1
|
Wtorek K, Ghidini A, Gentilucci L, Adamska-Bartłomiejczyk A, Piekielna-Ciesielska J, Ruzza C, Sturaro C, Calò G, Pieretti S, Kluczyk A, McDonald J, Lambert DG, Janecka A. Synthesis, Biological Activity and Molecular Docking of Chimeric Peptides Targeting Opioid and NOP Receptors. Int J Mol Sci 2022; 23:12700. [PMID: 36293553 PMCID: PMC9604311 DOI: 10.3390/ijms232012700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
Recently, mixed opioid/NOP agonists came to the spotlight for their favorable functional profiles and promising outcomes in clinical trials as novel analgesics. This study reports on two novel chimeric peptides incorporating the fragment Tyr-c[D-Lys-Phe-Phe]Asp-NH2 (RP-170), a cyclic peptide with high affinity for µ and κ opioid receptors (or MOP and KOP, respectively), conjugated with the peptide Ac-RYYRIK-NH2, a known ligand of the nociceptin/orphanin FQ receptor (NOP), yielding RP-170-RYYRIK-NH2 (KW-495) and RP-170-Gly3-RYYRIK-NH2 (KW-496). In vitro, the chimeric KW-496 gained affinity for KOP, hence becoming a dual KOP/MOP agonist, while KW-495 behaved as a mixed MOP/NOP agonist with low nM affinity. Hence, KW-495 was selected for further in vivo experiments. Intrathecal administration of this peptide in mice elicited antinociceptive effects in the hot-plate test; this action was sensitive to both the universal opioid receptor antagonist naloxone and the selective NOP antagonist SB-612111. The rotarod test revealed that KW-495 administration did not alter the mice motor coordination performance. Computational studies have been conducted on the two chimeras to investigate the structural determinants at the basis of the experimental activities, including any role of the Gly3 spacer.
Collapse
Affiliation(s)
- Karol Wtorek
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Alessia Ghidini
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Luca Gentilucci
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | | | | | - Chiara Ruzza
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| | - Chiara Sturaro
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| | - Girolamo Calò
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo Meneghetti 2, 35131 Padova, Italy
| | - Stefano Pieretti
- Istituto Superiore di Sanità, National Center for Drug Research and Evaluation, 00161 Rome, Italy
| | - Alicja Kluczyk
- Faculty of Chemistry, University of Wroclaw, 50-383 Wroclaw, Poland
| | - John McDonald
- Department of Cardiovascular Sciences, University of Leicester, Anaesthesia, Critical Care and Pain Management, Leicester Royal Infirmary, Leicester LE2 7LX, UK
| | - David G. Lambert
- Department of Cardiovascular Sciences, University of Leicester, Anaesthesia, Critical Care and Pain Management, Leicester Royal Infirmary, Leicester LE2 7LX, UK
| | - Anna Janecka
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| |
Collapse
|
2
|
Chen T, Sun T, Bian Y, Pei Y, Feng F, Chi H, Li Y, Tang X, Sang S, Du C, Chen Y, Chen Y, Sun H. The Design and Optimization of Monomeric Multitarget Peptides for the Treatment of Multifactorial Diseases. J Med Chem 2022; 65:3685-3705. [DOI: 10.1021/acs.jmedchem.1c01456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Tingkai Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Tianyu Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Yaoyao Bian
- College of Acupuncture and Massage, College of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing 210023, People’s Republic of China
| | - Yuqiong Pei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People’s Republic of China
| | - Feng Feng
- Food and Pharmaceutical Research Institute, Jiangsu Food and Pharmaceuticals Science College, Huaian 223003, People’s Republic of China
| | - Heng Chi
- Food and Pharmaceutical Research Institute, Jiangsu Food and Pharmaceuticals Science College, Huaian 223003, People’s Republic of China
| | - Yuan Li
- Department of Pharmaceutical Engineering, Jiangsu Food and Pharmaceuticals Science College, Huaian 223005, People’s Republic of China
| | - Xu Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People’s Republic of China
| | - Shenghu Sang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People’s Republic of China
| | - Chenxi Du
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Ying Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People’s Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| |
Collapse
|
3
|
Puls K, Schmidhammer H, Wolber G, Spetea M. Mechanistic Characterization of the Pharmacological Profile of HS-731, a Peripherally Acting Opioid Analgesic, at the µ-, δ-, κ-Opioid and Nociceptin Receptors. Molecules 2022; 27:919. [PMID: 35164182 PMCID: PMC8840597 DOI: 10.3390/molecules27030919] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 02/01/2023] Open
Abstract
Accumulated preclinical and clinical data show that peripheral restricted opioids provide pain relief with reduced side effects. The peripherally acting opioid analgesic HS-731 is a potent dual μ-/δ-opioid receptor (MOR/DOR) full agonist, and a weak, partial agonist at the κ-opioid receptor (KOR). However, its binding mode at the opioid receptors remains elusive. Here, we present a comprehensive in silico evaluation of HS-731 binding at all opioid receptors. We provide insights into dynamic interaction patterns explaining the different binding and activity of HS-731 on the opioid receptors. For this purpose, we conducted docking, performed molecular dynamics (MD) simulations and generated dynamic pharmacophores (dynophores). Our results highlight two residues important for HS-731 recognition at the classical opioid receptors (MOR, DOR and KOR), particular the conserved residue 5.39 (K) and the non-conserved residue 6.58 (MOR: K, DOR: W and KOR: E). Furthermore, we assume a salt bridge between the transmembrane helices (TM) 5 and 6 via K2275.39 and E2976.58 to be responsible for the partial agonism of HS-731 at the KOR. Additionally, we experimentally demonstrated the absence of affinity of HS-731 to the nociceptin/orphanin FQ peptide (NOP) receptor. We consider the morphinan phenol Y1303.33 responsible for this affinity lack. Y1303.33 points deep into the NOP receptor binding pocket preventing HS-731 binding to the orthosteric binding pocket. These findings provide significant structural insights into HS-731 interaction pattern with the opioid receptors that are important for understanding the pharmacology of this peripheral opioid analgesic.
Collapse
Affiliation(s)
- Kristina Puls
- Department of Pharmaceutical Chemistry, Institute of Pharmcy, Freie Universität Berlin, Königin-Luise-Str. 2+4, D-14195 Berlin, Germany;
| | - Helmut Schmidhammer
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria;
| | - Gerhard Wolber
- Department of Pharmaceutical Chemistry, Institute of Pharmcy, Freie Universität Berlin, Königin-Luise-Str. 2+4, D-14195 Berlin, Germany;
| | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria;
| |
Collapse
|
4
|
Santos JLS, Bezerra KS, Barbosa ED, Pereira ACL, Meurer YSR, Oliveira JIN, Gavioli EC, Fulco UL. In silico analysis of energy interactions between nociceptin/orfanin FQ receptor and two antagonists with potential antidepressive action. NEW J CHEM 2022. [DOI: 10.1039/d2nj00916a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study addresses the binding energies of NOPR-ligand complexes and presents the main amino acid residues involved in the interaction between these complexes.
Collapse
Affiliation(s)
- J. L. S. Santos
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970, Natal-RN, Brazil
| | - K. S. Bezerra
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970, Natal-RN, Brazil
| | - E. D. Barbosa
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970, Natal-RN, Brazil
| | - A. C. L. Pereira
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970, Natal-RN, Brazil
| | - Y. S. R. Meurer
- Departamento de Psicologia, Universidade Federal da Paraíba, 58051-900, João Pessoa-PB, Brazil
| | - J. I. N. Oliveira
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970, Natal-RN, Brazil
| | - E. C. Gavioli
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970, Natal-RN, Brazil
| | - U. L. Fulco
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970, Natal-RN, Brazil
| |
Collapse
|
5
|
De Neve J, Barlow TMA, Tourwé D, Bihel F, Simonin F, Ballet S. Comprehensive overview of biased pharmacology at the opioid receptors: biased ligands and bias factors. RSC Med Chem 2021; 12:828-870. [PMID: 34223156 PMCID: PMC8221262 DOI: 10.1039/d1md00041a] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/30/2021] [Indexed: 12/19/2022] Open
Abstract
One of the main challenges in contemporary medicinal chemistry is the development of safer analgesics, used in the treatment of pain. Currently, moderate to severe pain is still treated with the "gold standard" opioids whose long-term often leads to severe side effects. With the discovery of biased agonism, the importance of this area of pharmacology has grown exponentially over the past decade. Of these side effects, tolerance, opioid misuse, physical dependence and substance use disorder (SUD) stand out, since these have led to many deaths over the past decades in both USA and Europe. New therapeutic molecules that induce a biased response at the opioid receptors (MOR, DOR, KOR and NOP receptor) are able to circumvent these side effects and, consequently, serve as more advantageous therapies with great promise. The concept of biased signaling extends far beyond the already sizeable field of GPCR pharmacology and covering everything would be vastly outside the scope of this review which consequently covers the biased ligands acting at the opioid family of receptors. The limitation of quantifying bias, however, makes this a controversial subject, where it is dependent on the reference ligand, the equation or the assay used for the quantification. Hence, the major issue in the field of biased ligands remains the translation of the in vitro profiles of biased signaling, with corresponding bias factors to in vivo profiles showing the presence or the lack of specific side effects. This review comprises a comprehensive overview of biased ligands in addition to their bias factors at individual members of the opioid family of receptors, as well as bifunctional ligands.
