1
|
Gaborit M, Massotte D. Therapeutic potential of opioid receptor heteromers in chronic pain and associated comorbidities. Br J Pharmacol 2023; 180:994-1013. [PMID: 34883528 DOI: 10.1111/bph.15772] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/07/2021] [Accepted: 07/21/2021] [Indexed: 11/27/2022] Open
Abstract
Chronic pain affects 20% to 45% of the global population and is often associated with the development of anxio-depressive disorders. Treatment of this debilitating condition remains particularly challenging with opioids prescribed to alleviate moderate to severe pain. However, despite strong antinociceptive properties, numerous adverse effects limit opioid use in the clinic. Moreover, opioid misuse and abuse have become a major health concern worldwide. This prompted efforts to design original strategies that would efficiently and safely relieve pain. Targeting of opioid receptor heteromers is one of these. This review summarizes our current knowledge on the role of heteromers involving opioid receptors in the context of chronic pain and anxio-depressive comorbidities. It also examines how heteromerization in native tissue affects ligand binding, receptor signalling and trafficking properties. Finally, the therapeutic potential of ligands designed to specifically target opioid receptor heteromers is considered. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.
Collapse
Affiliation(s)
- Marion Gaborit
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Dominique Massotte
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| |
Collapse
|
2
|
Murayama MA, Shimizu J, Miyabe C, Yudo K, Miyabe Y. Chemokines and chemokine receptors as promising targets in rheumatoid arthritis. Front Immunol 2023; 14:1100869. [PMID: 36860872 PMCID: PMC9968812 DOI: 10.3389/fimmu.2023.1100869] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that commonly causes inflammation and bone destruction in multiple joints. Inflammatory cytokines, such as IL-6 and TNF-α, play important roles in RA development and pathogenesis. Biological therapies targeting these cytokines have revolutionized RA therapy. However, approximately 50% of the patients are non-responders to these therapies. Therefore, there is an ongoing need to identify new therapeutic targets and therapies for patients with RA. In this review, we focus on the pathogenic roles of chemokines and their G-protein-coupled receptors (GPCRs) in RA. Inflamed tissues in RA, such as the synovium, highly express various chemokines to promote leukocyte migration, tightly controlled by chemokine ligand-receptor interactions. Because the inhibition of these signaling pathways results in inflammatory response regulation, chemokines and their receptors could be promising targets for RA therapy. The blockade of various chemokines and/or their receptors has yielded prospective results in preclinical trials using animal models of inflammatory arthritis. However, some of these strategies have failed in clinical trials. Nonetheless, some blockades showed promising results in early-phase clinical trials, suggesting that chemokine ligand-receptor interactions remain a promising therapeutic target for RA and other autoimmune diseases.
Collapse
Affiliation(s)
- Masanori A Murayama
- Department of Animal Models for Human Diseases, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan
| | - Jun Shimizu
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Chie Miyabe
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kazuo Yudo
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Yoshishige Miyabe
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| |
Collapse
|
3
|
Jhumka ZA, Abdus-Saboor IJ. Next generation behavioral sequencing for advancing pain quantification. Curr Opin Neurobiol 2022; 76:102598. [PMID: 35780688 DOI: 10.1016/j.conb.2022.102598] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 05/17/2022] [Accepted: 05/24/2022] [Indexed: 11/28/2022]
Abstract
With symptoms such as spontaneous pain and pathologically heightened sensitivity to stimuli, chronic pain accounts for about 20% of physician visits and up to 2/3 of patients are dissatisfied with current treatments. Much of our knowledge on pain processing and analgesics has emerged from behavioral studies performed on animals presenting the same symptoms under pathological conditions. While humans can verbally describe their pain, studies on rodents have relied on behavioral assays providing non-exhaustive characterization or altering animals' original sensitivity through repetitive stimulations. The emergence of what we term "next-generation behavioral sequencing" is now permitting us to quantitatively describe behavioral features on millisecond to minutes long timescales that lie beyond easy detection with the unaided eye. Here, we summarize emerging videography and computational based behavioral approaches that have the potential to significantly improve pain research.
