1
|
Getsy PM, May WJ, Henderson F, Seckler JM, Grossfield A, Baby SM, Lewis SJ. Nitrosyl factors play a vital role in the ventilatory depressant effects of fentanyl in freely moving guinea pigs. Biomed Pharmacother 2025; 183:117847. [PMID: 39862705 DOI: 10.1016/j.biopha.2025.117847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/10/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
An understanding of intracellular mechanisms by which fentanyl and other synthetic opioids exert adverse effects on breathing is needed. Using freely moving adult male guinea pigs, we administered the nitric oxide synthase (NOS) inhibitor, L-NAME (NG-nitro-L-arginine methyl ester), to determine whether nitrosyl factors, such as nitric oxide and S-nitrosothiols, play a role in fentanyl-induced respiratory depression. Ventilatory parameters were recorded by whole body plethysmography to determine the effects of fentanyl (75 μg/kg, IV) in guinea pigs that had received a prior injection of vehicle (saline), L-NAME or the inactive D-isomer, D-NAME (both at 50 μmol/kg, IV), 15 min beforehand. L- and D-NAME elicited minor effects on most parameters, including frequency of breathing, tidal volume and minute ventilation, although L-NAME did decrease end expiratory pause and non-eupneic breathing index (NEBI). Subsequent injection of fentanyl in guinea pigs pre-treated with vehicle produced profound and sustained reductions in frequency, tidal volume, minute ventilation, peak inspiratory flow, and inspiratory and expiratory drives, while increasing inspiratory time, expiratory time, end inspiratory pause, and NEBI. These ventilatory depressant effects of fentanyl seen in guinea pigs pre-treated with vehicle were markedly diminished in guinea pigs pre-treated with L-NAME. Moreover, the adverse effects of fentanyl on many recorded breathing parameters were converted to stimulatory effects. In contrast, D-NAME did not alter any of the effects of fentanyl on breathing. This study is the first to characterize the role nitrosyl factors play in the intracellular mechanisms involved in fentanyl-induced respiratory depression in guinea pigs.
Collapse
Affiliation(s)
- Paulina M Getsy
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA.
| | - Walter J May
- Pediatric Respiratory Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Fraser Henderson
- Pediatric Respiratory Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - James M Seckler
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Alan Grossfield
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA
| | - Santhosh M Baby
- Section of Biology, Galleon Pharmaceuticals, Inc, Horsham, PA, USA
| | - Stephen J Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA; Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
2
|
Santana VC, Marmentini BM, Cruz GG, de Jesus LC, Walicheski L, Beffa FH, Maffei THP, Streg RV, Veiga-Junior VF, Andrighetti CR, Freitas de Lima MC, de Sousa Valladão DM, de Oliveira RC, Neyra MOC, de Araújo Berber RC, Falconi-Sobrinho LL, Coimbra NC, de Oliveira R. Copaifera langsdorffii Desf. tree oleoresin-induced antinociception recruits µ 1- and κ -opioid receptors in the ventrolateral columns of the periaqueductal gray matter. Behav Brain Res 2024; 461:114832. [PMID: 38142860 DOI: 10.1016/j.bbr.2023.114832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Popular medicine has been using oleoresin from several species of copaíba tree for the treatment of various diseases and its clinical administration potentially causes antinociception. Electrical stimulation of ventrolateral (vlPAG) and dorsolateral (dlPAG) columns of the periaqueductal gray matter also causes antinociception. The aim this study was to verify the antinociceptive effect of oleoresin extracted from Copaifera langsdorffii tree and to test the hypothesis that oleoresin-induced antinociception is mediated by µ1- and κ-opioid receptors in the vlPAG and dlPAG. Nociceptive thresholds were determined by the tail-flick test in Wistar rats. The copaíba tree oleoresin was administered at different doses (50, 100 and 200 mg/kg) through the gavage technique. After the specification of the most effective dose of copaíba tree oleoresin (200 mg/kg), rats were pretreated with either the µ1-opioid receptor selective antagonist naloxonazine (at 0.05, 0.5 and 5 µg/ 0.2 µl in vlPAG, and 5 µg/ 0.2 µl in dlPAG) or the κ-opioid receptor selective antagonist nor-binaltorphimine (at 1, 3 and 9 nmol/ 0.2 µl in vlPAG, and 9 nmol/ 0.2 µl in dlPAG). The blockade of µ1 and κ opioid receptors of vlPAG decreased the antinociception produced by copaíba tree oleoresin. However, the blockade of these receptors in dlPAG did not alter copaíba tree oleoresin-induced antinociception. These data suggest that vlPAG µ1 and κ opioid receptors are critically recruited in the antinociceptive effect produced by oleoresin extracted from Copaifera langsdorffii.
