1
|
Hastings LE, Frye EV, Carlson ER, Chuong V, Matthews AN, Koob GF, Vendruscolo LF, Marchette RCN. Cold nociception as a measure of hyperalgesia during spontaneous heroin withdrawal in mice. Pharmacol Biochem Behav 2024; 235:173694. [PMID: 38128767 PMCID: PMC10842911 DOI: 10.1016/j.pbb.2023.173694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Opioids are powerful analgesic drugs that are used clinically to treat pain. However, chronic opioid use causes compensatory neuroadaptations that result in greater pain sensitivity during withdrawal, known as opioid withdrawal-induced hyperalgesia (OWIH). Cold nociception tests are commonly used in humans, but preclinical studies often use mechanical and heat stimuli to measure OWIH. Thus, further characterization of cold nociception stimuli is needed in preclinical models. We assessed three cold nociception tests-thermal gradient ring (5-30 °C, 5-50 °C, 15-40 °C, and 25-50 °C), dynamic cold plate (4 °C to -1 °C at -1 °C/min, -1 °C to 4 °C at +1 °C/min), and stable cold plate (10 °C, 6 °C, and 2 °C)-to measure hyperalgesia in a mouse protocol of heroin dependence. On the thermal gradient ring, mice in the heroin withdrawal group preferred warmer temperatures, and the results depended on the ring's temperature range. On the dynamic cold plate, heroin withdrawal increased the number of nociceptive responses, with a temperature ramp from 4 °C to -1 °C yielding the largest response. On the stable cold plate, heroin withdrawal increased the number of nociceptive responses, and a plate temperature of 2 °C yielded the most significant increase in responses. Among the three tests, the stable cold plate elicited the most robust change in behavior between heroin-dependent and nondependent mice and had the highest throughput. To pharmacologically characterize the stable cold plate test, we used μ-opioid and non-opioid receptor-targeting drugs that have been previously shown to reverse OWIH in mechanical and heat nociception assays. The full μ-opioid receptor agonist methadone and μ-opioid receptor partial agonist buprenorphine decreased OWIH, whereas the preferential μ-opioid receptor antagonist naltrexone increased OWIH. Two N-methyl-d-aspartate receptor antagonists (ketamine, MK-801), a corticotropin-releasing factor 1 receptor antagonist (R121919), a β2-adrenergic receptor antagonist (butoxamine), an α2-adrenergic receptor agonist (lofexidine), and a 5-hydroxytryptamine-3 receptor antagonist (ondansetron) had no effect on OWIH. These data demonstrate that the stable cold plate at 2 °C yields a robust, reliable, and concise measure of OWIH that is sensitive to opioid agonists.
Collapse
Affiliation(s)
- Lyndsay E Hastings
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Emma V Frye
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Erika R Carlson
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Vicky Chuong
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA; Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Intitute on Drug Abuse, Intramural Research Program, and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, Baltimore, MD, USA
| | - Aniah N Matthews
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - George F Koob
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Leandro F Vendruscolo
- Stress and Addiction Neuroscience Unit, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, Baltimore, MD, USA
| | - Renata C N Marchette
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA.
| |
Collapse
|
2
|
Zheng W, Huang X, Wang J, Gao F, Chai Z, Zeng J, Li S, Yu C. The chronification mechanism of orofacial inflammatory pain: Facilitation by GPER1 and microglia in the rostral ventral medulla. Front Mol Neurosci 2023; 15:1078309. [PMID: 36683848 PMCID: PMC9853019 DOI: 10.3389/fnmol.2022.1078309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/12/2022] [Indexed: 01/08/2023] Open
Abstract
Background Chronic orofacial pain is a common and incompletely defined clinical condition. The role of G protein-coupled estrogen receptor 1 (GPER1) as a new estrogen receptor in trunk and visceral pain regulation is well known. Here, we researched the role of GPER1 in the rostral ventral medulla (RVM) during chronic orofacial pain. Methods and Results A pain model was established where rats were injected in the temporomandibular joint with complete Freund's adjuvant (CFA) to simulate chronic orofacial pain. Following this a behavioral test was performed to establish pain threshold and results showed that the rats injected with CFA had abnormal pain in the orofacial regions. Additional Immunostaining and blot analysis indicated that microglia were activated in the RVM and GPER1 and c-Fos were significantly upregulated in the rats. Conversely, when the rats were injected with G15 (a GPER1 inhibitor) the abnormal pain the CFA rats were experiencing was alleviated and microglia activation was prevented. In addition, we found that G15 downregulated the expression of phospholipase C (PLC) and protein kinase C (PKC), inhibited the expression of GluA1, restores aberrant synaptic plasticity and reduces the overexpression of the synapse-associated proteins PSD-95 and syb-2 in the RVM of CFA rats. Conclusion The findings indicate that GPER1 mediates chronic orofacial pain through modulation of the PLC-PKC signal pathway, sensitization of the RVM region and enhancement of neural plasticity. These results of this study therefore suggest that GPER1 may serve as a potential therapeutic target for chronic orofacial pain.
Collapse
Affiliation(s)
- Wenwen Zheng
- The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Xilu Huang
- The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Jing Wang
- The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Feng Gao
- The Sixth People’s Hospital of Chongqing, Anesthesiology, Chongqing, China
| | - Zhaowu Chai
- The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Jie Zeng
- The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Sisi Li
- The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Cong Yu
- The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China,*Correspondence: Cong Yu, ✉
| |
Collapse
|
3
|
Wang Z, Yao Y, Tao Y, Fan P, Yu Y, Xie K, Wang G. Spinal microRNA-134-5p targets glutamate receptor ionotropic kainate 3 to modulate opioid induced hyperalgesia in mice. Mol Pain 2023; 19:17448069231178271. [PMID: 37247385 DOI: 10.1177/17448069231178271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023] Open
Abstract
Background: Fentanyl and its analogs are extensively used for pain relief. However, their paradoxically pronociceptive effects often lead to increased opioids consumption and risk of chronic pain. Compared to other synthetic opioids, remifentanil has been strongly linked to acute opioid hyperalgesia after exposure [remifentanil-induced hyperalgesia (RIH)]. The epigenetic regulation of microRNAs (miRNAs) on targeted mRNAs has emerged as an important pathogenesis in pain. The current research aimed at exploring the significance and contributions of miR-134-5p to the development of RIH. Methods: Both the antinociceptive and pronociceptive effects of two commonly used opioids were assessed, and miRNA expression profiles in the spinal dorsal horn (SDH) of mice acutely exposed to remifentanil and remifentanil equianalgesic dose (RED) sufentanil were screened. Next, the candidate miRNA level, cellular distribution, and function were examined by qPCR, fluorescent in situ hybridization (FISH) and Argonaute-2 immunoprecipitation. Furthermore, bioinformatics analysis, luciferase assays, miRNA overexpression, behavioral tests, golgi staining, electron microscopy, whole-cell patch-clamp recording, and immunoblotting were employed to investigate the potential targets and mechanisms underlying RIH. Results: Remifentanil induced significant pronociceptive effects and a distinct miRNA-profile from sufentanil when compared to saline controls. Among top 30 differentially expressed miRNAs spectrum, spinal miR-134-5p was dramatically downregulated in RIH mice but remained comparative in mice subjected to sufentanil. Moreover, Glutamate Receptor Ionotropic Kainate 3 (Grik3) was a target of miR-134-5p. The overexpression of miR-134-5p attenuated the hyperalgesic phenotype, excessive dendritic spine remodeling, excitatory synaptic structural plasticity, and Kainate receptor-mediated miniature excitatory postsynaptic currents (mEPSCs) in SDH resulting from remifentanil exposure. Besides, intrathecal injection of selective KA-R antagonist was able to reverse the GRIK3 membrane trafficking and relieved RIH. Conclusion: The miR-134-5p contributes to remifentanil-induced pronociceptive features via directly targeting Grik3 to modulate dendritic spine morphology and synaptic plasticity in spinal neurons.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Anesthesiology, Tianjin Research Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yao Yao
- Department of Anesthesiology, Tianjin Research Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuzhu Tao
- Department of Anesthesiology, Tianjin Huanhu Hospital, Tianjin, China
| | - Peixin Fan
- Department of Animal and Dairy Science, Mississippi State University, Starkville, MS, USA
- Institute for Genomics, Biocomputing & Biotechnology, Mississippi State University, Starkville, MS, USA
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Research Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Guolin Wang
- Department of Anesthesiology, Tianjin Research Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
4
|
Kong Q, Tian S, Ma C, Wang G, Zhang M. Cannabinoid Receptor Type 2 Agonist Reduces Morphine Tolerance via Mitogen Activated Protein Kinase Phosphatase Induction and Mitogen Activated Protein Kinase Dephosphorylation. Neuroscience 2022; 480:56-64. [PMID: 34774714 DOI: 10.1016/j.neuroscience.2021.11.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/19/2022]
Abstract
Morphine is an opioid drug often used in treating moderate to severe pain. However, morphine tolerance in patients limits its used in clinical settings. Our previous study showed that a cannabinoid type 2 (CB2) receptor agonist attenuated morphine tolerance. However, the exact mechanism by which CB2 agonists reduce morphine tolerance remains unclear. In this study, we investigated the effect of mitogen activated protein kinase (MAPK) and mitogen activated protein kinase phosphatases 1 and 3 (MKP-1 and MKP-3) on the regulation of morphine tolerance by CB2 receptor agonist. Chronic morphine treatments for 7 days reduced the protein expression of MKP-1 and MKP-3 in the spinal cord and increased the phosphorylation of p38, ERK1/2 and the level of proinflammatory mediator, such as IL-1β, IL-6 and TNF-α. Coadministration of CB2 receptor agonist AM1241 alleviated the inhibition of MKP-1 and MKP-3 by chronic morphine administration and reduced the expression of phosphorylated MAPK and proinflammatory factors. The effect of the CB2 receptor agonist on morphine-induced downregulation of MKP-1 and MKP-3 was reversed by the MKP-1 and MKP-3 antagonist triptolide. Our findings suggested that CB2 receptor agonist may induce the expression of MKP-1 and MKP-3 to promote MAPK dephosphorylation and reduce the production of downstream cytokine, thereby reducing morphine tolerance. This finding suggested that MKPs may serve as a new target for alleviating morphine tolerance.
