1
|
Hohenwarter L, Puil E, Rouhollahi E, Bohrmann L, Lu S, Saatchi K, Häfeli UO, Barr A, Böttger R, Viswanadham KKD, Li SD. A Novel Leu-Enkephalin Prodrug Produces Pain-Relieving and Antidepressant Effects. Mol Pharm 2024; 21:688-703. [PMID: 38243899 DOI: 10.1021/acs.molpharmaceut.3c00807] [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] [Indexed: 01/22/2024]
Abstract
Persistent pain is a significant healthcare problem with limited treatment options. The high incidence of comorbid chronic pain and depression significantly reduces life quality and complicates the treatment of both conditions. Antidepressants are less effective for pain and depression than for depression alone and they induce severe side effects. Opioids are highly efficacious analgesics, but rapid development of tolerance, dependence, and debilitating side effects limit their efficacy and safe use. Leucine-enkephalin (Leu-ENK), the endogenous delta opioid receptor agonist, controls pain and mood and produces potent analgesia with reduced adverse effects compared to conventional opioids. High proteolytic instability, however, makes Leu-ENK ineffective after systemic administration and limits its clinical usefulness. KK-103, a Leu-ENK prodrug, was developed to overcome these limitations of Leu-ENK via markedly increased plasma stability in mice. We showed rapid and substantially increased systemic adsorption and blood plasma exposure of KK-103 compared to Leu-ENK. We also observed brain uptake of radiolabeled KK-103 after systemic administration, indicating a central effect of KK-103. We then established KK-103's prolonged antinociceptive efficacy in the ramped hot plate and formalin test. In both models, KK-103 produced a comparable dose to the maximum antinociceptive-effect relationship. The pain-alleviating effect of KK-103 primarily resulted from activating the delta opioid receptor after the likely conversion of KK-103 to Leu-ENK in vivo. Finally, KK-103 produced an antidepressant-like activity comparable to the antidepressant desipramine, but with minimal gastrointestinal inhibition and no incidence of sedation.
Collapse
Affiliation(s)
- Lukas Hohenwarter
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Ernest Puil
- Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Elham Rouhollahi
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Lennart Bohrmann
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Shawna Lu
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Katayoun Saatchi
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Urs O Häfeli
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Alasdair Barr
- Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Roland Böttger
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - K K DurgaRao Viswanadham
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|
2
|
Ramachandra V, Kang F, Kim C, Nova AS, Bajaj A, Hall FS, Uhl GR, Gonzales RA. The μ opioid receptor is not involved in ethanol-stimulated dopamine release in the ventral striatum of C57BL/6J mice. Alcohol Clin Exp Res 2011; 35:929-38. [PMID: 21294756 DOI: 10.1111/j.1530-0277.2010.01423.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND The mu opioid receptor (MOR) has previously been found to regulate ethanol-stimulated dopamine release under some, but not all, conditions. A difference in ethanol-evoked dopamine release between male and female mixed background C57BL/6J-129SvEv mice led to questions about its ubiquitous role in these effects of ethanol. Using congenic C57BL/6J MOR knockout (KO) mice and C57BL/6J mice pretreated with an irreversible MOR antagonist, we investigated the function of this receptor in ethanol-stimulated dopamine release. METHODS Microdialysis was used to monitor dopamine release and ethanol clearance in MOR -/-, +/+, and +/- . male and female mice after intraperitoneal (i.p.) injections of 1.0, 2.0, and 3.0 g/kg ethanol (or saline). We also measured the increase in dopamine release after 5 mg/kg morphine (i.p.) in male and female MOR+/+ and -/- mice. In a separate experiment, male C57BL/6J mice were pretreated with either the irreversible MOR antagonist beta funaltrexamine (BFNA) or vehicle, and dopamine levels were monitored after administration of 2 g/kg ethanol or 5 mg/kg morphine. RESULTS Although ethanol-stimulated dopamine release at all the 3 doses of alcohol tested, there were no differences between MOR+/+, -/-, and +/- mice in these effects. Female mice had a more prolonged effect compared to males at the 1 g/kg dose. Administration of 2 g/kg ethanol also caused a similar increase in dopamine levels in both saline-pretreated and BFNA-pretreated mice. Five mg/kg morphine caused a significant increase in dopamine levels in MOR+/+ mice but not in MOR-/- mice and in saline-pretreated mice but not in BFNA-pretreated mice. Intraperitoneal saline injections had a significant, albeit small and transient, effect on dopamine release when given in a volume equivalent to the ethanol doses, but not in a volume equivalent to the 5 mg/kg morphine dose. Ethanol pharmacokinetics were similar in all genotypes and both sexes at each dose and in both pretreatment groups. CONCLUSIONS MOR is not involved in ethanol-stimulated dopamine release in the ventral striatum of C57BL/6J mice.
Collapse
|
3
|
Bedini A, Baiula M, Gentilucci L, Tolomelli A, De Marco R, Spampinato S. Peripheral antinociceptive effects of the cyclic endomorphin-1 analog c[YpwFG] in a mouse visceral pain model. Peptides 2010; 31:2135-40. [PMID: 20713109 DOI: 10.1016/j.peptides.2010.08.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Revised: 08/02/2010] [Accepted: 08/02/2010] [Indexed: 11/29/2022]
Abstract
We previously described a novel cyclic endomorphin-1 analog c[Tyr-D-Pro-D-Trp-Phe-Gly] (c[YpwFG]), acting as a mu-opioid receptor (MOR) agonist. This study reports that c[YpwFG] is more lipophilic and resistant to enzymatic hydrolysis than endomorphin-1 and produces preemptive antinociception in a mouse visceral pain model when injected intraperitoneally (i.p.) or subcutaneously (s.c.) before 0.6% acetic acid, employed to evoke abdominal writhing (i.p. ED(50)=1.24 mg/kg; s.c. ED(50)=2.13 mg/kg). This effect is reversed by the selective MOR antagonist β-funaltrexamine and by a high dose of the mu(1)-opioid receptor-selective antagonist naloxonazine. Conversely, the kappa-opioid receptor antagonist nor-binaltorphimine and the delta-opioid receptor antagonist naltrindole are ineffective. c[YpwFG] produces antinociception when injected i.p. after acetic acid (ED(50)=4.80 mg/kg), and only at a dose of 20mg/kg did it elicit a moderate antinociceptive response in the mouse, evaluated by the tail flick assay. Administration of a lower dose of c[YpwFG] (10mg/kg i.p.) apparently produces a considerable part of antinociception on acetic acid-induced writhes through peripheral opioid receptors as this action is fully prevented by i.p. naloxone methiodide, which does not readily cross the blood-brain barrier; whereas this opioid antagonist injected intracerebroventricularly (i.c.v.) is not effective. Antinociception produced by a higher dose of c[YpwFG] (20mg/kg i.p.) is partially reversed by naloxone methiodide i.c.v. administered. Thus, only at the dose of 20mg/kg c[YpwFG] can produce antinociception through both peripheral and central opioid receptors. In conclusion, c[YpwFG] displays sufficient metabolic stability to be effective after peripheral administration and demonstrates the therapeutic potential of endomorphin derivatives as novel analgesic agents to control visceral pain.