Collapse
Affiliation(s)
- Vanessa Cristina Santana
- Laboratory of Experimental Neuropsychobiology and Toxicology, Institute of Health Sciences, Federal University of Mato Grosso (UFMT), Av. Alexandre Ferronato, 1200, Reserva 35, Setor Industrial, Sinop 78557-267, Mato Grosso, Brazil
| | - Bruna Magda Marmentini
- Laboratory of Experimental Neuropsychobiology and Toxicology, Institute of Health Sciences, Federal University of Mato Grosso (UFMT), Av. Alexandre Ferronato, 1200, Reserva 35, Setor Industrial, Sinop 78557-267, Mato Grosso, Brazil
| | - Geórgia Guedes Cruz
- Laboratory of Experimental Neuropsychobiology and Toxicology, Institute of Health Sciences, Federal University of Mato Grosso (UFMT), Av. Alexandre Ferronato, 1200, Reserva 35, Setor Industrial, Sinop 78557-267, Mato Grosso, Brazil
| | - Leila Camila de Jesus
- Laboratory of Experimental Neuropsychobiology and Toxicology, Institute of Health Sciences, Federal University of Mato Grosso (UFMT), Av. Alexandre Ferronato, 1200, Reserva 35, Setor Industrial, Sinop 78557-267, Mato Grosso, Brazil
| | - Luana Walicheski
- Laboratory of Experimental Neuropsychobiology and Toxicology, Institute of Health Sciences, Federal University of Mato Grosso (UFMT), Av. Alexandre Ferronato, 1200, Reserva 35, Setor Industrial, Sinop 78557-267, Mato Grosso, Brazil
| | - Fábio Henrique Beffa
- Laboratory of Experimental Neuropsychobiology and Toxicology, Institute of Health Sciences, Federal University of Mato Grosso (UFMT), Av. Alexandre Ferronato, 1200, Reserva 35, Setor Industrial, Sinop 78557-267, Mato Grosso, Brazil
| | - Talles Henrique Pichinelli Maffei
- Laboratory of Experimental Neuropsychobiology and Toxicology, Institute of Health Sciences, Federal University of Mato Grosso (UFMT), Av. Alexandre Ferronato, 1200, Reserva 35, Setor Industrial, Sinop 78557-267, Mato Grosso, Brazil
| | - Rafaela Vieira Streg
- Laboratory of Experimental Neuropsychobiology and Toxicology, Institute of Health Sciences, Federal University of Mato Grosso (UFMT), Av. Alexandre Ferronato, 1200, Reserva 35, Setor Industrial, Sinop 78557-267, Mato Grosso, Brazil
| | - Valdir Florêncio Veiga-Junior
- Chemical Engineering Section, Military Institute of Engineering, Praça General Tibúrcio, 80, Praia Vermelha, Urca, Rio de Janeiro, 22290-270 Rio de Janeiro, Brazil
| | - Carla Regina Andrighetti
- Laboratory of Pharmacognosy, Institute of Health Sciences, Mato Grosso Federal University (UFMT), Av. Alexandre Ferronato, 1200, Reserva 35, Setor Industrial, Sinop 78557-267, Mato Grosso, Brazil
| | - Milena Campelo Freitas de Lima
- Federal University of Amazonas, Department of Chemistry, Av. General Rodrigo Octávio Jordão Ramos, 1200, Coroado I, Manaus 69067-005, Amazonas, Brazil
| | - Dênia Mendes de Sousa Valladão
- Laboratory of Quality Control, Institute of Health Sciences, Federal University of Mato Grosso (UFMT), Av. Alexandre Ferronato, 1200, Reserva 35, Setor Industrial, Sinop 78557-267, Mato Grosso, Brazil
| | - Rithiele Cristina de Oliveira
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; Behavioural Neurosciences Institute (INeC), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil
| | - Milton Omar Cordova Neyra
- Laboratory of Experimental Neuropsychobiology and Toxicology, Institute of Health Sciences, Federal University of Mato Grosso (UFMT), Av. Alexandre Ferronato, 1200, Reserva 35, Setor Industrial, Sinop 78557-267, Mato Grosso, Brazil
| | - Rodolfo Cassimiro de Araújo Berber
- Laboratory of Experimental Neuropsychobiology and Toxicology, Institute of Health Sciences, Federal University of Mato Grosso (UFMT), Av. Alexandre Ferronato, 1200, Reserva 35, Setor Industrial, Sinop 78557-267, Mato Grosso, Brazil
| | - Luiz Luciano Falconi-Sobrinho
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; Behavioural Neurosciences Institute (INeC), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; NAP-USP-Neurobiology of Emotions Research Center (NuPNE), Ribeirão Preto Medical School of the University of São Paulo, Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil
| | - Norberto Cysne Coimbra
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; Behavioural Neurosciences Institute (INeC), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; NAP-USP-Neurobiology of Emotions Research Center (NuPNE), Ribeirão Preto Medical School of the University of São Paulo, Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil
| | - Ricardo de Oliveira
- Laboratory of Experimental Neuropsychobiology and Toxicology, Institute of Health Sciences, Federal University of Mato Grosso (UFMT), Av. Alexandre Ferronato, 1200, Reserva 35, Setor Industrial, Sinop 78557-267, Mato Grosso, Brazil; Behavioural Neurosciences Institute (INeC), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil.
| |
Collapse
|
3
|
Sakloth F, Sanchez-Reyes OB, Ruiz A, Nicolais A, Serafini RA, Pryce KD, Bertherat F, Torres-Berrío A, Gomes I, Devi LA, Wacker D, Zachariou V. A Regional and Projection-Specific Role of RGSz1 in the Ventrolateral Periaqueductal Grey in the Modulation of Morphine Reward. Mol Pharmacol 2023; 103:1-8. [PMID: 36310031 PMCID: PMC11033942 DOI: 10.1124/molpharm.122.000528] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 09/12/2022] [Accepted: 10/11/2022] [Indexed: 02/03/2023] Open
Abstract
Opioid analgesics exert their therapeutic and adverse effects by activating μ opioid receptors (MOPR); however, functional responses to MOPR activation are modulated by distinct signal transduction complexes within the brain. The ventrolateral periaqueductal gray (vlPAG) plays a critical role in modulation of nociception and analgesia, but the exact intracellular pathways associated with opioid responses in this region are not fully understood. We previously showed that knockout of the signal transduction modulator Regulator of G protein Signaling z1 (RGSz1) enhanced analgesic responses to opioids, whereas it decreased the rewarding efficacy of morphine. Here, we applied viral mediated gene transfer methodology and delivered adeno-associated virus (AAV) expressing Cre recombinase to the vlPAG of RGSz1fl\fl mice to demonstrate that downregulation of RGSz1 in this region decreases sensitivity to morphine in the place preference paradigm, under pain-free as well as neuropathic pain states. We also used retrograde viral vectors along with flippase-dependent Cre vectors to conditionally downregulate RGSz1 in vlPAG projections to the ventral tegmental area (VTA) and show that downregulation of RGSz1 prevents the development of place conditioning to low morphine doses. Consistent with the role for RGSz1 as a negative modulator of MOPR activity, RGSz1KO enhances opioid-induced cAMP inhibition in periaqueductal gray (PAG) membranes. Furthermore, using a new generation of bioluminescence resonance energy transfer (BRET) sensors, we demonstrate that RGSz1 modulates Gαz but not other Gαi family subunits and selectively impedes MOPR-mediated Gαz signaling events invoked by morphine and other opioids. Our work highlights a regional and circuit-specific role of the G protein-signaling modulator RGSz1 in morphine reward, providing insights on midbrain intracellular pathways that control addiction-related behaviors. SIGNIFICANCE STATEMENT: This study used advanced genetic mouse models to highlight the role of the signal transduction modulator named RGSz1 in responses to clinically used opioid analgesics. We show that RGSz1 controls the rewarding efficacy of opioids by actions in ventrolateral periaqueductal gray projections to the ventral tegmental area, a key component of the midbrain dopamine pathway. These studies highlight novel mechanisms by which pain-modulating structures control the rewarding efficacy of opioids.