Collapse
Affiliation(s)
- Qingling Kong
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Songyu Tian
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Chao Ma
- Department of Anesthesiology, The Fourth Hospital of Harbin Medical University, Harbin, China.
| | - Guonian Wang
- Department of Anesthesiology, The Fourth Hospital of Harbin Medical University, Harbin, China.
| | - Mingyue Zhang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
5
|
Abbasloo E, Abdollahi F, Saberi A, Esmaeili-Mahani S, Kaeidi A, Akhlaghinasab F, Sheibani V, Thomas TC, Kobeissy FH, Oryan S. Involvement of T-type calcium channels in the mechanism of low dose morphine-induced hyperalgesia in adult male rats. Neuropeptides 2021; 90:102185. [PMID: 34419803 DOI: 10.1016/j.npep.2021.102185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/19/2021] [Accepted: 08/09/2021] [Indexed: 11/23/2022]
Abstract
It has been shown that systemic and local administration of ultra-low dose morphine induced a hyperalgesic response via mu-opioid receptors. However, its exact mechanism(s) has not fully been clarified. It is documented that mu-opioid receptors functionally couple to T-type voltage dependent Ca+2 channels. Here, we investigated the role of T-type calcium channels, amiloride and mibefradil, on the induction of low-dose morphine hyperalgesia in male Wistar rats. The data showed that morphine (0.01 μg i.t. and 1 μg/kg i.p.) could elicit hyperalgesia as assessed by the tail-flick test. Administration of amiloride (5 and 10 μg i.t.) and mibefradil (2.5 and 5 μg i.t.) completely blocked low-dose morphine-induced hyperalgesia in spinal dorsal horn. Amiloride at doses of 1 and 5 mg/kg (i.p.) and mibefradil (9 mg/kg ip) 10 min before morphine (1 μg/kg i.p.) inhibited morphine-induced hyperalgesia. Our results indicate a role for T-type calcium channels in low dose morphine-induced hyperalgesia in rats.
Collapse
Affiliation(s)
- Elham Abbasloo
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| | - Farzaneh Abdollahi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences. Kerman, Iran
| | - Arezoo Saberi
- Neuroscience Research Center, Institute of neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Ayat Kaeidi
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Fereshteh Akhlaghinasab
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences. Kerman, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Institute of neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Theresa Currier Thomas
- College of Medicine-Phoenix, University of Arizona, Child Health, Phoenix, USA; BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix Children's Hospital, Phoenix, USA
| | - Firas Hosni Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Shahrbanoo Oryan
- Departments of Biology, Science and Research Branch, Islamic Azad University. Tehran, Iran
| |
Collapse
|
6
|
Assenov B, Pechlivanova D, Dzhambazova E, Peneva P, Todorov P. Antinociceptive Effects of VV-Hemorphin-5 Peptide Analogues Containing Amino phosphonate Moiety in Mouse Formalin Model of Pain. Protein Pept Lett 2021; 28:442-449. [PMID: 32798365 DOI: 10.2174/0929866527666200813200714] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Hemorphins are endogenous hemoglobin-derived peptides that belong to the family of "atypical" opioid peptides with both affinities to opioid receptors and ability to release other endogenous opioid peptides. OBJECTIVE In the present study, peptide analogues of Valorphin (VV-hemorphin-5) containing amino phosphonate moiety synthesized by solid-phase peptide synthesis (Fmoc-strategy) were investigated for their potential antinociceptive activities and compared to the reference VV-H in formalin- induced model of acute and inflammatory pain in mice. METHODS The hemorphin analogues were prepared by replacement of the one and/or two N-terminal Val in VV-hemorphin5 (VV-H) with ((dimethoxy phosphoryl) methyl)-L-valine and ((dimethoxy phosphoryl) methyl)-L-leucine to obtain the compounds pVV-H, pL-H, and pLV-H. Aiming to additionally prove the importance of amino acid valine, we introduced the ((dimethoxy phosphoryl) methyl)-L-leucine to the N-side of VV-hemorphin-5 (pLVV-H). The experiments were carried out on adult male ICR mice. All peptides were administered intracerebroventricularly at three doses (50, 25 and 12,5 μg/mouse). We have studied the effects of the peptides on acute (1st phase) and inflammatory (2nd phase) pain reaction using un experimental model with intraplantar formalin injection. RESULTS VV-H showed a significant antinociceptive effect both in the acute and inflammatory phases of the test. Although Valorphin hexa-, hepta-, and octapeptide analogs demonstrated a significant antinociceptive effect, they showed substantial differences considering their effective dose and the phase of the test as compared to the Valorphin. DISCUSSION Data showed that modified heptapeptides pVV-H and pLV-H exerted the same or better antinociception in acute and inflammatory pain, in comparison to the reference peptide, while pL-H and pLVV-H analogues were less effective. CONCLUSION Our study contributes to the elucidation of the role of Valine and the number of amino acid residues in the structure of hemorphin peptide analogs in their effectiveness in suppressing both acute and inflammatory experimental pain.
Collapse
Affiliation(s)
- Borislav Assenov
- Sofia University "St. Kliment Ohridski", Medical Faculty, Sofia 1407, Bulgaria
| | | | - Elena Dzhambazova
- Sofia University "St. Kliment Ohridski", Medical Faculty, Sofia 1407, Bulgaria
| | - Petia Peneva
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, Sofia 1756, Bulgaria
| | - Petar Todorov
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, Sofia 1756, Bulgaria
| |
Collapse
|
7
|
Murineddu G, Asproni B, Corona P, Gessi S, Merighi S, Battistello E, Sturaro C, Calò G, Galeotti N, Temml V, Herdlinger S, Schuster D, Pinna GA. Synthesis, biological evaluation and docking studies of a novel class of sulfur-bridged diazabicyclo[3.3.1]nonanes. Bioorg Chem 2020; 102:104072. [PMID: 32693307 DOI: 10.1016/j.bioorg.2020.104072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 05/06/2020] [Accepted: 07/01/2020] [Indexed: 11/27/2022]
Abstract
A small library of 3-thia-7,9-diazabicyclo[3.3.1]nonanes was synthesized and their opioid receptors affinity and selectivity evaluated. Among these novel sulfur-bridged compounds, the (E) 9-[3'-(3-chlorophenyl)-but-2'-en-1'-yl]-7-propionyl-3-thia-7,9-diazabicyclo[3.3.1]nonane 2i emerged as the derivative with the highest μ receptor affinity (Ki = 85 nM) and selectivity (Ki μ/δ = 58.8, Ki μ/κ > 117.6). The antinociceptive activity of 2i was also evaluated in acute thermal pain. Docking studies disclosed the specific pattern of interactions of these derivatives.
Collapse
Affiliation(s)
- Gabriele Murineddu
- Department of Chemistry and Pharmacy, University of Sassari, 07100 Sassari, Italy.
| | - Battistina Asproni
- Department of Chemistry and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Paola Corona
- Department of Chemistry and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Stefania Gessi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Stefania Merighi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Enrica Battistello
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Chiara Sturaro
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Girolamo Calò
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Nicoletta Galeotti
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Firenze, 50139 Firenze, Italy
| | - Veronika Temml
- Department of Pharmacy/Pharmacognosy, University of Innsbruck, 6020 Innsbruck, Austria
| | - Sonja Herdlinger
- Institute of Pharmacy/Pharmaceutical Chemistry, University of Innsbruck, 6020 Innsbruck, Austria
| | - Daniela Schuster
- Department of Pharmaceutical Chemistry, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Gerard A Pinna
- Department of Chemistry and Pharmacy, University of Sassari, 07100 Sassari, Italy
| |
Collapse
|
8
|
Zhang Y, Zhou P, Wang Z, Chen M, Fu F, Su R. Hsp90β positively regulates μ-opioid receptor function. Life Sci 2020; 252:117676. [PMID: 32304763 DOI: 10.1016/j.lfs.2020.117676] [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: 11/17/2019] [Revised: 03/29/2020] [Accepted: 04/13/2020] [Indexed: 01/19/2023]
Abstract
AIMS Many μ-opioid receptor (MOR)-associated proteins can regulate the MOR signaling pathway. Using a bacterial two-hybrid screen, we found that the C-terminal of the MOR associated with heat shock protein 90 isoform β (Hsp90β). Here, we explored the effect of Hsp90β on MOR signaling transduction and function. MAIN METHODS The interaction of Hsp90β with MOR was detected by co-immunoprecipitation and immunofluorescence. The effects of Hsp90β on MOR signaling induced by opioids were studied in vitro and in vivo. The effects of the Hsp90β inhibitor 17-N-allylamino-17-demethoxygeldanamycin (17-AAG) on morphine tolerance and dependence were studied via a hot plate test and CPP test. KEY FINDINGS Hsp90β, instead of Hsp90α, interacted with the MOR in HEK293 cells and SH-SY5Y cells, and the interaction was augmented after morphine pretreatment. The interaction of Hsp90β and MOR increased the inhibition of cAMP and decreased PKA activity under opioid treatment. The functional Hsp90β-MOR complex also promoted the phosphorylation and internalization of the MOR induced by DAMGO in MOR-CHO cells. 17-AAG blocked Hsp90β-MOR interactions and decreased the effect of Hsp90β on the MOR signal transduction. In C57BL/6 mice, 17-AAG decreased morphine-induced acute anti-nociception in the hot plate test, with an increase in phosphorylated PKA and phosphorylated JNK and a decrease in phosphorylated CREB and phosphorylated ERK in murine brains. Chronic morphine treatment induced tolerance, and dependence was inhibited by 17-AAG co-administration. SIGNIFICANCE Hsp90β is a positive co-regulator of the MOR via the activation of a G-protein-dependent and β-arrestin-dependent pathway. Hsp90β has the potential to improve the pharmacologic profile of existing opiates. It is conceivable that in future clinical treatments, the Hsp90β inhibitor, 17-AAG, could decrease the tolerance and dependence in cancer patients induced by opioids.