Collapse
Affiliation(s)
- Jolien De Neve
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| | - Thomas M A Barlow
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| | - Dirk Tourwé
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| | - Frédéric Bihel
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200, CNRS Université de Strasbourg Illkirch France
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR 7242, CNRS, Université de Strasbourg Illkirch France
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| |
Collapse
|
6
|
Wtorek K, Janecka A. Potential of Nociceptin/Orphanin FQ Peptide Analogs for Drug Development. Chem Biodivers 2021; 18:e2000871. [PMID: 33351271 DOI: 10.1002/cbdv.202000871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 11/27/2020] [Indexed: 12/23/2022]
Abstract
Nociceptin receptor (NOP) belongs to the family of opioid receptors but was discovered and characterized much later than the so called classical opioid receptors, μ, δ and κ (or MOP, DOP and KOP, resp.). Nociceptin/orphanin FQ (N/OFQ) is the endogenous ligand of this receptor and it controls numerous important functions in the central nervous system and in the periphery, so its analogs may be developed as innovative drugs for the treatment of a variety of conditions and pathological states. Availability of potent and selective ligands with high affinity to NOP receptor is essential to fully understand the role of NOP-N/OFQ system in the body, which in turn may lead to designing novel therapeutics. Here, we have focused on reviewing the structure of potent peptide-based agonists, antagonists, biased analogs and bivalent ligands that target NOP receptor.
Collapse
Affiliation(s)
- Karol Wtorek
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, PL-92-215 Lodz, Poland
| | - Anna Janecka
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, PL-92-215 Lodz, Poland
| |
Collapse
|
7
|
Erdei AI, Borbély A, Magyar A, Szűcs E, Ötvös F, Gombos D, Al-Khrasani M, Stefanucci A, Dimmito MP, Luisi G, Mollica A, Benyhe S. Biochemical and pharmacological investigation of novel nociceptin/OFQ analogues and N/OFQ-RYYRIK hybrid peptides. Peptides 2019; 112:106-113. [PMID: 30513351 DOI: 10.1016/j.peptides.2018.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/30/2018] [Accepted: 11/30/2018] [Indexed: 10/27/2022]
Abstract
The endogenous ligand nociceptin (N/OFQ) and a positively charged synthetic peptide RYYRIK are both selective for the nociceptin opioid receptor (NOPr). Despite their structural dissimilarity, N/OFQ and RYYRIK compete for the same binding site of NOP receptor possessing full and partial agonistic character, respectively. In the view of the message-address concept, hybrid peptide constructs were probed for the NOP receptor combining different regions of N/OFQ and RYYRIK related peptide sequences. Nine novel nociceptin- or Ac-RYYRIK-NH2 peptide variants or hybrid peptides were synthesized and characterized. Peptides P2 and P8 contain fragments of native N/OFQ. The other seven analogues (P1, P3-7, P9) are composed of Ac-RYYRIK-NH2 fragments and parts of the original nociceptin sequence. The analogues were characterized in receptor binding assays and G-protein activation experiments on rat brain membranes, as well as by electrically stimulated mouse vas deferens bioassay. In receptor binding assays ligands P2, P4, P6 (Ki 0.37 nM) and P7 showed higher affinity (Ki 0.65 nM, 0.6 nM, 0.37 nM and 0.44 nM, respectively) for NOP receptor than their parent compounds N/OFQ (Ki 2.8 nM) or Ac-RYYRIK-NH2 (Ki 4.2 nM). In [35S]GTPγS binding experiments P2 and P3 behaved as full agonists. The other variants exhibited partial agonist properties characterized by submaximal stimulatory effects. In mouse vas deferens bioassay only P2 showed agonist activity. P4, P5, P6 inhibited the biological activity of N/OFQ more effectively than the NOP receptor selective antagonist JTC-801. In summary, hybrid peptides P4, P5 and P6 proved to be NOP receptor partial agonists even antagonists, while P2 peptide retained the full agonist property.
Collapse
Affiliation(s)
- Anna I Erdei
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62., Hungary; Doctoral School of Theoretical Medicine, University of Szeged, Faculty of Medicine, Szeged, Hungary
| | - Adina Borbély
- MTA-ELTE Research Group of Peptide Chemistry, Hungarian Academy of Sciences, Eötvös Loránd University, H-1117, Budapest, Pázmány Péter sétány 1/A, Hungary
| | - Anna Magyar
- MTA-ELTE Research Group of Peptide Chemistry, Hungarian Academy of Sciences, Eötvös Loránd University, H-1117, Budapest, Pázmány Péter sétány 1/A, Hungary
| | - Edina Szűcs
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62., Hungary; Doctoral School of Theoretical Medicine, University of Szeged, Faculty of Medicine, Szeged, Hungary
| | - Ferenc Ötvös
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62., Hungary
| | - Dávid Gombos
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62., Hungary
| | - Mahmoud Al-Khrasani
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1445 Budapest, Nagyvárad tér 4., Hungary
| | - Azzurra Stefanucci
- Dipartimento di Farmacia, Università̀ degli Studi "G. d'Annunzio" di Chieti-Pescara, Via dei Vestini 31, Chieti, 66100, Italy
| | - Marilisa Pia Dimmito
- Dipartimento di Farmacia, Università̀ degli Studi "G. d'Annunzio" di Chieti-Pescara, Via dei Vestini 31, Chieti, 66100, Italy
| | - Grazia Luisi
- Dipartimento di Farmacia, Università̀ degli Studi "G. d'Annunzio" di Chieti-Pescara, Via dei Vestini 31, Chieti, 66100, Italy
| | - Adriano Mollica
- Dipartimento di Farmacia, Università̀ degli Studi "G. d'Annunzio" di Chieti-Pescara, Via dei Vestini 31, Chieti, 66100, Italy
| | - Sándor Benyhe
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62., Hungary.
| |
Collapse
|
8
|
Malfacini D, Simon K, Trapella C, Guerrini R, Zaveri NT, Kostenis E, Calo’ G. NOP receptor pharmacological profile - A dynamic mass redistribution study. PLoS One 2018; 13:e0203021. [PMID: 30161182 PMCID: PMC6117024 DOI: 10.1371/journal.pone.0203021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022] Open
Abstract
The Nociceptin/Orphanin FQ (N/OFQ) peptide NOP receptor is coupled to pertussis toxin (PTX)-sensitive G proteins (Gi/o) whose activation leads to the inhibition of both cAMP production and calcium channel activity, and to the stimulation of potassium currents. The label free dynamic mass redistribution (DMR) approach has been demonstrated useful for investigating the pharmacological profile of G protein-coupled receptors. Herein, we employ DMR technology to systematically characterize the pharmacology of a large panel of NOP receptor ligands. These are of peptide and non-peptide nature and display varying degrees of receptor efficacy, ranging from full agonism to pure antagonism. Using Chinese hamster ovary (CHO) cells expressing the human NOP receptor we provide rank orders of potency for full and partial agonists as well as apparent affinities for selective antagonists. We find the pharmacological profile of NOP receptor ligands to be similar but not identical to values reported in the literature using canonical assays for Gi/o-coupled receptors. Our data demonstrate that holistic label-free DMR detection can be successfully used to investigate the pharmacology of the NOP receptor and to characterize the cellular effects of novel NOP receptor ligands.
Collapse
Affiliation(s)
- Davide Malfacini
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
- Section of Pharmacology, Department of Medical Sciences, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy
- * E-mail:
| | - Katharina Simon
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Claudio Trapella
- Department of Chemical and Pharmaceutical Sciences and LTTA, University of Ferrara, Ferrara, Italy
| | - Remo Guerrini
- Department of Chemical and Pharmaceutical Sciences and LTTA, University of Ferrara, Ferrara, Italy
| | | | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Girolamo Calo’
- Section of Pharmacology, Department of Medical Sciences, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
9
|
Patent Highlights February-March 2018. Pharm Pat Anal 2018; 7:147-154. [PMID: 29882729 DOI: 10.4155/ppa-2018-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A snapshot of noteworthy recent developments in the patent literature of relevance to pharmaceutical and medical research development.
