1
|
Hovah ME, Holzgrabe U. Bivalent and bitopic ligands of the opioid receptors: The prospects of a dual approach. Med Res Rev 2024; 44:2545-2599. [PMID: 38751227 DOI: 10.1002/med.22050] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 04/09/2024] [Accepted: 05/05/2024] [Indexed: 10/05/2024]
Abstract
Opioid receptors belonging to the class A G-protein coupled receptors (GPCRs) are the targets of choice in the treatment of acute and chronic pain. However, their on-target side effects such as respiratory depression, tolerance and addiction have led to the advent of the 'opioid crisis'. In the search for safer analgesics, bivalent and more recently, bitopic ligands have emerged as valuable tool compounds to probe these receptors. The activity of bivalent and bitopic ligands rely greatly on the allosteric nature of the GPCRs. Bivalent ligands consist of two pharmacophores, each binding to the individual orthosteric binding site (OBS) of the monomers within a dimer. Bitopic or dualsteric ligands bridge the gap between the OBS and the spatially distinct, less conserved allosteric binding site (ABS) through the simultaneous occupation of these two sites. Bivalent and bitopic ligands stabilize distinct conformations of the receptors which ultimately translates into unique signalling and pharmacological profiles. Some of the interesting properties shown by these ligands include improved affinity and/or efficacy, subtype and/or functional selectivity and reduced side effects. This review aims at providing an overview of some of the bivalent and bitopic ligands of the opioid receptors and, their pharmacology in the hope of inspiring the design and discovery of the next generation of opioid analgesics.
Collapse
Affiliation(s)
- Marie Emilie Hovah
- Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany
| | - Ulrike Holzgrabe
- Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany
| |
Collapse
|
2
|
Bird MF, Hebbes CP, Tamang A, Willets JM, Thompson JP, Guerrini R, Calo G, Lambert DG. In vitro sepsis up-regulates Nociceptin/Orphanin FQ receptor expression and function on human T- but not B-cells. Br J Pharmacol 2023; 180:2298-2314. [PMID: 37021779 PMCID: PMC10953342 DOI: 10.1111/bph.16088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 02/24/2023] [Accepted: 03/16/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND AND PURPOSE In animal models of sepsis, increased activation of the Nociceptin/Orphanin FQ (N/OFQ) receptor NOP is associated with mortality and NOP antagonists improved survival. We have explored the role of the N/OFQ-NOP system in freshly isolated volunteer human B- and T-cells incubated with lipopolysaccharide (LPS) and peptidoglycan G (PepG) as a model of in vitro sepsis. EXPERIMENTAL APPROACH B- and T-cell NOP expression was measured using the NOP fluorescent probe N/OFQATTO594 , N/OFQ content was measured using immunofluorescence, N/OFQ release was tracked using a CHOhNOPGαiq5 biosensor assay and NOP function was measured using transwell migration and cytokine/chemokine release using a 25-plex assay format. Cells were challenged with LPS/PepG. KEY RESULTS CD19-positive B-cells bound N/OFQATTO594 ; they also contain N/OFQ. Stimulation with CXCL13/IL-4 increased N/OFQ release. N/OFQ trended to reduced migration to CXCL13/IL-4. Surface NOP expression was unaffected by LPS/PepG, but this treatment increased GM-CSF release in an N/OFQ sensitive manner. CD3-positive T-cells did not bind N/OFQATTO594 ; they did contain N/OFQ. Stimulation with CXCL12/IL-6 increased N/OFQ release. When incubated with LPS/PepG, NOP surface expression was induced leading to N/OFQATTO594 binding. In LPS/PepG-treated cells, N/OFQ reduced migration to CXCL12/IL-6. LPS/PepG increased GM-CSF release in an N/OFQ sensitive manner. CONCLUSIONS AND IMPLICATIONS We suggest both a constitutive and sepsis-inducible N/OFQ-NOP receptor autocrine regulation of B- and T-cell function, respectively. These NOP receptors variably inhibit migration and reduce GM-CSF release. These data provide mechanistic insights to the detrimental role for increased N/OFQ signalling in sepsis and suggest a potential role for NOP antagonists as treatments.