Collapse
Affiliation(s)
- Farhana Sakloth
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Omar B Sanchez-Reyes
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Anne Ruiz
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Andrew Nicolais
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Randal A Serafini
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Kerri D Pryce
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Feodora Bertherat
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Angélica Torres-Berrío
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Ivone Gomes
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Lakshmi A Devi
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Daniel Wacker
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Venetia Zachariou
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| |
Collapse
|
4
|
Hiroki T, Suto T, Ohta J, Saito S, Obata H. Spinal γ-Aminobutyric Acid Interneuron Plasticity Is Involved in the Reduced Analgesic Effects of Morphine on Neuropathic Pain. THE JOURNAL OF PAIN 2022; 23:547-557. [PMID: 34678470 DOI: 10.1016/j.jpain.2021.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/16/2021] [Accepted: 10/05/2021] [Indexed: 12/25/2022]
Abstract
Systemic administration of morphine increases serotonin (5-HT) in the spinal dorsal horn (SDH), which attenuates the analgesic effects of morphine on neuropathic pain through spinal 5-HT3 receptors. We hypothesized that dysfunction of the descending serotonergic system, including the periaqueductal gray (PAG), contributes to attenuate the efficacy of morphine on neuropathic pain through spinal 5-HT3 receptors and GABA neurons. Morphine (100 ng) injected into the PAG produced analgesic effects in normal rats, but not in spinal nerve ligation (SNL) rats. In vivo microdialysis showed that PAG morphine increased the SDH 5-HT concentration in both groups. Intrathecal injection of the 5-HT3 receptor antagonist ondansetron and the GABAA receptor antagonist bicuculline attenuated the analgesic effects of PAG morphine in normal rats, but increased the effects in SNL rats. The increased analgesic effect of PAG morphine induced by bicuculline was reversed by pretreatment with the tropomyosin receptor kinase B (TrkB) antagonist K252a. Activation of spinal 5-HT3 receptors by 2-methyl-5-HT increased the GABA concentration in both groups. Morphine activates GABAergic interneurons in the SDH by activating descending serotonergic neurons. Functional changes in GABAA receptors from inhibitory to facilitatory through the activation of TrkB receptors may contribute to the attenuated efficacy of morphine against neuropathic pain. PERSPECTIVE: Although morphine provides strong analgesia against acute pain, it has limited efficacy against neuropathic pain. This article demonstrates that functional changes in GABAA receptors in the spinal dorsal horn after nerve injury might strongly contribute to the attenuation of opioid-induced analgesia for neuropathic pain.
Collapse
Affiliation(s)
- Tadanao Hiroki
- Department of Anesthesiology, Gunma University Graduate School of Medicine, Maebashi, Japan.
| | - Takashi Suto
- Department of Anesthesiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Jo Ohta
- Department of Anesthesiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Shigeru Saito
- Department of Anesthesiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hideaki Obata
- Department of Anesthesiology, Saitama Medical Center, Saitama Medical University, Kawagoe, Japan
| |
Collapse
|
5
|
Rossi GC, Bodnar RJ. Interactive Mechanisms of Supraspinal Sites of Opioid Analgesic Action: A Festschrift to Dr. Gavril W. Pasternak. Cell Mol Neurobiol 2021; 41:863-897. [PMID: 32970288 DOI: 10.1007/s10571-020-00961-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/03/2020] [Indexed: 12/30/2022]
Abstract
Almost a half century of research has elaborated the discoveries of the central mechanisms governing the analgesic responses of opiates, including their receptors, endogenous peptides, genes and their putative spinal and supraspinal sites of action. One of the central tenets of "gate-control theories of pain" was the activation of descending supraspinal sites by opiate drugs and opioid peptides thereby controlling further noxious input. This review in the Special Issue dedicated to the research of Dr. Gavril Pasternak indicates his contributions to the understanding of supraspinal mediation of opioid analgesic action within the context of the large body of work over this period. This review will examine (a) the relevant supraspinal sites mediating opioid analgesia, (b) the opioid receptor subtypes and opioid peptides involved, (c) supraspinal site analgesic interactions and their underlying neurophysiology, (d) molecular (particularly AS) tools identifying opioid receptor actions, and (e) relevant physiological variables affecting site-specific opioid analgesia. This review will build on classic initial studies, specify the contributions that Gavril Pasternak and his colleagues did in this specific area, and follow through with studies up to the present.
Collapse
Affiliation(s)
- Grace C Rossi
- Department of Psychology, C.W. Post College, Long Island University, Post Campus, Brookville, NY, USA.
| | - Richard J Bodnar
- Department of Psychology, Queens College of the City University of New York, Flushing, NY, USA
- CUNY Neuroscience Collaborative, Graduate Center, CUNY, New York, NY, USA
| |
Collapse
|
6
|
Viisanen H, Lilius TO, Sagalajev B, Rauhala P, Kalso E, Pertovaara A. Neurophysiological response properties of medullary pain-control neurons following chronic treatment with morphine or oxycodone: modulation by acute ketamine. J Neurophysiol 2020; 124:790-801. [DOI: 10.1152/jn.00343.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Morphine and oxycodone are two clinically used strong opioids. Chronic treatment with oxycodone as well as morphine can lead to analgesic tolerance and paradoxical hyperalgesia. Here we show that an N-methyl-d-aspartate receptor-dependent pronociceptive change in discharge properties of rostroventromedial medullary neurons controlling spinal nociception has an important role in antinociceptive tolerance to morphine but not oxycodone. Interestingly, chronic oxycodone did not induce pronociceptive changes in the rostroventromedial medulla.
