinical) ,medicine.symptom ,μ-opioid receptor ,030217 neurology & neurosurgery ,medicine.drug - Abstract
The mu opioid receptor (MOR) and metabotropic glutamate receptor 5 (mGluR5) are well-established pharmacological targets in the management of chronic pain. Both receptors are expressed in the spinal cord. MMG22, a bivalent ligand containing 2 pharmacophores separated by 22 atoms, which simultaneously activates MOR and antagonizes mGluR5, has been shown to produce potent reversal of tactile hypersensitivity in rodent models of lipopolysaccharide (LPS)-and bone cancer-induced chronic pain. This study assessed whether intrathecal MMG22 also is effective in reducing pain of neuropathic origin. Furthermore, we theorized that MMG22 should reduce hyperalgesia in nerve-injured mice in a manner consistent with a synergistic interaction between MOR and mGluR5. Several weeks after spared nerve injury, tactile hypersensitivity was reversed in mice by the intrathecal injection of MMG22 (0.01-10 nmol) but also by its shorter spacer analog, MMG10, with similar potency. The potencies of the bivalent ligands were 10- to 14-fold higher than those of the compounds upon which the bivalent structure was based, the MOR agonist oxymorphone and the mGluR5 antagonist MPEP. Coadministration of oxymorphone and MPEP demonstrated analgesic synergism, an interaction confirmed by isobolographic analysis. This study indicates that in the spared nerve injury-induced model of neuropathic pain, the 2 pharmacophores of the bivalent ligands MMG22 and MMG10 target MOR and mGluR5 as separate receptor monomers. The observed increase in the potency of MMG22 and MMG10, compared with oxymorphone and MPEP, may reflect the synergistic interaction of the 2 pharmacophores of the bivalent ligand acting at their respective separate receptor monomers.
- Published
- 2017
- Full Text
- View/download PDF
9. Opioid receptors (version 2019.4) in the IUPHAR/BPS Guide to Pharmacology Database
- Author
-
Brian M. Cox, Eamonn Kelly, Girolamo Calo, Stephen M. Husbands, Mark Connor, Nurulain T. Zaveri, Jean-Claude Meunier, Anna Borsodi, Lee-Yuan Liu-Chen, Volker Höllt, Graeme Henderson, Eric J. Simon, Toni S. Shippenberg, Hiroshi Ueda, Olivier Civelli, Philip S. Portoghese, Mary Jeanne Kreek, Andreas Zimmer, MacDonald J. Christie, Yung Hou Wong, John R. Traynor, Stefan Schulz, Charles Chavkin, Brigitte L. Kieffer, Michael R. Bruchas, Ian Kitchen, Christopher J. Evans, Dominique Massot, Lawrence Toll, and Lakshmi A. Devi
- Subjects
chemistry.chemical_compound ,Nociceptin receptor ,Enkephalin ,chemistry ,Opioid ,Dynorphin B ,medicine ,Dynorphin A ,(+)-Naloxone ,Pharmacology ,Receptor ,Big dynorphin ,medicine.drug - Abstract
Opioid and opioid-like receptors are activated by a variety of endogenous peptides including [Met]enkephalin (met), [Leu]enkephalin (leu), β-endorphin (β-end), α-neodynorphin, dynorphin A (dynA), dynorphin B (dynB), big dynorphin (Big dyn), nociceptin/orphanin FQ (N/OFQ); endomorphin-1 and endomorphin-2 are also potential endogenous peptides. The Greek letter nomenclature for the opioid receptors, μ, δ and κ, is well established, and NC-IUPHAR considers this nomenclature appropriate, along with the symbols spelled out (mu, delta, and kappa), and the acronyms, MOP, DOP, and KOP. [116, 96, 88]. The human N/OFQ receptor, NOP, is considered 'opioid-related' rather than opioid because, while it exhibits a high degree of structural homology with the conventional opioid receptors [282], it displays a distinct pharmacology. Currently there are numerous clinically used drugs, such as morphine and many other opioid analgesics, as well as antagonists such as naloxone, however only for the μ receptor.
- Published
- 2019
- Full Text
- View/download PDF
10. Targeting MOR-mGluR(5) heteromers reduces bone cancer pain by activating MOR and inhibiting mGluR5
- Author
-
Samuel J. Erb, Catherine Harding-Rose, Sarah S. Shueb, Donald A. Simone, Philip S. Portoghese, Rebecca Speltz, Eyup Akgün, and Mary M. Lunzer
- Subjects
0301 basic medicine ,Agonist ,Male ,medicine.drug_class ,Fibrosarcoma ,Receptors, Opioid, mu ,Bone Neoplasms ,Pharmacology ,Ligands ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,0302 clinical medicine ,Receptors, Kainic Acid ,medicine ,Animals ,Mice, Inbred C3H ,Morphine ,Bone cancer ,business.industry ,Drug Administration Routes ,Cancer Pain ,medicine.disease ,Conditioned place preference ,Disease Models, Animal ,030104 developmental biology ,Mechanism of action ,Opioid ,Hyperalgesia ,Systemic administration ,medicine.symptom ,Cancer pain ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Pain is among the most common symptoms in cancer and approximately 90% of patients experience end-stage cancer pain. The management of cancer pain is challenging due to the significant side effects associated with opioids, and novel therapeutic approaches are needed. MMG22 is a bivalent ligand containing MOR agonist and mGluR5 antagonist pharmacophores joined by a 22-atom spacer. MMG22 exhibited extraordinary analgesia following intrathecal administration in a mouse model of bone cancer pain. Here, we assessed the effectiveness of systemic administration of MMG22 in reducing cancer pain and evaluated whether MMG22 displays side effects associated with opioids. Fibrosarcoma cells were injected into and around the calcaneus bone in C3H mice. Mechanical hyperalgesia was defined as an increase in the paw withdrawal frequencies (PWFs) evoked by application of a von Frey monofilament (3.9 mN bending force) applied to the plantar surface of the hind paw Subcutaneous (s.c.), intramuscular (i.m.), and oral (p.o.) administration of MMG22 produced robust dose-dependent antihyperalgesia, whose ED50 was orders of magnitude lower than morphine. Moreover, the ED50 for MMG22 decreased with disease progression. Importantly, s.c. administration of MMG22 did not produce acute (24 h) or long-term (9 days) tolerance, was not rewarding (conditioned place preference test), and did not produce naloxone-induced precipitated withdrawal or alter motor function. A possible mechanism of action of MMG22 is discussed in terms of inhibition of spinal NMDAR via antagonism of its co-receptor, mGluR5, and concomitant activation of neuronal MOR. We suggest that MMG22 may be a powerful alternative to traditional opioids for managing cancer pain. This article is part of the Special Issue entitled ‘New Vistas in Opioid Pharmacology’.
- Published
- 2019
11. Bivalent ligand MCC22 potently attenuates nociception in a murine model of sickle cell disease
- Author
-
Gregory M. Vercellotti, John D. Belcher, Julie K. Olson, Philip S. Portoghese, Donald A. Simone, Eyup Akgün, Mary M. Lunzer, and Giuseppe Cataldo
- Subjects
0301 basic medicine ,Male ,Nociception ,congenital, hereditary, and neonatal diseases and abnormalities ,Cell ,Mice, Transgenic ,Disease ,Anemia, Sickle Cell ,Pharmacology ,Bivalent (genetics) ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Animals ,Analgesics ,business.industry ,Chronic pain ,medicine.disease ,Chronic inflammatory disorder ,Analgesics, Opioid ,Disease Models, Animal ,030104 developmental biology ,Anesthesiology and Pain Medicine ,medicine.anatomical_structure ,Neurology ,Murine model ,Hyperalgesia ,Neurology (clinical) ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Sickle cell disease (SCD) is a chronic inflammatory disorder accompanied by chronic pain. In addition to ongoing pain and hyperalgesia, vaso-occlusive crises-induced pain can be chronic or episodic. Since analgesics typically used to treat pain are not very effective in SCD, opioids, including morphine, are a primary treatment for managing pain in SCD but are associated with many serious side effects, including constipation, tolerance, addiction, and respiratory depression. Thus, there is a need for the development of novel treatments for pain in SCD. In this study we used the Townes transgenic mouse model of SCD to investigate the anti-nociceptive efficacy of the bivalent ligand, MCC22, and compared its effectiveness to morphine. MCC22 consists of a mu opioid receptor (MOR) agonist and a chemokine receptor-5 (CCR5) antagonist that are linked through a 22-atom spacer. Our results show that intraperitoneal administration of MCC22 produced exceptionally potent dose-dependent anti-hyperalgesia as compared to morphine, dramatically decreased evoked responses of nociceptive dorsal horn neurons, and decreased expression of pro-inflammatory cytokines in the spinal cord. Moreover, tolerance did not develop to its analgesic effects following repeated administration. In view of the extraordinary potency of MCC22 without tolerance, MCC22 and similar compounds may vastly improve the management of pain associated with SCD.
- Published
- 2018
12. A bivalent compound targeting CCR5 and the mu opioid receptor treats inflammatory arthritis pain in mice without inducing pharmacologic tolerance
- Author
-
Philip S. Portoghese, Eyup Akgün, Bryce A. Binstadt, Mary M. Lunzer, Raini Dutta, and Jennifer L. Auger
- Subjects
0301 basic medicine ,Agonist ,lcsh:Diseases of the musculoskeletal system ,Receptors, CCR5 ,medicine.drug_class ,Chemokine receptor ,Inflammatory arthritis ,Receptors, Opioid, mu ,Arthritis ,Pain ,Mice, Transgenic ,Pharmacology ,Arthritis, Rheumatoid ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Piperidines ,Drug tolerance ,Opioid receptor ,Medicine ,Animals ,Rheumatoid arthritis ,Inflammation ,Analgesics ,business.industry ,Drug Tolerance ,medicine.disease ,Isoquinolines ,Arthritis, Experimental ,Mice, Inbred C57BL ,030104 developmental biology ,Opioid ,Heteromer ,Morphine ,μ-opioid receptor ,lcsh:RC925-935 ,Analgesia ,business ,CCR5 ,030217 neurology & neurosurgery ,medicine.drug ,Research Article - Abstract
Background Pain accompanies rheumatoid arthritis and other chronic inflammatory conditions and is difficult to manage. Although opioids provide potent analgesia, chronic opioid use can cause tolerance and addiction. Recent studies have demonstrated functional interactions between chemokine and opioid receptor signaling pathways. Reported heterodimerization of chemokine and opioid receptors led our group to develop bivalent compounds that bind both types of receptors, with the goal of targeting opioids to sites of inflammation. MCC22 is a novel bivalent compound containing a CCR5 antagonist and mu opioid receptor (MOR) agonist pharmacophores linked through a 22-atom spacer. We evaluated the efficacy of MCC22 in the K/B.g7 T-cell receptor transgenic mouse model of spontaneous inflammatory arthritis. Methods MCC22 or morphine was administered intraperitoneally at varying doses to arthritic K/B.g7 mice or nonarthritic control mice. Mechanical pain hypersensitivity was measured each day before and after drug administration, using the electronic von Frey test. The potency of MCC22 relative to that of morphine was calculated. Functional readouts of pain included grip strength and nesting behavior. A separate dosing regimen was used to determine whether the drugs induced pharmacologic tolerance. Results MCC22 provided ~ 3000-fold more potent analgesia than morphine in this model. Daily treatment with MCC22 also led to a cumulative analgesic effect, reducing the daily baseline pain level. MCC22 produced no observable analgesic effect in nonarthritic control mice. Importantly, repeated administration of MCC22 did not induce pharmacologic tolerance, whereas a similar regimen of morphine did. Both grip strength and nesting behaviors improved among arthritic mice treated with MCC22. Ankle thickness and arthritis scores were not affected by MCC22. The analgesic effect of MCC22 was abolished in K/B.g7 mice genetically lacking CCR5, demonstrating the receptor specificity of the antagonist pharmacophore. Conclusions MCC22 is a novel bivalent ligand that targets CCR5 and MOR. Our findings demonstrate that MCC22 provides highly potent analgesia and improved functional outcomes in a model of inflammatory arthritis, without inducing typical opioid tolerance. These findings suggest that MCC22 or similar compounds could be used to treat the pain associated with inflammatory arthritis and related conditions, while minimizing the risks typically associated with chronic opioid use. Electronic supplementary material The online version of this article (10.1186/s13075-018-1661-5) contains supplementary material, which is available to authorized users.