Collapse
Affiliation(s)
- Mark F. Bird
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain ManagementUniversity of LeicesterLeicesterUK
| | - Christopher P. Hebbes
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain ManagementUniversity of LeicesterLeicesterUK
| | | | | | - Jonathan P. Thompson
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain ManagementUniversity of LeicesterLeicesterUK
| | - Remo Guerrini
- Department of Chemical, Pharmaceutical and Agricultural SciencesUniversity of FerraraFerraraItaly
| | - Girolamo Calo
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPaduaItaly
| | - David G. Lambert
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain ManagementUniversity of LeicesterLeicesterUK
| |
Collapse
|
3
|
Lambert DG. Opioids and opioid receptors; understanding pharmacological mechanisms as a key to therapeutic advances and mitigation of the misuse crisis. BJA OPEN 2023; 6:100141. [PMID: 37588171 PMCID: PMC10430815 DOI: 10.1016/j.bjao.2023.100141] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/05/2023] [Accepted: 04/12/2023] [Indexed: 08/18/2023]
Abstract
Opioids are a mainstay in acute pain management and produce their effects and side effects (e.g., tolerance, opioid-use disorder and immune suppression) by interaction with opioid receptors. I will discuss opioid pharmacology in some controversial areas of enquiry of anaesthetic relevance. The main opioid target is the µ (mu,MOP) receptor but other members of the opioid receptor family, δ (delta; DOP) and κ (kappa; KOP) opioid receptors also produce analgesic actions. These are naloxone-sensitive. There is important clinical development relating to the Nociceptin/Orphanin FQ (NOP) receptor, an opioid receptor that is not naloxone-sensitive. Better understanding of the drivers for opioid effects and side effects may facilitate separation of side effects and production of safer drugs. Opioids bind to the receptor orthosteric site to produce their effects and can engage monomer or homo-, heterodimer receptors. Some ligands can drive one intracellular pathway over another. This is the basis of biased agonism (or functional selectivity). Opioid actions at the orthosteric site can be modulated allosterically and positive allosteric modulators that enhance opioid action are in development. As well as targeting ligand-receptor interaction and transduction, modulating receptor expression and hence function is also tractable. There is evidence for epigenetic associations with different types of pain and also substance misuse. As long as the opioid narrative is defined by the 'opioid crisis' the drive to remove them could gather pace. This will deny use where they are effective, and access to morphine for pain relief in low income countries.
Collapse
|
4
|
Giakomidi D, Khemiri S, Mahbuba W, McVey DG, Al-Janabi F, Guerrini R, Calo G, Ye S, Lambert DG. Nociceptin/Orphanin FQ receptor expression in primary human umbilical vein endothelial cells is not regulated by exposure to breast cancer cell media or angiogenic stimuli. BJA OPEN 2022; 4:100110. [PMID: 37588788 PMCID: PMC10430811 DOI: 10.1016/j.bjao.2022.100110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/20/2022] [Indexed: 08/18/2023]
Abstract
Background Opioid receptors are naloxone-sensitive (MOP [mu: μ], DOP [delta: δ], and KOP [kappa: κ]) and naloxone-insensitive Nociceptin/Orphanin FQ (N/OFQ) peptide receptor (NOP). Clinically, most opioid analgesics target MOP. Angiogenesis is the formation of new blood vessels and involves endothelial cell activation, proliferation, and migration. The effect of opioids on this process is controversial with no data for NOP receptor ligands. Methods We used patient-derived human umbilical vein endothelial cells (HUVECs) treated with media from the Michigan Cancer Foundation-7 (MCF-7) breast cancer cells or vascular endothelial growth factor (VEGF; 10 ng ml-1) and fibroblast growth factor (FGF; 10 ng ml-1) as angiogenic stimuli. We have measured (i) NOP/MOP messenger RNA, (ii) receptor protein using N/OFQATTO594 and DermorphinATTO488 as fluorescent probes for NOP and MOP, and (iii) NOP/MOP function in a wound healing assay (crude measure of migration that occurs during angiogenesis). Results HUVEC lines from 32 patients were used. Using all 32 lines, mRNA for NOP but not MOP was detected. This was unaffected by media from MCF-7 cells or VEGF/FGF. There was no binding of either N/OFQATTO594(NOP) or DermorphinATTO488(MOP) in the absence or presence of angiogenic stimuli (six lines tested). In the absence of MOP mRNA, this was expected. Whilst MCF-7 conditioned medium (not VEGF/FGF) reduced wound healing per se (14 lines tested), there was no effect of N/OFQ (NOP ligand) or morphine (MOP ligand). Conclusions Media from MCF-7 breast cancer cells or VEGF/FGF as angiogenic stimuli did not influence NOP translation into receptor protein. MOP was absent. In the absence of constitutive or inducible MOP/NOP, there was no effect on wound healing as a measure of angiogenesis.
Collapse
Affiliation(s)
- Despina Giakomidi
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain Management, Hodgkin Building, University of Leicester, Leicester, UK
| | - Sonja Khemiri
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain Management, Hodgkin Building, University of Leicester, Leicester, UK
| | - Wadhah Mahbuba
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain Management, Hodgkin Building, University of Leicester, Leicester, UK
| | - David G. McVey
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Fatin Al-Janabi
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Remo Guerrini
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Girolamo Calo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Shu Ye
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - David G. Lambert
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain Management, Hodgkin Building, University of Leicester, Leicester, UK
| |
Collapse
|
5
|
In vitro sepsis induces Nociceptin/Orphanin FQ receptor (NOP) expression in primary human vascular endothelial but not smooth muscle cells. PLoS One 2022; 17:e0274080. [PMID: 36107872 PMCID: PMC9477356 DOI: 10.1371/journal.pone.0274080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
Sepsis is a dysregulated host response to infection that can cause widespread effects on other organs including cardiovascular depression, hypotension and organ failure. The receptor for Nociceptin/Orphanin FQ (N/OFQ), NOP is expressed on immune cells and these cells can release the peptide. Exogenous N/OFQ can dilate blood vessels and this peptide is increased in animal and human sepsis. We hypothesise that NOP receptors are present on vascular endothelial cells and therefore provide the target for released N/OFQ to cause vasodilation and hence hypotension. Using human umbilical vein endothelial cells (HUVEC) and human vascular smooth muscle cells (HVSMC) freshly prepared from umbilical cords and up to passage 4, we assessed NOP mRNA expression by Polymerase Chain Reaction (PCR), NOP surface receptor expression using a fluorescent NOP selective probe (N/OFQATTO594) and NOP receptor function with N/OFQ stimulated ERK1/2 phosphorylation. As an in vitro sepsis mimic we variably incubated cells with 100ng/ml Lipopolysaccharide and Peptidoglycan G (LPS/PepG). HUVECs express NOP mRNA and this was reduced by ~80% (n = 49) after 24–48 hours treatment with LPS/PepG. Untreated cells do not express surface NOP receptors but when treated with LPS/PepG the reduced mRNA was translated into protein visualised by N/OFQATTO594 binding (n = 49). These NOP receptors in treated cells produced an N/OFQ (1μM) driven increase in ERK1/2 phosphorylation (n = 20). One (of 50) HUVEC lines expressed NOP mRNA and receptor protein in the absence of LPS/PepG treatment. In contrast, HVSMC expressed NOP mRNA and surface receptor protein (n = 10) independently of LPS/PepG treatment. These receptors were also coupled to ERK1/2 where N/OFQ (1μM) increased phosphorylation. Collectively these data show that an in vitro sepsis mimic (LPS/PepG) upregulates functional NOP expression in the vascular endothelium. Activation of these endothelial receptors as suggested from in vivo whole animal work may contribute to the hypotensive response seen in sepsis. Moreover, blockade of these receptors might be a useful adjunct in the treatment of sepsis.