Collapse
Affiliation(s)
- Hanna Viisanen
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tuomas O. Lilius
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Boriss Sagalajev
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pekka Rauhala
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eija Kalso
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Anaesthesiology, Intensive Care Medicine and Pain Medicine, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- SleepWell Research Programme, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Antti Pertovaara
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
7
|
μ-Opioid receptors in primary sensory neurons are involved in supraspinal opioid analgesia. Brain Res 2019; 1729:146623. [PMID: 31881186 DOI: 10.1016/j.brainres.2019.146623] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/20/2019] [Accepted: 12/23/2019] [Indexed: 11/21/2022]
Abstract
Both inhibiting ascending nociceptive transmission and activating descending inhibition are involved in the opioid analgesic effect. The spinal dorsal horn is a critical site for modulating nociceptive transmission by descending pathways elicited by opioids in the brain. μ-Opioid receptors (MORs, encoded by Oprm1) are highly expressed in primary sensory neurons and their central terminals in the spinal cord. In the present study, we tested the hypothesis that MORs expressed in primary sensory neurons contribute to the descending inhibition and supraspinal analgesic effect induced by centrally administered opioids. We generated Oprm1 conditional knockout (Oprm1-cKO) mice by crossing AdvillinCre/+ mice with Oprm1flox/flox mice. Immunocytochemical labeling in Oprm1-cKO mice showed that MORs are completely ablated from primary sensory neurons and are profoundly reduced in the superficial spinal dorsal horn. Intracerebroventricular injection of morphine or fentanyl produced a potent analgesic effect in wild-type mice, but such an effect was significantly attenuated in Oprm1-cKO mice. Furthermore, the analgesic effect produced by morphine or fentanyl microinjected into the periaqueductal gray was significantly greater in wild-type mice than in Oprm1-cKO mice. Blocking MORs at the spinal cord level diminished the analgesic effect of morphine and fentanyl microinjected into the periaqueductal gray in both groups of mice. Our findings indicate that MORs expressed at primary afferent terminals in the spinal cord contribute to the supraspinal opioid analgesic effect. These presynaptic MORs in the spinal cord may serve as an interface between ascending inhibition and descending modulation that are involved in opioid analgesia.
Collapse
|
8
|
Morgan MM, Tran A, Wescom RL, Bobeck EN. Differences in antinociceptive signalling mechanisms following morphine and fentanyl microinjections into the rat periaqueductal gray. Eur J Pain 2019; 24:617-624. [PMID: 31785128 DOI: 10.1002/ejp.1513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/08/2019] [Accepted: 11/25/2019] [Indexed: 01/13/2023]
Abstract
BACKGROUND Morphine and fentanyl are two of the most commonly used opioids to treat pain. Although both opioids produce antinociception by binding to mu-opioid receptors (MOR), they appear to act via distinct signalling pathways. OBJECTIVE This study will reveal whether differences in morphine and fentanyl antinociception are the result of selective activation of G-protein signalling and/or selective activation of pre- or postsynaptic MORs. METHODS The contribution of each mechanism to morphine and fentanyl antinociception was assessed by microinjecting drugs to alter G-protein signalling or block potassium channels linked to pre- and postsynaptic MORs in the ventrolateral periaqueductal gray (PAG) of male Sprague-Dawley rats. RESULTS Both morphine and fentanyl produced a dose-dependent antinociception when microinjected into the PAG. Enhancement of intracellular G-protein signalling by microinjection of the Regulator of G-protein Signalling 4 antagonist CCG-63802 into the PAG enhanced the antinociceptive potency of morphine, but not fentanyl. Microinjection of α-dendrotoxin into the PAG to block MOR activation of presynaptic Kv + channels caused a significant rightward shift in the dose-response curve of both morphine and fentanyl. Microinjection of tertiapin-Q to block MOR activation of postsynaptic GIRK channels caused a larger shift in the dose-response curve for fentanyl than morphine antinociception. CONCLUSIONS These findings reveal different PAG signalling mechanisms for morphine and fentanyl antinociception. In contrast with fentanyl, the antinociceptive effects of morphine are mediated by G-protein signalling primarily activated by presynaptic MORs. SIGNIFICANCE Microinjection of the opioids morphine and fentanyl into the periaqueductal gray (PAG) produce antinociception via mu-opioid receptor signalling. This study reveals differences in the signalling mechanisms underlying morphine and fentanyl antinociception in the PAG. In contrast with fentanyl, morphine antinociception is primarily mediated by presynaptic opioid receptors and is enhanced by blocking RGS proteins.