- Published
- 2017
13. Heteromer Induction: An Approach to Unique Pharmacology?
- Author
-
Eyup Akgün, Mary M. Lunzer, and Philip S. Portoghese
- Subjects
0301 basic medicine ,Physiology ,medicine.drug_class ,Cognitive Neuroscience ,Receptor, Metabotropic Glutamate 5 ,Heteromer ,Receptors, Opioid, mu ,Plasma protein binding ,Pharmacology ,Ligands ,Biochemistry ,Bivalent (genetics) ,03 medical and health sciences ,Structure-Activity Relationship ,0302 clinical medicine ,Opioid receptor ,Receptors, Opioid, delta ,mental disorders ,medicine ,Structure–activity relationship ,Animals ,Humans ,Receptor ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemistry ,Cell Biology ,General Medicine ,Analgesics, Opioid ,030104 developmental biology ,nervous system ,Models, Chemical ,Pharmacophore ,μ-opioid receptor ,Protein Multimerization ,030217 neurology & neurosurgery ,Protein Binding - Abstract
It is proposed that two types of opioid receptor heteromers exist: a) those that are constitutive and b) those that are induced by bivalent ligands. Mu opioid agonists interact with constitutive MOR-DOR heteromer to mediate tolerance and dependence. Bivalent ligand, MDAN21, is devoid of these adverse effects by virtue of its DOR antagonist pharmacophore. We propose that bivalent ligands MMG22 and MCC22 induce colocalized receptors to form heteromers (MOR-mGluR5 and MOR-CCR5, respectively) that do not occur naturally, thereby promoting unique pharmacology. Heteromer induction with bivalent ligands offers a general approach to unique pharmacology that complements traditional SAR.
- Published
- 2017
14. Targeting putative mu opioid/metabotropic glutamate receptor-5 heteromers produces potent antinociception in a chronic murine bone cancer model
- Author
-
Alvin J. Beitz, Philip S. Portoghese, Branden A. Smeester, Eyup Akgün, and Mary M. Lunzer
- Subjects
Male ,Nociception ,Agonist ,medicine.drug_class ,Receptor, Metabotropic Glutamate 5 ,Receptors, Opioid, mu ,Heteromer ,Pain ,Bone Neoplasms ,Pharmacology ,Ligands ,Article ,Mice ,Drug tolerance ,Cell Line, Tumor ,mental disorders ,medicine ,Animals ,Analgesics ,Mice, Inbred C3H ,Morphine ,Chemistry ,Metabotropic glutamate receptor 5 ,Chronic pain ,Drug Tolerance ,medicine.disease ,Hyperalgesia ,medicine.symptom ,μ-opioid receptor - Abstract
The therapeutic management of chronic pain associated with many cancers is problematic due to the development of tolerance and other adverse effects during the disease progression. Recently we reported on a bivalent ligand (MMG22) containing both mu agonist and mGluR5 antagonist pharmacophores that produced potent antinociception in mice with LPS-induced acute inflammatory pain via a putative MOR-mGluR5 heteromer. In the present study we have investigated the antinociception of MMG22 in a mouse model of bone cancer pain to determine its effectiveness in reducing this type of chronic nociception. There was a 572-fold increase in the potency of MMG22 over a period of 3-21 days that correlated with the progressive increase in hyperalgesia induced by bone tumor growth following implantation of fibrosarcoma cells in mice. The enhancement of antinociception with the progression of the cancer is possibly due to inhibition of NMDA receptor-mediated hyperalgesia via antagonism of mGluR5 and concomitant activation of MOR by the MMG22-occupied heteromer. Notably, MMG22 was 3.6-million-fold more potent than morphine at PID 21. Since MMG22 exhibited a 250,000-times greater potency than that of a mixture of the mu opioid (M19) agonist and mGluR5 antagonist (MG20) monovalent ligands, the data suggest that targeting the putative MOR-mGluR5 heteromer is far superior to univalent interaction with receptors in reducing tumor-induced nociception. In view of the high potency, long duration (>24h) of action and minimal side effects, MMG22 has the potential to be a superior pharmacological agent than morphine and other opiates in the treatment of chronic cancer pain and to serve as a novel pharmacologic tool.
- Published
- 2014
- Full Text
- View/download PDF
15. Putative Kappa Opioid Heteromers As Targets for Developing Analgesics Free of Adverse Effects
- Author
-
Philip S. Portoghese, Michael D. Powers, Mark J. Thomas, Alexander E. Kalyuzhny, Michael A. Benneyworth, Mary M. Lunzer, and Morgan Le Naour
- Subjects
Agonist ,Indoles ,medicine.drug_class ,Arrestins ,Substance-Related Disorders ,Receptors, Opioid, mu ,Pharmacology ,Naltrexone ,Article ,03 medical and health sciences ,Mice ,Structure-Activity Relationship ,0302 clinical medicine ,In vivo ,Receptors, Opioid, delta ,Drug Discovery ,medicine ,Avoidance Learning ,Animals ,Humans ,Receptor ,beta-Arrestins ,030304 developmental biology ,0303 health sciences ,Analgesics ,Chemistry ,Beta-Arrestins ,Receptors, Opioid, kappa ,HEK 293 cells ,Stereoisomerism ,Drug Tolerance ,Conditioned place preference ,3. Good health ,HEK293 Cells ,Opioid ,Molecular Medicine ,Calcium ,Protein Multimerization ,030217 neurology & neurosurgery ,medicine.drug - Abstract
It is now generally recognized that upon activation by an agonist, β-arrestin associates with G protein-coupled receptors and acts as a scaffold in creating a diverse signaling network that could lead to adverse effects. As an approach to reducing side effects associated with κ opioid agonists, a series of β-naltrexamides 3-10 was synthesized in an effort to selectively target putative κ opioid heteromers without recruiting β-arrestin upon activation. The most potent derivative 3 (INTA) strongly activated KOR-DOR and KOR-MOR heteromers in HEK293 cells. In vivo studies revealed 3 to produce potent antinociception, which, when taken together with antagonism data, was consistent with the activation of both heteromers. 3 was devoid of tolerance, dependence, and showed no aversive effect in the conditioned place preference assay. As immunofluorescence studies indicated no recruitment of β-arrestin2 to membranes in coexpressed KOR-DOR cells, this study suggests that targeting of specific putative heteromers has the potential to identify leads for analgesics devoid of adverse effects.
- Published
- 2014
16. Ligands that interact with putative MOR-mGluR5 heteromer in mice with inflammatory pain produce potent antinociception
- Author
-
Philip S. Portoghese, Branden A. Smeester, Muhammad I. Javed, Eyup Akgün, Mary M. Lunzer, and Al J Beitz
- Subjects
Agonist ,medicine.drug_class ,Receptor, Metabotropic Glutamate 5 ,Receptors, Opioid, mu ,Heteromer ,Pain ,Pharmacology ,Ligands ,Receptors, Metabotropic Glutamate ,Mice ,medicine ,Animals ,Inflammation ,Multidisciplinary ,Metabotropic glutamate receptor 5 ,Chemistry ,Antagonist ,Biological Sciences ,Analgesics, Opioid ,Metabotropic receptor ,nervous system ,Opioid ,Morphine ,μ-opioid receptor ,Protein Binding ,medicine.drug - Abstract
The low effectiveness of morphine and related mu opioid analgesics for the treatment of chronic inflammatory pain is a result of opioid-induced release of proinflammatory cytokines and glutamate that lower the pain threshold. In this regard, the use of opioids with metabotropic glutamate-5 receptor (mGluR 5 ) antagonist has been reported to increase the efficacy of morphine and prevent the establishment of adverse effects during chronic use. Given the presence of opioid receptors (MORs) and mGluR 5 in glia and neurons, together with reports that suggest coexpressed MOR/mGluR 5 receptors in cultured cells associate as a heteromer, the possibility that such a heteromer could be a target in vivo was addressed by the design and synthesis of a series of bivalent ligands that contain mu opioid agonist and mGluR 5 antagonist pharmacophores linked through spacers of varying length (10–24 atoms). The series was evaluated for antinociception using the tail-flick and von Frey assays in mice pretreated with lipopolysaccharide (LPS) or in mice with bone cancer. In LPS-pretreated mice, MMG22 (4c, 22-atom spacer) was the most potent member of the series (intrathecal ED 50 ∼9 fmol per mouse), whereas in untreated mice its ED 50 was more than three orders of magnitude higher. As members of the series with shorter or longer spacers have ≥500-fold higher ED 50s in LPS-treated mice, the exceptional potency of MMG22 may be a result of the optimal bridging of protomers in a putative MOR-mGluR 5 heteromer. The finding that MMG22 possesses a >10 6 therapeutic ratio suggests that it may be an excellent candidate for treatment of chronic, intractable pain via spinal administration.
- Published
- 2013
- Full Text
- View/download PDF
17. Bivalent Ligands That Target μ Opioid (MOP) and Cannabinoid1 (CB1) Receptors Are Potent Analgesics Devoid of Tolerance
- Author
-
Eyup Akgün, Philip S. Portoghese, Mary M. Lunzer, Mike D. Powers, Alexander E. Kalyuzhny, Morgan Le Naour, and Ajay S. Yekkirala
- Subjects
Male ,Agonist ,Cannabinoid receptor ,medicine.drug_class ,medicine.medical_treatment ,Receptors, Opioid, mu ,Heteromer ,Fluorescent Antibody Technique ,Pain ,Pharmacology ,Ligands ,Article ,Structure-Activity Relationship ,Receptor, Cannabinoid, CB1 ,Drug tolerance ,mental disorders ,Drug Discovery ,medicine ,Animals ,Humans ,Structure–activity relationship ,Receptor ,Injections, Spinal ,Injections, Intraventricular ,Mice, Inbred ICR ,Molecular Structure ,Chemistry ,Cell Membrane ,Drug Tolerance ,Endocytosis ,Analgesics, Opioid ,HEK293 Cells ,Models, Chemical ,nervous system ,Opioid ,Drug Design ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,Cannabinoid ,medicine.drug - Abstract
Given that μ opioid (MOP) and canabinoid (CB1) receptors are colocalized in various regions of the central nervous system and have been reported to associate as heteromer (MOP-CB1) in cultured cells, the possibility of functional, endogenous MOP-CB1 in nociception and other pharmacologic effects has been raised. As a first step in investigating this possibility, we have synthesized a series of bivalent ligands 1-5 that contain both μ agonist and CB1 antagonist pharmacophores for use as tools to study the functional interaction between MOP and CB1 receptors in vivo. Immunofluorescent studies on HEK293 cells coexpressing both receptors suggested 5 (20-atom spacer) to be the only member of the series that bridges the protomers of the heteromer. Antinociceptive testing in mice revealed 5 to be the most potent member of the series. As neither a mixture of monovalent ligands 9 + 10 nor bivalents 2-5 produced tolerance in mice, MOR-CB1 apparently is not an important target for reducing tolerance.