Collapse
|
6
|
Giakomidi D, Bird MF, Lambert DG. Opioids and cancer survival: are we looking in the wrong place? BJA OPEN 2022; 2:100010. [PMID: 37588274 PMCID: PMC10430855 DOI: 10.1016/j.bjao.2022.100010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 03/30/2022] [Indexed: 08/18/2023]
Abstract
There is a controversial narrative in the anaesthetic literature suggesting that anaesthetic technique (including opioids) may be detrimental to survival after tumour resection. The initial observations were retrospective. Several prospective studies are ongoing; one in breast cancer has reported no adverse outcome. The evidence for an effect of opioids stems from three pieces of information: (1) opioids depress the immune system, (2) opioids potentially promote angiogenesis, and (3) opioids potentially support tumour growth. Although the evidence for (2)/(3) is unclear, combinations of these effects are beneficial to tumours and potentially promote metastatic reseeding. Accepted wisdom suggests that opioid effects are driven by opioid receptor activation but the presence of opioid receptors on immune cells for example is unlikely. Immune cells, vascular endothelium and a range of tumour cells express Toll-like receptor 4 (TLR4) receptors (for Gram-negative bacterial wall components), and there is growing evidence for opioids interacting with this alternative receptor; and for some there is paradoxical naloxone sensitivity. Is the focus on opioid receptors and cancer the wrong target? TLR4 receptor activation produces immune activation, stimulates angiogenesis, and supports tumour survival. We know that some opioids are more immune suppressive than others (there is no such comparative information for angiogenesis and tumour survival); this may correlate with TLR4 activation. If there are clusters of opioids that have more opioid than TLR4 profiles and vice versa, this may influence outcome. If this is the case, then evidence-based advice could be given for perioperative use in the oncology-anaesthesia setting.
Collapse
Affiliation(s)
- Despina Giakomidi
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Hodgkin Building, Leicester, UK
| | - Mark F. Bird
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Hodgkin Building, Leicester, UK
| | - David G. Lambert
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Hodgkin Building, Leicester, UK
| |
Collapse
|
7
|
Al Yacoub ON, Awwad HO, Zhang Y, Standifer KM. Therapeutic potential of nociceptin/orphanin FQ peptide (NOP) receptor modulators for treatment of traumatic brain injury, traumatic stress, and their co-morbidities. Pharmacol Ther 2022; 231:107982. [PMID: 34480968 DOI: 10.1016/j.pharmthera.2021.107982] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/12/2021] [Accepted: 08/12/2021] [Indexed: 12/22/2022]
Abstract
The nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor is a member of the opioid receptor superfamily with N/OFQ as its endogenous agonist. Wide expression of the NOP receptor and N/OFQ, both centrally and peripherally, and their ability to modulate several biological functions has led to development of NOP receptor modulators by pharmaceutical companies as therapeutics, based upon their efficacy in preclinical models of pain, anxiety, depression, Parkinson's disease, and substance abuse. Both posttraumatic stress disorder (PTSD) and traumatic brain injury (TBI) are debilitating conditions that significantly affect the quality of life of millions of people around the world. PTSD is often a consequence of TBI, and, especially for those deployed to, working and/or living in a war zone or are first responders, they are comorbid. PTSD and TBI share common symptoms, and negatively influence outcomes as comorbidities of the other. Unfortunately, a lack of effective therapies or therapeutic agents limits the long term quality of life for either TBI or PTSD patients. Ours, and other groups, demonstrated that PTSD and TBI preclinical models elicit changes in the N/OFQ-NOP receptor system, and that administration of NOP receptor ligands alleviated some of the neurobiological and behavioral changes induced by brain injury and/or traumatic stress exposure. Here we review the past and most recent progress on understanding the role of the N/OFQ-NOP receptor system in PTSD and TBI neurological and behavioral sequelae. There is still more to understand about this neuropeptide system in both PTSD and TBI, but current findings warrant further examination of the potential utility of NOP modulators as therapeutics for these disorders and their co-morbidities. We advocate the development of standards for common data elements (CDE) reporting for preclinical PTSD studies, similar to current preclinical TBI CDEs. That would provide for more standardized data collection and reporting to improve reproducibility, interpretation and data sharing across studies.