Collapse
Affiliation(s)
- Z Anissa Jhumka
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA. https://twitter.com/AnissaJhumka
| | - Ishmail J Abdus-Saboor
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA. ia2458columbia.edu
| |
Collapse
|
4
|
Domínguez-Oliva A, Mota-Rojas D, Hernández-Avalos I, Mora-Medina P, Olmos-Hernández A, Verduzco-Mendoza A, Casas-Alvarado A, Whittaker AL. The neurobiology of pain and facial movements in rodents: Clinical applications and current research. Front Vet Sci 2022; 9:1016720. [PMID: 36246319 PMCID: PMC9556725 DOI: 10.3389/fvets.2022.1016720] [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: 08/11/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
One of the most controversial aspects of the use of animals in science is the production of pain. Pain is a central ethical concern. The activation of neural pathways involved in the pain response has physiological, endocrine, and behavioral consequences, that can affect both the health and welfare of the animals, as well as the validity of research. The strategy to prevent these consequences requires understanding of the nociception process, pain itself, and how assessment can be performed using validated, non-invasive methods. The study of facial expressions related to pain has undergone considerable study with the finding that certain movements of the facial muscles (called facial action units) are associated with the presence and intensity of pain. This review, focused on rodents, discusses the neurobiology of facial expressions, clinical applications, and current research designed to better understand pain and the nociceptive pathway as a strategy for implementing refinement in biomedical research.
Collapse
Affiliation(s)
- Adriana Domínguez-Oliva
- Master in Science Program “Maestría en Ciencias Agropecuarias”, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Daniel Mota-Rojas
- Neurophysiology, Behavior and Animal Welfare Assesment, DPAA, Universidad Autónoma Metropolitana, Mexico City, Mexico
- *Correspondence: Daniel Mota-Rojas
| | - Ismael Hernández-Avalos
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli, Mexico
| | - Patricia Mora-Medina
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli, Mexico
| | - Adriana Olmos-Hernández
- Division of Biotechnology-Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Antonio Verduzco-Mendoza
- Division of Biotechnology-Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Alejandro Casas-Alvarado
- Neurophysiology, Behavior and Animal Welfare Assesment, DPAA, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Alexandra L. Whittaker
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| |
Collapse
|
5
|
Ligand-Free Signaling of G-Protein-Coupled Receptors: Relevance to μ Opioid Receptors in Analgesia and Addiction. Molecules 2022; 27:molecules27185826. [PMID: 36144565 PMCID: PMC9503102 DOI: 10.3390/molecules27185826] [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: 07/26/2022] [Revised: 09/04/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Numerous G-protein-coupled receptors (GPCRs) display ligand-free basal signaling with potential physiological functions, a target in drug development. As an example, the μ opioid receptor (MOR) signals in ligand-free form (MOR-μ*), influencing opioid responses. In addition, agonists bind to MOR but can dissociate upon MOR activation, with ligand-free MOR-μ* carrying out signaling. Opioid pain therapy is effective but incurs adverse effects (ADRs) and risk of opioid use disorder (OUD). Sustained opioid agonist exposure increases persistent basal MOR-μ* activity, which could be a driving force for OUD and ADRs. Antagonists competitively prevent resting MOR (MOR-μ) activation to MOR-μ*, while common antagonists, such as naloxone and naltrexone, also bind to and block ligand-free MOR-μ*, acting as potent inverse agonists. A neutral antagonist, 6β-naltrexol (6BN), binds to but does not block MOR-μ*, preventing MOR-μ activation only competitively with reduced potency. We hypothesize that 6BN gradually accelerates MOR-μ* reversal to resting-state MOR-μ. Thus, 6BN potently prevents opioid dependence in rodents, at doses well below those blocking antinociception or causing withdrawal. Acting as a ‘retrograde addiction modulator’, 6BN could represent a novel class of therapeutics for OUD. Further studies need to address regulation of MOR-μ* and, more broadly, the physiological and pharmacological significance of ligand-free signaling in GPCRs.
Collapse
|
6
|
Chen BC, He HY, Niu K, Rui K, Huang JG, Xie YQ, Xiao M. Network pharmacology-based approach uncovers the JAK/STAT signaling mechanism underlying paederia scandens extract treatment of rheumatoid arthritis. Am J Transl Res 2022; 14:5295-5307. [PMID: 36105044 PMCID: PMC9452312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/19/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a common autoimmune disease. Paederia scandens (Lour.) Merr is a common folk remedy used in Hainan, China, to dispel the wind and dampness associated with RA. METHODS The active components of Paederia scandens were extracted using network pharmacology. The potential targets of active components were used to determine activated pathways, and the in vitro effects of Paederia scandens extracts were verified in RA fibroblast-like synoviocytes (HFLS-RA). RESULTS We identified 27 active components using ultra-high-performance liquid chromatography (UHPLC)-quadrupole time-of-flight (QTOF)-mass spectrometry (MS). Among the major target genes with high connectivity, IL-1β, PI3K, TNF, and JAK2 are known to play key roles in RA development. High-affinity interactions were identified between active compounds in Paederia scandens extract and Janus kinase JAK 2, which are key components of the JAK-signal transducer and activator of transcription (STAT) signaling pathway. In HFLS-RA cells, Paederia scandens extract treatment reduced the mRNA levels of IL-6, IL-1β, and IL-17. Paederia scandens extract treatment also significantly inhibited the phosphorylation of JAK 2 and STAT3, regulating cell proliferation. CONCLUSIONS Based on these results, we confirmed that Paederia scandens has potential for application as a therapeutic and preventive food and acts through the modulation and suppression of JAK-STAT pathway activation to control the inflammatory response in RA.