- Published
- 2013
- Full Text
- View/download PDF
18. Reduced Antinociception of Opioids in Rats and Mice by Vaccination with Immunogens Containing Oxycodone and Hydrocodone Haptens
- Author
-
Paul R. Pentel, Morgan Le Naour, Marco Pravetoni, Theresa Harmon, Tara M. Hawley, Philip S. Portoghese, and A.M. Tucker
- Subjects
Drug ,Immunogen ,media_common.quotation_subject ,Enzyme-Linked Immunosorbent Assay ,chemical and pharmacologic phenomena ,Pharmacology ,complex mixtures ,Article ,Mice ,Pharmacotherapy ,Drug Discovery ,medicine ,Animals ,Hydrocodone ,media_common ,Vaccines, Synthetic ,integumentary system ,biology ,Chemistry ,hemic and immune systems ,Rats ,Analgesics, Opioid ,Vaccination ,biology.protein ,Molecular Medicine ,Haptens ,Hapten ,Oxycodone ,Keyhole limpet hemocyanin ,medicine.drug - Abstract
Prescription opioids abuse and associated deaths are an emerging concern in the USA. Vaccination against prescription opioids may provide an alternative to pharmacotherapy. An oxycodone hapten containing a tetraglycine linker at the C6 position (6OXY(Gly)(4)OH) conjugated to keyhole limpet hemocyanin (KLH) has shown early proof-of-efficacy in rodents as a candidate immunogen (6OXY(Gly)(4)-KLH) for the treatment of oxycodone abuse. In this study, oxycodone-based and hydrocodone-based haptens were conjugated to KLH to generate immunogens that would recognize both oxycodone and hydrocodone. Vaccination with 6OXY(Gly)(4)-KLH increased drug binding in serum, reduced drug distribution to brain, and blunted analgesia for both oxycodone and hydrocodone. An analogous C6-linked hydrocodone vaccine blocked hydrocodone effects but less so than 6OXY(Gly)(4)-KLH. C8-Linked hydrocodone immunogens had only limited efficacy. Amide conjugation showed higher haptenation ratios and greater efficacy than thioether conjugation to maleimide activated KLH (mKLH). The 6OXY(Gly)(4)-KLH vaccine may be used for treatment of prescription opioid abuse.
- Published
- 2013
- Full Text
- View/download PDF
19. Modulation of Musculoskeletal Hyperalgesia by Brown Adipose Tissue Activity in Mice
- Author
-
Myra G. Nunez, Philip S. Portoghese, Katalin J. Kovács, Elizabeth M Goudie-DeAngelis, Casey L. Kissel, Ramy E. Abdelhamid, and Alice A. Larson
- Subjects
0301 basic medicine ,Male ,Pain Threshold ,Tail ,medicine.medical_specialty ,Adrenergic receptor ,Adipose tissue ,Article ,Body Temperature ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Adipose Tissue, Brown ,Musculoskeletal Pain ,Internal medicine ,Brown adipose tissue ,Adrenergic antagonist ,Reaction Time ,Medicine ,Animals ,Chronic stress ,Adrenergic agonist ,Muscle Strength ,Swimming ,Uncoupling Protein 1 ,Mice, Knockout ,business.industry ,Body Weight ,Adrenergic beta-Agonists ,Cold Temperature ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Anesthesiology and Pain Medicine ,Endocrinology ,medicine.anatomical_structure ,Neurology ,Ethanolamines ,Hyperalgesia ,Female ,Neurology (clinical) ,medicine.symptom ,business ,Thermogenesis ,030217 neurology & neurosurgery - Abstract
Cold exposure and a variety of types of mild stress increase pain in patients suffering from painful disorders such as fibromyalgia syndrome. Acutely, stress induces thermogenesis by increasing sympathetic activation of beta-3 (β3) adrenergic receptors in brown adipose tissue. Chronic stress leads to the hypertrophy of brown adipose, a phenomenon termed adaptive thermogenesis. Based on the innervation of skeletal muscle by collaterals of nerves projecting to brown adipose, we theorized an association between brown adipose tissue activity and musculoskeletal hyperalgesia and tested this hypothesis in mice. Exposure to a cold swim or injection of BRL37344 (β3 adrenergic agonist) each enhanced musculoskeletal hyperalgesia, as indicated by morphine-sensitive decreases in grip force responses, whereas SR59230A (β3 adrenergic antagonist) attenuated swim-induced hyperalgesia. Chemical ablation of interscapular brown adipose, using Rose Bengal, attenuated the development of hyperalgesia in response to either swim stress or BRL37344. In addition, elimination of the gene expressing uncoupling protein-1 (UCP1), the enzyme responsible for thermogenesis, prevented musculoskeletal hyperalgesia in response to either a swim or BRL37344, as documented in UCP1-knock out (UCP1-KO) mice compared to wild type controls. Together these data provide a convergence of evidence suggesting that activation of brown adipose contributes to stress-induced musculoskeletal hyperalgesia.
- Published
- 2016
20. Molecular Basis for Binding and Subtype Selectivity of 1,4-Benzodiazepine Antagonist Ligands of the Cholecystokinin Receptor
- Author
-
Andrew Orry, Erin E. Cawston, Mary Lou Augustine, Polo C.-H. Lam, Patrick M. Sexton, Ruben Abagyan, Eyup Akgün, Philip S. Portoghese, Maoqing Dong, Alicja M. Ball, Kaleeckal G. Harikumar, and Laurence J. Miller
- Subjects
Stereochemistry ,Molecular Sequence Data ,Allosteric regulation ,Peptide binding ,Ligands ,digestive system ,Biochemistry ,Cholecystokinin receptor ,Benzodiazepines ,Chlorocebus aethiops ,Animals ,Amino Acid Sequence ,Homology modeling ,Receptor ,Molecular Biology ,Sequence Homology, Amino Acid ,Ligand ,Chemistry ,digestive, oral, and skin physiology ,Cell Biology ,Small molecule ,Docking (molecular) ,COS Cells ,Receptors, Cholecystokinin ,hormones, hormone substitutes, and hormone antagonists ,Allosteric Site ,Signal Transduction - Abstract
Allosteric binding pockets in peptide-binding G protein-coupled receptors create opportunities for the development of small molecule drugs with substantial benefits over orthosteric ligands. To gain insights into molecular determinants for this pocket within type 1 and 2 cholecystokinin receptors (CCK1R and CCK2R), we prepared a series of receptor constructs in which six distinct residues in TM2, -3, -6, and -7 were reversed. Two novel iodinated CCK1R- and CCK2R-selective 1,4-benzodiazepine antagonists, differing only in stereochemistry at C3, were used. When all six residues within CCK1R were mutated to corresponding CCK2R residues, benzodiazepine selectivity was reversed, yet peptide binding selectivity was unaffected. Detailed analysis, including observations of gain of function, demonstrated that residues 6.51, 6.52, and 7.39 were most important for binding the CCK1R-selective ligand, whereas residues 2.61 and 7.39 were most important for binding CCK2R-selective ligand, although the effect of substitution of residue 2.61 was likely indirect. Ligand-guided homology modeling was applied to wild type receptors and those reversing benzodiazepine binding selectivity. The models had high predictive power in enriching known receptor-selective ligands from related decoys, indicating a high degree of precision in pocket definition. The benzodiazepines docked in similar poses in both receptors, with C3 urea substituents pointing upward, whereas different stereochemistry at C3 directed the C5 phenyl rings and N1 methyl groups into opposite orientations. The geometry of the binding pockets and specific interactions predicted for ligand docking in these models provide a molecular framework for understanding ligand selectivity at these receptor subtypes. Furthermore, the strong predictive power of these models suggests their usefulness in the discovery of lead compounds and in drug development programs.
- Published
- 2012
- Full Text
- View/download PDF
21. An Oxycodone Conjugate Vaccine Elicits Drug-Specific Antibodies that Reduce Oxycodone Distribution to Brain and Hot-Plate Analgesia
- Author
-
M. Le Naour, Philip S. Portoghese, Marco Pravetoni, A.M. Tucker, Paul R. Pentel, and Theresa Harmon
- Subjects
Male ,Hot Temperature ,Dose-Response Relationship, Immunologic ,Pain ,chemical and pharmacologic phenomena ,Pharmacology ,Antibodies ,Rats, Sprague-Dawley ,Drug Discovery and Translational Medicine ,Therapeutic index ,medicine ,Animals ,Bovine serum albumin ,Pain Measurement ,Vaccines, Conjugate ,integumentary system ,biology ,Chemistry ,Brain ,Rats ,Opioid ,Oxymorphone ,Antibody Formation ,biology.protein ,Morphine ,Molecular Medicine ,Cattle ,Analgesia ,Chickens ,Oxycodone ,Keyhole limpet hemocyanin ,Protein Binding ,medicine.drug ,Buprenorphine - Abstract
Opioid conjugate vaccines have shown promise in attenuating the behavioral effects of heroin or morphine in animals. The goal of this study was to extend this approach to oxycodone (OXY), a commonly abused prescription opioid. Haptens were generated by adding tetraglycine (Gly)(4) or hemisuccinate (HS) linkers at the 6-position of OXY. Immunization of rats with OXY(Gly)(4) conjugated to the carrier proteins bovine serum albumin (BSA) or keyhole limpet hemocyanin (KLH) produced high-titer antibodies to OXY and its metabolite oxymorphone with substantially lower affinities for other structurally related opioid agonists and antagonists. There was no measurable binding of antibody by the (Gly)(4) linker alone or off-target opioids methadone and buprenorphine. OXY(HS) conjugates were less immunogenic despite achieving protein haptenation ratios comparable to OXY(Gly)(4)-BSA. In rats given a single intravenous dose of OXY, immunization with OXY(Gly)(4)-KLH increased OXY protein binding and retention in serum while decreasing its unbound (free) concentration in plasma and distribution to brain. Vaccine efficacy correlated with serum antibody titers, and it was greatest in rats given the lowest OXY dose (0.05 mg/kg) but was significant even after a larger OXY dose (0.5 mg/kg), equivalent to the high end of the therapeutic range in humans. These effects of OXY(Gly)(4)-KLH on drug disposition were comparable to those of nicotine or cocaine vaccines that are in clinical trials as addiction treatments. Immunization with OXY(Gly)(4)-KLH also reduced OXY analgesia in a thermal nociception test. These data support further study of vaccination with the OXY(Gly)(4)-KLH immunogen as a potential treatment option for OXY abuse or addiction.