Collapse
Affiliation(s)
- Omar N Al Yacoub
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, OUHSC, Oklahoma City, OK 73117, United States of America
| | - Hibah O Awwad
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, OUHSC, Oklahoma City, OK 73117, United States of America
| | - Yong Zhang
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, OUHSC, Oklahoma City, OK 73117, United States of America
| | - Kelly M Standifer
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, OUHSC, Oklahoma City, OK 73117, United States of America.
| |
Collapse
|
8
|
Giakomidi D, Bird MF, Guerrini R, Calo G, Lambert DG. Fluorescent opioid receptor ligands as tools to study opioid receptor function. J Pharmacol Toxicol Methods 2021; 113:107132. [PMID: 34728348 DOI: 10.1016/j.vascn.2021.107132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/12/2021] [Accepted: 10/25/2021] [Indexed: 11/30/2022]
Abstract
Opioid receptors are divided into the three classical types: MOP(μ:mu), DOP(δ:delta) and KOP(κ:kappa) that are naloxone-sensitive and an additional naloxone-insensitive nociceptin/orphanin FQ(N/OFQ) peptide receptor(NOP). Studies to determine opioid receptor location and turnover variably rely on; (i) measuring receptor mRNA, (ii) genetically tagging receptors, (iii) labelling receptors with radioligands, (iv) use of antibodies in immunohistochemistry/Western Blotting or (v) measuring receptor function coupled with the use of selective antagonists. All have their drawbacks with significant issues relating to mRNA not necessarily predicting protein, poor antibody selectivity and utility of radiolabels in low expression systems. In this minireview we discuss use of fluorescently labelled opioid receptor ligands. To maintain the pharmacological properties of the corresponding parent ligand fluorescently labelled ligands must take into account fluorophore (brightness and propensity to bleach), linker length and chemistry, and site to which the linker (and hence probe) will be attached. Use of donor and acceptor fluorophores with spectral overlap facilitates use in FRET type assays to determine proximity of ligand or tagged receptor pairs. There is a wide range of probes of agonist and antagonist nature for all four opioid receptor types; caution is needed with agonist probes due to the possibility for internalization. We have produced two novel ATTO based probes; DermorphinATTO488 (MOP) and N/OFQATTO594 (NOP). These probes label MOP and NOP in a range of preparations and using N/OFQATTO594 we demonstrate internalization and ligand-receptor interaction by FRET. Fluorescent opioid probes offer potential methodological advantages over more traditional use of antibodies and radiolabels.
Collapse
Affiliation(s)
- Despina Giakomidi
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Hodgkin Building, Leicester LE1 9HN. UK
| | - Mark F Bird
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Hodgkin Building, Leicester LE1 9HN. UK
| | - Remo Guerrini
- Department of Chemical, Pharmaceutical and Agricultural Sciences and LTTA, University of Ferrara, Italy
| | - Girolamo Calo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - David G Lambert
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Hodgkin Building, Leicester LE1 9HN. UK.
| |
Collapse
|
9
|
Gornalusse GG, Vojtech LN, Levy CN, Hughes SM, Kim Y, Valdez R, Pandey U, Ochsenbauer C, Astronomo R, McElrath J, Hladik F. Buprenorphine Increases HIV-1 Infection In Vitro but Does Not Reactivate HIV-1 from Latency. Viruses 2021; 13:1472. [PMID: 34452338 PMCID: PMC8402857 DOI: 10.3390/v13081472] [Citation(s) in RCA: 8] [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: 04/27/2021] [Revised: 06/28/2021] [Accepted: 07/24/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND medication-assisted treatment (MAT) with buprenorphine is now widely prescribed to treat addiction to heroin and other illicit opioids. There is some evidence that illicit opioids enhance HIV-1 replication and accelerate AIDS pathogenesis, but the effect of buprenorphine is unknown. METHODS we obtained peripheral blood mononuclear cells (PBMCs) from healthy volunteers and cultured them in the presence of morphine, buprenorphine, or methadone. We infected the cells with a replication-competent CCR5-tropic HIV-1 reporter virus encoding a secreted nanoluciferase gene, and measured infection by luciferase activity in the supernatants over time. We also surveyed opioid receptor expression in PBMC, genital epithelial cells and other leukocytes by qPCR and western blotting. Reactivation from latency was assessed in J-Lat 11.1 and U1 cell lines. RESULTS we did not detect expression of classical opioid receptors in leukocytes, but did find nociception/orphanin FQ receptor (NOP) expression in blood and vaginal lymphocytes as well as genital epithelial cells. In PBMCs, we found that at physiological doses, morphine, and methadone had a variable or no effect on HIV infection, but buprenorphine treatment significantly increased HIV-1 infectivity (median: 8.797-fold increase with 20 nM buprenorphine, eight experiments, range: 3.570-691.9, p = 0.0078). Using latently infected cell lines, we did not detect reactivation of latent HIV following treatment with any of the opioid drugs. CONCLUSIONS our results suggest that buprenorphine, in contrast to morphine or methadone, increases the in vitro susceptibility of leukocytes to HIV-1 infection but has no effect on in vitro HIV reactivation. These findings contribute to our understanding how opioids, including those used for MAT, affect HIV infection and reactivation, and can help to inform the choice of MAT for people living with HIV or who are at risk of HIV infection.