Collapse
|
10
|
Analysis of natural product regulation of opioid receptors in the treatment of human disease. Pharmacol Ther 2018; 184:51-80. [DOI: 10.1016/j.pharmthera.2017.10.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
11
|
Della Longa S, Arcovito A. “In silico” study of the binding of two novel antagonists to the nociceptin receptor. J Comput Aided Mol Des 2018; 32:385-400. [DOI: 10.1007/s10822-017-0095-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/29/2017] [Indexed: 01/25/2023]
|
12
|
Gavioli EC, Holanda VAD, Ruzza C. NOP Ligands for the Treatment of Anxiety and Mood Disorders. Handb Exp Pharmacol 2018; 254:233-257. [PMID: 30535941 DOI: 10.1007/164_2018_188] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Many studies point toward the nociceptin/orphanin FQ (N/OFQ) and the N/OFQ peptide receptor (NOP) as targets for the development of innovative drugs for treating anxiety- and mood-related disorders. Evidence supports the view that the activation of NOP receptors with agonists elicits anxiolytic-like effects, while its blockade with NOP antagonists promotes antidepressant-like actions in rodents. Genetic studies showed that NOP receptor knockout mice display an antidepressant-like phenotype, and NOP antagonists are inactive in these animals. In contrast, the genetic blockade of NOP receptor signaling generally displays an increase of anxiety states in the elevated plus-maze test. In this chapter we summarized the most relevant findings of NOP receptor ligands in the modulation of anxiety and mood disorders, and the putative mechanisms of action are discussed.
Collapse
Affiliation(s)
- Elaine C Gavioli
- Behavioral Pharmacology Laboratory, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, Brazil.
| | - Victor A D Holanda
- Behavioral Pharmacology Laboratory, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Chiara Ruzza
- Department of Medical Sciences, Section of Pharmacology, and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| |
Collapse
|
13
|
Abstract
The opioid receptor system plays a major role in the regulation of mood, reward, and pain. The opioid receptors therefore make attractive targets for the treatment of many different conditions, including pain, depression, and addiction. However, stimulation or blockade of any one opioid receptor type often leads to on-target adverse effects that limit the clinical utility of a selective opioid agonist or antagonist. Literature precedent suggests that the opioid receptors do not act in isolation and that interactions among the opioid receptors and between the opioid receptors and other proteins may produce clinically useful targets. Multifunctional ligands have the potential to elicit desired outcomes with reduced adverse effects by allowing for the activation of specific receptor conformations and/or signaling pathways promoted as a result of receptor oligomerization or crosstalk. In this chapter, we describe several classes of multifunctional ligands that interact with at least one opioid receptor. These ligands have been designed for biochemical exploration and the treatment of a wide variety of conditions, including multiple kinds of pain, depression, anxiety, addiction, and gastrointestinal disorders. The structures, pharmacological utility, and therapeutic drawbacks of these classes of ligands are discussed.
Collapse
Affiliation(s)
- Jessica P Anand
- Department of Pharmacology, Medical School and the Edward F. Domino Research Center, University of Michigan, Ann Arbor, MI, USA.
| | - Deanna Montgomery
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
14
|
Ferrari F, Malfacini D, Journigan BV, Bird MF, Trapella C, Guerrini R, Lambert DG, Calo' G, Zaveri NT. In vitro pharmacological characterization of a novel unbiased NOP receptor-selective nonpeptide agonist AT-403. Pharmacol Res Perspect 2017; 5. [PMID: 28805972 PMCID: PMC5684865 DOI: 10.1002/prp2.333] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 06/04/2017] [Accepted: 06/06/2017] [Indexed: 12/29/2022] Open
Abstract
Nociceptin/orphanin FQ (N/OFQ) regulates several biological functions via selective activation of the N/OFQ receptor (NOP), a member of the opioid receptor family. We recently identified a new high affinity and highly selective NOP agonist AT-403. In this study, we characterized the functional profile of AT-403 and compared it to other known nonpeptide NOP agonists Ro 65-6570, Ro 2q, SCH-221510, MCOPPB, AT-202 and SCH-486757, using the following assays: GTPγ[35 S] stimulated binding, calcium mobilization assay in cells-expressing human NOP or classical opioid receptors and chimeric G proteins, bioluminescence resonance energy transfer (BRET) based assay for studying NOP receptor interaction with G protein and arrestin, and the electrically stimulated mouse vas deferens bioassay. All compounds behaved as NOP full agonists consistently showing the following rank order of potency MCOPPB > AT-403 > Ro 65-6570 = Ro 2q > SCH-221510 > AT-202 > SCH-486757. AT-403 and MCOPPB displayed the highest NOP selectivity both at human and murine receptors. Interestingly, while all the other nonpeptide NOP agonists displayed bias toward G protein-mediated signaling in the BRET assay, AT-403, similar to the natural ligand N/OFQ, behaved as an unbiased agonist, activating G-protein-mediated function as well as arrestin recruitment. AT-403 may be a useful nonpeptide tool compound to study the pharmacology of NOP activation in disease states.
Collapse
Affiliation(s)
- Federica Ferrari
- Section of Pharmacology, Department of Medical Sciences and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy
| | - Davide Malfacini
- Section of Pharmacology, Department of Medical Sciences and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy
| | - Blair V Journigan
- Astraea Therapeutics, LLC. 320 Logue Avenue, Mountain View, California
| | - Mark F Bird
- Division of Anaesthesia, Department of Cardiovascular Sciences, University of Leicester, Critical Care and Pain Management, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Claudio Trapella
- Department of Chemical and Pharmaceutical Sciences and LTTA, University of Ferrara, Ferrara, Italy
| | - Remo Guerrini
- Department of Chemical and Pharmaceutical Sciences and LTTA, University of Ferrara, Ferrara, Italy
| | - David G Lambert
- Division of Anaesthesia, Department of Cardiovascular Sciences, University of Leicester, Critical Care and Pain Management, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Girolamo Calo'
- Section of Pharmacology, Department of Medical Sciences and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy
| | - Nurulain T Zaveri
- Astraea Therapeutics, LLC. 320 Logue Avenue, Mountain View, California
| |
Collapse
|
15
|
Starnowska J, Guillemyn K, Makuch W, Mika J, Ballet S, Przewlocka B. Bifunctional opioid/nociceptin hybrid KGNOP1 effectively attenuates pain-related behaviour in a rat model of neuropathy. Eur J Pharm Sci 2017; 104:221-229. [PMID: 28347772 DOI: 10.1016/j.ejps.2017.03.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/21/2017] [Accepted: 03/23/2017] [Indexed: 10/19/2022]
Abstract
A bifunctional peptide containing an opioid and nociceptin receptor-binding pharmacophore, H-Dmt-D-Arg-Aba-β-Ala-Arg-Tyr-Tyr-Arg-Ile-Lys-NH2 (KGNOP1), was tested for its analgesic properties when administered intrathecally in naïve and chronic constriction injury (CCI)-exposed rats with neuropathy-like symptoms. KGNOP1 significantly increased the acute pain threshold, as measured by the tail-flick test, and also increased the threshold of a painful reaction to mechanical and thermal stimuli in CCI-exposed rats. Both of the effects could be blocked by pre-administration of [Nphe1]-Nociceptin (1-13)-NH2 (NPhe) or naloxone, antagonists for nociceptin and opioid receptors, respectively. This led us to conclude that KGNOP1 acts as a dual opioid and nociceptin receptor agonist in vivo. The analgesic effect of KGNOP1 proved to be more powerful than clinical drugs such as morphine and buprenorphine. Repeated daily intrathecal injections of KGNOP1 led to the development of analgesic tolerance, with the antiallodynic action being completely abolished on day 6. Nevertheless, the development of tolerance to the antihyperalgesic effect was delayed in comparison to morphine, which lost its efficacy as measured by the cold plate test after 3days of daily intrathecal administration, whereas KGNOP1 was efficient up to day 6. A single intrathecal injection of morphine to KGNOP1-tolerant rats did not raise the pain threshold in any of the behavioural tests; in contrast, a single intrathecal dose of KGNOP1 significantly suppressed allodynia and hyperalgesia in morphine-tolerant rats.
Collapse
Affiliation(s)
- Joanna Starnowska
- Institute of Pharmacology, Department of Pain Pharmacology, Krakow, Poland
| | - Karel Guillemyn
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Brussels, Belgium
| | - Wioletta Makuch
- Institute of Pharmacology, Department of Pain Pharmacology, Krakow, Poland
| | - Joanna Mika
- Institute of Pharmacology, Department of Pain Pharmacology, Krakow, Poland
| | - Steven Ballet
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Barbara Przewlocka
- Institute of Pharmacology, Department of Pain Pharmacology, Krakow, Poland.