- Published
- 2012
- Full Text
- View/download PDF
22. A κ Opioid Pharmacophore Becomes a Spinally Selective κ-δ Agonist When Modified with a Basic Extender Arm
- Author
-
Michael D. Powers, Jie Yang, Ye Tang, Philip S. Portoghese, and Mary M. Lunzer
- Subjects
Agonist ,medicine.drug_class ,Chemistry ,Organic Chemistry ,Pharmacology ,Biochemistry ,Nociception ,Opioid ,In vivo ,Opioid receptor ,Drug Discovery ,medicine ,Potency ,Pharmacophore ,Receptor ,medicine.drug - Abstract
We have explored the concept of a molecular extender arm attached to a κ opioid agonist pharmacophore 3 (ICI-199,441) in an effort to potentially interact with a complementary group on a neighboring opioid receptor. The molecular arm containing a terminal amine group was lengthened incrementally from 11 up to 18 atoms. Increasing the number of atoms in the arm produced virtually no change in the mouse intracerebroventricular (i.c.v.) antinociceptive potency. In contrast, the intrathecal (i.t.) potency of 6 (KDA-16) with a 16-atom arm was dramatically increased, as reflected by its antinociceptive i.c.v./i.t. ED50 ratio of ∼130. Further lengthening led to a decreased ED50 ratio. In vivo selective antagonist studies of KDA-16 revealed that κ and δ opioid receptors were responsible for the greatly enhanced i.t. potency. Calcium release experiments in HEK-293 cells suggested that KDA-16 selectively activate κ-δ heteromers. These data are consistent with the reported possible presence of heteromeric κ-δ opioid receptors in mouse spinal cord but not in the brain. The use of a molecular extender arm may be useful for developing spinally selective analgesics.
- Published
- 2010
- Full Text
- View/download PDF
23. Stereochemical Studies on Medicinal Agents. 24 Correlation of the Solid-State Conformations of 4-Phenylpiperidine Analgetic With the Torsional Relationship Between the Phenyl Group and Piperidine Ring in Solution, a Circular Dichroism Study
- Author
-
Jacques Poupaert and Philip S. Portoghese
- Subjects
chemistry.chemical_compound ,Circular dichroism ,chemistry ,Stereochemistry ,Solid-state ,Phenyl group ,General Chemistry ,4-Phenylpiperidine ,Piperidine ,Ring (chemistry) ,Cotton effect - Abstract
A CD study of 8 representatives of 4-phenylpiperidine analgetics is presented to correlate their solid-state conformations with their chiroptical properties in solution. In addition to the benzenoid 1Lb fingerprint, the studied compounds exhibit two waves: the former band around 230-223 nm is assigned to a 1La transition; the origin of the latter band is not determined. A quadrant rule is tentatively proposed to explain the sign of the 1La Cotton effect.
- Published
- 2010
- Full Text
- View/download PDF
24. Modulation of Cell Surface Expression of Nonactivated Cholecystokinin Receptors Using Bivalent Ligand-Induced Internalization
- Author
-
Philip S. Portoghese, Eyup Akgün, Kaleeckal G. Harikumar, and Laurence J. Miller
- Subjects
Agonist ,medicine.drug_class ,Stereochemistry ,media_common.quotation_subject ,Receptors, Opioid, mu ,Down-Regulation ,CHO Cells ,Immune receptor ,Ligands ,Cholecystokinin receptor ,Article ,Substrate Specificity ,Cricetulus ,Cricetinae ,Chlorocebus aethiops ,Drug Discovery ,Enzyme-linked receptor ,medicine ,Animals ,5-HT5A receptor ,Internalization ,Receptor ,media_common ,Chemistry ,Receptor antagonist ,Endocytosis ,Gene Expression Regulation ,COS Cells ,Biophysics ,Molecular Medicine ,Receptors, Cholecystokinin - Abstract
CCK(2) receptor antagonists potentiate pain relief by MOP receptor agonists. In an attempt to enhance this effect, we prepared bivalent ligands incorporating CCK(2) receptor antagonist and MOP receptor agonist pharmacophores. (9) Ligands with 16- to 22-atom spacers could simultaneously bind both receptors but provided no advantage in activity over individual ligands. We now examine the effect of these ligands on receptor internalization as a mechanism of receptor regulation. We prepared CHO cell lines expressing nonfluorescent halves (YN and YC) of yellow fluorescent protein attached to each receptor. Spatial approximation of constructs was needed to yield fluorescence. Monovalent MOP agonist 1 signaled normally and internalized the MOP receptor. Monovalent CCK(2) antagonist 2 did not stimulate receptor internalization. In the dual receptor-bearing cells, bivalent ligands 3a-c capable of simultaneously binding both receptors resulted in cell surface fluorescence and internalization of the fluorescent complex in a time- and temperature-dependent manner. Bivalent ligand 4 with spacer too short to occupy both receptors simultaneously yielded no signal. Receptor tethering with appropriate bivalent ligands can down-regulate signaling by moving a nonactivated receptor into the endocytic pathway.
- Published
- 2010
- Full Text
- View/download PDF
25. Standard Opioid Agonists Activate Heteromeric Opioid Receptors: Evidence for Morphine and [<scp>d</scp>-Ala2-MePhe4-Glyol5]Enkephalin as Selective μ−δ Agonists
- Author
-
Ajay S. Yekkirala, Alexander E. Kalyuzhny, and Philip S. Portoghese
- Subjects
Agonist ,medicine.medical_specialty ,Pyrrolidines ,Enkephalin ,Physiology ,medicine.drug_class ,Cognitive Neuroscience ,Benzeneacetamides ,Receptors, Opioid, mu ,Pharmacology ,Ligands ,Biochemistry ,OGFr ,chemistry.chemical_compound ,Receptors, Opioid, delta ,Internal medicine ,medicine ,Functional selectivity ,Humans ,Calcium Signaling ,Receptor ,Morphine ,Receptors, Opioid, kappa ,Cell Biology ,General Medicine ,Enkephalin, Ala(2)-MePhe(4)-Gly(5) ,Immunohistochemistry ,Analgesics, Opioid ,DAMGO ,HEK293 Cells ,Endocrinology ,chemistry ,Opioid ,Guanosine 5'-O-(3-Thiotriphosphate) ,Area Under Curve ,Receptors, Opioid ,Bremazocine ,Enkephalin, D-Penicillamine (2,5) ,medicine.drug - Abstract
Research in the opioid field has relied heavily on the use of standard agonist ligands such as morphine, [d-Ala(2)-MePhe(4)-Glyol(5)]enkephalin (DAMGO), U69593, bremazocine, [d-Pen(2)d-Pen(5)]enkephalin (DPDPE), and deltorphin-II as tools for investigating the three major types of opioid receptors, MOP (μ), KOP (κ), and DOP (δ), that mediate antinociception. The functional selectivity of these ligands has been based on the assumption that opioid receptors exist as homomers. As numerous studies in cultured cells have suggested that opioid receptors can associate both as homomers and heteromers, we have investigated the selectivity of these standard ligands using intracellular calcium release and [(35)S]GTPγS assays in HEK-293 cells that contain singly and coexpressed opioid receptors. The present study reveals that morphine and DAMGO, traditionally classified as μ selective agonists, selectively activate μ-δ heteromeric opioid receptors with greater efficacy than homomeric opioid receptors. Moreover, standard ligands that have been widely employed as κ- and δ-selective agonists display little or no differences in the activation of homomeric and heteromeric opioid receptors. The far-reaching implications of these results are discussed.
- Published
- 2009
- Full Text
- View/download PDF
26. Morphine Tolerance in Mice Changes Response of Heroin from μ to δ Opioid Receptors
- Author
-
Philip S. Portoghese, Blythe B. Holmes, James M. Fujimoto, and Jodie J. Rady
- Subjects
Agonist ,medicine.medical_specialty ,medicine.drug_class ,Chemistry ,Pharmacology ,Receptor antagonist ,General Biochemistry, Genetics and Molecular Biology ,Endocrinology ,Opioid ,Naltriben ,Opioid receptor ,Naltrindole ,Internal medicine ,Morphine ,medicine ,μ-opioid receptor ,medicine.drug - Abstract
Heroin produced antinociception in the tail flick test through mu receptors in the brain of ICR and CD-1 mice, a response inhibited by 3-O-methylnaltrexone. Tolerance to morphine was produced by subcutaneous morphine pellet implantation. By the third day, the heroin response was produced through delta opioid receptors. The response was inhibited by simultaneous intracerebroventricular (i.c. v.) administration of naltrindole, a delta opioid receptor antagonist. More specifically, delta1 rather than delta2 receptors were involved because 7-benzylidenenaltrexone, a delta1 receptor antagonist, inhibited but naltriben, a delta2 antagonist, did not. Also, antinociception produced by i.c.v. heroin was inhibited by intrathecal administration of bicuculline and picrotoxin consistent with the concept that delta1 receptors in the brain mediated the antinociceptive response through descending neuronal pathways to the spinal cord to activate GABAA and GABAB receptors rather than spinal alpha2-adrenergic and serotonergic receptors activated originally by the mu agonist action in naive mice. The mu response of 6-monoacetylmorphine, a metabolite of heroin, was changed by morphine pellet implantation to a delta2 response (inhibited by naltriben but not 7-benzylidenenaltrexone). The agonist action of morphine in these morphine-tolerant mice remained mu. Thus, the opioid receptor selectivity of heroin and 6-monoacetylmorphine in the brain is changed by production of tolerance to morphine. Such a change explains how morphine tolerant mice are not cross-tolerant to heroin.
- Published
- 2008
- Full Text
- View/download PDF
27. o-Naphthalenedicarboxaldehyde Derivative of 7‘-Aminonaltrindole as a Selective δ-Opioid Receptor Affinity Label
- Author
-
Yan Zhang, Bertrand Le Bourdonnec, Sarika Prabhu Haris, Christopher R. McCurdy, and Philip S. Portoghese
- Subjects
Conformational change ,Magnetic Resonance Spectroscopy ,Schiff base ,biology ,medicine.drug_class ,Chemistry ,Stereochemistry ,Narcotic Antagonists ,Affinity label ,Antagonist ,Affinity Labels ,Carboxamide ,Spectrometry, Mass, Fast Atom Bombardment ,Naltrexone ,chemistry.chemical_compound ,Opioid receptor ,Receptors, Opioid, delta ,Drug Discovery ,medicine ,biology.protein ,Molecular Medicine ,Moiety ,Derivative (chemistry) - Abstract
Incorporation of a naphthalene-dialdehyde moiety into the delta antagonist, 6'-aminonaltrindole afforded a potent, selective, irreversible delta-agonist 1. However, flow cytometry studies revealed no time-dependent specific fluorescence, suggesting that both Lys214 and Cys216 at the recognition site are not involved in covalent binding. Molecular simulation studies suggest that compound 1 may form a Schiff base with the epsilon-amino group of Lys214, which could explain its irreversibility and transformation into a delta-agonist through a conformational change of TM5.
- Published
- 2007
- Full Text
- View/download PDF
28. Synthesis and124I-labeling ofm-iodophenylpyrrolomorphinan as a potential PET imaging agent for delta opioid (DOP) receptors
- Author
-
Munawwar Sajjad, Eyup Akgün, Hani A. Nabi, and Philip S. Portoghese
- Subjects
Agonist ,medicine.drug_class ,Stereochemistry ,Organic Chemistry ,chemistry.chemical_element ,Biological activity ,Zinc ,Biochemistry ,Chemical synthesis ,Analytical Chemistry ,chemistry.chemical_compound ,Acetic acid ,chemistry ,Oxymorphone ,Drug Discovery ,medicine ,Radiology, Nuclear Medicine and imaging ,Receptor ,Sodium acetate ,Spectroscopy ,Nuclear chemistry ,medicine.drug - Abstract
Condensation of phenylazo-β-ketoamide 4 with oxymorphone 5 afforded an m-iodophenylpyrrolomorphinan (m-IPPM) 6 mediated by elemental zinc in acetic acid/sodium acetate buffer. m-IPPM 6 is a novel opioid receptor agonist (Ki = 4.53 nM for DOP) with high selectivity for DOP receptors. m-IPPM 6 was converted into the positron emitter m-[124I]PPM 8 via the stannylated intermediate 7. The final yield was 24.5 ± 1.9 % (n = 6) with a specific activity of 2.5 ± 1.2 Ci/µmol. Copyright © 2007 John Wiley & Sons, Ltd.