Collapse
Affiliation(s)
- Andrea Bedini
- Department of Pharmacology, University of Bologna, Irnerio 48, 4126 Bologna, Italy
| | | | | | | | | | | |
Collapse
|
4
|
Chevillard L, Mégarbane B, Baud FJ, Risède P, Declèves X, Mager D, Milan N, Ricordel I. Mechanisms of respiratory insufficiency induced by methadone overdose in rats. Addict Biol 2010; 15:62-80. [PMID: 20002023 DOI: 10.1111/j.1369-1600.2009.00184.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Methadone may cause respiratory depression. We aimed to understand methadone-related effects on ventilation as well as each opioid-receptor (OR) role. We studied the respiratory effects of intraperitoneal methadone at 1.5, 5, and 15 mg/kg (corresponding to 80% of the lethal dose-50%) in rats using arterial blood gases and plethysmography. OR antagonists, including intravenous 10 mg/kg-naloxonazine at 5 minutes (mu-OR antagonist), subcutaneous 30 mg/kg-naloxonazine at 24 hours (micro1-OR antagonist), 3 mg/kg-naltrindole at 45 minutes (delta-OR antagonist) and 5 mg/kg-Nor-binaltorphimine at 6 hours (kappa-OR antagonist) were pre-administered. Plasma concentrations of methadone enantiomers were measured using high-performance liquid chromatography coupled to mass-spectrometry. Methadone dose-dependent inspiratory time (T(I)) increase tended to be linear. Respiratory depression was observed only at 15 mg/kg and characterized by an increase in expiratory time (T(E)) resulting in hypoxemia and respiratory acidosis. Intravenous naloxonazine completely reversed all methadone-related effects on ventilation, while subcutaneous naloxonazine reduced its effects on pH (P < 0.05), PaCO(2) (P < 0.01) and T(E) (P < 0.001) but only partially on T(I) (P < 0.001). Naltrindole reduced methadone-related effects on T(E) (P < 0.001). Nor-binaltorphimine increased methadone-related effects on pH and PaO(2) (P < 0.05) Respiratory effects as a function of plasma R-methadone concentrations showed a decrease in PaO(2) (EC(50): 1.14 microg/ml) at lower concentrations than those necessary for PaCO(2) increase (EC(50): 3.35 microg/ml). Similarly, increased T(I) (EC(50): 0.501 microg/ml) was obtained at lower concentrations than those for T(E) (EC(50): 4.83 microg/ml). Methadone-induced hypoxemia is caused by mu-ORs and modulated by kappa-ORs. Additionally, methadone-induced increase in T(E) is caused by mu1- and delta-opioid receptors while increase in T(I) is caused by mu-ORs.
Collapse
MESH Headings
- Acidosis, Respiratory/chemically induced
- Acidosis, Respiratory/physiopathology
- Animals
- Dose-Response Relationship, Drug
- Drug Overdose/physiopathology
- Exhalation/drug effects
- Exhalation/physiology
- Hypoxia/chemically induced
- Hypoxia/physiopathology
- Injections, Intraperitoneal
- Injections, Intravenous
- Injections, Subcutaneous
- Male
- Methadone/pharmacokinetics
- Methadone/toxicity
- Narcotic Antagonists/pharmacology
- Narcotics/pharmacokinetics
- Narcotics/toxicity
- Oxygen/blood
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid/drug effects
- Receptors, Opioid/physiology
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/physiology
- Receptors, Opioid, kappa/drug effects
- Receptors, Opioid, kappa/physiology
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/physiology
- Respiratory Insufficiency/physiopathology
Collapse
Affiliation(s)
- Lucie Chevillard
- Université Paris-Descartes, Faculté de Pharmacie, Neuropsychopharmacologie des addictions, CNRS, UMR 7157, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Chevillard L, Mégarbane B, Risède P, Baud FJ. Characteristics and comparative severity of respiratory response to toxic doses of fentanyl, methadone, morphine, and buprenorphine in rats. Toxicol Lett 2009; 191:327-40. [PMID: 19819313 DOI: 10.1016/j.toxlet.2009.09.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Revised: 09/29/2009] [Accepted: 09/30/2009] [Indexed: 11/16/2022]
Abstract
Opioids are known to induce respiratory depression. We aimed to characterize in rats the effects of four opioids on arterial blood gases and plethysmography after intraperitoneal administration at 80% of their LD(50) in order to identify opioid molecule-specific patterns and classify response severity. Opioid-receptor (OR) antagonists, including intravenous 10 mg kg(-1)-naloxonazine at 5 min [mu-OR antagonist], subcutaneous 30 mg kg(-1)-naloxonazine at 24 h [mu1-OR antagonist], subcutaneous 3 mg kg(-1)-naltrindole at 45 min [delta-OR antagonist], and subcutaneous 5 mg kg(-1)-Nor-binaltorphimine at 6 h [kappa-OR antagonist] were pre-administered to test the role of each OR. Methadone, morphine, and fentanyl significantly decreased PaO(2) (P<0.001) and increased PaCO(2) (P<0.05), while buprenorphine only decreased PaO(2) (P<0.05). While all opioids significantly increased inspiratory time (T(I), P<0.001), methadone and fentanyl also increased expiratory time (T(E), P<0.05). Intravenous 10 mg kg(-1)-naloxonazine at 5 min completely reversed opioid-related effects on PaO(2) (P<0.05), PaCO(2) (P<0.001), T(I) (P<0.05), and T(E) (P<0.01) except in buprenorphine. Subcutaneous 30 mg kg(-1)-naloxonazine at 24 h completely reversed effects on PaCO(2) (P<0.01) and T(E) (P<0.001), partially reversed effects on T(I) (P<0.001), and did not reverse effects on PaO(2). Naltrindole reversed methadone-induced T(E) increases (P<0.01) but worsened fentanyl's effect on PaCO(2) (P<0.05) and T(I) (P<0.05). Nor-binaltorphimine reversed morphine- and buprenorphine-induced T(I) increases (P<0.001) but worsened methadone's effect on PaO(2) (P<0.05) and morphine (P<0.001) and buprenorphine's (P<0.01) effects on pH. In conclusion, opioid-related respiratory patterns are not uniform. Opioid-induced hypoxemia as well as increases in T(I) and T(E) are caused by mu-OR, while delta and kappa-OR roles appear limited, depending on the specific opioid. Regarding severity of opioid-induced respiratory effects at 80% of their LD(50), all drugs increased T(I). Methadone and fentanyl induced hypoxemia, hypercapnia, and T(E) increases, morphine caused both hypoxemia and hypercapnia while buprenorphine caused only hypoxemia.
Collapse
Affiliation(s)
- Lucie Chevillard
- Université Paris-Descartes, Faculté de Pharmacie, Neuropsychopharmacologie des addictions, CNRS, UMR7157, 75010 Paris, France
| | | | | | | |
Collapse
|
6
|
Sakurada S, Sawai T, Mizoguchi H, Watanabe H, Watanabe C, Yonezawa A, Morimoto M, Sato T, Komatsu T, Sakurada T. Possible involvement of dynorphin A release via mu1-opioid receptor on supraspinal antinociception of endomorphin-2. Peptides 2008; 29:1554-60. [PMID: 18571771 DOI: 10.1016/j.peptides.2008.04.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2008] [Revised: 04/17/2008] [Accepted: 04/18/2008] [Indexed: 11/26/2022]
Abstract
It has been demonstrated that the antinociception induced by i.t. or i.c.v. administration of endomorphins is mediated through mu-opioid receptors. Moreover, though endomorphins do not have appreciable affinity for kappa-opioid receptors, pretreatment with the kappa-opioid receptor antagonist nor-binaltorphimine markedly blocks the antinociception induced by i.c.v.- or i.t.-injected endomorphin-2, but not endomorphin-1. These evidences propose the hypothesis that endomorphin-2 may initially stimulate the mu-opioid receptors, which subsequently induces the release of dynorphins acting on kappa-opioid receptors to produce antinociception. The present study was performed to determine whether the release of dynorphins by i.c.v.-administered endomorphin-2 is mediated through mu-opioid receptors for producing antinociception. Intracerebroventricular pretreatment with an antiserum against dynorphin A, but not dynorphin B or alpha-neo-endorphin, and s.c. pretreatment with kappa-opioid receptor antagonist nor-binaltorphimine dose-dependently attenuated the antinociception induced by i.c.v.-administered endomorphin-2, but not endomorphin-1 and DAMGO. The attenuation of endomorphin-2-induced antinociception by pretreatment with antiserum against dynorphin A or nor-binaltorphimine was dose-dependently eliminated by additional s.c. pretreatment with a selective mu-opioid receptor antagonist beta-funaltrexamine or a selective mu1-opioid receptor antagonist naloxonazine at ultra low doses, which are inactive against micro-opioid receptor agonists in antinociception, suggesting that endomorphin-2 stimulates distinct subclass of micro1-opioid receptor that induces the release of dynorphin A acting on kappa-opioid receptors in the brain. It concludes that the antinociception induced by supraspinally administered endomorphin-2 is in part mediated through the release of endogenous kappa-opioid peptide dynorphin A, which is caused by the stimulation of distinct subclass of micro1-opioid receptor.