Collapse
Affiliation(s)
- Yixin Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China
| | - Peilan Zhou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China
| | - Zhen Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China; School of Pharmacy, Yantai University, Yantai 264005, China
| | - Ming Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China
| | - Fenghua Fu
- School of Pharmacy, Yantai University, Yantai 264005, China
| | - Ruibin Su
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China.
| |
Collapse
|
9
|
Ruan J, Chen L, Ma Z. Activation of spinal Extacellular Signal‐Regulated Kinases and c‐jun N‐terminal kinase signaling pathways contributes to morphine‐induced acute and chronic hyperalgesia in mice. J Cell Biochem 2019; 120:15045-15056. [PMID: 31016764 DOI: 10.1002/jcb.28766] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/27/2019] [Accepted: 01/30/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Jia‐Ping Ruan
- Department of Anesthesiology Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University Nanjing Jiangsu China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Ling Chen
- Laboratory of Reproductive Medicine Nanjing Medical University Nanjing Jiangsu China
| | - Zheng‐liang Ma
- Department of Anesthesiology Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University Nanjing Jiangsu China
| |
Collapse
|
10
|
Yin-and-yang bifurcation of opioidergic circuits for descending analgesia at the midbrain of the mouse. Proc Natl Acad Sci U S A 2018; 115:11078-11083. [PMID: 30297409 DOI: 10.1073/pnas.1806082115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In the descending analgesia pathway, opioids are known to disinhibit the projections from the periaqueductal gray (PAG) to the rostral ventromedial medulla (RVM), leading to suppression of pain signals at the spinal cord level. The locus coeruleus (LC) has been proposed to engage in the descending pathway through noradrenergic inputs to the spinal cord. Nevertheless, how the LC is integrated in the descending analgesia circuit has remained unknown. Here, we show that the opioidergic analgesia pathway is bifurcated in structure and function at the PAG. A knockout as well as a PAG-specific knockdown of phospholipase C β4 (PLCβ4), a signaling molecule for G protein-coupled receptors, enhanced swim stress-induced and morphine-induced analgesia in mice. Immunostaining after simultaneous retrograde labeling from the RVM and the LC revealed two mutually exclusive neuronal populations at the PAG, each projecting either to the LC or the RVM, with PLCβ4 expression only in the PAG-LC projecting cells that provide a direct synaptic input to LC-spinal cord (SC) projection neurons. The PAG-LC projection neurons in wild-type mice turned quiescent in response to opiates, but remained active in the PLCβ4 mutant, suggesting a possibility that an increased adrenergic function induced by the persistent PAG-LC activity underlies the enhanced opioid analgesia in the mutant. Indeed, the enhanced analgesia in the mutant was reversed by blocking α2-noradrenergic receptors. These findings indicate that opioids suppress descending analgesia through the PAG-LC pathway, while enhancing it through the PAG-RVM pathway, i.e., two distinct pathways with opposing effects on opioid analgesia. These results point to a therapeutic target in pain control.
Collapse
|
11
|
Sanna MD, Ghelardini C, Galeotti N. St. John's Wort Potentiates anti-Nociceptive Effects of Morphine in Mice Models of Neuropathic Pain. PAIN MEDICINE 2018; 18:1334-1343. [PMID: 27688309 DOI: 10.1093/pm/pnw241] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Objective In this study, we compared the efficacy of a combination of PKC-blocker St. John's Wort (SJW) and morphine in mice with painful antiretroviral (2,3-dideoxycitidine [ddC]) and chemotherapic (oxaliplatin) neuropathy. Methods Morphine (1 and 5 mg/Kg i.p.), SJW (1 and 5 mg/Kg o.s.), or their combination was administered by systemic injection, and antinociception was determined by using the hot and cold plate tests. Results Here we demonstrate the ability of SJW to relieve neuropathic pain in mice neuropathic models and a potentiation of morphine antinociception in thermal pain. The potentiating effect shown by SJW was not secondary to its antinociceptive activity as the increase of the morphine antinociceptive effect was produced at a dose (1mg/kg o.s.) devoid of any capability to modulate the pain threshold in neuropathic pain mice. Further examinations of the SJW main components revealed that hypericin was responsible for the potentiating properties whereas flavonoids were ineffective. Conclusions These results show that SJW has notable antinociceptive activity for both neuropathic pain models and could be used in neuropathic pain relief alone or in combination with morphine. These data support the utility of combination SJW/opioid therapy in pain management for antinociceptive efficacy by enhancing opioid analgesia.
Collapse
Affiliation(s)
- Maria Domenica Sanna
- Section of Pharmacology and Toxicology, Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Carla Ghelardini
- Section of Pharmacology and Toxicology, Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Nicoletta Galeotti
- Section of Pharmacology and Toxicology, Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| |
Collapse
|
12
|
The Complement System Component C5a Produces Thermal Hyperalgesia via Macrophage-to-Nociceptor Signaling That Requires NGF and TRPV1. J Neurosci 2017; 36:5055-70. [PMID: 27147658 DOI: 10.1523/jneurosci.3249-15.2016] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 03/21/2016] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED The complement cascade is a principal component of innate immunity. Recent studies have underscored the importance of C5a and other components of the complement system in inflammatory and neuropathic pain, although the underlying mechanisms are largely unknown. In particular, it is unclear how the complement system communicates with nociceptors and which ion channels and receptors are involved. Here we demonstrate that inflammatory thermal and mechanical hyperalgesia induced by complete Freund's adjuvant was accompanied by C5a upregulation and was markedly reduced by C5a receptor (C5aR1) knock-out or treatment with the C5aR1 antagonist PMX53. Direct administration of C5a into the mouse hindpaw produced strong thermal hyperalgesia, an effect that was absent in TRPV1 knock-out mice, and was blocked by the TRPV1 antagonist AMG9810. Immunohistochemistry of mouse plantar skin showed prominent expression of C5aR1 in macrophages. Additionally, C5a evoked strong Ca(2+) mobilization in macrophages. Macrophage depletion in transgenic macrophage Fas-induced apoptosis mice abolished C5a-dependent thermal hyperalgesia. Examination of inflammatory mediators following C5a injection revealed a rapid upregulation of NGF, a mediator known to sensitize TRPV1. Preinjection of an NGF-neutralizing antibody or Trk inhibitor GNF-5837 prevented C5a-induced thermal hyperalgesia. Notably, NGF-induced thermal hyperalgesia was unaffected by macrophage depletion. Collectively, these results suggest that complement fragment C5a induces thermal hyperalgesia by triggering macrophage-dependent signaling that involves mobilization of NGF and NGF-dependent sensitization of TRPV1. Our findings highlight the importance of macrophage-to-neuron signaling in pain processing and identify C5a, NGF, and TRPV1 as key players in this cross-cellular communication. SIGNIFICANCE STATEMENT This study provides mechanistic insight into how the complement system, a key component of innate immunity, regulates the development of pain hypersensitivity. We demonstrate a crucial role of the C5a receptor, C5aR1, in the development of inflammatory thermal and mechanical sensitization. By focusing on the mechanisms of C5a-induced thermal hyperalgesia, we show that this process requires recruitment of macrophages and initiation of macrophage-to-nociceptor signaling. At the molecular level, we demonstrate that this signaling depends on NGF and is mediated by the heat-sensitive nociceptive channel TRPV1. This deeper understanding of how immune cells and neurons interact to regulate pain processing is expected to facilitate mechanism-based approaches in the development of new analgesics.
Collapse
|
13
|
Calabrese EJ, Calabrese V, Giordano J. The role of hormesis in the functional performance and protection of neural systems. Brain Circ 2017; 3:1-13. [PMID: 30276298 PMCID: PMC6126232 DOI: 10.4103/2394-8108.203257] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/06/2016] [Accepted: 12/13/2016] [Indexed: 02/03/2023] Open
Abstract
This paper addresses how hormesis, a biphasic dose response, can protect and affect performance of neural systems. Particular attention is directed to the potential role of hormesis in mitigating age-related neurodegenerative diseases, genetically based neurological diseases, as well as stroke, traumatic brain injury, seizure, and stress-related conditions. The hormetic dose response is of particular significance since it mediates the magnitude and range of neuroprotective processes. Consideration of hormetic dose-response concepts can also enhance the quality of study designs, including sample size/statistical power strategies, selection of treatment groups, dose spacing, and temporal/repeat measures’ features.