| |
Collapse
|
16
|
Lagard C, Chevillard L, Guillemyn K, Risède P, Laplanche JL, Spetea M, Ballet S, Mégarbane B. Bifunctional peptide-based opioid agonist/nociceptin antagonist ligand for dual treatment of nociceptive and neuropathic pain. Pain 2017; 158:505-515. [PMID: 28135212 PMCID: PMC5302413 DOI: 10.1097/j.pain.0000000000000790] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/23/2016] [Accepted: 12/01/2016] [Indexed: 01/24/2023]
Abstract
Drugs able to treat both nociceptive and neuropathic pain effectively without major side effects are lacking. We developed a bifunctional peptide-based hybrid (KGNOP1) that structurally combines a mu-opioid receptor agonist (KGOP1) with antinociceptive activity and a weak nociceptin receptor antagonist (KGNOP3) with anti-neuropathic pain activity. We investigated KGNOP1-related behavioral effects after intravenous administration in rats by assessing thermal nociception, cold hyperalgesia in a model of neuropathic pain induced by chronic constriction injury of the sciatic nerve, and plethysmography parameters including inspiratory time (TI) and minute ventilation (VM) in comparison to the well-known opioid analgesics, tramadol and morphine. Time-course and dose-dependent effects were investigated for all behavioral parameters to determine the effective doses 50% (ED50). Pain-related effects on cold hyperalgesia were markedly increased by KGNOP1 as compared to KGNOP3 and tramadol (ED50: 0.0004, 0.32, and 12.1 μmol/kg, respectively), whereas effects on thermal nociception were significantly higher with KGNOP1 as compared to morphine (ED50: 0.41 and 14.7 μmol/kg, respectively). KGNOP1 and KGOP1 produced a larger increase in TI and deleterious decrease in VM in comparison to morphine and tramadol (ED50(TI): 0.63, 0.52, 12.2, and 50.9 μmol/kg; ED50(VM): 0.57, 0.66, 10.6, and 50.0 μmol/kg, respectively). Interestingly, the calculated ratios of anti-neuropathic pain/antinociceptive to respiratory effects revealed that KGNOP1 was safer than tramadol (ED50 ratio: 5.44 × 10 vs 0.24) and morphine (ED50 ratio: 0.72 vs 1.39). We conclude that KGNOP1 is able to treat both experimental neuropathic and nociceptive pain, more efficiently and safely than tramadol and morphine, respectively, and thus should be a candidate for future clinical developments.
Collapse
Affiliation(s)
- Camille Lagard
- Inserm, UMR-S 1144, Paris, France
- Paris-Descartes University, UMR-S 1144, Paris, France
- Paris-Diderot University, UMR-S 1144, Paris, France
| | - Lucie Chevillard
- Inserm, UMR-S 1144, Paris, France
- Paris-Descartes University, UMR-S 1144, Paris, France
- Paris-Diderot University, UMR-S 1144, Paris, France
| | - Karel Guillemyn
- Research Group of Organic Chemistry, Departments of Chemistry and Bio-engineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Patricia Risède
- Inserm, UMR-S 1144, Paris, France
- Paris-Descartes University, UMR-S 1144, Paris, France
- Paris-Diderot University, UMR-S 1144, Paris, France
| | - Jean-Louis Laplanche
- Inserm, UMR-S 1144, Paris, France
- Paris-Descartes University, UMR-S 1144, Paris, France
- Paris-Diderot University, UMR-S 1144, Paris, France
- Assistance Publique—Hôpitaux de Paris, Lariboisière Hospital, Laboratory of Biochemistry and Molecular Biology, Paris, France
| | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bio-engineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Bruno Mégarbane
- Inserm, UMR-S 1144, Paris, France
- Paris-Descartes University, UMR-S 1144, Paris, France
- Paris-Diderot University, UMR-S 1144, Paris, France
- Assistance Publique—Hôpitaux de Paris, Lariboisière Hospital, Department of Medical and Toxicological Critical Care, Paris, France
| |
Collapse
|
17
|
Ferrari F, Cerlesi MC, Malfacini D, Asth L, Gavioli EC, Journigan BV, Kamakolanu UG, Meyer ME, Yasuda D, Polgar WE, Rizzi A, Guerrini R, Ruzza C, Zaveri NT, Calo G. In vitro functional characterization of novel nociceptin/orphanin FQ receptor agonists in recombinant and native preparations. Eur J Pharmacol 2016; 793:1-13. [PMID: 27780725 DOI: 10.1016/j.ejphar.2016.10.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 10/18/2016] [Accepted: 10/21/2016] [Indexed: 12/21/2022]
Abstract
Nociceptin/Orphanin FQ (N/OFQ) regulates several biological functions via selective activation of the N/OFQ receptor (NOP). In this study novel nonpeptide NOP ligands were characterized in vitro in receptor binding and [35S]GTPγS stimulated binding in membranes of cells expressing human NOP and classical opioid receptors, calcium mobilization assay in cells coexpressing the receptors and chimeric G proteins, bioluminescence resonance energy transfer (BRET) based assay for studying NOP receptor interaction with G protein and arrestin, the electrically stimulated mouse vas deferens and the mouse colon bioassays. The action of the AT compounds were compared with standard NOP agonists (N/OFQ and Ro 65-6570) and the NOP selective antagonist SB-612111. AT compounds displayed high NOP affinity and behaved as NOP agonists in all the functional assays consistently showing the following rank order of potency AT-127≥AT-090≥AT-035>AT-004= AT-001. AT compounds behaved as NOP full agonists in the calcium mobilization and mouse colon assays and as partial agonists in the [35S]GTPγS and BRET assays. Interestingly AT-090 and AT-127, contrary to standard nonpeptide agonists that display G protein biased agonism, behaved as an unbiased agonists. AT-090 and AT-127 displayed higher NOP selectivity than Ro 65-6570 at native mouse receptors. AT-090 and AT-127 might be useful pharmacological tools for investigating the therapeutic potential of NOP partial agonists.
Collapse
Affiliation(s)
- Federica Ferrari
- Section of Pharmacology, Department of Medical Sciences, and National Institute of Neurosciences, University of Ferrara, Italy
| | - Maria Camilla Cerlesi
- Section of Pharmacology, Department of Medical Sciences, and National Institute of Neurosciences, University of Ferrara, Italy
| | - Davide Malfacini
- Section of Pharmacology, Department of Medical Sciences, and National Institute of Neurosciences, University of Ferrara, Italy
| | - Laila Asth
- Behavioral Pharmacology Laboratory, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Elaine C Gavioli
- Behavioral Pharmacology Laboratory, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | | | | - Michael E Meyer
- Astraea Therapeutics, LLC. 320 Logue Avenue, Mountain View, CA, USA
| | - Dennis Yasuda
- Astraea Therapeutics, LLC. 320 Logue Avenue, Mountain View, CA, USA
| | - Willma E Polgar
- SRI International, Biosciences Division, 333 Ravenswood Avenue, Menlo Park, CA, USA
| | - Anna Rizzi
- Section of Pharmacology, Department of Medical Sciences, and National Institute of Neurosciences, University of Ferrara, Italy
| | - Remo Guerrini
- Department of Chemical and Pharmaceutical Sciences and LTTA, University of Ferrara, Italy
| | - Chiara Ruzza
- Section of Pharmacology, Department of Medical Sciences, and National Institute of Neurosciences, University of Ferrara, Italy
| | | | - Girolamo Calo
- Section of Pharmacology, Department of Medical Sciences, and National Institute of Neurosciences, University of Ferrara, Italy.
| |
Collapse
|
18
|
Guillemyn K, Starnowska J, Lagard C, Dyniewicz J, Rojewska E, Mika J, Chung NN, Utard V, Kosson P, Lipkowski AW, Chevillard L, Arranz-Gibert P, Teixidó M, Megarbane B, Tourwé D, Simonin F, Przewlocka B, Schiller PW, Ballet S. Bifunctional Peptide-Based Opioid Agonist-Nociceptin Antagonist Ligands for Dual Treatment of Acute and Neuropathic Pain. J Med Chem 2016; 59:3777-92. [PMID: 27035422 DOI: 10.1021/acs.jmedchem.5b01976] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Herein, the opioid pharmacophore H-Dmt-d-Arg-Aba-β-Ala-NH2 (7) was linked to peptide ligands for the nociceptin receptor. Combination of 7 and NOP ligands (e.g., H-Arg-Tyr-Tyr-Arg-Ile-Lys-NH2) led to binding affinities in the low nanomolar domain. In vitro, the hybrids behaved as agonists at the opioid receptors and antagonists at the nociceptin receptor. Intravenous administration of hybrid 13a (H-Dmt-d-Arg-Aba-β-Ala-Arg-Tyr-Tyr-Arg-Ile-Lys-NH2) to mice resulted in potent and long lasting antinociception in the tail-flick test, indicating that 13a was able to permeate the BBB. This was further supported by a cell-based BBB model. All hybrids alleviated allodynia and hyperalgesia in neuropathic pain models. Especially with respect to hyperalgesia, they showed to be more effective than the parent compounds. Hybrid 13a did not result in significant respiratory depression, in contrast to an equipotent analgesic dose of morphine. These hybrids hence represent a promising avenue toward analgesics for the dual treatment of acute and neuropathic pain.