Collapse
Affiliation(s)
- Shinobu Sakurada
- Department of Physiology and Anatomy, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Sendai 981-8558, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Meyer PJ, Fossum EN, Ingram SL, Morgan MM. Analgesic tolerance to microinjection of the micro-opioid agonist DAMGO into the ventrolateral periaqueductal gray. Neuropharmacology 2007; 52:1580-5. [PMID: 17445843 PMCID: PMC1971241 DOI: 10.1016/j.neuropharm.2007.03.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Revised: 02/22/2007] [Accepted: 03/01/2007] [Indexed: 11/22/2022]
Abstract
Repeated administration of the relatively low-efficacy micro-opioid receptor agonist morphine induces tolerance to its antinociceptive effects. High-efficacy agonists such as D-Ala2NMePhe4,Gly-ol5 (DAMGO) have been shown to be less effective at producing tolerance, suggesting that different neural mechanisms underlie tolerance to these agonists. However, the correlation between agonist efficacy and tolerance development has not been examined within the ventrolateral periaqueductal gray (vPAG), a brain area known to be crucial for the development of morphine tolerance. The current studies examined whether tolerance to DAMGO occurs within the vPAG, and whether repeated treatment with DAMGO into the vPAG alters the development of morphine tolerance. The results showed that repeated vPAG microinjections of DAMGO induced robust tolerance and cross-tolerance to morphine. Further, co-administration of a low dose of DAMGO with morphine potentiated morphine tolerance. These findings indicate that similar mechanisms underlie tolerance to morphine and DAMGO within the vPAG.
Collapse
Affiliation(s)
- Paul J Meyer
- Washington State University Vancouver, 14204 NE Salmon Creek Ave, Vancouver, WA 98660, USA
| | | | | | | |
Collapse
|
8
|
Primeaux SD, Wilson SP, McDonald AJ, Mascagni F, Wilson MA. The role of delta opioid receptors in the anxiolytic actions of benzodiazepines. Pharmacol Biochem Behav 2006; 85:545-54. [PMID: 17109943 PMCID: PMC1892843 DOI: 10.1016/j.pbb.2006.09.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2006] [Revised: 09/12/2006] [Accepted: 09/26/2006] [Indexed: 11/22/2022]
Abstract
The anxiolytic effects of benzodiazepines appear to involve opioid processes in the amygdala. In previous experiments, overexpression of enkephalin in the amygdala enhanced the anxiolytic actions of the benzodiazepine agonist diazepam in the elevated plus maze. The effects of systemically administered diazepam are also blocked by injections of naltrexone into the central nucleus of the amygdala. The current studies investigated the role of delta opioid receptors in the anxiety-related effects of diazepam. Three days following bilateral stereotaxic injections of viral vectors containing cDNA encoding proenkephalin or beta-galactosidase (control vector), the delta opioid receptor antagonist naltrindole (10 mg/kg, s.c.) attenuated the enhanced anxiolytic effects of 1-2 mg/kg diazepam in rats overexpressing preproenkephalin in the amygdala. Despite this effect, naltrindole failed to attenuate the anxiolytic action of higher diazepam doses (3 mg/kg) in animals with normal amygdalar enkephalin expression. Similarly, the mu opioid receptor antagonist, beta-funaltrexamine (20 mg/kg, s.c.), had no effect on the anxiolytic effect of diazepam alone. These data support a role for delta opioid receptors in the opioid-enhanced anxiolytic effects of diazepam.
Collapse
Affiliation(s)
- Stefany D Primeaux
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Bldg 1, Columbia, SC 29208, USA
| | | | | | | | | |
Collapse
|
9
|
Mizoguchi H, Watanabe H, Hayashi T, Sakurada W, Sawai T, Fujimura T, Sakurada T, Sakurada S. Possible involvement of dynorphin A-(1-17) release via mu1-opioid receptors in spinal antinociception by endomorphin-2. J Pharmacol Exp Ther 2006; 317:362-8. [PMID: 16394196 DOI: 10.1124/jpet.105.098293] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The antinociception induced by i.t. or i.c.v. administration of endomorphins is mediated via mu-opioid receptors. However, although endomorphins do not have an appreciable affinity for kappa-opioid receptors, pretreatment with the kappa-opioid receptor antagonist norbinaltorphimine markedly reduces the antinociceptive response to i.c.v. or i.t. administered endomorphin-2 but not endomorphin-1. These results suggest that endomorphin-2 initially stimulates mu-opioid receptors, which subsequently induce the release of dynorphins that act on kappa-opioid receptors to produce antinociception. The present study was performed in mice to determine whether the release of dynorphins by i.t. administered endomorphin-2 is mediated through mu-opioid receptors to produce antinociception. Intrathecal pretreatment with an antiserum against dynorphin A-(1-17), but not against dynorphin B-(1-13) or alpha-neoendorphin, dose-dependently prevented the paw-withdrawal inhibition by endomorphin-2. The pretreatments with these antisera did not affect the endomorphin-1- or [D-Ala(2),MePhe(4),Gly(ol)(5)]enkephalin-induced paw-withdrawal inhibition. The attenuation of endomorphin-2-induced antinociception by i.t. pretreatment with an antiserum against dynorphin A-(1-17) or s.c. pretreatment with norbinaltorphimine was blocked dose-dependently by s.c. pretreatment with the mu-opioid receptor antagonist beta-funaltrexamine or the mu(1)-opioid receptor antagonist naloxonazine at ultra-low doses that are ineffective against mu-opioid receptor agonists. These results suggest that the spinal antinociception induced by endomorphin-2 is mediated through the stimulation of a distinct subtype of mu(1)-opioid receptor that induces the release of the endogenous kappa-opioid peptide dynorphin A-(1-17) in the spinal cord.
Collapse
Affiliation(s)
- Hirokazu Mizoguchi
- Department of Physiology and Anatomy, Tohoku Pharmaceutical University, Sendai, Japan
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Leng DN, Feng YP, Li YQ. Endomorphin-1 is more potent than endomorphin-2 in inhibition of synaptic transmission in substantia gelatinosa of adult rat spinal cord. Int J Neurosci 2006; 115:1515-26. [PMID: 16223698 DOI: 10.1080/00207450590957827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Effects of endomorphin-1 (EM-1) and endomorphin-2 (EM-2) on synaptic trans-mission were investigated on neurons in substantia gelatinosa (SG) of the spinal dorsal horn. Both EM-1 (1 microM) and EM-2 (1 microM) remarkably reduced the frequency but not the amplitude of miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs). These effects were antagonized by beta-funaltrexamine (beta-FNA, 10 microM), a selective mu-opioid receptor antagonist. Noticeably, EM-1 showed higher potency in decreasing the frequency of mEPSCs and mIPSCs than that of EM-2. These results indicate that EMs suppress both excitatory and inhibitory synaptic transmission by activating presynaptic mu-opioid receptors in the SG and EM-1, compared with EM-2, might be a more potent endogenous analgesic at the spinal cord level.