Collapse
Affiliation(s)
- Bo-Cen Chen
- Key Laboratory of Biochemistry and Molecular Biology, Hainan Medical UniversityHaikou 571000, Hainan, China
| | - He-Yu He
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical UniversityHaikou 571000, Hainan, China
- Osteoarthrosurgery Second Affiliated HospitalHaikou 571000, Hainan, China
| | - Kun Niu
- College of Chinese Traditional Medicine, Hainan Medical CollegeHaikou 571199, Hainan, China
| | - Kai Rui
- Key Laboratory of Biochemistry and Molecular Biology, Hainan Medical UniversityHaikou 571000, Hainan, China
| | - Ji-Gen Huang
- Key Laboratory of Biochemistry and Molecular Biology, Hainan Medical UniversityHaikou 571000, Hainan, China
| | - Yi-Qiang Xie
- College of Chinese Traditional Medicine, Hainan Medical CollegeHaikou 571199, Hainan, China
| | - Man Xiao
- Key Laboratory of Biochemistry and Molecular Biology, Hainan Medical UniversityHaikou 571000, Hainan, China
| |
Collapse
|
7
|
Single application of immersion cryotherapy in Wistar rats with experimental gout. J Therm Biol 2022; 107:103253. [DOI: 10.1016/j.jtherbio.2022.103253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/02/2022] [Accepted: 05/05/2022] [Indexed: 11/17/2022]
|
8
|
Chen Y, Xu E, Sang M, Wang Z, Zhang Y, Ye J, Zhou Q, Zhao C, Hu C, Lu W, Cao P. Makatoxin-3, a thermostable Nav1.7 agonist from Buthus martensii Karsch (BmK) scorpion elicits non-narcotic analgesia in inflammatory pain models. JOURNAL OF ETHNOPHARMACOLOGY 2022; 288:114998. [PMID: 35063590 DOI: 10.1016/j.jep.2022.114998] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/31/2021] [Accepted: 01/11/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chronic pain management represents a serious healthcare problem worldwide. The use of opioid analgesics for pain has always been hampered by their side effects; in particular, the addictive liability associated with chronic use. Finding a morphine replacement has been a long-standing goal in the field of analgesia. In traditional Chinese medicine, processed Buthus martensii Karsch (BmK) scorpion has been used as a painkiller to treat chronic inflammatory arthritis and spondylitis, so called "Scorpio-analgesia". However, the molecular basis and the underline mechanism for the Scorpio-analgesia are still unclear. AIM OF THE STUDY The study aims to investigate the molecular basis of "Scorpio analgesia" and identify novel analgesics from BmK scorpion. MATERIALS AND METHODS In this study, the analgesic abilities were determined using formalin-, acetic acid- and complete Freund's adjuvant-induced pain models. The effect of BmK venom and processed BmK venom on Nav1.7 were detected by whole-cell voltage-clamp recordings on HEK293-hNav1.7 stable cell line. Action potentials in Dorsal root ganglion (DRG) neurons induced by Makatoxin-3-R58A were recorded in current-clamp mode. The content of Makatoxin-3 was detected using competitive enzyme-linked immunosorbent assay based on the Makatoxin-3 antibody. High performance liquid chromatography, western blot and circular dichroism spectroscopy were used to analysis the stability of Makatoxin-3. RESULTS Here we demonstrate that Makatoxin-3, an α-like toxin in BmK scorpion venom targeting Nav1.7 is the critical component in Scorpio-analgesia. The analgesic effect of Makatoxin-3 could not be reversed by naloxone and is more potent than Nav1.7-selective inhibitors and non-steroidal anti-inflammatory drugs in inflammatory models. Moreover, a R58A mutant of Makatoxin-3 is capable of eliciting analgesia effect without inducing pain response. CONCLUSIONS This study advances ion channel biology and proposes Nav1.7 agonists, rather than the presumed Nav1.7-only blockers, for non-narcotic relief of chronic pain.