Collapse
Affiliation(s)
- Germán Gustavo Gornalusse
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Lucia N. Vojtech
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Claire N. Levy
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Sean M. Hughes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Yeseul Kim
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Rogelio Valdez
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
| | - Urvashi Pandey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Christina Ochsenbauer
- School of Medicine, Division of Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Rena Astronomo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
| | - Julie McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Pathobiology, Global Health and Laboratory Medicine, University of Washington, Seattle, WA 98195, USA
| | - Florian Hladik
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
10
|
Giakomidi D, Bird MF, McDonald J, Marzola E, Guerrini R, Chanoch S, Sabu N, Horley B, Calo G, Lambert DG. Evaluation of [Cys(ATTO 488)8]Dermorphin-NH2 as a novel tool for the study of μ-opioid peptide receptors. PLoS One 2021; 16:e0250011. [PMID: 33891604 PMCID: PMC8064508 DOI: 10.1371/journal.pone.0250011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/29/2021] [Indexed: 11/26/2022] Open
Abstract
The μ-opioid peptide (MOP) receptor is a member of the opioid receptor family and an important clinical target for analgesia. Measuring MOP receptor location and tracking its turnover traditionally used radiolabels or antibodies with attendant problems of utility of radiolabels in whole cells and poor antibody selectivity. To address these issues we have synthesized and characterised a novel ATTO488 based fluorescent Dermorphin analogue; [Cys(ATTO 488)8]Dermorphin-NH2 (DermATTO488). We initially assessed the binding profile of DermATTO488 in HEK cells expressing human MOP and CHO cells expressing human MOP, δ-opioid peptide (DOP), κ-opioid peptide (KOP) and Nociceptin/Orphanin FQ peptide (NOP) receptors using radioligand binding. Functional activity of the conjugated peptide was assessed by measuring (i) the ability of the ligand to engage G-protein by measuring the ability to stimulate GTPγ[35S] binding and (ii) the ability to stimulate phosphorylation of ERK1/2. Receptor location was visualised using confocal scanning laser microscopy. Dermorphin and DermATTO488 bound to HEKMOP (pKi: 8.29 and 7.00; p<0.05), CHOMOP (pKi: 9.26 and 8.12; p<0.05) and CHODOP (pKi: 7.03 and 7.16; p>0.05). Both ligands were inactive at KOP and NOP. Dermorphin and DermATTO488 stimulated the binding of GTPγ[35S] with similar pEC50 (7.84 and 7.62; p>0.05) and Emax (1.52 and 1.34fold p>0.05) values. Moreover, Dermorphin and DermATTO488 produced a monophasic stimulation of ERK1/2 phosphorylation peaking at 5mins (6.98 and 7.64-fold; p>0.05). Finally, in confocal microscopy DermATTO488 bound to recombinant MOP receptors on CHO and HEK cells in a concentration dependent manner that could be blocked by pre-incubation with unlabelled Dermorphin or Naloxone. Collectively, addition to ATTO488 to Dermorphin produced a ligand not dissimilar to Dermorphin; with ~10fold selectivity over DOP. This new ligand DermATTO488 retained functional activity and could be used to visualise MOP receptor location.
Collapse
Affiliation(s)
- Despina Giakomidi
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Mark F. Bird
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Leicester Royal Infirmary, Leicester, United Kingdom
| | - John McDonald
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Erika Marzola
- 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
| | - Serena Chanoch
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Nidhuna Sabu
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Barbara Horley
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Girolamo Calo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - David G. Lambert
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Leicester Royal Infirmary, Leicester, United Kingdom
- * E-mail:
| |
Collapse
|
11
|
Azevedo Neto J, Ruzza C, Sturaro C, Malfacini D, Pacifico S, Zaveri NT, Calò G. Functional Selectivity Does Not Predict Antinociceptive/Locomotor Impairing Potencies of NOP Receptor Agonists. Front Neurosci 2021; 15:657153. [PMID: 33859548 PMCID: PMC8042269 DOI: 10.3389/fnins.2021.657153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/10/2021] [Indexed: 11/13/2022] Open
Abstract
Nociceptin/orphanin FQ controls several functions, including pain transmission, via stimulation of the N/OFQ peptide (NOP) receptor. Here we tested the hypothesis that NOP biased agonism may be instrumental for identifying innovative analgesics. In vitro experiments were performed with the dynamic mass redistribution label free assay and the NOP non-peptide agonists Ro 65-6570, AT-403 and MCOPPB. In vivo studies were performed in wild type and β-arrestin 2 knockout mice using the formalin, rotarod and locomotor activity tests. In vitro all compounds mimicked the effects of N/OFQ behaving as potent NOP full agonists. In vivo Ro 65-6570 demonstrated a slightly higher therapeutic index (antinociceptive vs. motor impairment effects) in knockout mice. However, all NOP agonists displayed very similar therapeutic index in normal mice despite significant differences in G protein biased agonism. In conclusion the different ability of inducing G protein vs. β-arrestin 2 recruitment of a NOP agonist cannot be applied to predict its antinociceptive vs. motor impairment properties.