Collapse
Affiliation(s)
- Michael M Morgan
- Department of Psychology, Washington State University Vancouver, Vancouver, WA, USA
| | - Alexander Tran
- Department of Psychology, Washington State University Vancouver, Vancouver, WA, USA
| | - Rebecca L Wescom
- Department of Psychology, Washington State University Vancouver, Vancouver, WA, USA
| | - Erin N Bobeck
- Department of Biology, Utah State University, Logan, UT, USA
| |
Collapse
|
9
|
Riters LV, Kelm-Nelson CA, Spool JA. Why Do Birds Flock? A Role for Opioids in the Reinforcement of Gregarious Social Interactions. Front Physiol 2019; 10:421. [PMID: 31031641 PMCID: PMC6473117 DOI: 10.3389/fphys.2019.00421] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 03/27/2019] [Indexed: 12/16/2022] Open
Abstract
The formation of social groups provides safety and opportunities for individuals to develop and practice important social skills. However, joining a social group does not result in any form of obvious, immediate reinforcement (e.g., it does not result in immediate copulation or a food reward), and individuals often remain in social groups despite agonistic responses from conspecifics. Much is known about neural and endocrine mechanisms underlying the motivation to perform mate- or offspring-directed behaviors. In contrast, relatively little is known about mechanisms underlying affiliative behaviors outside of these primary reproductive contexts. Studies on flocking behavior in songbirds are beginning to fill this knowledge gap. Here we review behavioral evidence that supports the hypothesis that non-sexual affiliative, flocking behaviors are both (1) rewarded by positive social interactions with conspecifics, and (2) reinforced because affiliative contact reduces a negative affective state caused by social isolation. We provide evidence from studies in European starlings, Sturnus vulgaris, that mu opioid receptors in the medial preoptic nucleus (mPOA) play a central role in both reward and the reduction of a negative affective state induced by social interactions in flocks, and discuss potential roles for nonapeptide/opioid interactions and steroid hormones. Finally, we develop the case that non-sexual affiliative social behaviors may be modified by two complementary output pathways from mPOA, with a projection from mPOA to the periaqueductal gray integrating information during social interactions that reduces negative affect and a projection from mPOA to the ventral tegmental area integrating information leading to social approach and reward.
Collapse
Affiliation(s)
- Lauren V. Riters
- Department of Integrative Biology, University of Wisconsin–Madison, Madison, WI, United States
| | - Cynthia A. Kelm-Nelson
- Division of Otolaryngology-Head & Neck Surgery, Department of Surgery, University of Wisconsin–Madison, Madison, WI, United States
| | - Jeremy A. Spool
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| |
Collapse
|
10
|
Mascarenhas DC, Gomes KS, Sorregotti T, Nunes-de-Souza RL. Blockade of Cannabinoid CB1 Receptors in the Dorsal Periaqueductal Gray Unmasks the Antinociceptive Effect of Local Injections of Anandamide in Mice. Front Pharmacol 2017; 8:695. [PMID: 29046638 PMCID: PMC5632997 DOI: 10.3389/fphar.2017.00695] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 09/19/2017] [Indexed: 12/12/2022] Open
Abstract
Divergent results in pain management account for the growing number of studies aiming at elucidating the pharmacology of the endocannabinoid/endovanilloid anandamide (AEA) within several pain-related brain structures. For instance, the stimulation of both Transient Receptor Potential Vanilloid type 1 (TRPV1) and Cannabinoid type 1 (CB1) receptors led to paradoxical effects on nociception. Here, we attempted to propose a clear and reproducible methodology to achieve the antinociceptive effect of exogenous AEA within the dorsal periaqueductal gray (dPAG) of mice exposed to the tail-flick test. Accordingly, male Swiss mice received intra-dPAG injection of AEA (CB1/TRPV1 agonist), capsaicin (TRPV1 agonist), WIN (CB1 agonist), AM251 (CB1 antagonist), and 6-iodonordihydrocapsaicin (6-IODO) (TRPV1 selective antagonist) and their nociceptive response was assessed with the tail-flick test. In order to assess AEA effects on nociception specifically at vanilloid or cannabinoid (CB) substrates into the dPAG, mice underwent an intrinsically inactive dose of AM251 or 6-IODO followed by local AEA injections and were subjected to the same test. While intra-dPAG AEA did not change acute pain, local injections of capsaicin or WIN induced a marked TRPV1- and CB1-dependent antinociceptive effect, respectively. Regarding the role of AEA specifically at CB/vanilloid substrates, while the blockade of TRPV1 did not change the lack of effects of intra-dPAG AEA on nociception, local pre-treatment of AM251, a CB1 antagonist, led to a clear AEA-induced antinociception. It seems that the exogenous AEA-induced antinociception is unmasked when it selectively binds to vanilloid substrates, which might be useful to address acute pain in basic and perhaps clinical trials.
Collapse
Affiliation(s)
- Diego C Mascarenhas
- Joint Graduate Program in Physiological Sciences, Federal University of São Carlos and São Paulo State University, São Carlos, Brazil.,Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| | - Karina S Gomes
- Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| | - Tatiani Sorregotti
- Joint Graduate Program in Physiological Sciences, Federal University of São Carlos and São Paulo State University, São Carlos, Brazil.,Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| | - Ricardo L Nunes-de-Souza
- Joint Graduate Program in Physiological Sciences, Federal University of São Carlos and São Paulo State University, São Carlos, Brazil.,Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| |
Collapse
|
11
|
El Bitar N, Pollin B, Karroum E, Pincedé I, Le Bars D. Entanglement between thermoregulation and nociception in the rat: the case of morphine. J Neurophysiol 2016; 116:2473-2496. [PMID: 27605533 PMCID: PMC5133307 DOI: 10.1152/jn.00482.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/03/2016] [Indexed: 11/22/2022] Open
Abstract
In thermoneutral conditions, rats display cyclic variations of the vasomotion of the tail and paws, the most widely used target organs in current acute or chronic animal models of pain. Systemic morphine elicits their vasoconstriction followed by hyperthermia in a naloxone-reversible and dose-dependent fashion. The dose-response curves were steep with ED50 in the 0.5-1 mg/kg range. Given the pivotal functional role of the rostral ventromedial medulla (RVM) in nociception and the rostral medullary raphe (rMR) in thermoregulation, two largely overlapping brain regions, the RVM/rMR was blocked by muscimol: it suppressed the effects of morphine. "On-" and "off-" neurons recorded in the RVM/rMR are activated and inhibited by thermal nociceptive stimuli, respectively. They are also implicated in regulating the cyclic variations of the vasomotion of the tail and paws seen in thermoneutral conditions. Morphine elicited abrupt inhibition and activation of the firing of on- and off-cells recorded in the RVM/rMR. By using a model that takes into account the power of the radiant heat source, initial skin temperature, core body temperature, and peripheral nerve conduction distance, one can argue that the morphine-induced increase of reaction time is mainly related to the morphine-induced vasoconstriction. This statement was confirmed by analyzing in psychophysical terms the tail-flick response to random variations of noxious radiant heat. Although the increase of a reaction time to radiant heat is generally interpreted in terms of analgesia, the present data question the validity of using such an approach to build a pain index.