Collapse
Affiliation(s)
- Edward J Calabrese
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, Morrill I, N344, University of Massachusetts, Amherst, MA 01003, USA
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Viale Andrea Doria, Catania, Italy
| | - James Giordano
- Department of Neurology and Biochemistry, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
14
|
The dark side of opioids in pain management: basic science explains clinical observation. Pain Rep 2016; 1:e570. [PMID: 29392193 PMCID: PMC5741356 DOI: 10.1097/pr9.0000000000000570] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 07/21/2016] [Accepted: 07/25/2016] [Indexed: 12/03/2022] Open
Abstract
Although there is no doubt about opioids' ability to relieve pain in the short term, it is not always clear why longer-term analgesic efficacy seems to be impaired. Tolerance and hyperalgesia have been suggested as mechanisms for opioid analgesic deterioration. But could there also be an effect of opioids on pain itself? Introduction: In the past 2 decades, opioids have been used increasingly for the treatment of persistent pain, and doses have tended to creep up. As basic science elucidates mechanisms of pain and analgesia, the cross talk between central pain and opioid actions becomes clearer. Objectives: We aimed to examine the published literature on basic science explaining pronociceptive opioid actions, and apply this knowledge to clinical observation. Methods: We reviewed the existing literature on the pronociceptive actions of opioids, both preclinical and clinical studies. Results: Basic science provides a rationale for the clinical observation that opioids sometimes increase rather than decrease pain. Central sensitization (hyperalgesia) underlies pain chronification, but can also be produced by high dose and high potency opioids. Many of the same mechanisms account for both central pain and opioid hyperalgesia. Conclusion: Newly revealed basic mechanisms suggest possible avenues for drug development and new drug therapies that could alter pain sensitization through endogenous and exogenous opioid mechanisms. Recent changes in practice such as the introduction of titration-to-effect for opioids have resulted in higher doses used in the clinic setting than ever seen previously. New basic science knowledge hints that these newer dosing practices may need to be reexamined. When pain worsens in a patient taking opioids, can we be assured that this is not because of the opioids, and can we alter this negative effect of opioids through different dosing strategies or new drug intervention?
Collapse
|
15
|
Roeckel LA, Le Coz GM, Gavériaux-Ruff C, Simonin F. Opioid-induced hyperalgesia: Cellular and molecular mechanisms. Neuroscience 2016; 338:160-182. [PMID: 27346146 DOI: 10.1016/j.neuroscience.2016.06.029] [Citation(s) in RCA: 273] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/10/2016] [Accepted: 06/16/2016] [Indexed: 12/18/2022]
Abstract
Opioids produce strong analgesia but their use is limited by a paradoxical hypersensitivity named opioid-induced hyperalgesia (OIH) that may be associated to analgesic tolerance. In the last decades, a significant number of preclinical studies have investigated the factors that modulate OIH development as well as the cellular and molecular mechanisms underlying OIH. Several factors have been shown to influence OIH including the genetic background and sex differences of experimental animals as well as the opioid regimen. Mu opioid receptor (MOR) variants and interactions of MOR with different proteins were shown important. Furthermore, at the cellular level, both neurons and glia play a major role in OIH development. Several neuronal processes contribute to OIH, like activation of neuroexcitatory mechanisms, long-term potentiation (LTP) and descending pain facilitation. Increased nociception is also mediated by neuroinflammation induced by the activation of microglia and astrocytes. Neurons and glial cells exert synergistic effects, which contribute to OIH. The molecular actors identified include the Toll-like receptor 4 and the anti-opioid systems as well as some other excitatory molecules, receptors, channels, chemokines, pro-inflammatory cytokines or lipids. This review summarizes the intracellular and intercellular pathways involved in OIH and highlights some mechanisms that may be challenged to limit OIH in the future.
Collapse
Affiliation(s)
- Laurie-Anne Roeckel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
| | - Glenn-Marie Le Coz
- Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Université de Strasbourg, Illkirch, France
| | - Claire Gavériaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Ecole Supérieure de Biotechnologie de Strasbourg, Université de Strasbourg, France
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Université de Strasbourg, Illkirch, France.
| |
Collapse
|
16
|
Increase of neurofilament-H protein in sensory neurons in antiretroviral neuropathy: Evidence for a neuroprotective response mediated by the RNA-binding protein HuD. Pharmacol Res 2016; 111:23-33. [PMID: 27238228 DOI: 10.1016/j.phrs.2016.05.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 05/06/2016] [Accepted: 05/25/2016] [Indexed: 01/31/2023]
Abstract
Nucleoside reverse transcriptase inhibitors (NRTIs) are key components of HIV/AIDS treatment to reduce viral load. However, antiretroviral toxic neuropathy has become a common peripheral neuropathy among HIV/AIDS patients leading to discontinuation of antiretroviral therapy, for which the underlying pathogenesis is uncertain. This study examines the role of neurofilament (NF) proteins in the spinal dorsal horn, DRG and sciatic nerve after NRTI neurotoxicity in mice treated with zalcitabine (2',3'-dideoxycitidine; ddC). ddC administration up-regulated NF-M and pNF-H proteins with no effect on NF-L. The increase of pNF-H levels was counteracted by the silencing of HuD, an RNA binding protein involved in neuronal development and differentiation. Sciatic nerve sections of ddC exposed mice showed an increased axonal caliber, concomitantly to a pNF-H up-regulation. Both events were prevented by HuD silencing. pNF-H and HuD colocalize in DRG and spinal dorsal horn axons. However, the capability of HuD to bind NF mRNA was not demonstrated, indicating the presence of an indirect mechanism of control of NF expression by HuD. RNA immunoprecipitation experiments showed the capability of HuD to bind the BDNF mRNA and the administration of an anti-BDNF antibody prevented pNF-H increase. These data indicate the presence of a HuD - BDNF - NF-H pathway activated as a regenerative response to the axonal damage induced by ddC treatment to counteract the antiretroviral neurotoxicity. Since analgesics clinically used to treat neuropathic pain are ineffective on antiretroviral neuropathy, a neuroregenerative strategy might represent a new therapeutic opportunity to counteract neurotoxicity and avoid discontinuation or abandon of NRTI therapy.
Collapse
|
17
|
Ghelardini C, Di Cesare Mannelli L, Bianchi E. The pharmacological basis of opioids. CLINICAL CASES IN MINERAL AND BONE METABOLISM 2015; 12:219-21. [PMID: 26811699 DOI: 10.11138/ccmbm/2015.12.3.219] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
An opioid is a chemical that binds to opioid receptors, which are widely distributed in the central and peripheral nervous system and gastrointestinal tract. The different effects elicited by activation of these receptors are due to their specific neuronal and extraneuronal distribution. The painkiller effect of opioids is induced by the synergy of the two events, namely reduction of pain threshold and emotional detachment from pain. The opioid effects transcending analgesia include sedation, respiratory depression, constipation and a strong sense of euphoria. There are opioid-like substances endogenously produced by the body. Naturally occurring peptides, called enkephalins, have opioid-like activities but are not derived from opium and exert opioid-like effects by interacting with opioid receptors on cell membranes. Yet, animals do contain the same morphine precursors and metabolites as opium poppy and are able to synthesize endogenous morphine alkaloid. Experimental and clinical studies show that opioids, at doses comparable to those of endogenous opioids, can activate pronociceptive systems, leading to pain hypersensitivity and short-term tolerance, a phenomenon encountered in postoperative pain management by acute opioid administration. Whether endogenous opioids play a role in the acute pain necessary to the survival of the individual, remains an open question.
Collapse
Affiliation(s)
- Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Enrica Bianchi
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| |
Collapse
|
18
|
Kulyk VB, Chizhmakov IV, Volkova TM, Maximyuk OP, Krishtal OA. [ROLE PHOSPHOINOSITID SIGNALING PATHWAY IN OPIOIDS CONTROL OF P2X3 RECEPTORS IN THE PRIMARY SENSORY NEURONS]. ACTA ACUST UNITED AC 2015; 61:22-9. [PMID: 26552301 DOI: 10.15407/fz61.04.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Homomeric P2X3 receptors expressed in primary nociceptive neurons are crucial elements in the pain signal generation. In turn, opioid system regulates the intensity of this signal in both CNS and PNS. Here we describe the effects of opioids on P2X3 receptors in DRG neurons studied by using patch clamp technique. Activation of G-protein coupled opioid receptors by endogenous opioid Leu-enkephalin (Leu), resulted in the two opposite effects on P2X3 receptor-mediated currents (P2X3 currents). In particular, application of 1 µM Leu lead to the complete inhibition of P2X3 currents. However, after pretreatment of the neurons with a Gi/o-protein inhibitor pertussis toxin (PT), the same concentration of Leu caused facilitation of P2X3 currents. PLC inhibitor U-73122 at concentration of 1 µM completely eliminated both facilitating and inhibitory effects of Leu on P2X3 currents. Thus, opioid receptor agonists cause two oppositely directed effects on P2X3 receptors in DRG neurons of rats and both of them are mediated through PLC signaling pathway. Our results point to a possible molecular basis of the mechanism for the well-known transition inhibitory action of opioids (analgesia) to facilitating (hyperalgesia).
Collapse
|
19
|
Sanna MD, Mello T, Ghelardini C, Galeotti N. Inhibition of spinal ERK1/2–c-JUN signaling pathway counteracts the development of low doses morphine-induced hyperalgesia. Eur J Pharmacol 2015; 764:271-277. [DOI: 10.1016/j.ejphar.2015.07.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 07/06/2015] [Accepted: 07/08/2015] [Indexed: 01/18/2023]
|
20
|
Activation of JNK pathway in spinal astrocytes contributes to acute ultra–low-dose morphine thermal hyperalgesia. Pain 2015; 156:1265-1275. [DOI: 10.1097/j.pain.0000000000000164] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
21
|
Differential contribution of Gαi/o subunits in the response to food deprivation. Eur J Pharmacol 2015; 750:27-31. [PMID: 25620129 DOI: 10.1016/j.ejphar.2015.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 01/14/2015] [Accepted: 01/15/2015] [Indexed: 11/20/2022]
Abstract
Behavioral responses to food deprivation are a fundamental aspect of nervous system function in all animals. Several signaling molecules in the mammalian brain act through G proteins of the Gi/o family to mediate response to food restriction. The present study examined whether food intake changes under a condition of little stimulation to eat, such as that elicited by 4h of food deprivation, was altered by Gi/o isoform silencing induced by intracerebroventricular (i.c.v.) administration of antisense oligodeoxynucleotides (aODN) against the α subunit of Gi1, Gi2, Gi3, Go1 and Go2. The effect of aODN pretreatments on food intake was evaluated 15, 30, 45, and 60min after food re-administration. Selective effects were noted on food intake with anti-Giα1 (3.12-25nmol), Giα3 (1.56-12.5nmol) and Goα2 (3.12-25nmol) aODN exerting increase in food consumption, while anti-Giα2 (3.12-25nmol) and Goα1 (3.12-25nmol) aODN exerting decrease in food consumption. We observed the effect of the α-subunit silencing on food consumption as soon as 15min after food readministration, that was still significant after 60min. At the highest effective doses, different for each anti-Gαi/o subunit, any treatment did not impair motor coordination, nor modified spontaneous mobility and exploratory activity. These results indicate a relevant distinction between Gαi/o subunits on feeding behavior, and suggest that Gi/o proteins are critical for the integrative modulation of normal feeding behavior. Changes in Gi/o protein activity may be associated with modifications of feeding.