Collapse
Affiliation(s)
- Yonggen Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Erjin Xu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Ming Sang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Zhiheng Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Yuxin Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Juan Ye
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Qian Zhou
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Chenglei Zhao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Chunping Hu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Wuguang Lu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China.
| | - Peng Cao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China.
| |
Collapse
|
9
|
Vincenzi M, Milella MS, D’Ottavio G, Caprioli D, Reverte I, Maftei D. Targeting Chemokines and Chemokine GPCRs to Enhance Strong Opioid Efficacy in Neuropathic Pain. Life (Basel) 2022; 12:life12030398. [PMID: 35330149 PMCID: PMC8955776 DOI: 10.3390/life12030398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/06/2022] [Accepted: 03/07/2022] [Indexed: 12/21/2022] Open
Abstract
Neuropathic pain (NP) originates from an injury or disease of the somatosensory nervous system. This heterogeneous origin and the possible association with other pathologies make the management of NP a real challenge. To date, there are no satisfactory treatments for this type of chronic pain. Even strong opioids, the gold-standard analgesics for nociceptive and cancer pain, display low efficacy and the paradoxical ability to exacerbate pain sensitivity in NP patients. Mounting evidence suggests that chemokine upregulation may be a common mechanism driving NP pathophysiology and chronic opioid use-related consequences (analgesic tolerance and hyperalgesia). Here, we first review preclinical studies on the role of chemokines and chemokine receptors in the development and maintenance of NP. Second, we examine the change in chemokine expression following chronic opioid use and the crosstalk between chemokine and opioid receptors. Then, we examine the effects of inhibiting specific chemokines or chemokine receptors as a strategy to increase opioid efficacy in NP. We conclude that strong opioids, along with drugs that block specific chemokine/chemokine receptor axis, might be the right compromise for a favorable risk/benefit ratio in NP management.
Collapse
Affiliation(s)
- Martina Vincenzi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy;
- Correspondence: (M.V.); (I.R.)
| | - Michele Stanislaw Milella
- Toxicology and Poison Control Center Unit, Department of Emergency, Anesthesia and Critical Care, Policlinico Umberto I Hospital-Sapienza University of Rome, 00161 Rome, Italy;
| | - Ginevra D’Ottavio
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00143 Rome, Italy; (G.D.); (D.C.)
- Laboratory Affiliated to Institute Pasteur Italia-Fondazione Cenci Bolognetti, Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy
| | - Daniele Caprioli
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00143 Rome, Italy; (G.D.); (D.C.)
- Laboratory Affiliated to Institute Pasteur Italia-Fondazione Cenci Bolognetti, Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy
| | - Ingrid Reverte
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy;
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00143 Rome, Italy; (G.D.); (D.C.)
- Correspondence: (M.V.); (I.R.)
| | - Daniela Maftei
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy;
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00143 Rome, Italy; (G.D.); (D.C.)
| |
Collapse
|
10
|
Iriah SC, Borges C, Shalev U, Cai X, Madularu D, Kulkarni PP, Ferris CF. The utility of maraviroc, an antiretroviral agent used to treat HIV, as treatment for opioid abuse? Data from MRI and behavioural testing in rats. J Psychiatry Neurosci 2021; 46:E548-E558. [PMID: 34625487 PMCID: PMC8526136 DOI: 10.1503/jpn.200191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 03/04/2021] [Accepted: 07/02/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Maraviroc is an antiretroviral agent and C-C chemokine coreceptor 5 (CCR5) antagonist that is currently used to treat human immunodeficiency virus. CCR5/μ-opioid receptor heterodimerization suggests that maraviroc could be a treatment for oxycodone abuse. We treated rats with maraviroc to explore its effect on oxycodone-seeking and its interference with the analgesic effects of oxycodone. We used resting-state blood-oxygen-level-dependent functional connectivity to assess the effect of maraviroc on oxycodone-enhanced coupling in the reward circuitry and performed behavioural tests to evaluate the effect of maraviroc on oxycodone rewarding properties and on oxycodone-seeking after prolonged abstinence. METHODS Two groups of rats were exposed to 8 consecutive days of oxycodone-conditioned place preference training and treatment with maraviroc or vehicle. Two additional groups were trained to self-administer oxycodone for 10 days and then tested for drug seeking after 14 days of abstinence with or without daily maraviroc treatment. We tested the effects of maraviroc on oxycodone analgesia using a tail-flick assay. We analyzed resting-state functional connectivity data using a rat 3-dimensional MRI atlas of 171 brain areas. RESULTS Maraviroc significantly decreased conditioned place preference and attenuated oxycodone-seeking behaviour after prolonged abstinence. The analgesic effect of oxycodone was maintained after maraviroc treatment. Oxycodone increased functional coupling with the accumbens, ventral pallidum and olfactory tubercles, but this was reduced with maraviroc treatment. LIMITATIONS All experiments were performed in male rats only. CONCLUSION Maraviroc treatment attenuated oxycodone-seeking in abstinent rats and reduced functional coupling in the reward circuitry. The analgesic effects of oxycodone were not affected by maraviroc.