Collapse
Affiliation(s)
- Joaquim Azevedo Neto
- Section of Pharmacology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Chiara Ruzza
- Section of Pharmacology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy.,Technopole of Ferrara, LTTA Laboratory for Advanced Therapies, Ferrara, Italy
| | - Chiara Sturaro
- Section of Pharmacology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Davide Malfacini
- Section of Pharmacology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Salvatore Pacifico
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| | | | - Girolamo Calò
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| |
Collapse
|
12
|
Topical Treatments and Their Molecular/Cellular Mechanisms in Patients with Peripheral Neuropathic Pain-Narrative Review. Pharmaceutics 2021; 13:pharmaceutics13040450. [PMID: 33810493 PMCID: PMC8067282 DOI: 10.3390/pharmaceutics13040450] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 12/25/2022] Open
Abstract
Neuropathic pain in humans results from an injury or disease of the somatosensory nervous system at the peripheral or central level. Despite the considerable progress in pain management methods made to date, peripheral neuropathic pain significantly impacts patients' quality of life, as pharmacological and non-pharmacological methods often fail or induce side effects. Topical treatments are gaining popularity in the management of peripheral neuropathic pain, due to excellent safety profiles and preferences. Moreover, topical treatments applied locally may target the underlying mechanisms of peripheral sensitization and pain. Recent studies showed that peripheral sensitization results from interactions between neuronal and non-neuronal cells, with numerous signaling molecules and molecular/cellular targets involved. This narrative review discusses the molecular/cellular mechanisms of drugs available in topical formulations utilized in clinical practice and their effectiveness in clinical studies in patients with peripheral neuropathic pain. We searched PubMed for papers published from 1 January 1995 to 30 November 2020. The key search phrases for identifying potentially relevant articles were "topical AND pain", "topical AND neuropathic", "topical AND treatment", "topical AND mechanism", "peripheral neuropathic", and "mechanism". The result of our search was 23 randomized controlled trials (RCT), 9 open-label studies, 16 retrospective studies, 20 case (series) reports, 8 systematic reviews, 66 narrative reviews, and 140 experimental studies. The data from preclinical studies revealed that active compounds of topical treatments exert multiple mechanisms of action, directly or indirectly modulating ion channels, receptors, proteins, and enzymes expressed by neuronal and non-neuronal cells, and thus contributing to antinociception. However, which mechanisms and the extent to which the mechanisms contribute to pain relief observed in humans remain unclear. The evidence from RCTs and reviews supports 5% lidocaine patches, 8% capsaicin patches, and botulinum toxin A injections as effective treatments in patients with peripheral neuropathic pain. In turn, single RCTs support evidence of doxepin, funapide, diclofenac, baclofen, clonidine, loperamide, and cannabidiol in neuropathic pain states. Topical administration of phenytoin, ambroxol, and prazosin is supported by observational clinical studies. For topical amitriptyline, menthol, and gabapentin, evidence comes from case reports and case series. For topical ketamine and baclofen, data supporting their effectiveness are provided by both single RCTs and case series. The discussed data from clinical studies and observations support the usefulness of topical treatments in neuropathic pain management. This review may help clinicians in making decisions regarding whether and which topical treatment may be a beneficial option, particularly in frail patients not tolerating systemic pharmacotherapy.
Collapse
|
13
|
Abstract
This paper is the forty-first consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2018 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (2), the roles of these opioid peptides and receptors in pain and analgesia in animals (3) and humans (4), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (5), opioid peptide and receptor involvement in tolerance and dependence (6), stress and social status (7), learning and memory (8), eating and drinking (9), drug abuse and alcohol (10), sexual activity and hormones, pregnancy, development and endocrinology (11), mental illness and mood (12), seizures and neurologic disorders (13), electrical-related activity and neurophysiology (14), general activity and locomotion (15), gastrointestinal, renal and hepatic functions (16), cardiovascular responses (17), respiration and thermoregulation (18), and immunological responses (19).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY, 11367, United States.