Collapse
Affiliation(s)
- Nabil El Bitar
- Sorbonne Universités, Université Pierre et Marie Curie, Faculté de Médecine, Paris, France; and
- Neurosciences Paris-Seine, Institut National de la Santé et de la Recherche Médicale UMRS-1130, Centre National de la Recherche Scientifique UMR-8246, Paris, France
| | - Bernard Pollin
- Sorbonne Universités, Université Pierre et Marie Curie, Faculté de Médecine, Paris, France; and
- Neurosciences Paris-Seine, Institut National de la Santé et de la Recherche Médicale UMRS-1130, Centre National de la Recherche Scientifique UMR-8246, Paris, France
| | - Elias Karroum
- Sorbonne Universités, Université Pierre et Marie Curie, Faculté de Médecine, Paris, France; and
- Neurosciences Paris-Seine, Institut National de la Santé et de la Recherche Médicale UMRS-1130, Centre National de la Recherche Scientifique UMR-8246, Paris, France
| | - Ivanne Pincedé
- Sorbonne Universités, Université Pierre et Marie Curie, Faculté de Médecine, Paris, France; and
- Neurosciences Paris-Seine, Institut National de la Santé et de la Recherche Médicale UMRS-1130, Centre National de la Recherche Scientifique UMR-8246, Paris, France
| | - Daniel Le Bars
- Sorbonne Universités, Université Pierre et Marie Curie, Faculté de Médecine, Paris, France; and
- Neurosciences Paris-Seine, Institut National de la Santé et de la Recherche Médicale UMRS-1130, Centre National de la Recherche Scientifique UMR-8246, Paris, France
| |
Collapse
|
12
|
Kandasamy R, Calsbeek JJ, Morgan MM. Analysis of inflammation-induced depression of home cage wheel running in rats reveals the difference between opioid antinociception and restoration of function. Behav Brain Res 2016; 317:502-507. [PMID: 27746208 DOI: 10.1016/j.bbr.2016.10.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/11/2016] [Accepted: 10/12/2016] [Indexed: 02/06/2023]
Abstract
Opioids are effective at inhibiting responses to noxious stimuli in rodents, but have limited efficacy and many side effects in chronic pain patients. One reason for this disconnect is that nociception is typically assessed using withdrawal from noxious stimuli in animals, whereas chronic pain patients suffer from abnormal pain that disrupts normal activity. We hypothesized that assessment of home cage wheel running in rats would provide a much more clinically relevant method to assess opioid efficacy to restore normal behavior. Intraplantar injection of Complete Freund's Adjuvant (CFA) into the right hindpaw depressed wheel running and caused mechanical allodynia measured with the von Frey test in both male and female rats. Administration of an ED50 dose of morphine (3.2mg/kg) reversed mechanical allodynia, but did not reverse CFA-induced depression of wheel running. In contrast, administration of a low dose of morphine (1.0mg/kg) restored running for one hour in both sexes, but had no effect on mechanical allodynia. Administration of the atypical opioid buprenorphine had no effect on inflammation-induced depression of wheel running in male or female rats, but attenuated mechanical allodynia in male rats. Administration of buprenorphine and higher doses of morphine depressed wheel running in non-inflamed rats, suggesting that the side effects of opioids interfere with restoration of function. These data indicate that restoration of pain-depressed function requires antinociception in the absence of disruptive side effects. The disruptive side effects of opioids are consistent with the major limitation of opioid use in human pain patients.
Collapse
Affiliation(s)
- Ram Kandasamy
- Graduate Program in Neuroscience, Washington State University, Pullman, WA, United States.
| | - Jonas J Calsbeek
- Department of Psychology, Washington State University Vancouver, Vancouver, WA, United States
| | - Michael M Morgan
- Graduate Program in Neuroscience, Washington State University, Pullman, WA, United States; Department of Psychology, Washington State University Vancouver, Vancouver, WA, United States
| |
Collapse
|
13
|
Roles of prefrontal cortex and paraventricular thalamus in affective and mechanical components of visceral nociception. Pain 2016; 156:2479-2491. [PMID: 26262826 DOI: 10.1097/j.pain.0000000000000318] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Visceral pain represents a major clinical challenge in the management of many gastrointestinal disorders, eg, pancreatitis. However, cerebral neurobiological mechanisms underlying visceral nociception are poorly understood. As a representative model of visceral nociception, we applied cerulein hyperstimulation in C57BL6 mice to induce acute pancreatitis and performed a behavioral test battery and c-Fos staining of brains. We observed a specific pain phenotype and a significant increase in c-Fos immunoreactivity in the paraventricular nucleus of the thalamus (PVT), the periaqueductal gray, and the medial prefrontal cortex (mPFC). Using neuronal tracing, we observed projections of the PVT to cortical layers of the mPFC with contacts to inhibitory GABAergic neurons. These inhibitory neurons showed more activation after cerulein treatment suggesting thalamocortical "feedforward inhibition" in visceral nociception. The activity of neurons in pancreatitis-related pain centers was pharmacogenetically modulated by designer receptors exclusively activated by designer drugs, selectively and cell type specifically expressed in target neurons using adeno-associated virus-mediated gene transfer. Pharmacogenetic inhibition of PVT but not periaqueductal gray neurons attenuated visceral pain and induced an activation of the descending inhibitory pain pathway. Activation of glutamatergic principle neurons in the mPFC, but not inhibitory neurons, also reversed visceral nociception. These data reveal novel insights into central pain processing that underlies visceral nociception and may trigger the development of novel, potent centrally acting analgesic drugs.