Collapse
Affiliation(s)
- Sade C Iriah
- From the Centre for Translational Neuroimaging, Northeastern University, Boson, Mass., USA (Iriah, Cai, Madularu, Kulkarni, Ferris); and Concordia University, Montreal, Que., Canada (Borges, Shalev).
| | - Catarina Borges
- From the Centre for Translational Neuroimaging, Northeastern University, Boson, Mass., USA (Iriah, Cai, Madularu, Kulkarni, Ferris); and Concordia University, Montreal, Que., Canada (Borges, Shalev)
| | - Uri Shalev
- From the Centre for Translational Neuroimaging, Northeastern University, Boson, Mass., USA (Iriah, Cai, Madularu, Kulkarni, Ferris); and Concordia University, Montreal, Que., Canada (Borges, Shalev)
| | - Xuezhu Cai
- From the Centre for Translational Neuroimaging, Northeastern University, Boson, Mass., USA (Iriah, Cai, Madularu, Kulkarni, Ferris); and Concordia University, Montreal, Que., Canada (Borges, Shalev)
| | - Dan Madularu
- From the Centre for Translational Neuroimaging, Northeastern University, Boson, Mass., USA (Iriah, Cai, Madularu, Kulkarni, Ferris); and Concordia University, Montreal, Que., Canada (Borges, Shalev)
| | - Praveen P Kulkarni
- From the Centre for Translational Neuroimaging, Northeastern University, Boson, Mass., USA (Iriah, Cai, Madularu, Kulkarni, Ferris); and Concordia University, Montreal, Que., Canada (Borges, Shalev)
| | - Craig F Ferris
- From the Centre for Translational Neuroimaging, Northeastern University, Boson, Mass., USA (Iriah, Cai, Madularu, Kulkarni, Ferris); and Concordia University, Montreal, Que., Canada (Borges, Shalev)
| |
Collapse
|
11
|
Pelechas E, Voulgari PV, Drosos AA. Recent advances in the opioid mu receptor based pharmacotherapy for rheumatoid arthritis. Expert Opin Pharmacother 2020; 21:2153-2160. [PMID: 33135514 DOI: 10.1080/14656566.2020.1796969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Opioids are used for severe forms of acute and cancer pain. Over the last years, their potential use in patients with noncancer pain such as those with rheumatoid arthritis (RA) has been postulated. A recent population-based comparative study showed that chronic opioid use was 12% vs. 4% among RA and non-RA patients, respectively. Another study showed an increase from 7.4% to 16.9% (2002 to 2015). In general, there has been an increasing tendency to use opioids in recent years. AREAS COVERED The authors have performed an extensive literature search using PubMed for articles including noncancer pain and the use of the mu opioid receptor (MOR) agonists in patients with RA. EXPERT OPINION Data is not sufficient to support opioid use for the treatment of chronic pain in patients with RA. Data is scarce and inconclusive. Rheumatologists should think and ponder the question: Why is this patient in pain? Differential diagnosis should include a disease flare, degenerative changes of the musculoskeletal system, and fibromyalgia. And while there are new strategies for opioid administration currently being researched, unfortunately, they are far from being applied to human subjects in the everyday clinical setting, and are still being evaluated at an experimental level. CNS: Central nervous system; DORs: delta opioid receptor agonists; GI: Gastrointestinal; GPCRs: G protein-coupled receptors; IL: Interleukin; JAK: Janus kinase; KORs: kappa opioid receptor agonists; MCPs: Metacarpophalangeal joints; MORs: Mu opioid receptor agonists; MTPs: Metatarsophalangeal joints; NSAIDs: Non-steroidal anti-inflammatory drugsOA: Osteoarthritis; ORs: Opioid receptors; PD: Pharmacodynamic; PIPs: Proximal interphalangeal joints; PK: Pharmacokinetic; PNS: Peripheral nervous system; RA: Rheumatoid arthritis; RGS: Regulator of G protein signaling; SSRIs: Selective serotonin reuptake inhibitors; TNF: Tumor necrosis factor.