- Published
- 2007
- Full Text
- View/download PDF
29. Inhibition of Inflammatory and Neuropathic Pain by Targeting a Mu Opioid Receptor/Chemokine Receptor5 Heteromer (MOR-CCR5)
- Author
-
Eyup Akgün, Mary M. Lunzer, Yoshikazu Watanabe, Michael D. Powers, Yuk Y. Sham, Philip S. Portoghese, and Muhammad I. Javed
- Subjects
Agonist ,Male ,Models, Molecular ,Chemokine ,Receptors, CCR5 ,medicine.drug_class ,Heteromer ,Receptors, Opioid, mu ,CCR5 receptor antagonist ,Pharmacology ,Article ,Chemokine receptor ,Mice ,Drug Discovery ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Inflammation ,Analgesics ,biology ,Chemistry ,HEK293 Cells ,Opioid ,CCR5 Receptor Antagonists ,Chronic Disease ,Morphine ,biology.protein ,Molecular Medicine ,Neuralgia ,μ-opioid receptor ,medicine.drug - Abstract
Chemokine release promotes cross-talk between opioid and chemokine receptors that in part leads to reduced efficacy of morphine in the treatment of chronic pain. On the basis of the possibility that a MOR-CCR5 heteromer is involved in such cross-talk, we have synthesized bivalent ligands (MCC series) that contain mu opioid agonist and CCR5 antagonist pharmacophores linked through homologous spacers (14–24 atoms). When tested on lipopolysaccharide-inflamed mice, a member of the series (MCC22; 3e) with a 22-atom spacer exhibited profound antinociception (i.t. ED50 = 0.0146 pmol/mouse) that was 2000× greater than morphine. Moreover, MCC22 was ∼3500× more potent than a mixture of mu agonist and CCR5 antagonist monovalent ligands. These data strongly suggest that MCC22 acts by bridging the protomers of a MOR-CCR5 heteromer having a TM5,6 interface. Molecular simulation studies are consistent with such bridging. This study supports the MOR-CCR5 heteromer as a novel target for the treatment of chronic pain.
- Published
- 2015
30. Opioid-induced tolerance and dependence in mice is modulated by the distance between pharmacophores in a bivalent ligand series
- Author
-
Chris L. Etienne, Sandra C. Roerig, Philip S. Portoghese, Ping-Yee Law, David J. Daniels, and Natalie R. Lenard
- Subjects
Male ,Narcotics ,Agonist ,Time Factors ,Stereochemistry ,medicine.drug_class ,Chemistry, Pharmaceutical ,Narcotic Antagonists ,Receptors, Opioid, mu ,Physical dependence ,Pharmacology ,Ligands ,Naltrexone ,Mice ,Naltrindole ,Opioid receptor ,Receptors, Opioid, delta ,medicine ,Animals ,Analgesics ,Mice, Inbred ICR ,Models, Statistical ,Multidisciplinary ,Dose-Response Relationship, Drug ,Morphine ,Chemistry ,Biological Sciences ,Analgesics, Opioid ,Models, Chemical ,Opioid ,Oxymorphone ,medicine.symptom ,medicine.drug - Abstract
Given the mounting evidence for involvement of δ opioid receptors in the tolerance and physical dependence of μ opioid receptor agonists, we have investigated the possible physical interaction between μ and δ opioid receptors by using bivalent ligands. Based on reports of suppression of antinociceptive tolerance by the δ antagonist naltrindole (NTI), bivalent ligands [μ-δ agonist-antagonist (MDAN) series] that contain different length spacers, and pharmacophores derived from NTI and the μ agonist oxymorphone, have been synthesized and evaluated by intracerebroventricular (i.c.v.) administration in the tail-flick test in mice. In acute i.c.v. studies, the bivalent ligands functioned as agonists with potencies ranging from 1.6- to 45-fold greater than morphine. In contrast, the monovalent μ agonist analogues were substantially more potent than the MDAN congeners and were essentially equipotent with one another and oxymorphone. Pretreatment with NTI decreased the ED 50 values for MDAN-19 to a greater degree than for MDAN-16 but had no effect on MDAN-21. Chronic i.c.v. studies revealed that MDAN ligands whose spacer was 16 atoms or longer produced less dependence than either morphine or μ monovalent control MA-19. On the other hand, both physical dependence and tolerance were suppressed at MDAN spacer lengths of 19 atoms or greater. These data suggest that physical interaction between the μ and δ opioid receptors modulates μ-mediated tolerance and dependence. Because MDAN-21 was found to be 50-fold more potent than morphine by the i.v. route (i.v.), it offers a previously uncharacterized approach for the development of analgesics devoid of tolerance and dependence.
- Published
- 2005
- Full Text
- View/download PDF
31. Criteria for the safety evaluation of flavoring substances
- Author
-
Samuel M. Cohen, Victor J. Feron, T. B. Adams, Robert L. Smith, Bernard M. Wagner, Jay I. Goodman, Philip S. Portoghese, William J. Waddell, I. C. Munro, John Doull, and Lawrence J. Marnett
- Subjects
food.ingredient ,business.industry ,Food additive ,food and beverages ,Flavoring Agents ,Context (language use) ,General Medicine ,Toxicology ,Biotechnology ,Human health ,food ,Chemical groups ,Medicine ,Degree of confidence ,business ,S typhimurium ,Flavor ,Food Science - Abstract
The current status of the GRAS evaluation program of flavoring substances operated by the Expert Panel of FEMA is discussed. The Panel maintains a rigorous rotating 10-year program of continuous review of scientific data related to the safety evaluation of flavoring substances. The Panel concluded a comprehensive review of the GRAS (GRASa) status of flavors in 1985 and began a second comprehensive review of the same substances and any recently GRAS materials in 1994. This second re-evaluation program of chemical groups of flavor ingredients, recognized as the GRAS reaffirmation (GRASr) program, is scheduled to be completed in 2005. The evaluation criteria used by the Panel during the GRASr program reflects the significant impact of advances in biochemistry, molecular biology and toxicology that have allowed for a more complete understanding of the molecular events associated with toxicity. The interpretation of novel data on the relationship of dose to metabolic fate, formation of protein and DNA adducts, enzyme induction, and the cascade of cellular events leading to toxicity provides a more comprehensive basis upon which to evaluate the safety of the intake of flavor ingredients under conditions of intended use. The interpretation of genotoxicity data is evaluated in the context of other data such as in vivo animal metabolism and lifetime animal feeding studies that are more closely related to actual human experience. Data are not viewed in isolation, but comprise one component that is factored into the Panel's overall safety assessment. The convergence of different methodologies that assess intake of flavoring substances provides a greater degree of confidence in the estimated intake of flavor ingredients. When these intakes are compared to dose levels that in some cases result in related chemical and biological effects and the subsequent toxicity, it is clear that exposure to these substances through flavor use presents no significant human health risk.
- Published
- 2005
- Full Text
- View/download PDF
32. The FEMA GRAS assessment of benzyl derivatives used as flavor ingredients
- Author
-
Jay I. Goodman, Philip S. Portoghese, Samuel M. Cohen, William J. Waddell, Robert L. Smith, T. B. Adams, Victor J. Feron, John Doull, Lawrence J. Marnett, I. C. Munro, and Bernard M. Wagner
- Subjects
food.ingredient ,Flavoring Agents ,Context (language use) ,Toxicology ,chemistry.chemical_compound ,food ,Benzyl benzoate ,Toxicity Tests ,Animals ,Food Industry ,Humans ,Food science ,Flavor ,United States Food and Drug Administration ,business.industry ,Food additive ,food and beverages ,General Medicine ,Benzoic Acid ,equipment and supplies ,Food safety ,United States ,Food Analysis ,Benzyl acetate ,Biotechnology ,chemistry ,Benzaldehydes ,business ,Benzyl Alcohol ,Food Science - Abstract
This publication is the eighth in a series of safety evaluations performed by the Expert Panel of the Flavor and Extract Manufacturers Association (FEMA). In 1993, the panel initiated a comprehensive program to re-evaluate the safety of more than 1700 GRAS flavoring substances under conditions of intended use. Elements that are fundamental to the safety evaluation of flavor ingredients include exposure, structural analogy, metabolism, pharmacokinetics and toxicology. Flavor ingredients are evaluated individually and in the context of the available scientific information on the group of structurally related substances. Scientific data relevant to the safety evaluation of the use of benzyl derivatives as flavoring ingredients is evaluated. The group of benzyl derivatives was reaffirmed as GRAS (GRASr) based, in part, on their self-limiting properties as flavoring substances in food; their rapid absorption, metabolic detoxication, and excretion in humans and other animals, their low level of flavor use, the wide margins of safety between the conservative estimates of intake and the no-observed-adverse effect levels determined from subchronic and chronic studies and the lack of significant genotoxic and mutagenic potential. This evidence of safety is supported by the fact that the intake of benzyl derivatives as natural components of traditional foods is greater than their intake as intentionally added flavoring substances.
- Published
- 2005
- Full Text
- View/download PDF
33. The FEMA GRAS assessment of phenethyl alcohol, aldehyde, acid, and related acetals and esters used as flavor ingredients
- Author
-
Bernard M. Wagner, Robert L. Smith, John Doull, Philip S. Portoghese, Lawrence J. Marnett, Samuel M. Cohen, William J. Waddell, I. C. Munro, Victor J. Feron, Jay I. Goodman, and T. B. Adams
- Subjects
Phenylacetates ,medicine.medical_treatment ,Flavoring Agents ,Context (language use) ,Acetaldehyde ,Toxicology ,chemistry.chemical_compound ,Acetals ,Detoxification (alternative medicine) ,Toxicity Tests ,medicine ,Animals ,Food Industry ,Humans ,Organic chemistry ,Flavor ,United States Food and Drug Administration ,food and beverages ,Esters ,General Medicine ,Phenylethyl Alcohol ,equipment and supplies ,United States ,Food Analysis ,chemistry ,Phenethyl alcohol ,Food Science - Abstract
This publication is the ninth in a series of safety evaluations performed by the Expert Panel of the Flavor and Extract Manufacturers Association (FEMA). In 1993, the Panel initiated a comprehensive program to re-evaluate the safety of more than 1700 GRAS flavoring substances under conditions of intended use. Elements that are fundamental to the safety evaluation of flavor ingredients include exposure, structural analogy, metabolism, pharmacokinetics and toxicology. Flavor ingredients are evaluated individually and in the context of the available scientific information on the group of structurally related substances. Scientific data relevant to the safety evaluation of the use of phenethyl alcohol, aldehyde, acid, and related acetals and esters as flavoring ingredients is evaluated. The group of phenethylalcohol, aldehyde, acid, and related acetals and esters was reaffirmed as GRAS (GRASr) based, in part, on their self-limiting properties as flavoring substances in food, their rapid absorption, metabolic detoxication, and excretion in humans and other animals, their low level of flavor use, the wide margins of safety between the conservative estimates of intake and the no-observed-adverse effect levels determined from subchronic and chronic studies and the lack of significant genotoxic and mutagenic potential. This evidence of safety is supported by the fact that the intake of phenethyl alcohol, aldehyde, acid, and related acetals and esters as natural components of traditional foods is greater than their intake as intentionally added flavoring substances.