Collapse
Affiliation(s)
- Dong-Ni Leng
- Department of Anatomy and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, PR China
| | | | | |
Collapse
|
11
|
Raehal KM, Walker JKL, Bohn LM. Morphine side effects in beta-arrestin 2 knockout mice. J Pharmacol Exp Ther 2005; 314:1195-201. [PMID: 15917400 DOI: 10.1124/jpet.105.087254] [Citation(s) in RCA: 482] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Morphine is a potent analgesic, yet, like most opioid narcotics, it exerts unwanted side effects such as constipation and respiratory suppression, thereby limiting its clinical utility. Pharmacological approaches taken to preserve the analgesic properties, while eliminating the unwanted side effects, have met with very limited success. Here, we provide evidence that altering mu opioid receptor regulation may provide a novel approach to discriminate morphine's beneficial and deleterious effects in vivo. We have previously reported that mice lacking the G protein-coupled receptor regulatory protein, beta-arrestin 2, display profoundly altered morphine responses. beta-Arrestin 2 knockout mice have enhanced and prolonged morphine analgesia with very little morphine tolerance. In this report, we examine whether the side effects of morphine treatment are also augmented in this animal model. Surprisingly, the genetic disruption of opioid receptor regulation, while enhancing and prolonging analgesia, dramatically attenuates the respiratory suppression and acute constipation caused by morphine.
Collapse
Affiliation(s)
- Kirsten M Raehal
- Department of Pharmacology and Psychiatry, Ohio State University College of Medicine, Columbus, 43210-1239, USA
| | | | | |
Collapse
|
12
|
Temsamani J, Bonnafous C, Rousselle C, Fraisse Y, Clair P, Granier LA, Rees AR, Kaczorek M, Scherrmann JM. Improved brain uptake and pharmacological activity profile of morphine-6-glucuronide using a peptide vector-mediated strategy. J Pharmacol Exp Ther 2005; 313:712-9. [PMID: 15647327 DOI: 10.1124/jpet.104.081000] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Morphine-6-glucuronide (M6G), an active metabolite of morphine, has been shown to have significantly attenuated brain penetration relative to that of morphine. Recently, we have demonstrated that conjugation of various drugs to peptide vectors significantly enhances their brain uptake. In this study, we have conjugated morphine-6-glucuronide to a peptide vector SynB3 to enhance its brain uptake and its analgesic potency after systemic administration. We show by in situ brain perfusion that vectorization of M6G (Syn1001) markedly enhances the brain uptake of M6G. This enhancement results in a significant improvement in the pharmacological activity of M6G in several models of nociception. Syn1001 was about 4 times more potent than free M6G (ED(50) of 1.87 versus 8.74 micromol/kg). Syn1001 showed also a prolonged duration of action compared with free M6G (300 and 120 min, respectively). Furthermore, the conjugation of M6G results in a lowered respiratory depression, as measured in a rat model. Taken together, these data strongly support the utility of peptide-mediated strategies for improving the efficacy of drugs such as M6G for the treatment of pain.
Collapse
|
13
|
Berrocoso E, Rojas-Corrales MO, Micó JA. Non-selective opioid receptor antagonism of the antidepressant-like effect of venlafaxine in the forced swimming test in mice. Neurosci Lett 2004; 363:25-8. [PMID: 15157989 DOI: 10.1016/j.neulet.2004.03.041] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2003] [Revised: 03/06/2004] [Accepted: 03/06/2004] [Indexed: 11/26/2022]
Abstract
The opioid system has been implicated in mood disorders as well as in the mechanism of action of antidepressants. Since the opioid component in venlafaxine (VLX) is still a matter of discussion, we investigated the role of opioid receptors in the antidepressant-like effect of VLX in the forced swimming test in mice. The non-selective opiate antagonist naloxone at high dose (2 mg/kg, s.c.) antagonized the effect of VLX. In contrast, beta-funaltrexamine (40 mg/kg, s.c.), which preferentially blocks mu(1)/mu(2) opioid receptors, naloxonazine (35 mg/kg, s.c.), a selective mu(1) opioid antagonist, naltrindole (10 mg/kg, s.c.), a selective delta opioid antagonist, and Nor-binaltorphimine (10 mg/kg, s.c.), which selectively blocks kappa-opioid receptors, were all ineffective. Thus, although apparently mediated by the opioid system, the behavioural effect of VLX does not involve specific opioid receptors.
Collapse
Affiliation(s)
- Esther Berrocoso
- Pharmacology and Neuroscience Research Group (PAI-510), Department of Neuroscience (Pharmacology and Psychiatry), Faculty of Medicine, University of Cádiz, Plaza Falla, 9, 11003 Cadiz, Spain
| | | | | |
Collapse
|
14
|
Wu HE, Sun HS, Darpolar M, Leitermann RJ, Kampine JP, Tseng LF. Dynorphinergic mechanism mediating endomorphin-2-induced antianalgesia in the mouse spinal cord. J Pharmacol Exp Ther 2003; 307:1135-41. [PMID: 14557378 DOI: 10.1124/jpet.103.056242] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have previously demonstrated that both endomorphin-1 (EM-1) and endomorphin-2 (EM-2) at high doses (1.75-35 nmol) given intrathecally (i.t.) or intracerebroventricularly produce antinociception by stimulation of mu-opioid receptors. Now, we report that EM-2 at small doses (0.05-1.75 nmol), which injected alone did not produce antinociception, produces anti-analgesia against opioid agonist-induced antinociception. The tail-flick (TF) response was used to test the antinociception in male CD-1 mice. Intrathecal pretreatment with EM-2 (0.02-1.75 nmol) 45 min before i.t. morphine (3.0 nmol) injection dose dependently attenuated morphine-induced TF inhibition. On the other hand, a similar dose of EM-1 (1.64 nmol) failed to produce any antianalgesic effect. The EM-2 (1.75 nmol)-produced anti-analgesia against morphine-induced TF inhibition was blocked by i.t. pretreatment with the mu-opioid antagonist naloxone or 3-methoxynaltrexone, but not delta-opioid receptor antagonist naltrindole, kappa-opioid receptor antagonist nor-binaltorphimine, or N-methyl-d-aspartate (NMDA) receptor antagonist MK-801. The EM-2-induced antianalgesic effect against morphine-induced TF inhibition was blocked by i.t. pretreatment with antiserum against dynorphin A(1-17), but not beta-endorphin, [Met]-enkephalin, [Leu]-enkephalin, or cholecystokinin antiserum (200 microg each). The i.t. EM-2 pretreatment also attenuated the TF inhibition induced by other mu-opioid agonists, [d-Ala2,N-Me-Phe4,Gly-ol5]-enkephalin, EM-1 and EM-2, delta-opioid agonist deltorphin II, and kappa-opioid agonist (trans)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)-cyclohexyl]benzeneacetamide methane-sulfonate hydrate (U50,488H). It is concluded that EM-2 at subanalgesic doses presumably stimulates a subtype of mu-opioid receptor and subsequently induces the release of dynorphin A(1-17) to produce antianalgesic effects against mu-, delta-, or kappa-agonists-induced antinociception. The EM-2-induced antianalgesia is not mediated by the release of [Met]-enkephalin, [Leu]-enkephalin, beta-endorphin, or cholecystokinin, nor does it involve kappa- or delta-opioid or NMDA receptors in the spinal cord.
Collapse
Affiliation(s)
- Hsiang-En Wu
- Department of Anesthesiology, Medical College of Wisconsin, Medical Education Building, Room M4308, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | | | | | | | | | | |
Collapse
|
15
|
Masocha W, Horvath G, Agil A, Ocana M, Del Pozo E, Szikszay M, Baeyens JM. Role of Na(+), K(+)-ATPase in morphine-induced antinociception. J Pharmacol Exp Ther 2003; 306:1122-8. [PMID: 12756273 DOI: 10.1124/jpet.103.052977] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We evaluated the modulation by Na+,K+-ATPase inhibitors of morphine-induced antinociception in the tail-flick test and [3H]naloxone binding to forebrain membranes. The antinociception induced by morphine (1-32 mg/kg, s.c.) in mice was dose-dependently antagonized by ouabain (1-10 ng/mouse, i.c.v.), which produced a significant shift to the right of the morphine dose-response curve. The i.c.v. administration of three Na+,K+-ATPase inhibitors (ouabain at 0.1-100, digoxin at 1-1000, and digitoxin at 10-10000 ng/mouse) dose-dependently antagonized the antinociceptive effect of morphine (4 mg/kg, s.c.) in mice, with the following order of potency: ouabain > digoxin > digitoxin. This effect cannot be explained by any interaction at opioid receptors, since none of these Na+,K+-ATPase inhibitors displaced [3H]naloxone from its binding sites, whereas naloxone did so in a concentration-dependent manner. The antinociception induced by morphine (5 mg/kg, s.c.) in rats was antagonized by the i.c.v. administration of ouabain at 10 ng/rat, whereas it was not significantly modified by intrathecally administered ouabain (10 and 100 ng/rat). These results suggest that the activation of Na+,K+-ATPase plays a role in the supraspinal, but not spinal, antinociceptive effect of morphine.