Collapse
Affiliation(s)
- Eleftherios Pelechas
- Rheumatology Clinic, Department of Internal Medicine, Medical School, University of Ioannina , Ioannina, Greece
| | - Paraskevi V Voulgari
- Rheumatology Clinic, Department of Internal Medicine, Medical School, University of Ioannina , Ioannina, Greece
| | - Alexandros A Drosos
- Rheumatology Clinic, Department of Internal Medicine, Medical School, University of Ioannina , Ioannina, Greece
| |
Collapse
|
12
|
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
|
13
|
Qi B, Fang Q, Liu S, Hou W, Li J, Huang Y, Shi J. Advances of CCR5 antagonists: From small molecules to macromolecules. Eur J Med Chem 2020; 208:112819. [PMID: 32947226 DOI: 10.1016/j.ejmech.2020.112819] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/10/2020] [Accepted: 09/03/2020] [Indexed: 02/08/2023]
Abstract
C-C chemokine receptor 5(CCR5) is a cell membrane protein from G protein-coupled receptors (GPCR) family, which is an important modulator for leukocyte activation and mobilization. In the 1980s, several reports suggest that lack of the HIV-1 co-receptor, the chemokine receptor CCR5, offers protection against HIV infection. Later, it was shown that CCR5 was confirmed to be the most common co-receptor for the HIV-1 virus R5 strain. In recent years, many studies have shown that CCR5 is closely related to the development of various cancers and inflammations to facilitate the discovery of CCR5 antagonists. There are many types of CCR5 antagonists, mainly including chemokine derivatives, non-peptide small molecule compounds, monoclonal antibodies, and peptide compounds. This review focus on the recent research processes and pharmacological effects of CCR5 antagonists such as Maraviroc, TAK-779 and PRO 140. After focusing on the therapeutic effect of CCR5 antagonists on AIDS, it also discusses the therapeutic prospect of CCR5 in other diseases such as inflammation and tumor.
Collapse
Affiliation(s)
- Baowen Qi
- Chengdu Kanghua Biological Products Co., Ltd, Chengdu, China; College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Qiang Fang
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Shiyuan Liu
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Wenli Hou
- Chengdu Kanghua Biological Products Co., Ltd, Chengdu, China
| | - Jian Li
- Department of Pharmacy, West China Hospital Sichuan University, Chengdu, 610041, China.
| | - Yingchun Huang
- Beijing Key Laboratory of Biomass Waste Resource Utilization, College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China.
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
14
|
Shchepinova MM, Hanyaloglu AC, Frost GS, Tate EW. Chemical biology of noncanonical G protein-coupled receptor signaling: Toward advanced therapeutics. Curr Opin Chem Biol 2020; 56:98-110. [PMID: 32446179 DOI: 10.1016/j.cbpa.2020.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 12/20/2022]
Abstract
G protein-coupled receptors (GPCRs), the largest family of signaling membrane proteins, are the target of more than 30% of the drugs on the market. Recently, it has become clear that GPCR functions are far more multidimensional than previously thought, with multiple noncanonical aspects coming to light, including biased, oligomeric, and compartmentalized signaling. These additional layers of functional selectivity greatly expand opportunities for advanced therapeutic interventions, but the development of new chemical biology tools is absolutely required to improve our understanding of noncanonical GPCR regulation and pave the way for future drugs. In this opinion, we highlight the most notable examples of chemical and chemogenetic tools addressing new paradigms in GPCR signaling, discuss their promises and limitations, and explore future directions.
Collapse
Affiliation(s)
- Maria M Shchepinova
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, 80 Wood Lane, London, W12 0BZ, UK.
| | - Aylin C Hanyaloglu
- Institute of Reproductive and Developmental Biology, Dept. Surgery and Cancer, Imperial College, London, UK
| | - Gary S Frost
- Department of Medicine, Faculty of Medicine, Nutrition and Dietetic Research Group, Imperial College, London, UK
| | - Edward W Tate
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, 80 Wood Lane, London, W12 0BZ, UK.
| |
Collapse
|
15
|
Reiss D, Maduna T, Maurin H, Audouard E, Gaveriaux-Ruff C. Mu opioid receptor in microglia contributes to morphine analgesic tolerance, hyperalgesia, and withdrawal in mice. J Neurosci Res 2020; 100:203-219. [PMID: 32253777 DOI: 10.1002/jnr.24626] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/23/2020] [Accepted: 03/18/2020] [Indexed: 12/17/2022]
Abstract
A major challenge in medicine is developing potent pain therapies without the adverse effects of opiates. Neuroinflammation and in particular microglial activation have been shown to contribute to these effects. However, the implication of the microglial mu opioid receptor (MOR) is not known. We developed a novel conditional knockout (cKO) mouse line, wherein MOR is deleted in microglia. Morphine analgesic tolerance was delayed in both sexes in cKO mice in the hot plate assay. Opioid-induced hyperalgesia (OIH) as measured in the tail immersion assay was abolished in male cKO mice, and physical dependence to morphine as assessed by naloxone-induced withdrawal was attenuated in female cKO mice. Our results show a sex-dependent contribution of microglial MOR in morphine analgesic tolerance, OIH, and physical dependence. In conclusion, our data suggest that blockade of microglial MOR could represent a therapeutic target for opiate analgesia without the opiate adverse effects.