Collapse
Affiliation(s)
- Karel Guillemyn
- Research Group of Organic Chemistry, Departments of Chemistry and Bio-engineering Sciences, Vrije Universiteit Brussel , Pleinlaan 2, 1050 Brussels, Belgium
| | - Joanna Starnowska
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences , Smetna 12, PL 31-343 Kraków, Poland
| | - Camille Lagard
- Assistance Publique-Hôpitaux de Paris, Hôpital Lariboisière, Réanimation Médicale et Toxicologique, Inserm U1144, Université Paris Descartes UMR-S 1144, Université Paris Didero, UMR-S 1144 , Paris, France
| | - Jolanta Dyniewicz
- Neuropeptide Laboratory, Medical Research Centre, Polish Academy of Sciences , 5 Pawinskiego Street, PL 02-106 Warsaw, Poland
| | - Ewelina Rojewska
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences , Smetna 12, PL 31-343 Kraków, Poland
| | - Joanna Mika
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences , Smetna 12, PL 31-343 Kraków, Poland
| | - Nga N Chung
- Department of Chemical Biology and Peptide Research, Clinical Research Institute , 110 Avenue Des Pins Ouest, Montreal, Quebec H2W 1R7, Canada
| | - Valérie Utard
- University of Strasbourg, CNRS, UMR7242, ESBS , 67412 Illkirch-Graffenstaden, France
| | - Piotr Kosson
- Neuropeptide Laboratory, Medical Research Centre, Polish Academy of Sciences , 5 Pawinskiego Street, PL 02-106 Warsaw, Poland
| | - Andrzej W Lipkowski
- Neuropeptide Laboratory, Medical Research Centre, Polish Academy of Sciences , 5 Pawinskiego Street, PL 02-106 Warsaw, Poland
| | - Lucie Chevillard
- Assistance Publique-Hôpitaux de Paris, Hôpital Lariboisière, Réanimation Médicale et Toxicologique, Inserm U1144, Université Paris Descartes UMR-S 1144, Université Paris Didero, UMR-S 1144 , Paris, France
| | - Pol Arranz-Gibert
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST) , Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Meritxell Teixidó
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST) , Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Bruno Megarbane
- Assistance Publique-Hôpitaux de Paris, Hôpital Lariboisière, Réanimation Médicale et Toxicologique, Inserm U1144, Université Paris Descartes UMR-S 1144, Université Paris Didero, UMR-S 1144 , Paris, France
| | - Dirk Tourwé
- Research Group of Organic Chemistry, Departments of Chemistry and Bio-engineering Sciences, Vrije Universiteit Brussel , Pleinlaan 2, 1050 Brussels, Belgium
| | - Frédéric Simonin
- University of Strasbourg, CNRS, UMR7242, ESBS , 67412 Illkirch-Graffenstaden, France
| | - Barbara Przewlocka
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences , Smetna 12, PL 31-343 Kraków, Poland
| | - Peter W Schiller
- Department of Chemical Biology and Peptide Research, Clinical Research Institute , 110 Avenue Des Pins Ouest, Montreal, Quebec H2W 1R7, Canada
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bio-engineering Sciences, Vrije Universiteit Brussel , Pleinlaan 2, 1050 Brussels, Belgium
| |
Collapse
|
19
|
Toll L, Bruchas MR, Calo' G, Cox BM, Zaveri NT. Nociceptin/Orphanin FQ Receptor Structure, Signaling, Ligands, Functions, and Interactions with Opioid Systems. Pharmacol Rev 2016; 68:419-57. [PMID: 26956246 PMCID: PMC4813427 DOI: 10.1124/pr.114.009209] [Citation(s) in RCA: 219] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The NOP receptor (nociceptin/orphanin FQ opioid peptide receptor) is the most recently discovered member of the opioid receptor family and, together with its endogenous ligand, N/OFQ, make up the fourth members of the opioid receptor and opioid peptide family. Because of its more recent discovery, an understanding of the cellular and behavioral actions induced by NOP receptor activation are less well developed than for the other members of the opioid receptor family. All of these factors are important because NOP receptor activation has a clear modulatory role on mu opioid receptor-mediated actions and thereby affects opioid analgesia, tolerance development, and reward. In addition to opioid modulatory actions, NOP receptor activation has important effects on motor function and other physiologic processes. This review discusses how NOP pharmacology intersects, contrasts, and interacts with the mu opioid receptor in terms of tertiary structure and mechanism of receptor activation; location of receptors in the central nervous system; mechanisms of desensitization and downregulation; cellular actions; intracellular signal transduction pathways; and behavioral actions with respect to analgesia, tolerance, dependence, and reward. This is followed by a discussion of the agonists and antagonists that have most contributed to our current knowledge. Because NOP receptors are highly expressed in brain and spinal cord and NOP receptor activation sometimes synergizes with mu receptor-mediated actions and sometimes opposes them, an understanding of NOP receptor pharmacology in the context of these interactions with the opioid receptors will be crucial to the development of novel therapeutics that engage the NOP receptor.
Collapse
Affiliation(s)
- Lawrence Toll
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Michael R Bruchas
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Girolamo Calo'
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Brian M Cox
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Nurulain T Zaveri
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| |
Collapse
|
20
|
Statnick MA, Chen Y, Ansonoff M, Witkin JM, Rorick-Kehn L, Suter TM, Song M, Hu C, Lafuente C, Jiménez A, Benito A, Diaz N, Martínez-Grau MA, Toledo MA, Pintar JE. A Novel Nociceptin Receptor Antagonist LY2940094 Inhibits Excessive Feeding Behavior in Rodents: A Possible Mechanism for the Treatment of Binge Eating Disorder. J Pharmacol Exp Ther 2015; 356:493-502. [PMID: 26659925 DOI: 10.1124/jpet.115.228221] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 12/02/2015] [Indexed: 11/22/2022] Open
Abstract
Nociceptin/orphanin FQ (N/OFQ), a 17 amino acid peptide, is the endogenous ligand of the ORL1/nociceptin-opioid-peptide (NOP) receptor. N/OFQ appears to regulate a variety of physiologic functions including stimulating feeding behavior. Recently, a new class of thienospiro-piperidine-based NOP antagonists was described. One of these molecules, LY2940094 has been identified as a potent and selective NOP antagonist that exhibited activity in the central nervous system. Herein, we examined the effects of LY2940094 on feeding in a variety of behavioral models. Fasting-induced feeding was inhibited by LY2940094 in mice, an effect that was absent in NOP receptor knockout mice. Moreover, NOP receptor knockout mice exhibited a baseline phenotype of reduced fasting-induced feeding, relative to wild-type littermate controls. In lean rats, LY2940094 inhibited the overconsumption of a palatable high-energy diet, reducing caloric intake to control chow levels. In dietary-induced obese rats, LY2940094 inhibited feeding and body weight regain induced by a 30% daily caloric restriction. Last, in dietary-induced obese mice, LY2940094 decreased 24-hour intake of a high-energy diet made freely available. These are the first data demonstrating that a systemically administered NOP receptor antagonist can reduce feeding behavior and body weight in rodents. Moreover, the hypophagic effect of LY2940094 is NOP receptor dependent and not due to off-target or aversive effects. Thus, LY2940094 may be useful in treating disorders of appetitive behavior such as binge eating disorder, food choice, and overeating, which lead to obesity and its associated medical complications and morbidity.
Collapse
Affiliation(s)
- Michael A Statnick
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.A.S., Y.C., J.M.W., L.R.K., T.M.S., M.S., C.H.); Eli Lilly and Company, Madrid, Spain (C.L., A.J., A.B., N.D., M.A.M.G., M.A.T.); and Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 (M.A., J.E.P.)
| | - Yanyun Chen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.A.S., Y.C., J.M.W., L.R.K., T.M.S., M.S., C.H.); Eli Lilly and Company, Madrid, Spain (C.L., A.J., A.B., N.D., M.A.M.G., M.A.T.); and Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 (M.A., J.E.P.)
| | - Michael Ansonoff
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.A.S., Y.C., J.M.W., L.R.K., T.M.S., M.S., C.H.); Eli Lilly and Company, Madrid, Spain (C.L., A.J., A.B., N.D., M.A.M.G., M.A.T.); and Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 (M.A., J.E.P.)
| | - Jeffrey M Witkin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.A.S., Y.C., J.M.W., L.R.K., T.M.S., M.S., C.H.); Eli Lilly and Company, Madrid, Spain (C.L., A.J., A.B., N.D., M.A.M.G., M.A.T.); and Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 (M.A., J.E.P.)
| | - Linda Rorick-Kehn
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.A.S., Y.C., J.M.W., L.R.K., T.M.S., M.S., C.H.); Eli Lilly and Company, Madrid, Spain (C.L., A.J., A.B., N.D., M.A.M.G., M.A.T.); and Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 (M.A., J.E.P.)
| | - Todd M Suter
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.A.S., Y.C., J.M.W., L.R.K., T.M.S., M.S., C.H.); Eli Lilly and Company, Madrid, Spain (C.L., A.J., A.B., N.D., M.A.M.G., M.A.T.); and Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 (M.A., J.E.P.)
| | - Min Song
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.A.S., Y.C., J.M.W., L.R.K., T.M.S., M.S., C.H.); Eli Lilly and Company, Madrid, Spain (C.L., A.J., A.B., N.D., M.A.M.G., M.A.T.); and Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 (M.A., J.E.P.)
| | - Charlie Hu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.A.S., Y.C., J.M.W., L.R.K., T.M.S., M.S., C.H.); Eli Lilly and Company, Madrid, Spain (C.L., A.J., A.B., N.D., M.A.M.G., M.A.T.); and Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 (M.A., J.E.P.)