Collapse
|
14
|
Tryon VL, Mizumori SJY, Morgan MM. Analysis of morphine-induced changes in the activity of periaqueductal gray neurons in the intact rat. Neuroscience 2016; 335:1-8. [PMID: 27545314 DOI: 10.1016/j.neuroscience.2016.08.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 07/09/2016] [Accepted: 08/11/2016] [Indexed: 11/29/2022]
Abstract
Microinjection of morphine into the periaqueductal gray (PAG) produces antinociception. In vitro slice recordings indicate that all PAG neurons are sensitive to morphine either by direct inhibition or indirect disinhibition. We tested the hypothesis that all PAG neurons respond to opioids in vivo by examining the extracellular activity of PAG neurons recorded in lightly anesthetized and awake rats. Spontaneous activity was less than 1Hz in most neurons. Noxious stimuli (heat, pinch) caused an increase in activity in 57% and 75% of the neurons recorded in anesthetized and awake rats, respectively. The same noxious stimuli caused a decrease in activity in only 17% and 6% of neurons recorded in anesthetized and awake rats. Systemic administration of morphine caused approximately equal numbers of neurons to increase, decrease, or show no change in activity in lightly anesthetized rats. In contrast, administration of morphine caused an increase in the activity of 22 of the 27 neurons recorded in awake rats. No change in activity was evident in the remaining five neurons. Changes in activity caused by morphine were surprisingly modest (a median increase from 0.7 to 1.3Hz). The small inconsistent effects of morphine are in stark contrast to the large changes produced by morphine on the activity of rostral ventromedial medulla (RVM) neurons or the widespread inhibition and excitation of PAG neurons treated with opioids in in vitro slice experiments. The relatively modest effects of morphine in the present study suggest that morphine produces antinociception by causing small changes in the activity of many PAG neurons.
Collapse
Affiliation(s)
- Valerie L Tryon
- Department of Psychology, University of Washington, Guthrie Hall, Room 119A, UW Box 351525, Seattle, WA 98195, USA.
| | - Sheri J Y Mizumori
- Department of Psychology, University of Washington, Guthrie Hall, Room 119A, UW Box 351525, Seattle, WA 98195, USA.
| | - Michael M Morgan
- Department of Psychology, Washington State University, Vancouver, 14204 NE Salmon Creek Avenue, Vancouver, WA 98686, USA.
| |
Collapse
|
15
|
Biagioni AF, de Oliveira RC, de Oliveira R, da Silva JA, dos Anjos-Garcia T, Roncon CM, Corrado AP, Zangrossi H, Coimbra NC. 5-Hydroxytryptamine 1A receptors in the dorsomedial hypothalamus connected to dorsal raphe nucleus inputs modulate defensive behaviours and mediate innate fear-induced antinociception. Eur Neuropsychopharmacol 2016; 26:532-45. [PMID: 26749090 DOI: 10.1016/j.euroneuro.2015.12.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 12/09/2015] [Accepted: 12/14/2015] [Indexed: 02/04/2023]
Abstract
The dorsal raphe nucleus (DRN) is an important brainstem source of 5-hydroxytryptamine (5-HT), and 5-HT plays a key role in the regulation of panic attacks. The aim of the present study was to determine whether 5-HT1A receptor-containing neurons in the medial hypothalamus (MH) receive neural projections from DRN and to then determine the role of this neural substrate in defensive responses. The neurotracer biotinylated dextran amine (BDA) was iontophoretically microinjected into the DRN, and immunohistochemical approaches were then used to identify 5HT1A receptor-labelled neurons in the MH. Moreover, the effects of pre-treatment of the dorsomedial hypothalamus (DMH) with 8-OH-DPAT and WAY-100635, a 5-HT1A receptor agonist and antagonist, respectively, followed by local microinjections of bicuculline, a GABAA receptor antagonist, were investigated. We found that there are many projections from the DRN to the perifornical lateral hypothalamus (PeFLH) but also to DMH and ventromedial (VMH) nuclei, reaching 5HT1A receptor-labelled perikarya. DMH GABAA receptor blockade elicited defensive responses that were followed by antinociception. DMH treatment with 8-OH-DPAT decreased escape responses, which strongly suggests that the 5-HT1A receptor modulates the defensive responses. However, DMH treatment with WAY-100635 failed to alter bicuculline-induced defensive responses, suggesting that 5-HT exerts a phasic influence on 5-HT1A DMH neurons. The activation of the inhibitory 5-HT1A receptor had no effect on antinociception. However, blockade of the 5-HT1A receptor decreased fear-induced antinociception. The present data suggest that the ascending pathways from the DRN to the DMH modulate panic-like defensive behaviours and mediate antinociceptive phenomenon by recruiting 5-HT1A receptor in the MH.
Collapse
Affiliation(s)
- Audrey Franceschi Biagioni
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo 14049-900, Brazil; Behavioural Neurosciences Institute (INeC), Av. do Café, 2450, Monte Alegre, Ribeirão Preto, São Paulo 14050-220, Brazil
| | - Rithiele Cristina de Oliveira
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo 14049-900, Brazil; Behavioural Neurosciences Institute (INeC), Av. do Café, 2450, Monte Alegre, Ribeirão Preto, São Paulo 14050-220, Brazil
| | - Ricardo de Oliveira
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo 14049-900, Brazil; Behavioural Neurosciences Institute (INeC), Av. do Café, 2450, Monte Alegre, Ribeirão Preto, São Paulo 14050-220, Brazil; Mato Grosso Federal University Medical School (UFMT), Av. Alexandre Ferronato, 1200, Reserva 35, Setor Industrial, 78550-000 Sinop, Mato Grosso, Brazil
| | - Juliana Almeida da Silva
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo 14049-900, Brazil; Behavioural Neurosciences Institute (INeC), Av. do Café, 2450, Monte Alegre, Ribeirão Preto, São Paulo 14050-220, Brazil
| | - Tayllon dos Anjos-Garcia
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo 14049-900, Brazil; Behavioural Neurosciences Institute (INeC), Av. do Café, 2450, Monte Alegre, Ribeirão Preto, São Paulo 14050-220, Brazil
| | - Camila Marroni Roncon
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo 14049-900, Brazil; Behavioural Neurosciences Institute (INeC), Av. do Café, 2450, Monte Alegre, Ribeirão Preto, São Paulo 14050-220, Brazil
| | - Alexandre Pinto Corrado
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Hélio Zangrossi
- Behavioural Neurosciences Institute (INeC), Av. do Café, 2450, Monte Alegre, Ribeirão Preto, São Paulo 14050-220, Brazil; NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), Ribeirão Preto Medical School of the University of São Paulo, Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo 14049-900, Brazil; Laboratory of Neuropsychopharmacology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo, Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Norberto Cysne Coimbra
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo 14049-900, Brazil; Behavioural Neurosciences Institute (INeC), Av. do Café, 2450, Monte Alegre, Ribeirão Preto, São Paulo 14050-220, Brazil; NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), Ribeirão Preto Medical School of the University of São Paulo, Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo 14049-900, Brazil.