| |
Collapse
|
14
|
Zhao C, Bobe G, Wang Y, Zhang X, Zhao Z, Zhang S, Sun G, Yuan X, Li X, Liu G. Potential Role of SLC5A8 Expression in the Etiology of Subacute Ruminal Acidosis. Front Vet Sci 2020; 7:394. [PMID: 32850999 PMCID: PMC7406710 DOI: 10.3389/fvets.2020.00394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/02/2020] [Indexed: 12/23/2022] Open
Abstract
Rumen fluid of cows with subacute ruminal acidosis (SARA) has high concentrations of short chain fatty acids (SCFA). However, the mechanism of SCFA accumulation is unknown. The solute-linked carrier 5a8 (SLC5A8) plays a key role in the transportation and absorption of SCFA in the intestinal epithelium. The objective of the current study was to investigate (1) SLC5A8 gene and protein expression in various parts of the bovine gastrointestinal tract, (2) the effect of SCFA on SLC5A8 expression in rumen epithelial cells, and (3) SLC5A8 gene and protein expression in SARA and healthy cows. A total of 10 dairy cows, 84 ± 26 days in milk and in their second to fourth parity were allocated to control (n = 5) and SARA groups (n = 5). Three cows from the control group and three calves (1-day-old, female, 45–50 kg, healthy, fasting) were chosen to collect a total of 10 sections of digestive tract, from rumen to rectum, and then bovine ruminal epithelial cells were isolated from the three calves. Gene and protein expression of SLC5A8 was detected in all tested regions of the gastrointestinal tract in calves and adult cows by Western blot and quantitative real-time PCR and were both highest in the rumen. Gene and protein expression of SLC5A8 was more than 50% lower in the rumen epithelium of SARA vs. control cows and was partly restored after therapy of SARA cows. Compared with SCFA concentrations typical for control cows (60 mM acetate, 30 mM propionate, and 20 mM butyrate), gene and protein expression of SLC5A8 in rumen epithelium was lower at elevated SCFA concentrations typical for SARA cows (90 mM acetate, 40 mM propionate, and 30 mM butyrate), specifically for elevated concentrations of propionate or butyrate in contrast to elevated concentrations of acetate increased gene and protein expression of SLC5A8 in rumen epithelium. In conclusion, the elevated concentrations of propionate and butyrate inhibit ruminal absorption of SCFA via downregulation of SLC5A8 in SARA cows; the expression of SLC5A8 plays an important role in the etiology of SARA.
Collapse
Affiliation(s)
- Chenxu Zhao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Gerd Bobe
- Department of Animal Sciences, Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Yazhou Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xinyue Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhibo Zhao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shiqi Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Guoquan Sun
- College of Animal Science and Technology, Inner Mongolia National University, Tongliao, China
| | - Xue Yuan
- College of Animal Science and Technology, Inner Mongolia National University, Tongliao, China
| | - Xinwei Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Guowen Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
15
|
Machelska H, Celik MÖ. Opioid Receptors in Immune and Glial Cells-Implications for Pain Control. Front Immunol 2020; 11:300. [PMID: 32194554 PMCID: PMC7064637 DOI: 10.3389/fimmu.2020.00300] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 02/06/2020] [Indexed: 11/23/2022] Open
Abstract
Opioid receptors comprise μ (MOP), δ (DOP), κ (KOP), and nociceptin/orphanin FQ (NOP) receptors. Opioids are agonists of MOP, DOP, and KOP receptors, whereas nociceptin/orphanin FQ (N/OFQ) is an agonist of NOP receptors. Activation of all four opioid receptors in neurons can induce analgesia in animal models, but the most clinically relevant are MOP receptor agonists (e.g., morphine, fentanyl). Opioids can also affect the function of immune cells, and their actions in relation to immunosuppression and infections have been widely discussed. Here, we analyze the expression and the role of opioid receptors in peripheral immune cells and glia in the modulation of pain. All four opioid receptors have been identified at the mRNA and protein levels in immune cells (lymphocytes, granulocytes, monocytes, macrophages) in humans, rhesus monkeys, rats or mice. Activation of leukocyte MOP, DOP, and KOP receptors was recently reported to attenuate pain after nerve injury in mice. This involved intracellular Ca2+-regulated release of opioid peptides from immune cells, which subsequently activated MOP, DOP, and KOP receptors on peripheral neurons. There is no evidence of pain modulation by leukocyte NOP receptors. More good quality studies are needed to verify the presence of DOP, KOP, and NOP receptors in native glia. Although still questioned, MOP receptors might be expressed in brain or spinal cord microglia and astrocytes in humans, mice, and rats. Morphine acting at spinal cord microglia is often reported to induce hyperalgesia in rodents. However, most studies used animals without pathological pain and/or unconventional paradigms (e.g., high or ultra-low doses, pain assessment after abrupt discontinuation of chronic morphine treatment). Therefore, the opioid-induced hyperalgesia can be viewed in the context of dependence/withdrawal rather than pain management, in line with clinical reports. There is convincing evidence of analgesic effects mediated by immune cell-derived opioid peptides in animal models and in humans. Together, MOP, DOP, and KOP receptors, and opioid peptides in immune cells can ameliorate pathological pain. The relevance of NOP receptors and N/OFQ in leukocytes, and of all opioid receptors, opioid peptides and N/OFQ in native glia for pain control is yet to be clarified.
Collapse
Affiliation(s)
- Halina Machelska
- Department of Experimental Anesthesiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Melih Ö Celik
- Department of Experimental Anesthesiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
16
|
Abstract
The nociceptin/orphanin FQ (N/OFQ) peptide receptor (NOP) is a G protein-coupled receptor involved in the regulation of several physiological functions and pathological conditions. Thus, researchers from academia and industry are pursuing NOP to discover and study novel pharmacological entities. In a multidisciplinary effort of pharmacologists, medicinal chemists, and molecular and structural biologists the mechanisms of NOP activation and inhibition have been, at least partially, disentangled. Here, we review the in vitro methodologies employed, which have contributed to our understanding of this target. We hope this chapter guides the reader through the mostly established assay platforms to investigate NOP pharmacology, and gives some hints taking advantage from what has already illuminated the function of other GPCRs. We analyzed the pharmacological results obtained with a large panel of NOP ligands investigated in several assays including receptor binding, stimulation of GTPγS binding, decrease of cAMP levels, calcium flux stimulation via chimeric G proteins, NOP/G protein and NOP/β-arrestin interaction, label-free assays such as dynamic mass redistribution, and bioassays such as the electrically stimulated mouse vas deferens.