Collapse
Affiliation(s)
- David Reiss
- Translational Medicine and Neurogenetics Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,IGBMC, Université de Strasbourg, Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique, Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Tando Maduna
- Translational Medicine and Neurogenetics Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,IGBMC, Université de Strasbourg, Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique, Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale, Illkirch, France.,Neurology Research Group, Department of Physiology, Stellenbosch University, Stellenbosch, South Africa
| | - Hervé Maurin
- Translational Medicine and Neurogenetics Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,IGBMC, Université de Strasbourg, Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique, Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Emilie Audouard
- Translational Medicine and Neurogenetics Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,IGBMC, Université de Strasbourg, Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique, Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Claire Gaveriaux-Ruff
- Translational Medicine and Neurogenetics Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,IGBMC, Université de Strasbourg, Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique, Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale, Illkirch, France.,Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| |
Collapse
|
16
|
González-Cano R, Montilla-García Á, Ruiz-Cantero MC, Bravo-Caparrós I, Tejada MÁ, Nieto FR, Cobos EJ. The search for translational pain outcomes to refine analgesic development: Where did we come from and where are we going? Neurosci Biobehav Rev 2020; 113:238-261. [PMID: 32147529 DOI: 10.1016/j.neubiorev.2020.03.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/06/2020] [Accepted: 03/04/2020] [Indexed: 12/11/2022]
Abstract
Pain measures traditionally used in rodents record mere reflexes evoked by sensory stimuli; the results thus may not fully reflect the human pain phenotype. Alterations in physical and emotional functioning, pain-depressed behaviors and facial pain expressions were recently proposed as additional pain outcomes to provide a more accurate measure of clinical pain in rodents, and hence to potentially enhance analgesic drug development. We aimed to review how preclinical pain assessment has evolved since the development of the tail flick test in 1941, with a particular focus on a critical analysis of some nonstandard pain outcomes, and a consideration of how sex differences may affect the performance of these pain surrogates. We tracked original research articles in Medline for the following periods: 1973-1977, 1983-1987, 1993-1997, 2003-2007, and 2014-2018. We identified 606 research articles about alternative surrogate pain measures, 473 of which were published between 2014 and 2018. This indicates that preclinical pain assessment is moving toward the use of these measures, which may soon become standard procedures in preclinical pain laboratories.
Collapse
Affiliation(s)
- Rafael González-Cano
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, Granada, Spain.
| | - Ángeles Montilla-García
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, Granada, Spain.
| | - M Carmen Ruiz-Cantero
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, Granada, Spain.
| | - Inmaculada Bravo-Caparrós
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, Granada, Spain.
| | - Miguel Á Tejada
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, Granada, Spain; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria.
| | - Francisco R Nieto
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, Granada, Spain.
| | - Enrique J Cobos
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; Teófilo Hernando Institute for Drug Discovery, Madrid, Spain.
| |
Collapse
|
17
|
Abstract
Research on the effects of opioids on immune responses was stimulated in the 1980s by the intersection of use of intravenous heroin and HIV infection, to determine if opioids were enhancing HIV progression. The majority of experiments administering opioid alkaloids (morphine and heroin) in vivo, or adding these drugs to cell cultures in vitro, showed that they were immunosuppressive. Immunosuppression was reported as down-regulation: of Natural Killer cell activity; of responses of T and B cells to mitogens; of antibody formation in vivo and in vitro; of depression of phagocytic and microbicidal activity of neutrophils and macrophages; of cytokine and chemokine production by macrophages, microglia, and astrocytes; by sensitization to various infections using animal models; and by enhanced replication of HIV in vitro. The specificity of the receptor involved in the immunosuppression was shown to be the mu opioid receptor (MOR) by using pharmacological antagonists and mice genetically deficient in MOR. Beginning with a paper published in 2005, evidence was presented that morphine is immune-stimulating via binding to MD2, a molecule associated with Toll-like Receptor 4 (TLR4), the receptor for bacterial lipopolysaccharide (LPS). This concept was pursued to implicate inflammation as a mechanism for the psychoactive effects of the opioid. This review considers the validity of this hypothesis and concludes that it is hard to sustain. The experiments demonstrating immunosuppression were carried out in vivo in rodent strains with normal levels of TLR4, or involved use of cells taken from animals that were wild-type for expression of TLR4. Since engagement of TLR4 is universally accepted to result in immune activation by up-regulation of NF-κB, if morphine were binding to TLR4, it would be predicted that opioids would have been found to be pro-inflammatory, which they were not. Further, morphine is immunosuppressive in mice with a defective TLR4 receptor. Morphine and morphine withdrawal have been shown to permit leakage of Gram-negative bacteria and LPS from the intestinal lumen. LPS is the major ligand for TLR4. It is proposed that an occult variable in experiments where morphine is being proposed to activate TLR4 is actually underlying sepsis induced by the opioid.