Collapse
Affiliation(s)
- Willias Masocha
- Department of Pharmacology and Neuroscience Institute, Faculty of Medicine, University of Granada, Granada, Spain
| | | | | | | | | | | | | |
Collapse
|
16
|
Okada Y, Tsuda Y, Bryant SD, Lazarus LH. Endomorphins and related opioid peptides. VITAMINS AND HORMONES 2003; 65:257-79. [PMID: 12481550 DOI: 10.1016/s0083-6729(02)65067-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Abstract
Opioid peptides and their G-protein-coupled receptors (delta, kappa, mu) are located in the central nervous system and peripheral tissues. The opioid system has been studied to determine the intrinsic mechanism of modulation of pain and to develop uniquely effective pain-control substances with minimal abuse potential and side effects. Two types of endogenous opioid peptides exist, one containing Try-Gly-Gly-Phe as the message domain (enkephalins, endorphins, dynorphins) and the other containing the Tyr-Pro-Phe/Trp sequence (endomorphins-1 and -2). Endomorphin-1 (Tyr-Pro-Trp-Phe-NH2), which has high mu receptor affinity (Ki = 0.36 nM) and remarkable selectivity (4000- and 15,000-fold preference over the delta and kappa receptors, respectively), was isolated from bovine and human brain. In addition, endomorphin-2 (Tyr-Pro-Phe-Phe-NH2), isolated from the same sources, exhibited high mu receptor affinity (Ki = 0.69 nM) and very high selectivity (13,000- and 7500-fold preference relative to delta and kappa receptors, respectively). Both opioids bind to mu-opioid receptors, thereby activating G-proteins, resulting in regulation of gastrointestinal motility, manifestation of antinociception, and effects on the vascular systems and memory. To develop novel analgesics with less addictive properties, evaluation of the structure-activity relationships of the endomorphins led to the design of more potent and stable analgesics. Opioidmimetics and opioid peptides containing the amino acid sequence of the message domain of endomorphins, Tyr-Pro-Phe/Trp, could exhibit unique binding activity and lead to the development of new therapeutic drugs for controlling pain.
Collapse
Affiliation(s)
- Yoshio Okada
- Faculty of Pharmaceutical Sciences, High Technology Research Center, Kobe Gakuin University, Kobe 651-2180, Japan
| | | | | | | |
Collapse
|
17
|
Masocha W, González LG, Baeyens JM, Agil A. Mechanisms involved in morphine-induced activation of synaptosomal Na+,K+-ATPase. Brain Res 2002; 957:311-9. [PMID: 12445973 DOI: 10.1016/s0006-8993(02)03663-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Morphine through mu-opioid receptors and G(i/o) proteins modulates several cellular effector systems; however, the mechanisms involved in the regulation of Na(+),K(+)-ATPase are not well known. We evaluated the effect of two mu-opioid receptor agonists on ouabain-sensitive Na(+),K(+)-ATPase activity in mice forebrain synaptosomes, and examined the modulation of this effect by antagonists of opioid receptors and a blocker of G(i/o) proteins. Incubation of synaptosomes with morphine (10(-9) to 10(-4) M) or buprenorphine (10(-10) to 10(-5) M) concentration-dependently stimulated Na(+),K(+)-ATPase activity, morphine being less potent but more efficacious than buprenorphine. Morphine did not displace [3H]ouabain from its binding site (Na(+),K(+)-ATPase) to forebrain membranes, whereas ouabain did so in a concentration-dependent manner. Naloxone, an opioid antagonist (10(-6) M), added to the synaptosomal medium, antagonized the enhancement of Na(+),K(+)-ATPase activity induced by morphine, producing a parallel shift to the right of the morphine concentration-response curve. Treatment with beta-funaltrexamine, a mu antagonist (2.5 and 10 microg/mouse, i.c.v.) and naloxonazine, a mu1 antagonist (35 mg/kg, s.c.), 24 h before the synaptosomes were obtained, produced a dose-dependent reduction in the E(max) of the morphine-induced increase in Na(+),K(+)-ATPase activity in vitro, but did not significantly modify its EC(50). Pertussis toxin (G(i/o) protein blocker) treatment at a dose of 0.5 microg/mouse, administered i.c.v. 5 days before the synaptosomes were obtained, completely abolished the enhancement of Na(+),K(+)-ATPase activity induced by morphine. A lower dose (0.25 microg/mouse) decreased the E(max) of morphine by 50% but did not significantly affect its EC(50). These results suggest that morphine indirectly enhances Na(+),K(+)-ATPase activity in the brain by activating mu-opioid receptors and G(i/o) proteins.
Collapse
Affiliation(s)
- Willias Masocha
- Department of Pharmacology and Neuroscience Institute, Faculty of Medicine, University of Granada, Avenida de Madrid 11, Granada, Spain
| | | | | | | |
Collapse
|
18
|
Sakurada S, Watanabe H, Hayashi T, Yuhki M, Fujimura T, Murayama K, Sakurada C, Sakurada T. Endomorphin analogues containing D-Pro2 discriminate different mu-opioid receptor mediated antinociception in mice. Br J Pharmacol 2002; 137:1143-6. [PMID: 12466222 PMCID: PMC1573622 DOI: 10.1038/sj.bjp.0705047] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2002] [Accepted: 10/14/2002] [Indexed: 11/08/2022] Open
Abstract
The antagonistic actions of D-Pro(2)-endomorphins on inhibition of the paw withdrawal response by endomorphins were studied in mice. D-Pro(2)-endomorphin-1 and D-Pro(2)-endomorphin-2, injected intrathecally (i.t.), had no significant effect on the nociceptive thermal threshold alone. When D-Pro(2)-endomorphin-1 (0.05-0.1 pmol) was injected simultaneously with i.t. endomorphin-1 (5.0 nmol) or endomorphin-2 (5.0 nmol), antinociception induced by endomoprhin-1 was reduced significantly, whereas endomorphin-2-induced antinociception was not affected by D-Pro(2)-endomorphin-1. Antinociception induced by i.t. endomorphin-2 (5.0 nmol) was reduced significantly by its analogue, D-Pro(2)-endomorphin-2 (100 pmol), but not by D-Pro(2)-endomorphin-1. D-Pro(2)-endomorphin-1. D-Pro(2)-endomorphin-1 also antagonized the antinociceptive effect of i.t. DAMGO, a mu-opioid receptor agonist, whereas D-Pro(2)-endomorphin-2 failed to reduce the effect of DAMGO. These results suggest that endomorphin analogues containing D-Pro(2) are able to discriminate the antinociceptive actions of mu(1)- and mu(2)-opioid receptor agonists at the spinal cord level.