Collapse
Affiliation(s)
- Davide Malfacini
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Girolamo Caló
- Section of Pharmacology, Department of Medical Sciences, National Institute of Neurosciences, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
17
|
Kadhim S, Bird MF, Lambert DG. N/OFQ-NOP System in Peripheral and Central Immunomodulation. Handb Exp Pharmacol 2019; 254:297-311. [PMID: 30771012 DOI: 10.1007/164_2018_203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Classical opioids (μ: mu, MOP; δ: delta, DOP and κ: kappa, KOP) variably affect immune function; they are immune depressants and there is good clinical evidence in the periphery. In addition, there is evidence for a central role in the control of a number of neuropathologies, e.g., neuropathic pain. Nociceptin/Orphanin FQ (N/OFQ) is the endogenous ligand for the N/OFQ peptide receptor, NOP; peripheral and central activation can modulate immune function. In the periphery, NOP activation generally depresses immune function, but unlike classical opioids this is in part driven by NOP located on circulating immune cells. Peripheral activation has important implications in pathologies like asthma and sepsis. NOP is expressed on central neurones and glia where activation can modulate glial function. Microglia, as resident central 'macrophages', increase/infiltrate in pain and following trauma; these changes can be reduced by N/OFQ. Moreover, the interaction with other glial cell types such as the ubiquitous astrocytes and their known cross talk with microglia open a wealth of possibilities for central immunomodulation. At the whole animal level, clinical ligands with wide central and peripheral distribution have the potential to modulate immune function, and defining the precise nature of that interaction is important in mitigating or even harnessing the adverse effect profile of these important drugs.
Collapse
Affiliation(s)
- Salim Kadhim
- Department of Cardiovascular Sciences, Anaesthesia Critical Care and Pain Management, University of Leicester, Leicester Royal Infirmary, Leicester, UK
| | - Mark F Bird
- Department of Cardiovascular Sciences, Anaesthesia Critical Care and Pain Management, University of Leicester, Leicester Royal Infirmary, Leicester, UK
| | - David G Lambert
- Department of Cardiovascular Sciences, Anaesthesia Critical Care and Pain Management, University of Leicester, Leicester Royal Infirmary, Leicester, UK.
| |
Collapse
|
18
|
Bird MF, Guerrini R, Willets JM, Thompson JP, Caló G, Lambert DG. Nociceptin/Orphanin FQ (N/OFQ) conjugated to ATTO594: a novel fluorescent probe for the N/OFQ (NOP) receptor. Br J Pharmacol 2018; 175:4496-4506. [PMID: 30276802 PMCID: PMC6255954 DOI: 10.1111/bph.14504] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/31/2018] [Accepted: 09/04/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE The nociceptin/orphanin FQ (N/OFQ) receptor (NOP) is a member of the opioid receptor family and is involved in a number of physiological responses, pain and immune regulation as examples. In this study, we conjugated a red fluorophore-ATTO594 to the peptide ligand N/OFQ (N/OFQATTO594 ) for the NOP receptor and explored NOP receptor function at high (in recombinant systems) and low (on immune cells) expression. EXPERIMENTAL APPROACH We assessed N/OFQATTO594 receptor binding, selectivity and functional activity in recombinant (CHO) cell lines. Live cell N/OFQATTO594 binding was measured in (i) HEK cells expressing NOP and NOPGFP receptors, (ii) CHO cells expressing the hNOPGαqi5 chimera (to force coupling to measurable Ca2+ responses) and (iii) freshly isolated human polymorphonuclear cells (PMN). KEY RESULTS N/OFQATTO594 bound to NOP receptor with nM affinity and high selectivity. N/OFQATTO594 activated NOP receptor by reducing cAMP formation and increasing Ca2+ levels in CHOhNOPGαqi5 cells. N/OFQATTO594 was also able to visualize NOP receptors at low expression levels on PMN cells. In NOP-GFP-tagged receptors, N/OFQATTO594 was used in a FRET protocol where GFP emission activated ATTO, visualizing ligand-receptor interaction. When the NOPGFP receptor is activated by N/OFQATTO594 , movement of ligand and receptor from the cell surface to the cytosol can be measured. CONCLUSIONS AND IMPLICATIONS In the absence of validated NOP receptor antibodies and issues surrounding the use of radiolabels (especially in low expression systems), these data indicate the utility of N/OFQATTO594 to study a wide range of N/OFQ-driven cellular responses.
Collapse
Affiliation(s)
- M F Bird
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain Management, Leicester Royal InfirmaryUniversity of LeicesterLeicesterUK
| | - R Guerrini
- Department of Chemical and Pharmaceutical Sciences and LTTAUniversity of FerraraFerraraItaly
| | - J M Willets
- Department of Molecular and Cell BiologyUniversity of LeicesterLeicesterUK
| | - J P Thompson
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain Management, Leicester Royal InfirmaryUniversity of LeicesterLeicesterUK
| | - G Caló
- Department of Medical Sciences, Section of Pharmacology and National Institute of NeuroscienceUniversity of FerraraFerraraItaly
| | - D G Lambert
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain Management, Leicester Royal InfirmaryUniversity of LeicesterLeicesterUK
| |
Collapse
|