Collapse
|
22
|
Chizhmakov I, Kulyk V, Khasabova I, Khasabov S, Simone D, Bakalkin G, Gordienko D, Verkhratsky A, Krishtal O. Molecular mechanism for opioid dichotomy: bidirectional effect of μ-opioid receptors on P2X₃ receptor currents in rat sensory neurones. Purinergic Signal 2015; 11:171-81. [PMID: 25592684 DOI: 10.1007/s11302-015-9443-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/05/2015] [Indexed: 12/28/2022] Open
Abstract
Here, we describe a molecular switch associated with opioid receptors-linked signalling cascades that provides a dual opioid control over P2X3 purinoceptor in sensory neurones. Leu-enkephalin inhibited P2X3-mediated currents with IC50 ~10 nM in ~25% of small nociceptive rat dorsal root ganglion (DRG) neurones. In contrast, in neurones pretreated with pertussis toxin leu-enkephalin produced stable and significant increase of P2X3 currents. All effects of opioid were abolished by selective μ-opioid receptor antagonist D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP), nonselective inhibitor naloxone, and by PLC inhibitor U73122. Thus, we discovered a dual link between purinoceptors and μ-opioid receptors: the latter exert both inhibitory (pertussis toxin-sensitive) and stimulatory (pertussis toxin-insensitive) actions on P2X3 receptors through phospholipase C (PLC)-dependent pathways. This dual opioid control of P2X3 receptors may provide a molecular explanation for dichotomy of opioid therapy. Pharmacological control of this newly identified facilitation/inhibition switch may open new perspectives for the adequate medical use of opioids, the most powerful pain-killing agents known today.
Collapse
Affiliation(s)
- Igor Chizhmakov
- State Key Laboratory for Molecular Biology, Bogomoletz Institute of Physiology, 4 Bogomoletz str., Kiev, 01024, Ukraine
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Sanna MD, Ghelardini C, Galeotti N. Regionally selective activation of ERK and JNK in morphine paradoxical hyperalgesia: A step toward improving opioid pain therapy. Neuropharmacology 2014; 86:67-77. [DOI: 10.1016/j.neuropharm.2014.06.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 06/03/2014] [Accepted: 06/05/2014] [Indexed: 12/30/2022]
|
24
|
Sanna M, Quattrone A, Mello T, Ghelardini C, Galeotti N. The RNA-binding protein HuD promotes spinal GAP43 overexpression in antiretroviral-induced neuropathy. Exp Neurol 2014; 261:343-53. [DOI: 10.1016/j.expneurol.2014.05.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/10/2014] [Accepted: 05/16/2014] [Indexed: 01/20/2023]
|
25
|
Younger J, Parkitny L, McLain D. The use of low-dose naltrexone (LDN) as a novel anti-inflammatory treatment for chronic pain. Clin Rheumatol 2014; 33:451-9. [PMID: 24526250 PMCID: PMC3962576 DOI: 10.1007/s10067-014-2517-2] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 01/22/2014] [Accepted: 01/26/2014] [Indexed: 02/08/2023]
Abstract
Low-dose naltrexone (LDN) has been demonstrated to reduce symptom severity in conditions such as fibromyalgia, Crohn's disease, multiple sclerosis, and complex regional pain syndrome. We review the evidence that LDN may operate as a novel anti-inflammatory agent in the central nervous system, via action on microglial cells. These effects may be unique to low dosages of naltrexone and appear to be entirely independent from naltrexone's better-known activity on opioid receptors. As a daily oral therapy, LDN is inexpensive and well-tolerated. Despite initial promise of efficacy, the use of LDN for chronic disorders is still highly experimental. Published trials have low sample sizes, and few replications have been performed. We cover the typical usage of LDN in clinical trials, caveats to using the medication, and recommendations for future research and clinical work. LDN may represent one of the first glial cell modulators to be used for the management of chronic pain disorders.
Collapse
|
26
|
PKC-mediated HuD–GAP43 pathway activation in a mouse model of antiretroviral painful neuropathy. Pharmacol Res 2014; 81:44-53. [DOI: 10.1016/j.phrs.2014.02.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 02/11/2014] [Accepted: 02/12/2014] [Indexed: 11/23/2022]
|
27
|
Galeotti N, Farzad M, Bianchi E, Ghelardini C. PKC-Mediated Potentiation of Morphine Analgesia by St. John’s Wort in Rodents and Humans. J Pharmacol Sci 2014; 124:409-17. [DOI: 10.1254/jphs.13226fp] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
28
|
Inhibition of Gβγ-subunit signaling potentiates morphine-induced antinociception but not respiratory depression, constipation, locomotion, and reward. Behav Pharmacol 2013; 24:144-52. [PMID: 23412114 DOI: 10.1097/fbp.0b013e32835f3d2f] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Inhibition of Gβγ-subunit signaling to phospholipase C β3 has been shown to potentiate morphine-mediated antinociception while attenuating the development of tolerance and dependence in mice. The objective of this study was to determine the effect of Gβγ-subunit inhibition on antinociception and other pharmacological effects, such as respiratory depression, constipation, and hyperlocomotion, mediated by the μ-opioid receptor. The Gβγ-subunit inhibitor, gallein, was administered to C57BL/6J mice by intraperitoneal injection before morphine, and data were compared with mice treated with vehicle, morphine, or gallein alone. Morphine-induced antinociception was measured using the 55°C warm-water tail-withdrawal test. Pretreatment with gallein produced a dose-dependent potentiation of morphine-mediated antinociception, producing up to a 10-fold leftward shift in the morphine dose-response curve and extending the duration of antinociception induced by a single dose of morphine. Gallein pretreatment also prevented acute antinociceptive tolerance induced by morphine. In contrast, the dose-dependent respiratory depression and hyperlocomotion induced by morphine were not potentiated by gallein pretreatment. Similarly, gallein pretreatment did not potentiate morphine-conditioned place preference responses or morphine-induced constipation, as measured as a reduction in excreta. These results suggest that selectively inhibiting Gβγ-mediated signaling may selectively increase μ-opioid receptor-mediated antinociception without matching increases in adverse physiological effects.
Collapse
|
29
|
Merighi S, Gessi S, Varani K, Fazzi D, Mirandola P, Borea PA. Cannabinoid CB(2) receptor attenuates morphine-induced inflammatory responses in activated microglial cells. Br J Pharmacol 2012; 166:2371-85. [PMID: 22428664 DOI: 10.1111/j.1476-5381.2012.01948.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Among several pharmacological properties, analgesia is the most common feature shared by either opioid or cannabinoid systems. Cannabinoids and opioids are distinct drug classes that have been historically used separately or in combination to treat different pain states. In the present study, we characterized the signal transduction pathways mediated by cannabinoid CB(2) and µ-opioid receptors in quiescent and LPS-stimulated murine microglial cells. EXPERIMENTAL APPROACH We examined the effects of µ-opioid and CB(2) receptor stimulation on phosphorylation of MAPKs and Akt and on IL-1β, TNF-α, IL-6 and NO production in primary mouse microglial cells. KEY RESULTS Morphine enhanced release of the proinflammatory cytokines, IL-1β, TNF-α, IL-6, and of NO via µ-opioid receptor in activated microglial cells. In contrast, CB(2) receptor stimulation attenuated morphine-induced microglial proinflammatory mediator increases, interfering with morphine action by acting on the Akt-ERK1/2 signalling pathway. CONCLUSIONS AND IMPLICATIONS Because glial activation opposes opioid analgesia and enhances opioid tolerance and dependence, we suggest that CB(2) receptors, by inhibiting microglial activity, may be potential targets to increase clinical efficacy of opioids.