Collapse
Affiliation(s)
- Shinobu Sakurada
- Department of Physiology and Anatomy, Tohoku Pharmaceutical University, 4-4-1 Komatushima, Sendai 981-8558, Japan
| | - Hiroyuki Watanabe
- Department of Physiology and Anatomy, Tohoku Pharmaceutical University, 4-4-1 Komatushima, Sendai 981-8558, Japan
| | - Takafumi Hayashi
- Department of Physiology and Anatomy, Tohoku Pharmaceutical University, 4-4-1 Komatushima, Sendai 981-8558, Japan
| | - Masayuki Yuhki
- Department of Physiology and Anatomy, Tohoku Pharmaceutical University, 4-4-1 Komatushima, Sendai 981-8558, Japan
| | - Tsutomu Fujimura
- Division of Biochemical Analysis, Central Laboratory of Medical Sciences, Juntendo University School of Medicine, 2-1-1 Hongo, Tokyo 113-8421, Japan
| | - Kimie Murayama
- Division of Biochemical Analysis, Central Laboratory of Medical Sciences, Juntendo University School of Medicine, 2-1-1 Hongo, Tokyo 113-8421, Japan
| | - Chikai Sakurada
- Department of Biochemistry, Daiichi College of Pharmaceutical Sciences, 22-1 Tamagawa-cho, Minami-ku, Fukuoka 815-8511, Japan
| | - Tsukasa Sakurada
- Department of Biochemistry, Daiichi College of Pharmaceutical Sciences, 22-1 Tamagawa-cho, Minami-ku, Fukuoka 815-8511, Japan
| |
Collapse
|
19
|
Sakurada S, Hayashi T, Yuhki M, Fujimura T, Murayama K, Yonezawa A, Sakurada C, Takeshita M, Sato T, Zadina JE, Kastin AJ, Sakurada T. Differential antagonism of endomorphin-1 and endomorphin-2 supraspinal antinociception by naloxonazine and 3-methylnaltrexone. Peptides 2002; 23:895-901. [PMID: 12084520 DOI: 10.1016/s0196-9781(02)00016-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
To determine if different subtypes of mu-opioid receptors were involved in antinociception induced by endomorphin-1 and endomorphin-2, the effect of pretreatment with various mu-opioid receptor antagonists beta-funaltrexamine, naloxonazine and 3-methylnaltrexone on the inhibition of the paw-withdrawal induced by endomorphin-1 and endomorphin-2 given intracerebroventricularly (i.c.v.) were studied in ddY male mice. The inhibition of the paw-withdrawal induced by i.c.v. administration of endomorphin-1, endomorphin-2 or DAMGO was completely blocked by the pretreatment with a selective mu-opioid receptor antagonist beta-funaltrexamine (40 mg/kg), indicating that the antinociception induced by all these peptides are mediated by the stimulation of mu-opioid receptors. However, naloxonazine, a mu1-opioid receptor antagonist pretreated s.c. for 24h was more effective in blocking the antinociception induced by endomorphin-2, than by endomorphin-1 or DAMGO given i.c.v. Pretreatment with a selective morphine-6 beta-glucuronide blocker 3-methylnaltrexone 0.25mg/kg given s.c. for 25 min or co-administration of 3-methylnaltrexone 2.5 ng given i.c.v. effectively attenuated the antinociception induced by endomorphin-2 given i.c.v. and co-administration of 3-methylnaltrexone shifted the dose-response curves for endomorphin-2 induced antinociception to the right by 4-fold. The administration of 3-methylnaltrexone did not affect the antinociception induced by endomorphin-1 or DAMGO given i.c.v. Our results indicate that the antinociception induced by endomorphin-2 is mediated by the stimulation of subtypes of mu-opioid receptor, which is different from that of mu-opioid receptor subtype stimulation by endomorphin-1 and DAMGO.
Collapse
Affiliation(s)
- Shinobu Sakurada
- Department of Physiology and Anatomy, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Sendai, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Narita M, Imai S, Itou Y, Yajima Y, Suzuki T. Possible involvement of mu1-opioid receptors in the fentanyl- or morphine-induced antinociception at supraspinal and spinal sites. Life Sci 2002; 70:2341-54. [PMID: 12150199 DOI: 10.1016/s0024-3205(01)01550-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Fentanyl has been shown to be a potent analgesic with a lower propensity to produce tolerance and physical dependence in the clinical setting. The present study was designed to investigate the mechanisms of fentanyl- or morphine-induced antinociception at both supraspinal and spinal sites. In the mouse tail-flick test, the antinociceptive effects induced by both fentanyl and morphine were blocked by either the mu1-opioid receptor antagonist naloxonazine or the mu1/mu2-opioid receptor antagonist beta-funaltrexamine (beta-FNA) after s.c., i.c.v. or i.t. injection. In contrast, both fentanyl and morphine given i.c.v. or i.t. failed to produce antinociception in mu1-deficient CXBK mice. These findings indicate that like morphine, the antinociception induced by fentanyl may be mediated predominantly through mu1-opioid receptors at both supraspinal and spinal sites in mice. We also determined the ED50 values for s.c.-, i.c.v.- and i.t.-administered fentanyl- or morphine-induced antinociception in mice. The ED50 values for s.c.-, i.c.v.- and i.t.-administered fentanyl-induced antinociception were 73.7, 18.5 and 1.2-fold lower than that of morphine, respectively. The present data clearly suggest the usefulness of peripheral treatment with fentanyl for the control of pain.
Collapse
MESH Headings
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/pharmacology
- Animals
- Dose-Response Relationship, Drug
- Enkephalin, D-Penicillamine (2,5)-/pharmacology
- Fentanyl/administration & dosage
- Fentanyl/pharmacology
- Injections, Intraventricular
- Injections, Spinal
- Injections, Subcutaneous
- Male
- Mice
- Mice, Inbred C57BL
- Morphine/administration & dosage
- Morphine/pharmacology
- Naloxone/analogs & derivatives
- Naloxone/pharmacology
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Pain Measurement
- Receptors, Opioid, mu/drug effects
- Spinal Cord/drug effects
Collapse
Affiliation(s)
- Minoru Narita
- Department of Toxicology, School of Pharmacy, Hoshi University, Tokyo, Japan
| | | | | | | | | |
Collapse
|
21
|
Sakurada S, Hayashi T, Yuhki M, Orito T, Zadina JE, Kastin AJ, Fujimura T, Murayama K, Sakurada C, Sakurada T, Narita M, Suzuki T, Tan-no K, Tseng LF. Differential antinociceptive effects induced by intrathecally administered endomorphin-1 and endomorphin-2 in the mouse. Eur J Pharmacol 2001; 427:203-10. [PMID: 11567650 DOI: 10.1016/s0014-2999(01)01238-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Two highly selective mu-opioid receptor agonists, endomorphin-1 and endomorphin-2, have been identified and postulated to be endogenous ligands for mu-opioid receptors. Intrathecal (i.t.) administration of endomorphin-1 and endomorphin-2 at doses from 0.039 to 5 nmol dose-dependently produced antinociception with the paw-withdrawal test. The paw-withdrawal inhibition rapidly reached its peak at 1 min, rapidly declined and returned to the pre-injection levels in 20 min. The inhibition of the paw-withdrawal responses to endomorphin-1 and endomorphin-2 at a dose of 5 nmol observed at 1 and 5 min after injection was blocked by pretreatment with a non-selective opioid receptor antagonist naloxone (1 mg/kg, s.c.). The antinociceptive effect of endomorphin-2 was more sensitive to the mu (1)-opioid receptor antagonist, naloxonazine than that of endomorphin-1. The endomorphin-2-induced paw-withdrawal inhibition at both 1 and 5 min after injection was blocked by pretreatment with kappa-opioid receptor antagonist nor-binaltorphimine (10 mg/kg, s.c.) or the delta(2)-opioid receptor antagonist naltriben (0.6 mg/kg, s.c.) but not the delta(1)-opioid receptor antagonist 7-benzylidine naltrexone (BNTX) (0.6 mg/kg s.c.). In contrast, the paw-withdrawal inhibition induced by endomorphin-1 observed at both 1 and 5 min after injection was not blocked by naloxonazine (35 mg/kg, s.c.), nor-binaltorphimine (10 mg/kg, s.c.), naltriben (0.6 mg/kg, s.c.) or BNTX (0.6 mg/kg s.c.). The endomorphin-2-induced paw-withdrawal inhibition was blocked by the pretreatment with an antiserum against dynorphin A-(1-17) or [Met(5)]enkephalin, but not by antiserum against dynorphin B-(1-13). Pretreatment with these antisera did not affect the endomorphin-1-induced paw-withdrawal inhibition. Our results indicate that endomorphin-2 given i.t. produces its antinociceptive effects via the stimulation of mu (1)-opioid receptors (naloxonazine-sensitive site) in the spinal cord. The antinociception induced by endomophin-2 contains additional components, which are mediated by the release of dynorphin A-(1-17) and [Met(5)]enkephalin which subsequently act on kappa-opioid receptors and delta(2)-opioid receptors to produce antinociception.