- Published
- 2005
- Full Text
- View/download PDF
34. Interaction of Bivalent Ligand KDN21 with Heterodimeric δ-κ Opioid Receptors in Human Embryonic Kidney 293 Cells
- Author
-
David J. Daniels, Philip S. Portoghese, Rashmi G. Bhushan, and Zhihua Xie
- Subjects
Pharmacology ,Chemistry ,medicine.drug_class ,Stereochemistry ,Receptors, Opioid, kappa ,Molecular Sequence Data ,Kidney metabolism ,Kidney ,Ligands ,κ-opioid receptor ,Naltrexone ,Cell Line ,Naltriben ,Naltrindole ,Receptors, Opioid, delta ,medicine ,Molecular Medicine ,Amino Acid Sequence ,Phosphorylation ,Diprenorphine ,Norbinaltorphimine ,Receptor ,Oligopeptides ,Opioid antagonist ,medicine.drug - Abstract
KDN21 is a bivalent ligand that contains delta and kappa opioid antagonist pharmacophores linked through a 21-atom spacer. It has been reported that KDN21 bridges delta and kappa receptors that are organized as heterodimers. We have shown previously that when using [(3)H]diprenorphine as radioligand, KDN21 displayed greatly enhanced affinity in this series for coexpressed delta and kappa opioid receptors (CDK). The present study used in vitro expression systems to investigate interactions of members of the KDN series with delta-kappa heterodimers through competition binding using selective ligands and the mitogen-activated protein kinase (MAPK) assay. In this regard, the use of the selective radioligands [(3)H]naltrindole and [(3)H]norbinaltorphimine (nor-BNI) in competition binding studies revealed that KDN21 has much higher affinity than other KDN members for CDK and bound to CDK more selectively relative to mixed delta and kappa opioid receptors or singly expressed delta and kappa opioid receptors. Other experiments revealed that the binding of naltrindole to delta opioid receptors could increase the binding of nor-BNI to kappa opioid receptors and vice versa, suggesting reciprocal allosteric modulation of receptors in the heterodimer. Regarding the selectivity of KDN21 for phenotypic delta and kappa opioid receptors, we investigated the effect of KDN21 on the activation of MAPKs [extracellular signal-regulated kinases 1 and 2 (ERK1/2)] by delta- or kappa-selective agonists. KDN21 inhibited the activation of ERK1/2 by [D-Pen(2),D-Pen(5)]-enkephalin (delta(1)) and bremazocine (kappa(2)) but had no effect on the activation by deltorphin II (delta(2)) and (+)-(5alpha,7alpha,8beta)-N-methyl-N-[7-(1-pyrrolidinyl)-1-oxaspiro[4.5]dec-8-yl]benzeneacetamide (U69593, kappa(1)). 7-Benzylidenenaltrexone (delta(1)) and bremazocine (kappa(2)) significantly reduced the binding of KDN21 to CDK, whereas naltriben (delta(2)) and U69593 produced no such change. Taken together, these data support the idea that the organization of delta and kappa receptors as heterodimers gives rise to delta(1) and kappa(2) phenotypes.
- Published
- 2005
- Full Text
- View/download PDF
35. Homology Modeling and Molecular Dynamics Simulations of the Mu Opioid Receptor in a Membrane-Aqueous System
- Author
-
Yan Zhang, Philip S. Portoghese, Ramkumar Rajamani, Jiali Gao, and Yuk Y. Sham
- Subjects
Models, Molecular ,Rhodopsin ,Molecular model ,Stereochemistry ,Receptors, Opioid, mu ,Sequence Homology ,Crystallography, X-Ray ,Biochemistry ,Naltrexone ,Protein structure ,medicine ,Animals ,Computer Simulation ,Homology modeling ,Lipid bilayer ,Molecular Biology ,G protein-coupled receptor ,Binding Sites ,biology ,Chemistry ,Cell Membrane ,Organic Chemistry ,Water ,Lipid Metabolism ,Lipids ,Protein Structure, Tertiary ,Structural Homology, Protein ,Solvents ,biology.protein ,Molecular Medicine ,Cattle ,μ-opioid receptor ,medicine.drug - Abstract
Three types of opioid receptors-mu, delta, and kappa-belong to the rhodopsin subfamily in the G protein-coupled receptor superfamily. With the recent characterization of the high-resolution X-ray crystal structure of bovine rhodopsin, considerable attention has been focused on molecular modeling of these transmembrane proteins. In this study, a homology model of the mu opioid receptor was constructed based on the X-ray crystal structure of bovine rhodopsin. A phospholipid bilayer was built around the receptor, and two water layers were placed on both surfaces of the lipid bilayer. Molecular-dynamics simulations were carried out by using CHARMM for the entire system, which consisted of 316 amino acid residues, 92 phospholipid molecules, 8327 water molecules, and 11 chloride counter ions-40 931 atoms altogether. The whole system was equilibrated for 250 ps followed by another 2 ns dynamic simulation. The opioid ligand naltrexone was docked into the optimized model, and the critical amino acid residues for binding were identified. The mu opioid receptor homology model optimized in a complete membrane-aqueous system should provide a good starting point for further characterization of the binding modes for opioid ligands. Furthermore, the method developed herein will be applicable to molecular model building to other opioid receptors as well as other GPCRs.
- Published
- 2005
- Full Text
- View/download PDF
36. A Bivalent Ligand (KDAN-18) Containing δ-Antagonist and κ-Agonist Pharmacophores Bridges δ2 and κ1 Opioid Receptor Phenotypes
- Author
-
Zhihua Xie, David J. Daniels, Rashmi G. Bhushan, Philip S. Portoghese, and Amol Kulkarni
- Subjects
Agonist ,medicine.drug_class ,Chemistry ,Stereochemistry ,Antagonist ,Opioid ,Opioid receptor ,Naltrindole ,Drug Discovery ,medicine ,Molecular Medicine ,Pharmacophore ,Receptor ,Kappa ,medicine.drug - Abstract
To characterize delta- and kappa-opioid receptor phenotypes, bivalent ligands (KDAN series) containing delta-antagonist (naltrindole) and kappa(1)-agonist (ICI-199,441) pharmacophores were synthesized and evaluated by the intrathecal route using the mouse tail-flick assay and binding studies. The data have suggested that KDAN-18 (2) bridges phenotypic delta(2)- and kappa(1)-receptors. A conceptual model is presented to explain the organizational differences between the opioid receptors that give rise to the phenotypes (delta(1), delta(2), kappa(1), kappa(2)).
- Published
- 2004
- Full Text
- View/download PDF
37. A Bivalent Ligand (KDN-21) Reveals Spinal δ and κ Opioid Receptors Are Organized as Heterodimers That Give Rise to δ1 and κ2 Phenotypes. Selective Targeting of δ−κ Heterodimers
- Author
-
Shiv Kumar Sharma, David J. Daniels, Rashmi G. Bhushan, Zhihua Xie, and Philip S. Portoghese
- Subjects
medicine.drug_class ,Chemistry ,Ligand ,Stereochemistry ,Central nervous system ,Bivalent (genetics) ,medicine.anatomical_structure ,Opioid ,Opioid receptor ,Drug Discovery ,medicine ,Molecular Medicine ,Structure–activity relationship ,Pharmacophore ,Receptor ,medicine.drug - Abstract
In view of recent pharmacological studies suggesting the existence of δ−κ opioid receptor heterodimers/oligomers in the spinal cord, we have synthesized and evaluated (intrathecally in mice) a series of bivalent ligands (KDN series) containing κ and δ antagonist pharmacophores. Pharmacological and binding data have provided evidence for the bridging of spinal δ−κ receptor heterodimers by KDN-21 and for their identification as δ1 and κ2. The selectivity profile of KDN-21 and the apparent absence of coupled δ1−κ2 phenotypes in the brain suggest a new approach for targeting receptors.
- Published
- 2004
- Full Text
- View/download PDF
38. Characterization of Specific Opioid Binding Sites in Neural Membranes from the Myenteric Plexus of Porcine Small Intestine
- Author
-
David R. Brown, Philip S. Portoghese, and DeWayne Townsend
- Subjects
Male ,Narcotics ,Agonist ,medicine.medical_specialty ,Enkephalin ,Swine ,Stereochemistry ,medicine.drug_class ,Narcotic Antagonists ,Receptors, Opioid, mu ,Diprenorphine ,Myenteric Plexus ,(+)-Naloxone ,Tritium ,Receptors, Opioid, delta ,Internal medicine ,Intestine, Small ,medicine ,Animals ,Drug Interactions ,Binding site ,Receptor ,Myenteric plexus ,Pharmacology ,Binding Sites ,Chemistry ,Receptors, Opioid, kappa ,Cell Membrane ,Sodium ,Endocrinology ,Naltriben ,Molecular Medicine ,Female ,Enkephalin, D-Penicillamine (2,5) ,HEPES ,medicine.drug - Abstract
Delta- and kappa-opioid receptors (OPRs), but not micro-OPRs, are expressed in the myenteric plexus of the porcine distal small intestine. In a subpopulation of myenteric neurons, delta- and kappa-OPRs seem to be colocalized and may functionally interact. In this study, radioligand binding was used to characterize myenteric OPR populations in detail. The nonselective OPR antagonist [3H]diprenorphine bound to a single, high-affinity site in myenteric neural membrane homogenates. Naloxone displaced 65 and 59% of [3H]diprenorphine binding from this site in Na(+)-free Tris and Krebs-HEPES buffers, respectively. Naltrexone-derived delta- and kappa-OPR antagonists, including naltriben, 7-benzylidenenaltrexone, nor-binaltorphimine, and 5'-guanidinonaltrindole, displaced [3H]diprenorphine from two distinct binding sites to levels similar to that of naloxone. The selective delta-OPR ligands Tyr-1,2,3,4-tetrahydroisoquinoline-Phe-Phe-OH (TIPP), [D-Pen2,D-Pen5]enkephalin (DPDPE), [D-Ala2, Glu4]deltorphin II, and (+)-4-[(alphaR)-alpha((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl-3-methoxybenzyl)-N,N-diethylbenzamide (SNC-80) and the kappa-OPR agonist (D-(5alpha,7alpha,8beta)-(-)-N-methyl-N-(7-(1-pyrrolidinyl)-1-oxoaspiro-(4,5)dec-8-yl) benzeneacetamide (U-69,593) displaced [3H]diprenorphine from three independent binding sites; these included high-affinity delta- and kappa-OPR sites, and a residual binding site. Residual [3H]diprenorphine binding was displaced by the selective kappa-OPR antagonist nor-binaltorphimine after saturation of delta and kappa sites, respectively, with DPDPE and U-69,593. The residual binding site displayed low affinity for delta- and kappa-OPR agonists and TIPP, as well as moderate affinity for naltrexone-derived ligands, properties reminiscent of delta-/kappa-OPR heterodimers.