Collapse
Affiliation(s)
- Stefania Merighi
- Department of Clinical and Experimental Medicine, Pharmacology Section and Interdisciplinary Center for the Study of Inflammation, University of Ferrara, Ferrara, Italy
| | | | | | | | | | | |
Collapse
|
30
|
Schindler EAD, Harvey JA, Aloyo VJ. Phospholipase C mediates (±)-1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI)-, but not lysergic acid diethylamide (LSD)-elicited head bobs in rabbit medial prefrontal cortex. Brain Res 2012; 1491:98-108. [PMID: 23123701 DOI: 10.1016/j.brainres.2012.10.057] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 10/20/2012] [Accepted: 10/27/2012] [Indexed: 11/24/2022]
Abstract
The phenethylamine and indoleamine classes of hallucinogens demonstrate distinct pharmacological properties, although they share a serotonin(2A) (5-HT(2A)) receptor mechanism of action (MOA). The 5-HT(2A) receptor signals through phosphatidylinositol (PI) hydrolysis, which is initiated upon activation of phospholipase C (PLC). The role of PI hydrolysis in the effects of hallucinogens remains unclear. In order to better understand the role of PI hydrolysis in the MOA of hallucinogens, the PLC inhibitor, 1-[6-((17β-3-methoxyestra-1,3,5(10)-trien-17-yl)amino)hexyl]-1H-pyrrole-2,5-dione (U73122), was used to study the effects of two hallucinogens, the phenethylamine, (±)-1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI), and the indoleamine, lysergic acid diethylamide (LSD). PI hydrolysis was quantified through release of [3H]inositol-4-phosphate from living rabbit frontocortical tissue prisms. Head bobs were counted after hallucinogens were infused into the medial prefrontal cortex (mPFC) of rabbits. Both DOI and LSD stimulated PI hydrolysis in frontocortical tissue through activation of PLC. DOI-stimulated PI hydrolysis was blocked by 5-HT(2A/2C) receptor antagonist, ketanserin, whereas the LSD signal was blocked by 5-HT(2B/2C) receptor antagonist, SB206553. When infused into the mPFC, both DOI- and LSD-elicited head bobs. Pretreatment with U73122 blocked DOI-, but not LSD-elicited head bobs. The two hallucinogens investigated were distinct in their activation of the PI hydrolysis signaling pathway. The serotonergic receptors involved with DOI and LSD signals in frontocortical tissue were different. Furthermore, PLC activation in mPFC was necessary for DOI-elicited head bobs, whereas LSD-elicited head bobs were independent of this pathway. These novel findings urge closer investigation into the intracellular mechanism of action of these unique compounds.
Collapse
Affiliation(s)
- Emmanuelle A D Schindler
- Drexel University College of Medicine, Department of Pharmacology & Physiology, 245 N. 15th Street, Philadelphia, PA 19102, United States.
| | | | | |
Collapse
|
31
|
Intrathecal PLC(β3) oligodeoxynucleotides antisense potentiates acute morphine efficacy and attenuates chronic morphine tolerance. Brain Res 2012; 1472:38-44. [PMID: 22771399 DOI: 10.1016/j.brainres.2012.06.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 03/21/2012] [Accepted: 06/24/2012] [Indexed: 11/20/2022]
Abstract
Morphine is a mainstay for chronic pain treatment, but its efficacy has been hampered by physical tolerance. The underlying mechanism for chronic morphine induced tolerance is complicated and not well understood. PLC(β3) is regarded as an important factor in the morphine tolerance signal pathway. In this study, we determined intrathecal (i.t.) administration of an antisense oligodeoxynucleotide (ODN) of PLC(β3) could quicken the on-set antinociceptive efficacy of acute morphine treatment and prolong the maximum effect up to 4h. The antisense could also attenuate the development of morphine-induced tolerance and left shift the ED50 after 7 day of coadministration with morphine. These results probably were contributed by the PLC(β3) antisense ODN as they successfully knocked down protein expression levels and reduced activity of PLC(β3) in spinal cord in rats. The mismatch group had no such effects. The results confirmed the important involvement of PLC(β3) in both acute morphine efficacy and chronic morphine tolerance at spinal level in rats. This study may provide an idea for producing a novel adjuvant for morphine treatment.
Collapse
|
32
|
Aira Z, Buesa I, Del Caño GG, Salgueiro M, Mendiable N, Mingo J, Aguilera L, Bilbao J, Azkue JJ. Selective impairment of spinal mu-opioid receptor mechanism by plasticity of serotonergic facilitation mediated by 5-HT2A and 5-HT2B receptors. Pain 2012; 153:1418-1425. [PMID: 22520172 DOI: 10.1016/j.pain.2012.03.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 01/19/2012] [Accepted: 03/14/2012] [Indexed: 01/11/2023]
Abstract
Opioid analgesia is compromised by intracellular mediators such as protein kinase C (PKC). The phosphatidylinositol hydrolysis-coupled serotonin receptor 5-HT2 is ideally suited to promote PKC activation. We test the hypothesis that 5-HT2A and 5-HT2B receptors, which have been previously shown to become pro-excitatory after spinal nerve ligation (SNL), can negatively influence the ability of opioids to depress spinal excitation evoked by noxious input. Spinal superfusion with (100 nM) mu-opioid receptor (MOR)-agonist DAMGO significantly depressed C fiber-evoked spinal field potentials. Simultaneous administration of subclinical 5-HT2AR antagonist 4F 4PP (100 nM) or 5-HT2BR antagonist SB 204741 (100 nM) significantly reduced the IC50 value for DAMGO in nerve-ligated rats (97.56 nM ± 1.51 and 1.20 nM ± 1.28 respectively, relative to 104 nM ± 1.08 at the baseline condition), but not in sham-operated rats. Both antagonists failed to alter depression induced by delta-opioid receptor (DOR)-agonist D-ala2-deltorphin II after SNL as well as in the sham condition. Western blot analysis of dorsal horn homogenates revealed bilateral upregulation of 5-HT2AR and 5-HT2BR protein band densities after SNL. As assessed from double immunofluorescence labeling for confocal laser scanning microscopy, scarce dorsal horn cell processes showed co-localization color overlay for 5-HT2AR/MOR, 5-HT2BR/MOR, 5-HT2AR/DOR, or 5-HT2BR/DOR in sham-operated rats. Intensity correlation-based analyses showed significant increases in 5-HT2AR/MOR and 5-HT2BR/MOR co-localizations after SNL. These results indicate that plasticity of spinal serotonergic neurotransmission can selectively reduce spinal MOR mechanisms via 5-HT2A and 5-HT2B receptors, including upregulation of the latter and increased expression in dorsal horn neurons containing MOR.
Collapse
Affiliation(s)
- Zigor Aira
- Department of Neurosciences, School of Medicine and Dentistry, University of the Basque Country, Leioa, Spain Department of Neurosciences, School of Pharmacy, University of the Basque Country, Vitoria-Gasteiz, Spain Department of Surgery, Radiology and Physical Medicine, University of the Basque Country, Bilbao, Spain Department of Preventive Medicine and Public Health, School of Medicine and Dentistry, University of the Basque Country, Leioa, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Xin W, Chun W, Ling L, Wei W. Role of melatonin in the prevention of morphine-induced hyperalgesia and spinal glial activation in rats: protein kinase C pathway involved. Int J Neurosci 2011; 122:154-63. [PMID: 22050217 DOI: 10.3109/00207454.2011.635828] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSES Morphine can induce tolerance and hyperalgesia after long-term administration. Glial activation is believed to cause and maintain a state of morphine-induced pain hypersensitivity. The present study examines the effect of melatonin on tolerance, hyperalgesia, and reactive gliosis induced by morphine in rats. METHODS The study examines the effect of melatonin on morphine-induced hyperalgesia using tail-flick test. Immunohistochemistry and Western blot was performed to investigate the expression of glial fibrillary acidic protein (GFAP) indicative of spinal glial activity. This study also measures protein kinase C (PKC) activity and cyclic adenosine monophosphate (cAMP) levels in spinal cords to investigate the mechanisms which melatonin involved. RESULTS When coadministered intragastrically (i.g.) with morphine, melatonin in doses of 50 or 100 mg/kg significantly prevented hyperalgesia after termination of morphine. Immunohistochemistry and Western blot with GFAP revealed that melatonin significantly decreased morphine-induced over-expression of GFAP in spinal cord (p < .05). By measuring PKC activity and cAMP levels, the upregulated PKC activity and cAMP levels induced by morphine were significantly inhibited by melatonin. CONCLUSIONS Melatonin can prevent morphine-withdrawal-induced hyperalgesia and glial reactivity. This effect of melatonin after morphine administration may mediated by inhibiting PKC activity and cAMP upregulation.
Collapse
Affiliation(s)
- Wei Xin
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Antiinflammatory and Immunopharmacology, Ministry of Education, Hefei, P. R. China
| | | | | | | |
Collapse
|
34
|
Wei X, Wei W. Role of gabapentin in preventing fentanyl- and morphine-withdrawal-induced hyperalgesia in rats. J Anesth 2011; 26:236-41. [PMID: 22048285 DOI: 10.1007/s00540-011-1272-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 10/18/2011] [Indexed: 11/30/2022]
Abstract
PURPOSE This study was undertaken to examine the effect of gabapentin for preventing hyperalgesia induced by morphine and fentanyl withdrawal in rats. METHODS To induce hyperalgesia, Sprague Dawley (SD) rats were subcutaneously injected with fentanyl four times at 15-min intervals (60 μg/kg per injection), resulting in total dose of 240 μg/kg over 1 h, and morphine 10 mg/kg twice daily for 7 days. The effect of gabapentin was detected with behavioral tail-flick and paw-withdrawal tests. RESULTS Drug termination produced significant decrease in antinociception thresholds (P < 0.05 vs. saline group), indicating that the rats became sensitive to thermal stimuli. In rats that received combined treatment with fentanyl/morphine and gabapentin (25/50 mg/kg), results demonstrated that there were no significant decreases in antinociception thresholds (vs. saline group) after opioid withdrawal. Gabapentin (50 mg/kg) could also prevent morphine tolerance. The 50% effective dose (ED50) value was 12.5 mg/kg in tail-flick and 13.6 mg/kg in paw-withdrawal tests. CONCLUSIONS The study showed that gabapentin can significantly prevented opioid-induced hyperalgesia (OIH) induced caused by fentanyl and morphine, suggesting a role for the addition of gabapentin in the perioperative period and during chronic pain treatment as an effective drug to prevent OIH.