Collapse
Affiliation(s)
- S Sakurada
- Department of Physiology and Anatomy, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Sendai 981-8558, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Colman AS, Miller JH. Modulation of breathing by mu1 and mu2 opioid receptor stimulation in neonatal and adult rats. RESPIRATION PHYSIOLOGY 2001; 127:157-72. [PMID: 11504587 DOI: 10.1016/s0034-5687(01)00240-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Opioid modulation of breathing during postnatal development through to the adult was investigated in the rat. Respiratory frequency, tidal volume and minute volume were recorded in unanesthetized, unrestrained rat pups and adults using barometric plethysmography. Subjects were administered the highly selective mu opioid agonists dermorphin and fentanyl. Fentanyl, which readily crosses the blood-brain barrier, was included to ensure that developmental changes in blood-brain barrier restrictions did not mask some of the dermorphin effects in older neonates. Drugs were administered subcutaneously in neonates and adults, although dermorphin was given by intracerebroventricular route only in adults. In neonates, mu agonist administration caused a gasping-like pattern of breathing, characterized by a marked fall in frequency and a smaller increase in tidal volume. The gasping response was prevented by pre-treatment with the long-acting mu1 antagonist naloxonazine (NALZ). In the presence of NALZ, mu agonists elicited only a small, but significant, reduction in tidal volume. Both dermorphin and fentanyl showed more potent activity in younger pups than in older pups, possibly in the case of dermorphin because of developmental maturation of blood-brain barrier function. In adults, fentanyl and dermorphin both caused a reduction in frequency and minute volume. The response of adults to fentanyl, but not dermorphin, was prevented by NALZ. These results suggest that both mu1 and mu2 receptors contribute to opioid-induced respiratory depression during neonatal and adult life.
Collapse
Affiliation(s)
- A S Colman
- School of Biological Sciences, Victoria University of Wellington, P.O. Box 600, Wellington, New Zealand
| | | |
Collapse
|
23
|
Kozaki Y, Tadaki E, Kumazawa T. Morphine inhibits resting respiration, but it attenuates reflexive respiratory suppression in anesthetized cat through kappa-receptor. THE JAPANESE JOURNAL OF PHYSIOLOGY 2000; 50:615-24. [PMID: 11173557 DOI: 10.2170/jjphysiol.50.615] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Noxious stimulation of thin-fiber muscular afferents induces a reflexive respiratory suppression that we call "poststimulus respiratory suppression." In anesthetized, vagotomized, paralyzed, and artificially ventilated cats, morphine depressed the level of resting respiration (inhibitory effect on resting respiration) and attenuated the magnitude of the poststimulus respiratory suppression (excitatory effect on the reflexively modified respiration). These two kinds of morphine effects were antagonized by naloxone, suggesting the participation of opioid receptors. To clarify the opioid receptor subtypes responsible for these effects of morphine, three type-selective opioid antagonists-naltrindole (delta antagonist), gamma-funaltrexamine (mu antagonist), and Mr2266 (kappa antagonist)-were tested. The morphine-induced depression in the resting respiration was antagonized by pretreatment with the kappa antagonist, not with the mu or delta antagonist. Furthermore, the morphine-induced attenuation in the magnitude of the poststimulus suppression was also blocked by the kappa antagonist, but not by the mu or delta antagonist. In conclusion, (1) morphine inhibits resting respiration, but it attenuates the magnitude of the poststimulus respiratory suppression; (2) both these morphine effects are mediated by kappa opioid receptors. The possibility that the kappa(3) receptor, one of the kappa receptors subtypes, mediates the two kinds of morphine effects has been discussed.
Collapse
Affiliation(s)
- Y Kozaki
- Department of Neural Regulation, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601 Japan.
| | | | | |
Collapse
|
24
|
Sakurada S, Hayashi T, Yuhki M, Fujimura T, Murayama K, Yonezawa A, Sakurada C, Takeshita M, Zadina JE, Kastin AJ, Sakurada T. Differential antagonism of endomorphin-1 and endomorphin-2 spinal antinociception by naloxonazine and 3-methoxynaltrexone. Brain Res 2000; 881:1-8. [PMID: 11033087 DOI: 10.1016/s0006-8993(00)02770-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
To determine the role of spinal mu-opioid receptor subtypes in antinociception induced by intrathecal (i.t.) injection of endomorphin-1 and -2, we assessed the effects of beta-funaltrexamine (a selective mu-opioid receptor antagonist) naloxonazine (a selective antagonist at the mu(1)-opioid receptor) and a novel receptor antagonist (3-methoxynaltrexone) using the paw-withdrawal test. Antinociception of i.t. endomorphins and [D-Ala(2), MePhe(4), Gly(ol)(5)]enkephalin (DAMGO) was completely reversed by pretreatment with beta-funaltrexamine (40 mg/kg s.c.). Pretreatment with a variety of doses of i.t. or s.c. naloxonazine 24 h before testing antagonized the antinociception of endomorphin-1, -2 and DAMGO. Judging from the ID(50) values of naloxonazine, the antinociceptive effect of endomorphin-2 was more sensitive to naloxonazine than that of endomorphin-1 or DAMGO. The selective morphine-6beta-glucuronide antagonist, 3-methoxynaltrexone, which blocked endomorphin-2-induced antinociception at each dose (0.25 mg/kg s.c. or 2.5 ng i.t.) that was inactive against DAMGO, did not affect endomorphin-1-induced antinociception but shifted the dose-response curve of endomorphin-2 3-fold to the right. These findings may be interpreted as indicative of the existence of a novel mu-opioid receptor subtype in spinal sites, where antinociception of morphine-6beta-glucuronide and endomorphin-2 are antagonized by 3-methoxynaltrexone. The present results suggest that endomorphin-1 and endomorphin-2 may produce antinociception through different subtypes of mu-opioid receptor.
Collapse
Affiliation(s)
- S Sakurada
- Department of Physiology and Anatomy, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, 981-8558, Sendai, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Rady JJ, Holmes BB, Portoghese PS, Fujimoto JM. Morphine tolerance in mice changes response of heroin from mu to delta opioid receptors. PROCEEDINGS OF THE SOCIETY FOR EXPERIMENTAL BIOLOGY AND MEDICINE. SOCIETY FOR EXPERIMENTAL BIOLOGY AND MEDICINE (NEW YORK, N.Y.) 2000; 224:93-101. [PMID: 10806416 DOI: 10.1046/j.1525-1373.2000.22406.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Heroin produced antinociception in the tail flick test through mu receptors in the brain of ICR and CD-1 mice, a response inhibited by 3-O-methylnaltrexone. Tolerance to morphine was produced by subcutaneous morphine pellet implantation. By the third day, the heroin response was produced through delta opioid receptors. The response was inhibited by simultaneous intracerebroventricular (i.c. v.) administration of naltrindole, a delta opioid receptor antagonist. More specifically, delta1 rather than delta2 receptors were involved because 7-benzylidenenaltrexone, a delta1 receptor antagonist, inhibited but naltriben, a delta2 antagonist, did not. Also, antinociception produced by i.c.v. heroin was inhibited by intrathecal administration of bicuculline and picrotoxin consistent with the concept that delta1 receptors in the brain mediated the antinociceptive response through descending neuronal pathways to the spinal cord to activate GABAA and GABAB receptors rather than spinal alpha2-adrenergic and serotonergic receptors activated originally by the mu agonist action in naive mice. The mu response of 6-monoacetylmorphine, a metabolite of heroin, was changed by morphine pellet implantation to a delta2 response (inhibited by naltriben but not 7-benzylidenenaltrexone). The agonist action of morphine in these morphine-tolerant mice remained mu. Thus, the opioid receptor selectivity of heroin and 6-monoacetylmorphine in the brain is changed by production of tolerance to morphine. Such a change explains how morphine tolerant mice are not cross-tolerant to heroin.