| | - Celia Lafuente
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.A.S., Y.C., J.M.W., L.R.K., T.M.S., M.S., C.H.); Eli Lilly and Company, Madrid, Spain (C.L., A.J., A.B., N.D., M.A.M.G., M.A.T.); and Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 (M.A., J.E.P.)
| | - Alma Jiménez
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.A.S., Y.C., J.M.W., L.R.K., T.M.S., M.S., C.H.); Eli Lilly and Company, Madrid, Spain (C.L., A.J., A.B., N.D., M.A.M.G., M.A.T.); and Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 (M.A., J.E.P.)
| | - Ana Benito
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.A.S., Y.C., J.M.W., L.R.K., T.M.S., M.S., C.H.); Eli Lilly and Company, Madrid, Spain (C.L., A.J., A.B., N.D., M.A.M.G., M.A.T.); and Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 (M.A., J.E.P.)
| | - Nuria Diaz
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.A.S., Y.C., J.M.W., L.R.K., T.M.S., M.S., C.H.); Eli Lilly and Company, Madrid, Spain (C.L., A.J., A.B., N.D., M.A.M.G., M.A.T.); and Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 (M.A., J.E.P.)
| | - Maria Angeles Martínez-Grau
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.A.S., Y.C., J.M.W., L.R.K., T.M.S., M.S., C.H.); Eli Lilly and Company, Madrid, Spain (C.L., A.J., A.B., N.D., M.A.M.G., M.A.T.); and Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 (M.A., J.E.P.)
| | - Miguel A Toledo
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.A.S., Y.C., J.M.W., L.R.K., T.M.S., M.S., C.H.); Eli Lilly and Company, Madrid, Spain (C.L., A.J., A.B., N.D., M.A.M.G., M.A.T.); and Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 (M.A., J.E.P.)
| | - John E Pintar
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.A.S., Y.C., J.M.W., L.R.K., T.M.S., M.S., C.H.); Eli Lilly and Company, Madrid, Spain (C.L., A.J., A.B., N.D., M.A.M.G., M.A.T.); and Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 (M.A., J.E.P.)
| |
Collapse
|
21
|
The Importance of Ligand-Receptor Conformational Pairs in Stabilization: Spotlight on the N/OFQ G Protein-Coupled Receptor. Structure 2015; 23:2291-2299. [PMID: 26526853 DOI: 10.1016/j.str.2015.07.024] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 06/15/2015] [Accepted: 07/31/2015] [Indexed: 11/24/2022]
Abstract
Understanding the mechanism by which ligands affect receptor conformational equilibria is key in accelerating membrane protein structural biology. In the case of G protein-coupled receptors (GPCRs), we currently pursue a brute-force approach for identifying ligands that stabilize receptors and facilitate crystallogenesis. The nociceptin/orphanin FQ peptide receptor (NOP) is a member of the opioid receptor subfamily of GPCRs for which many structurally diverse ligands are available for screening. We observed that antagonist potency is correlated with a ligand's ability to induce receptor stability (Tm) and crystallogenesis. Using this screening strategy, we solved two structures of NOP in complex with top candidate ligands SB-612111 and C-35. Docking studies indicate that while potent, stabilizing antagonists strongly favor a single binding orientation, less potent ligands can adopt multiple binding modes, contributing to their low Tm values. These results suggest a mechanism for ligand-aided crystallogenesis whereby potent antagonists stabilize a single ligand-receptor conformational pair.
Collapse
|
22
|
Malfacini D, Ambrosio C, Gro’ MC, Sbraccia M, Trapella C, Guerrini R, Bonora M, Pinton P, Costa T, Calo’ G. Pharmacological Profile of Nociceptin/Orphanin FQ Receptors Interacting with G-Proteins and β-Arrestins 2. PLoS One 2015; 10:e0132865. [PMID: 26248189 PMCID: PMC4527783 DOI: 10.1371/journal.pone.0132865] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 06/18/2015] [Indexed: 01/11/2023] Open
Abstract
Nociceptin/orphanin FQ (N/OFQ) controls several biological functions by selectively activating an opioid like receptor named N/OFQ peptide receptor (NOP). Biased agonism is emerging as an important and therapeutically relevant pharmacological concept in the field of G protein coupled receptors including opioids. To evaluate the relevance of this phenomenon in the NOP receptor, we used a bioluminescence resonance energy transfer technology to measure the interactions of the NOP receptor with either G proteins or β-arrestin 2 in the absence and in presence of increasing concentration of ligands. A large panel of receptor ligands was investigated by comparing their ability to promote or block NOP/G protein and NOP/arrestin interactions. In this study we report a systematic analysis of the functional selectivity of NOP receptor ligands. NOP/G protein interactions (investigated in cell membranes) allowed a precise estimation of both ligand potency and efficacy yielding data highly consistent with the known pharmacological profile of this receptor. The same panel of ligands displayed marked differences in the ability to promote NOP/β-arrestin 2 interactions (evaluated in whole cells). In particular, full agonists displayed a general lower potency and for some ligands an inverted rank order of potency was noted. Most partial agonists behaved as pure competitive antagonists of receptor/arrestin interaction. Antagonists displayed similar values of potency for NOP/Gβ1 or NOP/β-arrestin 2 interaction. Using N/OFQ as reference ligand we computed the bias factors of NOP ligands and a number of agonists with greater efficacy at G protein coupling were identified.
Collapse
Affiliation(s)
- D. Malfacini
- Department of Medical Sciences, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - C. Ambrosio
- Department of Pharmacology, Istituto Superiore di Sanità, Rome, Italy
| | - M. C. Gro’
- Department of Pharmacology, Istituto Superiore di Sanità, Rome, Italy
| | - M. Sbraccia
- Department of Pharmacology, Istituto Superiore di Sanità, Rome, Italy
| | - C. Trapella
- Department of Chemical and Pharmaceutical Sciences and LTTA, University of Ferrara, Ferrara, Italy
| | - R. Guerrini
- Department of Chemical and Pharmaceutical Sciences and LTTA, University of Ferrara, Ferrara, Italy
| | - M. Bonora
- Department of Morphology, Surgery, and Experimental Medicine, Section of Pathology, Oncology, and Experimental Biology and LTTA, University of Ferrara, Ferrara, Italy
| | - P. Pinton
- Department of Morphology, Surgery, and Experimental Medicine, Section of Pathology, Oncology, and Experimental Biology and LTTA, University of Ferrara, Ferrara, Italy
| | - T. Costa
- Department of Pharmacology, Istituto Superiore di Sanità, Rome, Italy
| | - G. Calo’
- Department of Medical Sciences, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
- * E-mail:
| |
Collapse
|
23
|
Zaveri NT, Journigan VB, Polgar WE. Discovery of the first small-molecule opioid pan antagonist with nanomolar affinity at mu, delta, kappa, and nociceptin opioid receptors. ACS Chem Neurosci 2015; 6:646-57. [PMID: 25635572 PMCID: PMC4401318 DOI: 10.1021/cn500367b] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The trans-(3R,4R)-dimethyl-4-(3-hydroxyphenyl)piperidine scaffold is a known pharmacophore for mu opioid (MOP), kappa opioid (KOP), and delta opioid (DOP) receptor antagonists; however, it has not been explored in nociceptin opioid (NOP/ORL-1) receptor ligands. We recently found that the selective KOP antagonist JDTic, (3R)-7-hydroxy-N-((1S)-1-{[(3R,4R)-4-(3-hydroxyphenyl)-3,4-dimethyl-1-piperidinyl]methyl}-2-methylpropyl)-1,2,3,4-tetrahydro-3-isoquinolinecarboxamide, containing this opioid antagonist pharmacophore, has significant binding affinity at the NOP receptor (Ki 16.67 ± 0.76 nM), with no intrinsic activity in the [(35)S]GTPγS functional assay. Since this is the first ligand containing the trans-(3R,4R)-dimethyl-4-(3-hydroxyphenyl)piperidine opioid antagonist pharmacophore to have affinity for the NOP receptor, we explored the structural determinants of its NOP binding affinity. When rational chemical modifications of JDTic were carried out, based on our previously established NOP pharmacophoric structure-activity relationship (SAR) model, most modifications led to a significant decrease in NOP and opioid binding affinity compared to JDTic. Interestingly, however, removal of the 3,4-dimethyl groups of the trans-(3R,4R)-dimethyl-4-(3-hydroxyphenyl)piperidine antagonist scaffold of JDTic increased the binding affinity at NOP by 10-fold (Ki 1.75 ± 0.74 nM) while maintaining comparable affinity for KOP, MOP, and DOP receptors (Ki 1.14 ± 0.63, 1.67 ± 0.6, and 19.6 ± 1.3 nM, respectively). In vitro functional efficacy studies using the [(35)S]GTPγS assay showed that this compound AT-076 functions as an antagonist at all four opioid receptors. Detailed characterization of the antagonist activity of AT-076 shows that it has a noncompetitive antagonist profile at the NOP and KOP receptors (insurmountable antagonism), but is a potent competitive antagonist at the MOP and DOP receptors, with Ke values 3-6-fold more potent than those of JDTic. AT-076 is the first opioid pan antagonist with high affinity at all four opioid receptor subtypes. Our SAR studies show that the 3,4-dimethyl groups of the well-known trans-(3R,4R)-dimethyl-4-(3-hydroxyphenyl)piperidine opioid antagonist scaffold may be removed without significant loss in binding affinity or antagonist potency to obtain an opioid pan antagonist such as AT-076.