Collapse
Affiliation(s)
- Xin Wei
- Department of Anesthesiology, Anhui Medical University Affiliated Auhui Provincial Hospital, 230001 Hefei, People's Republic of China
| | | |
Collapse
|
35
|
Bianchi E, Galeotti N, Menicacci C, Ghelardini C. Contribution of G inhibitory protein alpha subunits in paradoxical hyperalgesia elicited by exceedingly low doses of morphine in mice. Life Sci 2011; 89:918-25. [PMID: 22008476 DOI: 10.1016/j.lfs.2011.09.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 08/26/2011] [Accepted: 09/15/2011] [Indexed: 11/30/2022]
Abstract
AIMS Although morphine, at higher doses, induces analgesia, it may also enhance sensitivity to pain at extremely low doses as shown in studies for testing an animal's sensitivity to pain. We used an antisense approach capable of selectively down-regulating in vivo G(i)(G inhibitory protein),G(o) and G(s) members of the G(α) sub-family protein subunits in order to establish if these proteins might be implicated in the effects induced by extremely low morphine doses on acute thermonociception. MAIN METHODS Mice pretreated with a morphine hyperalgesic dose (1μg/kg) were submitted to hot plate test after pre-treatment with antisense oligodeoxynucleotides (aODNs) targeting G(iα), G(oα) and G(sα) regulatory proteins. The association of G-protein (guanine nucleotide-binding regulatory protein) coupled receptors with G protein was investigated using co-immunoprecipitation procedure. KEY FINDINGS The downregulation of the G(iα1-3) and G(oα1) proteins reversed the licking latency responses induced by 1μg/kg morphine administration toward the basal value whereas downregulation of the G(oα2) and G(sα) proteins did not significantly modify the hyperalgesic response. SIGNIFICANCE These results suggest that G inhibitory proteins play a role in the production of low dose evoked morphine hyperalgesia in mouse. Immunoprecipitation studies revealed that both μ opioid receptor (μOR) and α(2) adrenoreceptor (α(2) AR) are bound to G inhibitory proteins in hyperalgesic response to morphine extremely low dose. Both μOR and α(2) AR appear to be necessary for low morphine dose induced hyperalgesic response through G inhibitory proteins.
Collapse
Affiliation(s)
- Enrica Bianchi
- Department of Neuroscience, University of Siena, Siena, Italy.
| | | | | | | |
Collapse
|
36
|
Gupta LK, Gupta R, Tripathi CD. N-Methyl-d-aspartate receptor modulators block hyperalgesia induced by acute low-dose morphine. Clin Exp Pharmacol Physiol 2011; 38:592-7. [DOI: 10.1111/j.1440-1681.2011.05556.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
37
|
Buprenorphine-induced hyperalgesia in the rat. Eur J Pharmacol 2011; 651:89-95. [DOI: 10.1016/j.ejphar.2010.10.083] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Revised: 10/13/2010] [Accepted: 10/31/2010] [Indexed: 11/19/2022]
|
38
|
Lee FA, Baiamonte BA, Spano D, Lahoste GJ, Soignier RD, Harrison LM. Mice lacking rhes show altered morphine analgesia, tolerance, and dependence. Neurosci Lett 2010; 489:182-6. [PMID: 21163334 DOI: 10.1016/j.neulet.2010.12.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 10/12/2010] [Accepted: 12/07/2010] [Indexed: 02/05/2023]
Abstract
Rhes, the Ras Homolog Enriched in Striatum, is an intermediate-size GTP binding protein. Although its full functions are not yet known, it has been shown to affect signaling and behaviors mediated by G protein-coupled receptors. Here we have tested whether Rhes affects behaviors mediated by opioid receptors. Wild type and rhes-deficient mice were administered morphine and tested for analgesia in formalin and tail flick tests. Rhes⁻/⁻ mice showed significantly enhanced analgesia in both tests relative to rhes+/+ mice. Furthermore, rhes⁻/⁻ mice did not display tolerance to repeated morphine administration and displayed significantly less withdrawal than rhes+/+ mice. These findings indicate that Rhes is involved in behaviors mediated by mu opioid receptors and in the adaptive response to repeated morphine administration.
Collapse
Affiliation(s)
- Franklin A Lee
- Department of Psychology, University of New Orleans, New Orleans, LA 70148, USA
| | | | | | | | | | | |
Collapse
|
39
|
Song Z, Zou W, Liu C, Guo Q. Gene knockdown with lentiviral vector-mediated intrathecal RNA interference of protein kinase C gamma reverses chronic morphine tolerance in rats. J Gene Med 2010; 12:873-80. [PMID: 21105149 DOI: 10.1002/jgm.1514] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Revised: 08/14/2010] [Accepted: 10/10/2010] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Although morphine is a widely used opioid analgesic, morphine tolerance (MT) has limited the use of the drug because it creates the necessity for high doses. Protein kinase C (PKC), especially the PKCγ isoform, is considered to play a key role in the development of MT. Because RNA interference provides a powerful method for the investigation of gene function, and lentiviral delivery systems have been approved for human use, this present study examined rats tolerant to morphine to determine whether an intrathecal injection of a lentiviral vector of PKCγ short hairpin RNA (LV-shPKCγ) down-regulated the expression of the PKCγ gene and reversed MT. METHODS MT was induced by intrathecal morphine (10 µg b.i.d.) for six consecutive days. A lentiviral-mediated short hairpin RNA (shRNA) system was synthesized to deliver the PKCγ shRNAs to the spinal cord of the rats with MT. Mechanical and thermal paw withdrawal threshold were assessed to determine the analgesic effects of morphine. Expression of PKCγ mRNA and protein was determined by reverse transcriptase-polymerase chain reaction and western blotting analysis, respectively. RESULTS The chronic administration of morphine induced a stabilized analgesic tolerance. A single injection of LV-shPKCγ significantly reversed morphine antinociceptive tolerance. Compared to the control group, PKCγ mRNA and protein levels were dramatically down-regulated in the LV-shPKCγ group. CONCLUSIONS A single injection of LV-shPKCγ reversed MT by reducing the expression of PKCγ in the spinal cord. These findings indicate that the use of LV-shPKCγ might be a potential strategy for therapy in MT.
Collapse
Affiliation(s)
- Zongbin Song
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | | | | | | |
Collapse
|
40
|
Molecular assays for characterization of alternatively spliced isoforms of the u opioid receptor (MOR). Methods Mol Biol 2010; 617:421-35. [PMID: 20336438 DOI: 10.1007/978-1-60327-323-7_30] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mu-opioid receptor (MOR) belongs to a family of heptahelical G-protein-coupled receptors (GPCRs). Studies in humans and rodents demonstrated that the OPRM1 gene coding for MOR undergoes extensive alternative splicing afforded by the genetic complexity of OPRM1. Evidence from rodent studies also demonstrates an important role of these alternatively spliced forms in mediating opiate analgesia via their differential signaling properties. MOR signaling is predominantly G(ia) coupled. Release of the alpha subunit from G-protein complex results in the inhibition of adenylyl cyclase/cAMP pathway, whereas release of the betagamma subunits activates G-protein-activated inwardly rectifying potassium channels and inhibits voltage-dependent calcium channels. These molecular events result in the suppression of cellular activities that diminish pain sensations. Recently, a new isoform of OPRM1, MOR3, has been identified that shows an increase in the production of nitric oxide (NO) upon stimulation with morphine. Hence, there is a need to describe molecular techniques that enable the functional characterization of MOR isoforms. In this review, we describe the methodologies used to assay key mediators of MOR activation including cellular assays for cAMP, free Ca(2+), and NO, all of which have been implicated in the pharmacological effects of MOR agonists.
Collapse
|
41
|
Bianchi E, Lehmann D, Vivoli E, Norcini M, Ghelardini C. Involvement of PLC-beta3 in the effect of morphine on memory retrieval in passive avoidance task. J Psychopharmacol 2010; 24:891-6. [PMID: 19282422 DOI: 10.1177/0269881108102013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Phospholipase C (PLC) is one signalling effector enzyme whose activity is directly modulated by opioids. Several physiological studies have implicated PLC-linked pathways in in-vivo pain regulation and opioid tolerance. Co-administration of PLC-beta(2/3) activity blocker M119 with morphine resulted in a dramatic increase in morphine-induced amnesic effect in mice, proving a role for beta subunit of PLC enzyme in these processes. Administration of morphine to mice at amnesic dose increased PLC-beta(3) activity, with respect to basal value, in the membrane-soluble material from anterior cortex and hippocampal formation in brain areas. PLC-beta(3) appears to be simultaneously implicated in both analgesic and amnesic effects induced by administration of morphine to mice suggesting a commonality in the molecular mechanisms of morphine-induced analgesia and memory impairment.
Collapse
Affiliation(s)
- E Bianchi
- Department of Neuroscience, University of Siena, Siena, Italy.
| | | | | | | | | |
Collapse
|
42
|
|
43
|
Berger AC, Whistler JL. How to design an opioid drug that causes reduced tolerance and dependence. Ann Neurol 2010; 67:559-69. [PMID: 20437553 DOI: 10.1002/ana.22002] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Mu opioid receptor (MOR) agonists such as morphine are extremely effective treatments for acute pain. In the setting of chronic pain, however, their long-term utility is limited by the development of tolerance and physical dependence. Drug companies have tried to overcome these problems by simply "dialing up" signal transduction at the receptor, designing more potent and efficacious agonists and more long-lasting formulations. Neither of these strategies has proven to be successful, however, because the net amount of signal transduction, particularly over extended periods of drug use, is a product of much more than the pharmacokinetic properties of potency, efficacy, half-life, and bioavailability, the mainstays of traditional pharmaceutical screening. Both the quantity and quality of signal transduction are influenced by many regulated processes, including receptor desensitization, trafficking, and oligomerization. Importantly, the efficiency with which an agonist first stimulates signal transduction is not necessarily related to the efficiency with which it stimulates these other processes. Here we describe recent findings that suggest MOR agonists with diminished propensity to cause tolerance and dependence can be identified by screening drugs for the ability to induce MOR desensitization, endocytosis, and recycling. We also discuss preliminary evidence that heteromers of the delta opioid receptor and the MOR are pronociceptive, and that drugs that spare such heteromers may also induce reduced tolerance.