| |
Collapse
|
16
|
Zhang H, Wu L, Liu Y, Peng S, Wang W. The antinociceptive effect and mechanism of action of SY0916. Int Immunopharmacol 2016; 32:16-23. [PMID: 26780232 DOI: 10.1016/j.intimp.2016.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 12/16/2015] [Accepted: 01/04/2016] [Indexed: 12/16/2022]
Abstract
Pain greatly affects the quality of life of people worldwide. Despite their demonstrated efficacy, currently used opioid drugs and nonsteroidal anti-inflammatory drugs (NSAIDs) are frequently associated with several adverse events. The identification of new therapeutic targets and the development of corresponding analgesics may represent novel approaches for effectively treating pain. SY0916 is a novel compound that was designed and synthesized by the Institute of Materia Medica, Chinese Academy of Medical Sciences. As demonstrated by the hot plate test, tail-flick test and the formalin test, SY0916 exerted strong peripheral and central antinociceptive effects. Western blot, immunohistochemistry and enzyme-linked immunosorbent assay (ELISA) results indicate that SY0916 induces its peripheral antinociceptive effect by suppressing the peripheral activity of inflammatory mediators such as prostaglandin E2 (PGE2), tumor necrosis factor-alpha (TNF-α) and 5-hydroxytryptamine (5-HT). Moreover, its central antinociceptive effect might be mediated by the down-regulation of PGE2 and TNF-α expression and the inhibition of p-p38 and NF-κB pathway signaling in glial cells. These findings demonstrate that SY0916 may serve as a promising analgesic candidate drug.
Collapse
Affiliation(s)
- Haijing Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lianqiu Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yang Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shanying Peng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wenjie Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
17
|
Abstract
This paper is the thirty-seventh consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2014 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (endogenous opioids and receptors), and the roles of these opioid peptides and receptors in pain and analgesia (pain and analgesia); stress and social status (human studies); tolerance and dependence (opioid mediation of other analgesic responses); learning and memory (stress and social status); eating and drinking (stress-induced analgesia); alcohol and drugs of abuse (emotional responses in opioid-mediated behaviors); sexual activity and hormones, pregnancy, development and endocrinology (opioid involvement in stress response regulation); mental illness and mood (tolerance and dependence); seizures and neurologic disorders (learning and memory); electrical-related activity and neurophysiology (opiates and conditioned place preferences (CPP)); general activity and locomotion (eating and drinking); gastrointestinal, renal and hepatic functions (alcohol and drugs of abuse); cardiovascular responses (opiates and ethanol); respiration and thermoregulation (opiates and THC); and immunological responses (opiates and stimulants). This paper is the thirty-seventh consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2014 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (endogenous opioids and receptors), and the roles of these opioid peptides and receptors in pain and analgesia (pain and analgesia); stress and social status (human studies); tolerance and dependence (opioid mediation of other analgesic responses); learning and memory (stress and social status); eating and drinking (stress-induced analgesia); alcohol and drugs of abuse (emotional responses in opioid-mediated behaviors); sexual activity and hormones, pregnancy, development and endocrinology (opioid involvement in stress response regulation); mental illness and mood (tolerance and dependence); seizures and neurologic disorders (learning and memory); electrical-related activity and neurophysiology (opiates and conditioned place preferences (CPP)); general activity and locomotion (eating and drinking); gastrointestinal, renal and hepatic functions (alcohol and drugs of abuse); cardiovascular responses (opiates and ethanol); respiration and thermoregulation (opiates and THC); and immunological responses (opiates and stimulants).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
18
|
The role of nicotinic acetylcholine and opioid systems of the ventral orbital cortex in modulation of formalin-induced orofacial pain in rats. Eur J Pharmacol 2015; 758:147-52. [DOI: 10.1016/j.ejphar.2015.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 03/30/2015] [Accepted: 04/02/2015] [Indexed: 12/16/2022]
|
19
|
Jokinen V, Lilius T, Laitila J, Niemi M, Rauhala P, Kalso E. Pregabalin enhances the antinociceptive effect of oxycodone and morphine in thermal models of nociception in the rat without any pharmacokinetic interactions. Eur J Pain 2015; 20:297-306. [DOI: 10.1002/ejp.728] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2015] [Indexed: 12/26/2022]
Affiliation(s)
- V. Jokinen
- Department of Pharmacology; Faculty of Medicine; University of Helsinki; Finland
| | - T.O. Lilius
- Department of Pharmacology; Faculty of Medicine; University of Helsinki; Finland
| | - J. Laitila
- Department of Clinical Pharmacology; Faculty of Medicine; University of Helsinki; Finland
| | - M. Niemi
- Department of Clinical Pharmacology; Faculty of Medicine; University of Helsinki; Finland
- HUSLAB; Helsinki University Central Hospital; Finland
| | - P.V. Rauhala
- Department of Pharmacology; Faculty of Medicine; University of Helsinki; Finland
| | - E.A. Kalso
- Department of Pharmacology; Faculty of Medicine; University of Helsinki; Finland
- Department of Anaesthesia and Intensive Care Medicine; Pain Clinic; Helsinki University Central Hospital; Finland
| |
Collapse
|