Collapse
Affiliation(s)
- Nurulain T. Zaveri
- Astraea
Therapeutics, 320 Logue Avenue, Suite
142, Mountain View, California 94043, United States
| | - V. Blair Journigan
- Astraea
Therapeutics, 320 Logue Avenue, Suite
142, Mountain View, California 94043, United States
| | - Willma E. Polgar
- SRI International,
Biosciences, 333 Ravenswood Avenue, Menlo Park, California 94025, United States
| |
Collapse
|
24
|
Daga PR, Polgar WE, Zaveri NT. Structure-based virtual screening of the nociceptin receptor: hybrid docking and shape-based approaches for improved hit identification. J Chem Inf Model 2014; 54:2732-43. [PMID: 25148595 PMCID: PMC4210177 DOI: 10.1021/ci500291a] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
![]()
The
antagonist-bound crystal structure of the nociceptin receptor
(NOP), from the opioid receptor family, was recently reported along
with those of the other opioid receptors bound to opioid antagonists.
We recently reported the first homology model of the ‘active-state’
of the NOP receptor, which when docked with ‘agonist’
ligands showed differences in the TM helices and residues, consistent
with GPCR activation after agonist binding. In this study, we explored
the use of the active-state NOP homology model for structure-based
virtual screening to discover NOP ligands containing new chemical
scaffolds. Several NOP agonist and antagonist ligands previously reported
are based on a common piperidine scaffold. Given the structure–activity
relationships for known NOP ligands, we developed a hybrid method
that combines a structure-based and ligand-based approach, utilizing
the active-state NOP receptor as well as the pharmacophoric features
of known NOP ligands, to identify novel NOP binding scaffolds by virtual
screening. Multiple conformations of the NOP active site including
the flexible second extracellular loop (EL2) loop were generated by
simulated annealing and ranked using enrichment factor (EF) analysis
and a ligand–decoy dataset containing known NOP agonist ligands.
The enrichment factors were further improved by combining shape-based
screening of this ligand–decoy dataset and calculation of consensus
scores. This combined structure-based and ligand-based EF analysis
yielded higher enrichment factors than the individual methods, suggesting
the effectiveness of the hybrid approach. Virtual screening of the
CNS Permeable subset of the ZINC database was carried out using the
above-mentioned hybrid approach in a tiered fashion utilizing a ligand
pharmacophore-based filtering step, followed by structure-based virtual
screening using the refined NOP active-state models from the enrichment
analysis. Determination of the NOP receptor binding affinity of a
selected set of top-scoring hits resulted in identification of several
compounds with measurable binding affinity at the NOP receptor, one
of which had a new chemotype for NOP receptor binding. The hybrid
ligand-based and structure-based methodology demonstrates an effective
approach for virtual screening that leverages existing SAR and receptor
structure information for identifying novel hits for NOP receptor
binding. The refined active-state NOP homology models obtained from
the enrichment studies can be further used for structure-based optimization
of these new chemotypes to obtain potent and selective NOP receptor
ligands for therapeutic development.
Collapse
Affiliation(s)
- Pankaj R Daga
- Astraea Therapeutics, LLC. , 320 Logue Avenue, Mountain View, California 94043, United States
| | | | | |
Collapse
|
25
|
Corrado S, Sorbi C, Tait A, Battisti UM, Camarda V, Malfacini D, Calò G, Brasili L. 1,4-Dioxolane-triazaspirodecanone derivatives as nociceptin/orphanin FQ receptor ligands. Med Chem Res 2014. [DOI: 10.1007/s00044-014-1032-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
26
|
Toledo MA, Pedregal C, Lafuente C, Diaz N, Martinez-Grau MA, Jiménez A, Benito A, Torrado A, Mateos C, Joshi EM, Kahl SD, Rash KS, Mudra DR, Barth VN, Shaw DB, McKinzie D, Witkin JM, Statnick MA. Discovery of a novel series of orally active nociceptin/orphanin FQ (NOP) receptor antagonists based on a dihydrospiro(piperidine-4,7'-thieno[2,3-c]pyran) scaffold. J Med Chem 2014; 57:3418-29. [PMID: 24678969 DOI: 10.1021/jm500117r] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Nociceptin/OFQ (N/OFQ) is a 17 amino acid peptide that is the endogenous ligand for the ORL1/NOP receptor. Nociceptin appears to regulate a host of physiological functions such as biological reactions to stress, anxiety, mood, and drug abuse, in addition to feeding behaviors. To develop tools to study the function of nociceptin and NOP receptor, our research effort sought to identify orally available NOP antagonists. Our effort led to the discovery of a novel chemical series based on the dihydrospiro(piperidine-4,7'-thieno[2,3-c]pyran) scaffold. Herein we show that dihydrospiro(piperidine-4,7'-thieno[2,3-c]pyran)-derived compounds are potent NOP antagonists with high selectivity versus classical opioid receptors (μ, δ, and κ). Moreover, these compounds exhibit sufficient bioavailability to produce a high level of NOP receptor occupancy in the brain following oral administration in rats.
Collapse
Affiliation(s)
- Miguel A Toledo
- Centro de Investigación Lilly, Avenida de la Industria 30, 28108-Alcobendas, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Gavioli EC, Calo' G. Nociceptin/orphanin FQ receptor antagonists as innovative antidepressant drugs. Pharmacol Ther 2013; 140:10-25. [PMID: 23711793 DOI: 10.1016/j.pharmthera.2013.05.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 05/07/2013] [Indexed: 12/21/2022]
Abstract
Nociceptin/orphanin FQ (N/OFQ) and its receptor (NOP) were identified in the mid 90s as a novel peptidergic system structurally related to opioids. A growing body of preclinical evidence suggests that blockade of NOP receptors evokes antidepressant-like actions. These have been explored using a range of compounds (peptide and non peptide antagonists), across different species (rat and mouse) and assays (behavioral despair and chronic mild stress) suggesting a robust and consistent antidepressant-like effect. Moreover, rats and mice knockout for the NOP receptor gene display an antidepressant-like phenotype in behavioral despair assays. Electrophysiological, immunohistochemical and neurochemical studies point to an important role played by monoaminergic systems, particularly 5-HTergic, in mediating the antidepressant-like properties of NOP antagonists. However other putative mechanisms of action, including modulation of the CRF system, circadian rhythm and a possible neuroendocrine-immune control might be involved. A close relationship between the N/OFQ-NOP receptor system and stress responses is well described in the literature. Stressful situations also alter endocrine, behavioral and neurochemical parameters in rats and chronic administration of a NOP antagonist restored these alterations. Interestingly, clinical findings showed that plasma N/OFQ levels were significantly altered in major and post-partum depression, and bipolar disease patients. Collectively, data in the literature support the notion that blockade of NOP receptor signaling could be a novel and interesting strategy for the development of innovative antidepressants.
Collapse
Affiliation(s)
- Elaine Cristina Gavioli
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, 59078-970 Natal-RN, Brazil.
| | | |
Collapse
|
28
|
Distrutti E, Cipriani S, Mencarelli A, Renga B, Fiorucci S. Probiotics VSL#3 protect against development of visceral pain in murine model of irritable bowel syndrome. PLoS One 2013; 8:e63893. [PMID: 23691109 PMCID: PMC3655059 DOI: 10.1371/journal.pone.0063893] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Accepted: 04/07/2013] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND AIMS Irritable bowel syndrome (IBS) is linked to post-inflammatory and stress-correlated factors that cause changes in the perception of visceral events. Probiotic bacteria may be effective in treating IBS symptoms. Here, we have investigated whether early life administration of VSL#3, a mixture of 8 probiotic bacteria strains, protects against development of visceral hypersensitivity driven by neonatal maternal separation (NMS), a rat model of IBS. METHODS Male NMS pups were treated orally with placebo or VSL#3 from days 3 to 60, while normal, not separated rats were used as controls. After 60 days from birth, perception of painful sensation induced by colorectal distension (CRD) was measured by assessing the abdominal withdrawal reflex (score 0-4). The colonic gene expression was assessed by using the Agilent Whole Rat Genome Oligo Microarrays platform and confirmed by real time PCR. RESULTS NMS rats exhibited both hyperalgesia and allodynia when compared to control rats. VSL#3 had a potent analgesic effect on CRD-induced pain without changing the colorectal compliance. The microarray analysis demonstrated that NMS induces a robust change in the expression of subsets of genes (CCL2, NOS3, THP1, NTRK1, CCR2, BDRKRB1, IL-10, TNFRSF1B, TRPV4, CNR1 and OPRL1) involved in pain transmission and inflammation. TPH1, tryptophan hydroxylase 1, a validated target gene in IBS treatment, was markedly upregulated by NMS and this effect was reversed by VSL#3 intervention. CONCLUSIONS Early life administration of VSL#3 reduces visceral pain perception in a model of IBS and resets colonic expression of subsets of genes mediating pain and inflammation. TRANSCRIPT PROFILING Accession number of repository for expression microarray data is GSE38942 (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE38942).