- Published
- 2003
- Full Text
- View/download PDF
39. Antidepressant-Like Effects of κ-Opioid Receptor Antagonists in the Forced Swim Test in Rats
- Author
-
William A. Carlezon, William C. Stevens, Mark S. Todtenkopf, Philip S. Portoghese, Hilarie C. Tomasiewicz, Stephen D. Mague, Andrea M. Pliakas, Robert M. Jones, and Yan Zhang
- Subjects
Male ,medicine.medical_specialty ,medicine.drug_class ,Narcotic Antagonists ,Dynorphin ,Motor Activity ,Nucleus accumbens ,Citalopram ,Pharmacology ,CREB ,Rats, Sprague-Dawley ,Opioid receptor ,Internal medicine ,Desipramine ,medicine ,Animals ,Swimming ,biology ,Receptors, Opioid, kappa ,Antidepressive Agents ,Rats ,Endocrinology ,Brain stimulation ,Models, Animal ,biology.protein ,Molecular Medicine ,Psychology ,Behavioural despair test ,medicine.drug - Abstract
We showed previously that cAMP response element-binding protein (CREB) within the nucleus accumbens (NAc) of rats regulates immobility in the forced swim test (FST), an assay used to study depression. Because CREB regulates expression of dynorphin (which acts at kappa-opioid receptors) in NAc neurons, these findings raised the possibility that kappa-receptors mediate immobility behaviors in the FST. Here, we report that i.c.v. administration of the kappa-antagonist nor-binaltorphimine dose dependently decreased immobility in the FST, suggesting that it has antidepressant-like effects. Implicating a specific effect at kappa-receptors, similar antidepressant-like effects were seen after treatment with either of two novel, structurally dissimilar kappa-antagonists: 5'-guanidinonaltrindole, which was effective after i.c.v. but not systemic treatment, and 5'-acetamidinoethylnaltrindole (ANTI), which was potent and effective after systemic treatment. The behavioral effects of the kappa-antagonists resembled those of tricyclic antidepressants (desipramine) and selective serotonin reuptake inhibitors (fluoxetine and citalopram). Conversely, systemic administration of the kappa-agonist [5alpha,7alpha,8beta]-N-methyl-N-[7-[1-pyrrolidinyl]-1-oxaspiro[4.5]dec8-yl]-benzenacetamide (U-69593) dose dependently increased immobility in the FST, consistent with prodepressant-like effects. The effects of the kappa-ligands in the FST were not correlated with nonspecific effects on locomotor activity. Furthermore, the most potent and effective kappa-antagonist (ANTI) did not affect the rewarding impact of lateral hypothalamic brain stimulation at a dose with strong antidepressant-like effects. These findings are consistent with the hypothesis that CREB-mediated induction of dynorphin in the NAc "triggers" immobility behavior in the FST. Furthermore, they raise the possibility that kappa-antagonists may have efficacy as antidepressants, but lack stimulant or reward-related effects.
- Published
- 2003
- Full Text
- View/download PDF
40. Kappa opioid antagonist effects of the novel kappa antagonist 5?-guanidinonaltrindole (GNTI) in an assay of schedule-controlled behavior in rhesus monkeys
- Author
-
Robert M. Jones, Philip S. Portoghese, David C. Linsenmayer, S. Stevens Negus, and Nancy K. Mello
- Subjects
Male ,Narcotics ,Agonist ,medicine.medical_specialty ,Pyrrolidines ,Reinforcement Schedule ,Time Factors ,medicine.drug_class ,Narcotic Antagonists ,Benzeneacetamides ,Pharmacology ,Guanidines ,κ-opioid receptor ,Piperazines ,chemistry.chemical_compound ,Receptors, Opioid, delta ,Internal medicine ,5'-Guanidinonaltrindole ,medicine ,Animals ,Drug Interactions ,Analgesics ,Dose-Response Relationship, Drug ,Morphine ,business.industry ,Receptors, Opioid, kappa ,3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer ,Antagonist ,Analgesics, Non-Narcotic ,JDTic ,Macaca mulatta ,Naltrexone ,Endocrinology ,Morphinans ,Opioid ,chemistry ,Benzamides ,Conditioning, Operant ,Female ,business ,Opioid antagonist ,medicine.drug - Abstract
Rationale: Opioid receptors are divided into three types: kappa, mu, and delta receptors. Receptor-selective antagonists are useful experimental tools for evaluation of opioid receptor-mediated processes. 5′-Guanidinonaltrindole (GNTI) was recently developed as a novel kappa-selective antagonist. Objectives: To evaluate the potency, time course, and selectivity of GNTI's opioid antagonist effects in rhesus monkeys in an assay of schedule-controlled responding. Methods: Five rhesus monkeys were trained to respond under a fixed ratio 30 schedule of food reinforcement. The rate-decreasing effects of the kappa agonists U50,488 and U69,593, the mu agonist morphine, and the delta agonist SNC80 were examined alone and after pretreatment with GNTI (0.1 and 1.0 mg/kg i.m.; 1 h to 14 days). Results: U50,488, U69,593, morphine, and SNC80 dose-dependently decreased response rates in this procedure. GNTI produced a dose- and time-dependent antagonism of the rate-decreasing effects of U50,488. The kappa antagonist effects of GNTI had a slow onset and a long duration of action, and peak antagonist effects were observed after 24 h. A higher dose of 3.2 mg/kg GNTI eliminated responding in one monkey and was not studied further. The antagonist effects of GNTI were kappa selective, because 1.0 mg/kg GNTI also antagonized the effects of U69,593, but not those of morphine or SNC80. Conclusions: These results suggest that GNTI is a potent and selective kappa antagonist with a slow onset and long duration of action in rhesus monkeys. Relative to the prototype kappa antagonist nor-binaltorphimine, GNTI may have some advantages as a tool for the study of kappa receptor-mediated processes.
- Published
- 2002
- Full Text
- View/download PDF
41. The FEMA GRAS assessment of pyrazine derivatives used as flavor ingredients
- Author
-
Philip S. Portoghese, P.M. Newberne, William J. Waddell, Bernard M. Wagner, Victor J. Feron, John Doull, Lawrence J. Marnett, Jay I. Goodman, T. B. Adams, Robert L. Smith, and I. C. Munro
- Subjects
Computer science ,Reference values ,General Medicine ,Food science ,Toxicology ,Flavor ,Food Science - Abstract
This is the fifth in a series of safety evaluations performed by the Expert Panel of the Flavor and Extract Manufacturers Association (FEMA). In 1993, the Panel initiated a comprehensive program to re-evaluate the safety of more than 1700 GRAS flavoring substances under conditions of intended use. Elements that are fundamental to the safety evaluation of flavor ingredients include exposure, structural analogy, metabolism, pharmacokinetics and toxicology. Flavor ingredients are evaluated individually taking into account the available scientific information on the group of structurally related substances. Scientific data relevant to the safety evaluation of the use of pyrazine derivatives as flavoring ingredients is evaluated.
- Published
- 2002
- Full Text
- View/download PDF
42. Methadone and Heroin Antinociception: Predominant £-Opioid-Receptor Responses in Methadone-Tolerant Mice
- Author
-
James M. Fujimoto, Philip S. Portoghese, and Jodie J. Rady
- Subjects
Agonist ,Methadone maintenance ,medicine.drug_class ,Receptors, Opioid, mu ,Pharmacology ,Receptors, N-Methyl-D-Aspartate ,Mice ,Species Specificity ,Opioid receptor ,Receptors, Opioid, delta ,medicine ,Animals ,5-HT receptor ,Injections, Intraventricular ,Drug Implants ,Dose-Response Relationship, Drug ,Chemistry ,Drug Tolerance ,Bicuculline ,Analgesics, Opioid ,Heroin ,Naltriben ,Morphine ,Methadone ,medicine.drug - Abstract
Antinociceptive tail flick responses to heroin and 6-monoacetylmorphine mediated in the brain by mu-opioid receptor are switched by morphine pellet implantation to delta1- and delta2-opioid-receptors mediation, respectively. Present results showed that the mu-receptor response (inhibited by beta-funaltrexamine) to methadone was changed by morphine pellet implantation to delta1 (inhibited by 7-benzylidenenaltrexone)- and delta2 (inhibited by naltriben)-opioid-receptor responses. Methadone pellet implantation likewise changed mediation from mu- to delta-opioid receptors for heroin and methadone but not for morphine (beta-funaltrexamine continued to inhibit). Methadone mu action in the brain was linked through a descending system to activate spinal serotonin receptors (inhibited by methysergide), but this link was gone in the methadone-pellet-implanted group. In the latter group, the new delta1- and delta2-receptor responses were mediated by spinal GABAA (inhibited by bicuculline) and GABAB (inhibited by 2-hydroxysaclofen) receptors. These shifts in neuronal systems meant that mu receptors on a given neuron were not changed into delta receptors. Preliminary results showed that delta-agonist action for methadone was prevented from appearing by MK801, a NMDA-receptor antagonist, and did not occur in 129S6/SvEv mice which lack NMDA responsiveness. Could methadone maintenance treatment in humans uncover delta-agonist actions?
- Published
- 2002
- Full Text
- View/download PDF
43. Transformation of a κ-Opioid Receptor Antagonist to a κ-Agonist by Transfer of a Guanidinium Group from the 5‘- to 6‘-Position of Naltrindole
- Author
-
David M. Ferguson, Shiv Kumar Sharma, Philip S. Portoghese, Robert M. Jones, and Thomas G. Metzger
- Subjects
Conformational change ,Stereochemistry ,medicine.drug_class ,Narcotic Antagonists ,Guinea Pigs ,Molecular Conformation ,In Vitro Techniques ,Transfection ,Cell Line ,Structure-Activity Relationship ,Opioid receptor ,Naltrindole ,Drug Discovery ,medicine ,Animals ,Humans ,Structure–activity relationship ,Cloning, Molecular ,Receptor ,Guanidine ,Chemistry ,Receptors, Opioid, kappa ,Antagonist ,Muscle, Smooth ,Biological activity ,Naltrexone ,Rats ,Transmembrane domain ,Mutagenesis, Site-Directed ,Molecular Medicine ,Muscle Contraction ,medicine.drug - Abstract
The importance of the indole scaffold of GNTI 3 in directing its address (5'-guanidinium group) to associate with the Glu297 residue of the kappa-opioid receptor was investigated by the synthesis and biological evaluation of its 4'- (4a), 6'- (4b), and 7'- (4c) regioisomers. The finding that only the 5'-regioisomer (GNTI) possessed potent kappa-opioid antagonist activity and high affinity at kappa-receptors illustrates the importance of the 5'-position in orienting the guanidinium group to the proper recognition locus (Glu 297) for potent kappa-antagonist activity. The discovery that the 6'-regioisomer of GNTI was a potent kappa-agonist, together with the results of site-directed mutagenesis studies that are consistent with association between the 6'-guanidinium group and Glu297, suggest that the transition from an inactive to an active state of the kappa-receptor involves a conformational change of TM6. We propose that association of the 6'-guanidinium group of 4b with Glu297 promotes axial rotational motion of transmembrane helix VI which leads to receptor activation via a conformational change of inner loop 3.