Collapse
Affiliation(s)
- Amy Chang Berger
- Department of Neurology, University of California, San Francisco, Ernest Gallo Clinic and Research Center, Emeryville, CA, USA
| | | |
Collapse
|
44
|
Abstract
Clinical pain conditions may remain responsive to opiate analgesics for extended periods, but such persistent acute pain can undergo a transition to an opiate-resistant chronic pain state that becomes a much more serious clinical problem. To test the hypothesis that cellular mechanisms of chronic pain in the primary afferent also contribute to the development of opiate resistance, we used a recently developed model of the transition of from acute to chronic pain, hyperalgesic priming. Repeated intradermal administration of the potent and highly selective mu-opioid agonist, [d-Ala(2),N-MePhe(4),gly-ol]-enkephalin (DAMGO), to produce tolerance for its inhibition of prostaglandin E(2) hyperalgesia, simultaneously produced hyperalgesic priming. Conversely, injection of an inflammogen, carrageenan, used to produce priming produced DAMGO tolerance. Both effects were prevented by inhibition of protein kinase Cepsilon (PKCepsilon). Carrageenan also induced opioid dependence, manifest as mu-opioid receptor antagonist (d-Phe-Cys-Tyr-d-Trp-Orn-Thr-Pen-Thr-NH(2))-induced hyperalgesia that, like priming, was PKCepsilon and G(i) dependent. These findings suggest that the transition from acute to chronic pain, and development of mu-opioid receptor tolerance and dependence may be linked by common cellular mechanisms in the primary afferent.
Collapse
|
45
|
Contralateral electroacupuncture pretreatment suppresses carrageenan-induced inflammatory pain via the opioid-mu receptor. Rheumatol Int 2010; 31:725-30. [PMID: 20130880 DOI: 10.1007/s00296-010-1364-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Accepted: 09/13/2009] [Indexed: 10/19/2022]
Abstract
Acupuncture has been used to treat various clinical diseases in Eastern medicine. To investigate the analgesic effect of electroacupuncture (EA) pretreatment on carrageenan-induced inflammatory pain, we studied on the effect of EA parameters on an animal model of acute arthritic pain. Pretreatment with 1 mA, 10 Hz EA prior to carrageenan injection under halothane anesthesia suppressed carrageenan-induced pain. Interestingly, EA stimulation of the 'Zu-San-Li' (ST36) acupuncture point (1 mA, 10 Hz) contralateral to the site of the carrageenan injection in the rat synovial cavity produced significantly greater improvement of the weight-bearing force compared with EA stimulation of the 'San-Yin-Jiao' acupuncture point. To determine how ST36 EA treatment suppresses carrageenan-induced inflammatory pain, we examined the effect of a mu opioid receptor antagonist on ST36 EA-induced analgesia. The selective antagonist of the mu opioid receptor (OR) significantly suppressed contralateral ST36 EA-induced analgesia against carrageenan-induced inflammation. These results suggested that the analgesic effect mediated by the mu OR during low-frequency contralateral EA pretreatment has an anti-nociceptive action against inflammatory pain and that it may provide a potential strategy to treat inflammatory arthritic pain.
Collapse
|
46
|
Systemic Lidocaine Inhibits Remifentanil-induced Hyperalgesia via the Inhibition of cPKCgamma Membrane Translocation in Spinal Dorsal Horn of Rats. J Neurosurg Anesthesiol 2009; 21:318-25. [DOI: 10.1097/ana.0b013e3181abbde5] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
47
|
Bianchi E, Norcini M, Smrcka A, Ghelardini C. Supraspinal Gbetagamma-dependent stimulation of PLCbeta originating from G inhibitory protein-mu opioid receptor-coupling is necessary for morphine induced acute hyperalgesia. J Neurochem 2009; 111:171-80. [PMID: 19656263 DOI: 10.1111/j.1471-4159.2009.06308.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Although alterations in micro-opioid receptor (microOR) signaling mediate excitatory effects of opiates in opioid tolerance, the molecular mechanism for the excitatory effect of acute low dose morphine, as it relates to microOR coupling, is presently unknown. A pronounced coupling of microOR to the alpha subunit of G inhibitory protein emerged in periaqueductal gray (PAG) from mice systemically administered with morphine at a dose producing acute thermal hyperalgesia. This coupling was abolished in presence of the selective microOR antagonist d-Phe-Cys-Tyr-d-Trp-Orn-Thr-Pen-Thr-NH(2) administered at the PAG site, showing that the low dose morphine effect is triggered by microOR activated G inhibitory protein at supraspinal level. When Gbetagamma downstream signalling was blocked by intra-PAG co-administration of 2-(3,4,5-trihydroxy-6-oxoxanthen-9-yl)cyclohexane-1-carboxylic acid, a compound that inhibits Gbetagamma dimer-dependent signaling, a complete prevention of low dose morphine induced acute thermal hyperalgesia was obtained. Phospholipase C beta3, an enzyme necessary to morphine hyperalgesia, was revealed to be associated with Gbetagamma in PAG. Although opioid administration induces a shift in microOR-G protein coupling from Gi to Gs after chronic administration, our data support that this condition is not realized in acute treatment providing evidence that a separate molecular mechanism underlies morphine induced acute excitatory effect.
Collapse
Affiliation(s)
- Enrica Bianchi
- Department of Neuroscience, University of Siena, Siena, Italy.
| | | | | | | |
Collapse
|
48
|
A novel Gbetagamma-subunit inhibitor selectively modulates mu-opioid-dependent antinociception and attenuates acute morphine-induced antinociceptive tolerance and dependence. J Neurosci 2009; 28:12183-9. [PMID: 19020012 DOI: 10.1523/jneurosci.2326-08.2008] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The Gbetagamma subunit has been implicated in many downstream signaling events associated with opioids. We previously demonstrated that a small molecule inhibitor of Gbetagamma-subunit-dependent phospholipase (PLC) activation potentiated morphine-induced analgesia (Bonacci et al., 2006). Here, we demonstrate that this inhibitor, M119 (cyclohexanecarboxylic acid [2-(4,5,6-trihydroxy-3-oxo-3H-xanthen-9-yl)-(9Cl)]), is selective for mu-opioid receptor-dependent analgesia and has additional efficacy in mouse models of acute tolerance and dependence. When administered by an intracerebroventricular injection in mice, M119 caused 10-fold and sevenfold increases in the potencies of morphine and the mu-selective peptide, DAMGO, respectively. M119 had little or no effect on analgesia induced by the kappa agonist U50,488 or delta agonists DPDPE or Deltorphin II. Similar results were obtained in vitro, as only activation of the mu-opioid receptor stimulated PLC activation, whereas no effect was seen with the kappa- and delta-opioid receptors. M119 inhibited mu-receptor-dependent PLC activation. In studies to further explore the in vivo efficacy of M119, systemic administration M119 also resulted in a fourfold shift increase in potency of systemically administered morphine. Of particular interest, M119 was also able to attenuate acute, antinociceptive tolerance and dependence in mice treated concomitantly with both M119 and morphine. These studies suggest that small organic molecules, such as M119, that specifically regulate Gbetagamma subunit signaling may have important therapeutic applications in enhancing opioid analgesia, while attenuating the development of tolerance and dependence.
Collapse
|
49
|
Shabalina SA, Zaykin DV, Gris P, Ogurtsov AY, Gauthier J, Shibata K, Tchivileva IE, Belfer I, Mishra B, Kiselycznyk C, Wallace MR, Staud R, Spiridonov NA, Max MB, Goldman D, Fillingim RB, Maixner W, Diatchenko L. Expansion of the human mu-opioid receptor gene architecture: novel functional variants. Hum Mol Genet 2008; 18:1037-51. [PMID: 19103668 PMCID: PMC2649019 DOI: 10.1093/hmg/ddn439] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The μ-opioid receptor (OPRM1) is the principal receptor target for both endogenous and exogenous opioid analgesics. There are substantial individual differences in human responses to painful stimuli and to opiate drugs that are attributed to genetic variations in OPRM1. In searching for new functional variants, we employed comparative genome analysis and obtained evidence for the existence of an expanded human OPRM1 gene locus with new promoters, alternative exons and regulatory elements. Examination of polymorphisms within the human OPRM1 gene locus identified strong association between single nucleotide polymorphism (SNP) rs563649 and individual variations in pain perception. SNP rs563649 is located within a structurally conserved internal ribosome entry site (IRES) in the 5′-UTR of a novel exon 13-containing OPRM1 isoforms (MOR-1K) and affects both mRNA levels and translation efficiency of these variants. Furthermore, rs563649 exhibits very strong linkage disequilibrium throughout the entire OPRM1 gene locus and thus affects the functional contribution of the corresponding haplotype that includes other functional OPRM1 SNPs. Our results provide evidence for an essential role for MOR-1K isoforms in nociceptive signaling and suggest that genetic variations in alternative OPRM1 isoforms may contribute to individual differences in opiate responses.
Collapse
Affiliation(s)
- Svetlana A Shabalina
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Phospholipase C{beta}3 in mouse and human dorsal root ganglia and spinal cord is a possible target for treatment of neuropathic pain. Proc Natl Acad Sci U S A 2008; 105:20004-8. [PMID: 19066214 DOI: 10.1073/pnas.0810899105] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Treatment of neuropathic pain is a major clinical problem. This study shows expression of phospholipase ss3 (PLCss3) in mouse and human DRG neurons, mainly in small ones and mostly with a nonpeptidergic phenotype. After spared nerve injury, the pain threshold was strongly reduced, and systemic treatment of such animals with the unselective PLC inhibitor U73122 caused a rapid and long-lasting (48-h) increase in pain threshold. Thus, inhibition of PLC may provide a way to treat neuropathic pain.
Collapse
|