Collapse
Affiliation(s)
- Toby K. Eisenstein
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
18
|
Akgün E, Lunzer MM, Portoghese PS. Combined Glia Inhibition and Opioid Receptor Agonism Afford Highly Potent Analgesics without Tolerance. ACS Chem Neurosci 2019; 10:2004-2011. [PMID: 30110531 DOI: 10.1021/acschemneuro.8b00323] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Commonly prescribed opioid analgesics produce tolerance upon chronic use due in part to induction of hyperalgesia. Given that two reported bivalent ligands (MMG22 and MCC22) produce potent antinociception without tolerance only in inflamed mice, we have investigated the possible cellular and receptor targets of these ligands. The selective microglia inhibitors, minocycline and SB290157, antagonized intrathecal (i.t.) MCC22 antinociception orders of magnitude more potently than MMG22, suggesting that MCC22 selectively targets activated microglia. The astrocyte toxin, l-α-aminoadipic acid antagonized MMG22 antinociception 126-fold without reducing the potency of MCC22, indicating that activated astrocytes are targets of MMG22. MK-801 and Ro25-6981 antagonism of MMG22 antinociception, but not MCC22, is consistent with selective inhibition of activated NMDAR in astrocytes. The antinociception produced by i.t. MMG22 or MCC22 were both antagonized by the selective mu opioid receptor antagonist, β-FNA, implicating interaction of these ligands with MOR in spinal afferent neurons. MCC22 antinociception was potently blocked by kainate or AMPA ion channel antagonists (LY382884; NBQX), in contrast to MMG22. It is concluded that i.t. MMG22 and MCC22 produce exceptional antinociception via potent inhibition of activated spinal glia, thereby leading to desensitization of spinal neurons and enhanced activation of neuronal MOR. Thus, the present study suggests a new approach to treatment of chronic inflammatory pain without tolerance through a single molecular entity that simultaneously inhibits activated glia and stimulates MOR in spinal neurons.
Collapse
Affiliation(s)
- Eyup Akgün
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mary M. Lunzer
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Philip S. Portoghese
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
19
|
Cataldo G, Erb SJ, Lunzer MM, Luong N, Akgün E, Portoghese PS, Olson JK, Simone DA. The bivalent ligand MCC22 potently attenuates hyperalgesia in a mouse model of cisplatin-evoked neuropathic pain without tolerance or reward. Neuropharmacology 2019; 158:107598. [PMID: 30970233 DOI: 10.1016/j.neuropharm.2019.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/26/2019] [Accepted: 04/03/2019] [Indexed: 10/27/2022]
Abstract
Cisplatin and other widely employed platinum-based anticancer agents produce chemotherapy-induced peripheral neuropathy (CIPN) that often results in pain and hyperalgesia that are difficult to manage. We investigated the efficacy of a novel bivalent ligand, MCC22, for the treatment of pain arising from CIPN. MCC22 consists of mu opioid receptor (MOR) agonist and chemokine receptor 5 (CCR5) antagonist pharmacophores connected through a 22-atom spacer and was designed to target a putative MOR-CCR5 heteromer localized in pain processing areas. Mice received once daily intraperitoneal (i.p.) injections of cisplatin (1 mg/kg) for seven days and behavior testing began 7 days later. Cisplatin produced mechanical hyperalgesia that was decreased dose-dependently by MCC22 given by intrathecal (ED50 = 0.004 pmol) or i.p. (3.07 mg/kg) routes. The decrease in hyperalgesia was associated with decreased inflammatory response by microglia in the spinal cord. Unlike morphine, MCC22 given daily for nine days did not exhibit tolerance to its analgesic effect and its characteristic antihyperalgesic activity was fully retained in morphine-tolerant mice. Furthermore, MCC22 did not alter motor function and did not exhibit rewarding properties. Given the exceptional potency of MCC22 without tolerance or reward, MCC22 has the potential to vastly improve management of chronic pain due to CIPN. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.
Collapse
Affiliation(s)
- Giuseppe Cataldo
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Samuel J Erb
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Mary M Lunzer
- Department of Medicinal Chemistry, College of Pharmacy University of Minnesota, Minneapolis, MN, USA
| | - Nhungoc Luong
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Eyup Akgün
- Department of Medicinal Chemistry, College of Pharmacy University of Minnesota, Minneapolis, MN, USA
| | - Philip S Portoghese
- Department of Medicinal Chemistry, College of Pharmacy University of Minnesota, Minneapolis, MN, USA
| | - Julie K Olson
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Donald A Simone
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|