Collapse
Affiliation(s)
- Eleonora Distrutti
- S.C. di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, Perugia, Italy.
| | | | | | | | | |
Collapse
|
29
|
Tariq S, Nurulain SM, Tekes K, Adeghate E. Deciphering intracellular localization and physiological role of nociceptin and nocistatin. Peptides 2013; 43:174-83. [PMID: 23454174 DOI: 10.1016/j.peptides.2013.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 02/18/2013] [Accepted: 02/18/2013] [Indexed: 01/16/2023]
Abstract
Nociceptin and nocistatin are endogenous ligands of G protein coupled receptor family. Numerous techniques have been used to study the diverse parameters including, localization, distribution and ultrastructure of these peptides. The majority of the study parameters are based on their physiological roles in different organ systems. The present study presents an overview of the different methods used for the study of nociceptin, nocistatin and their receptors. Nociceptin has been implicated in many physiological functions including, nociception, locomotion, stressed-induced analgesia, learning and memory, neurotransmitter and hormone release, renal function, neuronal differentiation, sexual and reproductive behavior, uterine contraction, feeding, anxiety, gastrointestinal motility, cardiovascular function, micturition, cough, hypoxic-ischemic brain injury, diuresis and sodium balance, temperature regulation, vestibular function, and mucosal transport. It has been noted that the use of light and electron microscopy was less frequent, though it may be one of the most promising tools to study the intracellular localization of these neuropeptides. In addition, more studies on the level of circulating nociceptin and nocistatin are also necessary for investigating their clinical roles in health and disease. A variety of modern tools including physiological, light and electron microscopy (EM) are needed to decipher the extent of intracellular localization, tissue distribution and function of these peptides. The intracellular localization of nociceptin and nocistatin will require a high resolution transmission EM capable of identifying these peptides and other supporting molecules that co-localize with them. A tracing technique could also elucidate a possible migratory ability of nociceptin and nocistatin from one cellular compartment to the other.
Collapse
Affiliation(s)
- Saeed Tariq
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | | | | | | |
Collapse
|
30
|
Calo’ G, Guerrini R. Medicinal Chemistry, Pharmacology, and Biological Actions of Peptide Ligands Selective for the Nociceptin/Orphanin FQ Receptor. ACS SYMPOSIUM SERIES 2013. [DOI: 10.1021/bk-2013-1131.ch015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Girolamo Calo’
- Department of Experimental and Clinical Medicine, Section of Pharmacology, University of Ferrara and National Institute of Neuroscience, Italy
- Department of Pharmaceutical Sciences and LTTA (Laboratorio per le Tecnologie delle Terapie Avanzate), University of Ferrara, Italy
| | - Remo Guerrini
- Department of Experimental and Clinical Medicine, Section of Pharmacology, University of Ferrara and National Institute of Neuroscience, Italy
- Department of Pharmaceutical Sciences and LTTA (Laboratorio per le Tecnologie delle Terapie Avanzate), University of Ferrara, Italy
| |
Collapse
|
31
|
Molinari S, Camarda V, Rizzi A, Marzola G, Salvadori S, Marzola E, Molinari P, McDonald J, Ko MC, Lambert DG, Calo' G, Guerrini R. [Dmt1]N/OFQ(1-13)-NH2: a potent nociceptin/orphanin FQ and opioid receptor universal agonist. Br J Pharmacol 2013; 168:151-62. [PMID: 22827708 PMCID: PMC3570011 DOI: 10.1111/j.1476-5381.2012.02115.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 06/21/2012] [Accepted: 06/28/2012] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Intrathecally (i.t.) administered nociceptin/orphanin FQ (N/OFQ) evokes antinociceptive effects in rodents. Recent studies in monkeys demonstrated that i.t. co-application of N/OFQ and morphine elicits synergistic antinociceptive actions suggesting mixed N/OFQ peptide (NOP) and μ opioid receptor agonists as innovative spinal analgesics. Thus, novel N/OFQ related peptides were synthesized in order to identify and pharmacologically characterize a mixed NOP/ μ opioid receptor agonist. EXPERIMENTAL APPROACH The following in vitro assays were used: calcium mobilization in cells expressing the human NOP or classical opioid receptors and chimeric G proteins, receptor and [(35)S]-GTPγS binding, [(35)S]-GTPγS binding in rat spinal cord membranes, guinea pig ileum bioassay. In vivo experiments were performed in monkeys using the tail withdrawal assay. KEY RESULTS From calcium mobilization studies [Dmt(1)]N/OFQ(1-13)-NH(2) was selected as the most potent and least selective compound. The mixed NOP/opioid full agonist activity and high affinity of [Dmt(1)]N/OFQ(1-13)-NH(2) was confirmed at human recombinant receptors in receptor binding, calcium mobilization and/or [(35)S]-GTPγS binding studies, at rat spinal cord receptors in [(35)S]-GTPγS binding experiments, and at guinea pig receptors inhibiting neurogenic contractions in the ileum. In vivo in the tail withdrawal assay in monkeys i.t. [Dmt(1) ]N/OFQ(1-13)-NH(2) was able to elicit robust and long-lasting antinociceptive effects. CONCLUSIONS AND IMPLICATIONS Collectively, these results demonstrate that [Dmt(1)]N/OFQ(1-13)-NH(2) behaves as NOP/opioid receptor universal agonist and substantiate the suggestion that such mixed ligands are worthy of development as innovative spinal analgesics.
Collapse
Affiliation(s)
- S Molinari
- Department of Experimental and Clinical Medicine, Section of Pharmacology, University of Ferrara, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Structural requirements of N-substituted spiropiperidine analogues as agonists of nociceptin/orphanin FQ receptor. Int J Mol Sci 2011; 12:8961-81. [PMID: 22272114 PMCID: PMC3257111 DOI: 10.3390/ijms12128961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 11/10/2011] [Accepted: 11/21/2011] [Indexed: 11/17/2022] Open
Abstract
The nociceptin/orphanin FQ (NOP) receptor is involved in a wide range of biological functions, including pain, anxiety, depression and drug abuse. Especially, its agonists have great potential to be developed into anxiolytics. In this work, both the ligand- and receptor-based three-dimensional quantitative structure–activity relationship (3D-QSAR) studies were carried out using comparative molecular field analysis (CoMFA) and comparative molecular similarity indices analysis (CoMSIA) techniques on 103 N-substituted spiropiperidine analogues as NOP agonists. The resultant optimal ligand-based CoMSIA model exhibited Q2 of 0.501, R2ncv of 0.912 and its predictive ability was validated by using an independent test set of 26 compounds which gave R2pred value of 0.818. In addition, docking analysis and molecular dynamics simulation (MD) were also applied to elucidate the probable binding modes of these agonists. Interpretation of the 3D contour maps, in the context of the topology of the active site of NOP, provided insight into the NOP-agonist interactions. The information obtained from this work can be used to accurately predict the binding affinity of related agonists and also facilitate the future rational design of novel agonists with improved activity.
Collapse
|
33
|
Harrison RS, Ruiz-Gómez G, Hill TA, Chow SY, Shepherd NE, Lohman RJ, Abbenante G, Hoang HN, Fairlie DP. Novel helix-constrained nociceptin derivatives are potent agonists and antagonists of ERK phosphorylation and thermal analgesia in mice. J Med Chem 2010; 53:8400-8. [PMID: 21067234 DOI: 10.1021/jm101139f] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The nociceptin opioid peptide receptor (NOP, NOR, ORL-1) is a GPCR that recognizes nociceptin, a 17-residue peptide hormone. Nociceptin regulates pain transmission, learning, memory, anxiety, locomotion, cardiovascular and respiratory stress, food intake, and immunity. Nociceptin was constrained using an optimized helix-inducing cyclization strategy to produce the most potent NOP agonist (EC50 = 40 pM) and antagonist (IC50 = 7.5 nM) known. Alpha helical structures were measured in water by CD and 2D (1)H NMR spectroscopy. Agonist and antagonist potencies, evaluated by ERK phosphorylation in mouse neuroblastoma cells natively expressing NOR, increased 20-fold and 5-fold, respectively, over nociceptin. Helix-constrained peptides with key amino acid substitutions had much higher in vitro activity, serum stability, and thermal analgesic activity in mice, without cytotoxicity. The most potent agonist increased hot plate contact time from seconds up to 60 min; the antagonist prevented this effect. Such helix-constrained peptides may be valuable physiological probes and therapeutics for treating some forms of pain.
Collapse
Affiliation(s)
- Rosemary S Harrison
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|