- Published
- 2001
- Full Text
- View/download PDF
44. From Models to Molecules: Opioid Receptor Dimers, Bivalent Ligands, and Selective Opioid Receptor Probes
- Author
-
Philip S. Portoghese
- Subjects
Molecular model ,Chemistry ,medicine.drug_class ,Stereochemistry ,Morphine derivatives ,Bivalent (genetics) ,Opioid receptor ,Drug Discovery ,medicine ,Molecular Medicine ,Molecule ,Norbinaltorphimine ,Morphinan derivatives ,medicine.drug - Published
- 2001
- Full Text
- View/download PDF
45. κ-Opioid receptor agonist suppression of HIV-1 expression in CD4+ lymphocytes
- Author
-
An Chih Chang, Phillip K. Peterson, Genya Gekker, Xingin Fang, Jean M. Bidlack, Philip S. Portoghese, and James R. Lokensgard
- Subjects
CD4-Positive T-Lymphocytes ,Agonist ,medicine.medical_specialty ,medicine.drug_class ,Lymphocyte ,Cell ,U-50488 ,In Vitro Techniques ,Pharmacology ,Biology ,Biochemistry ,Internal medicine ,Gene expression ,medicine ,Humans ,Receptor ,Receptors, Opioid, kappa ,3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer ,Biological activity ,Analgesics, Non-Narcotic ,Flow Cytometry ,In vitro ,medicine.anatomical_structure ,Endocrinology ,HIV-1 - Abstract
Synthetic κ-opioid receptor (KOR) agonists have been shown to suppress HIV-1 expression in acutely infected macrophages. In the present study, we examined the effects of the KOR ligand trans -3,4-dichloro- N -methyl- N [2-(1-pyrolidinyl)cyclohexyl]benzeneaceamide methanesulfonate (U50,488) on HIV-1 expression in CD4 + lymphocytes, the main target cell of this virus. When U50,488 was added to activated CD4 + lymphocytes, HIV-1 expression was inhibited in a concentration- and time-dependent manner with maximal suppression (≈60%) at 10 -7 M U50,488. The KOR selective antagonist nor-binaltorphimine (nor-BNI) had no effect by itself on viral expression but blocked the antiviral property of U50,488, suggesting that U50,488 was acting via a KOR-related mechanism. Support for the involvement of KOR was provided by the findings that 34% of activated CD4 + lymphocytes were positive for KOR, using an immunofluorescence technique, and that seven additional synthetic KOR ligands also inhibited HIV-1 expression. The results of this study broaden understanding of the antiviral properties of KOR ligands to include cells outside of the nervous system and suggest a potential role for these agents in the treatment of HIV-1 infection.
- Published
- 2001
- Full Text
- View/download PDF
46. Affinity labels as tools for the identification of opioid receptor recognition sites
- Author
-
Rachid El Kouhen, Philip S. Portoghese, Ping Y. Law, Bertrand Le Bourdonnec, and Horace H. Loh
- Subjects
Binding Sites ,biology ,Chemistry ,Stereochemistry ,medicine.drug_class ,Affinity label ,Pharmaceutical Science ,Affinity Labels ,Biochemistry ,Opioid receptor ,Receptors, Opioid ,Drug Discovery ,medicine ,biology.protein ,Humans ,μ-opioid receptor ,Binding site ,Pharmacophore ,Receptor ,Opioid antagonist ,Endogenous agonist - Abstract
Affinity labels have proven to be useful tools in opioid research. We review experiments carried out with the mu opioid receptor affinity label, beta-funaltrexamine (2), that support the concept of different recognition sites for mu opioid agonists and antagonists. The data are interpreted in the context of a dimeric receptor that contains two allosterically coupled binding sites: one that binds endogenous agonist, and the second that functions as an inhibitory modulator of agonism. It is proposed that exogenous antagonists bind selectively to the second site. The first of a new class of affinity labels, PGNA (5), that contains the phthaldehyde moiety attached to an opioid antagonist pharmacophore, is described. This class of ligands has been named 'reporter affinity labels' because covalent association leads to the formation of a fluorescent isoindole that is diagnostic for cross-linking of lysine and cysteine residues. PGNA binds opioid receptors covalently, as suggested by (a) irreversible binding to cloned opioid receptors, (b) irreversible opioid antagonism in the guinea pig ileum preparation, and (c) ultra-long opioid antagonism in mice. Since flow cytometry experiments revealed specific enhancement of fluorescence in cloned mu receptors after a 1 min exposure to 5, it is concluded that covalent binding has occurred via the formation of an isoindole, presumably by cross-linking neighboring lysine and cysteine residues in the vicinity of the receptor recognition site.
- Published
- 2001
- Full Text
- View/download PDF
47. Covalently Induced Activation of the δ Opioid Receptor by a Fluorogenic Affinity Label, 7‘-(Phthalaldehydecarboxamido)naltrindole (PNTI)
- Author
-
Gennady Poda, Horace H. Loh, Philip S. Portoghese, Ping Y. Law, David M. Ferguson, Rachid El Kouhen, and Bertrand Le Bourdonnec
- Subjects
Male ,Models, Molecular ,Agonist ,Stereochemistry ,medicine.drug_class ,Affinity label ,Guinea Pigs ,CHO Cells ,In Vitro Techniques ,Ligands ,Chemical synthesis ,δ-opioid receptor ,Mice ,Vas Deferens ,Ileum ,Naltrindole ,Cricetinae ,Receptors, Opioid, delta ,Drug Discovery ,medicine ,Animals ,Fluorescent Dyes ,Analgesics ,biology ,Chemistry ,Affinity Labels ,Biological activity ,Naltrexone ,In vitro ,Covalent bond ,biology.protein ,Molecular Medicine ,medicine.drug - Published
- 2001
- Full Text
- View/download PDF
48. Potent and Selective Indolomorphinan Antagonists of the Kappa-Opioid Receptor
- Author
-
William C. Stevens, Thomas G. Metzger, Robert M. Jones, Philip S. Portoghese, David M. Ferguson, and Govindan Subramanian
- Subjects
Models, Molecular ,Indoles ,Stereochemistry ,Narcotic Antagonists ,Transfection ,Guanidines ,κ-opioid receptor ,Cell Line ,Structure-Activity Relationship ,chemistry.chemical_compound ,Naltrindole ,Drug Discovery ,medicine ,Humans ,Moiety ,Indole test ,Morphine Derivatives ,Receptors, Opioid, kappa ,Antagonist ,JDTic ,Naltrexone ,Morphinans ,chemistry ,Molecular Medicine ,Norbinaltorphimine ,Kappa ,medicine.drug - Abstract
The indole moiety in the delta-opioid antagonist, naltrindole (2, NTI), was employed as a scaffold to hold an "address" for interaction with the kappa-opioid receptor. The attachment of the address to the 5'-position of the indole moiety was based on superposition of NTI upon the kappa antagonist, norbinaltorphimine (1, norBNI). A variety of cationic groups were employed as a kappa address in an effort to investigate its interaction with the anionic address subsite, Glu297, on the kappa receptor. Some of the groups that were employed for this purpose were amines, amidines, guanidines, and quaternary ammonium. Members of the series were found to have a varying degree of kappa antagonist potency and kappa selectivity when tested in smooth muscle preparations. The 5'-guanidine derivative 12a (GNTI) was the most potent member of the series and had the highest kappa selectivity ratio. GNTI was 2 times more potent and 6-10-fold more selective than norBNI (1). In general, the order of potency in the series was: guanidinesamidines approximately quaternary ammoniumamines. The kappa antagonist potency appeared to be a function of a combination of the pK(a) and distance constraint of the cationic substituent of the ligand. Receptor binding studies were qualitatively in agreement with the pharmacological data. Molecular modeling studies on 12a suggested that the protonated N-17 and guanidinium groups of GNTI are associated with Asp138 (TM3) and Glu297 (TM6), respectively, while the phenolic hydroxyl may be involved in donor-acceptor interactions with the imidazole ring of His291. It was concluded that the basis for the high kappa selectivity of GNTI is related both to association with the nonconserved Glu297 residue and to unfavorable interactions with an equivalent position in mu- and delta-opioid receptors.
- Published
- 2000
- Full Text
- View/download PDF
49. 5′-Guanidinonaltrindole, a highly selective and potent κ-opioid receptor antagonist
- Author
-
Robert M. Jones and Philip S. Portoghese
- Subjects
Male ,medicine.medical_specialty ,medicine.drug_class ,Guinea Pigs ,CHO Cells ,In Vitro Techniques ,Pharmacology ,Ligands ,Guanidines ,Mice ,chemistry.chemical_compound ,Opioid receptor ,Cricetinae ,Internal medicine ,5'-Guanidinonaltrindole ,medicine ,Animals ,Humans ,Mice, Inbred ICR ,Membranes ,Chemistry ,Receptors, Opioid, kappa ,Antagonist ,Muscle, Smooth ,Enkephalin, Ala(2)-MePhe(4)-Gly(5) ,JDTic ,Receptor antagonist ,Naltrexone ,Endocrinology ,Morphinans ,Competitive antagonist ,Enkephalin, D-Penicillamine (2,5) ,Norbinaltorphimine ,Opioid antagonist ,medicine.drug - Abstract
5′-Guanidinonaltrindole (GNTI) possesses 5-fold greater opioid antagonist potency ( K e =0.04 nM) and an order of magnitude greater selectivity (selectivity ratios >500) than the prototypical κ-opioid receptor antagonist, norbinaltorphimine, in smooth muscle preparations. Binding and functional studies conducted on cloned human opioid receptors expressed in Chinese hamster ovarian (CHO) cells afforded pA 2 values that were comparable to the smooth muscle data. In view of the high selectivity and potency of GNTI, it is a potentially valuable pharmacological tool for opioid research.
- Published
- 2000
- Full Text
- View/download PDF
50. Molecular Docking Reveals a Novel Binding Site Model for Fentanyl at the μ-Opioid Receptor
- Author
-
Govindan Subramanian, M G Paterlini, David M. Ferguson, and Philip S. Portoghese
- Subjects
Models, Molecular ,Molecular model ,Stereochemistry ,Molecular Sequence Data ,Molecular Conformation ,Receptors, Opioid, mu ,Ligands ,Carfentanil ,Structure-Activity Relationship ,chemistry.chemical_compound ,Receptors, Opioid, delta ,Drug Discovery ,medicine ,Humans ,R-30490 ,Amino Acid Sequence ,Binding site ,Conformational isomerism ,Binding Sites ,Chemistry ,Receptors, Opioid, kappa ,Stereoisomerism ,Ligand (biochemistry) ,Fentanyl ,Searching the conformational space for docking ,Docking (molecular) ,Molecular Medicine ,medicine.drug - Abstract
The ligand binding modes of a series of fentanyl derivatives are examined using a combination of conformational analysis and molecular docking to the mu-opioid receptor. Condensed-phase molecular dynamics simulations are applied to evaluate potential relationships between ligand conformation and fentanyl substitution and to generate probable "bioactive" structures for the ligand series. Automated docking of the largely populated solution conformers identified a common binding site orientation that places the N-phenethyl group of fentanyl deep in a crevice between transmembrane (TM) helices II and III while the N-phenylpropanamide group projected toward a pocket formed by TM-III, -VI, and -VII domains. An analysis of the binding modes indicates the most potent fentanyl derivatives adopt an extended conformation both in solution and in the bound state, suggesting binding affinity may depend on the conformational preferences of the ligands. The results are consistent with ligand binding data derived from chimeric and mutant receptor studies as well as structure-activity relationship data reported on a wide range of fentanyl analogues. The binding site model is also compared to that of N-phenethylnormorphine. An overlay of the bound conformation of the opiate and cis-3-methylfentanyl shows the N-phenethyl groups occupy equivalent binding domains in the receptor. While the cationic amines of both ligand classes were found docked to an established anchor site (D149 in TM-III), no overlap was observed between the N-phenylpropanamide group and the remaining components of the opiate scaffold. The unique binding mode(s) proposed for the fentanyl series may, in part, explain the difficulties encountered in defining models of recognition at the mu-receptor and suggest opioid receptors may display multiple binding epitopes. Furthermore, the results provide new insight to the design of experiments aimed at understanding the structural basis to the differential selectivities of ligands at the mu-, delta-, and kappa-opioid receptors.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.