Collapse
Affiliation(s)
- J J Rady
- Research Service and Department of Pharmacology and Toxicology, VA Medical Center and Medical College of Wisconsin, Milwaukee, Wisconsin 53295, USA
| | | | | | | |
Collapse
|
26
|
Sakurada S, Takeda S, Sato T, Hayashi T, Yuki M, Kutsuwa M, Tan-No K, Sakurada C, Kisara K, Sakurada T. Selective antagonism by naloxonazine of antinociception by Tyr-D-Arg-Phe-beta-Ala, a novel dermorphin analogue with high affinity at mu-opioid receptors. Eur J Pharmacol 2000; 395:107-12. [PMID: 10794815 DOI: 10.1016/s0014-2999(00)00166-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
To examine the role of mu-opioid receptor subtypes, we assessed the antinociceptive effect of H-Tyr-D-Arg-Phe-beta-Ala-OH (TAPA), an analogue of dermorphin N-terminal peptide in mice, using the tail-flick test. Intracerebroventricularly (i.c.v.) or intrathecally (i.t.) injected TAPA produced potent antinociception with tail-flick as a thermal noxious stimulus. The selective mu(1)-opioid receptor antagonist, naloxonazine (35 mg/kg, s.c.), or the selective mu-opioid receptor antagonist, beta-funaltrexamine, 24 h before testing antagonized the antinociceptive effect of i.t. or i.c.v. TAPA on the response to noxious stimuli. Pretreatment with beta-funaltrexamine completely antagonized the antinociception by both i.c.v. and i.t. administered TAPA and [D-Ala(2), Me-Phe(4), Gly(ol)(5)]enkephalin (DAMGO). Especially in the tail-flick test, pretreatment with naloxonazine produced a marked rightward displacement of the i.t. TAPA dose-response curve for antinociception. Though DAMGO is a highly selective mu-opioid receptor agonist, pretreatment with naloxonazine partially blocked the antinociceptive response to DAMGO after i.c.v., but not after i. t. injection. These results indicate that TAPA can act as a highly selective mu(1)-opioid receptor agonist (notable naloxonazine-sensitive receptor agonist) at not only the supraspinal level, but also the spinal level. These data also reveal different antinociceptive mechanisms for DAMGO and for TAPA.
Collapse
Affiliation(s)
- S Sakurada
- Department of Physiology and Anatomy, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Crain SM, Shen K. Enhanced analgesic potency and reduced tolerance of morphine in 129/SvEv mice: evidence for a deficiency in GM1 ganglioside-regulated excitatory opioid receptor functions. Brain Res 2000; 856:227-35. [PMID: 10677630 DOI: 10.1016/s0006-8993(99)02446-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
10-fold higher doses in SW mice. Furthermore, cotreatment of 129/SvEv mice with morphine plus a low dose of naltrexone (ca. 0.1 microgram/kg) that markedly enhances and prolongs morphine's antinociceptive effects in SW mice did not enhance, and often attenuated6 h. The marked GM1-induced attenuation of morphine's antinociceptive effects in 129/SvEv mice may be due to conversion of some of the opioid receptors in these mice from an inhibitory Gi/Go-coupled to an excitatory Gs-coupled mode. Exogenous GM1 supplementation can, therefore, reverse the anomalous lack of morphine tolerance displayed by this mouse strain in comparison to SW and other mice. The present study may provide insights into factors that regulate the marked variability in nociceptive sensitivity and opioid tolerance/dependence liability among individual humans.
Collapse
Affiliation(s)
- S M Crain
- Department of Neuroscience, Albert Einstein College of Medicine, Yeshiva University, 1300 Morris Park Ave., Bronx, NY, USA.
| | | |
Collapse
|
28
|
Verborgh C, Meert TF. Antagonistic effects of naloxone and naloxonazine on sufentanil-induced antinociception and respiratory depression in rats. Pain 1999; 83:17-24. [PMID: 10506668 DOI: 10.1016/s0304-3959(99)00068-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Several binding studies in rodent brain homogenates have revealed two distinct micro-opiate binding sites based on differences in binding affinity of several opiate peptides and opiate alkaloids. Naloxonazine (NLZ), which preferentially binds to the high affinity micro(1) sites, is often used to discriminate between pharmacological effects mediated by micro(1) and micro(2) binding sites. The present series of experiments were undertaken to compare the opioid antagonistic properties of naloxonazine and naloxone (NLX) (a non-selective micro(1)-antagonist) on intravenous (i.v.) and intrathecal (i.t.) sufentanil (SUF)-induced antinociception and respiratory depression. The opioid antagonists were given either intravenously at 5 min after SUF, or subcutaneously (s.c.) 24 h prior to the opioid. Intravenous NLX and NLZ reduced the i.v. and i. t. SUF-induced antinociception, hypercapnia and hypoxia when given directly after the opioid. There were no major differences in activity between both antagonists. Pretreatment with 30 mg/kg NLX did not reverse the i.v. or i.t. SUF-induced antinociception and respiratory depression. Subcutaneous pretreatment with doses up to 30 mg/kg NLX only partially antagonized the i.v. SUF-induced antinociception, while a complete reversal was present of the opioid-induced hypercapnia and hypoxia. With regard to i.t. SUF, doses up to 30 mg/kg NLZ were unable to reduce the antinociception. The respiratory depression was partially affected; with 30 mg/kg NLZ, the i.t. SUF-induced hypercapnia returned to baseline levels, whereas the SUF-induced hypoxia was only minimally affected. These results challenge the classical view of the selectivity of NLZ for the high affinity micro(1) binding sites. They further fail to conform an exclusive role for micro(2) receptor sites in the respiratory depression and spinal analgesia induced by a strong lipophilic opioid such as SUF in rats.
Collapse
Affiliation(s)
- C Verborgh
- Departement Anesthesiologie, Akademisch Ziekenhuis Vrije Universiteit Brussel, Laarbeeklaan 101, B-1090, Brussels, Belgium
| | | |
Collapse
|
29
|
Izenwasser S, Staley JK, Cohn S, Mash DC. Characterization of kappa1-opioid receptor binding in human insular cortex. Life Sci 1999; 65:857-62. [PMID: 10465345 DOI: 10.1016/s0024-3205(99)00315-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Mesolimbic dopaminergic neurotransmission is modulated by dynorphin peptides binding to kappa-opioid receptors. The interaction between dynorphin and dopamine systems makes the kappa-opioid receptor a potential drug discovery target for the development of therapeutic agents for schizophrenia and drug abuse. This study reports the specificity and parameters of [3H]U69593 binding in the insular cortex, a representative corticolimbic area of the human brain. The results demonstrate that the radioligand [3H]U69593 labels a single population of receptors in human insular cortex with an affinity in the low nanomolar range. The pharmacological profile for inhibition of [3H]U69593 binding was determined in this brain region using drugs known to bind to mu, kappa and delta opioid receptors. The results show that kappa-opioid selective agonists and antagonists inhibit binding of this ligand in human brain with comparable affinities and rank order as previously described for rat and guinea pig brain and the cloned kappa1-opioid receptor subtype.
Collapse
MESH Headings
- Animals
- Benzeneacetamides
- Binding, Competitive
- Cerebral Cortex/metabolism
- Humans
- Pyrrolidines/metabolism
- Rats
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/metabolism
- Stereoisomerism
Collapse
Affiliation(s)
- S Izenwasser
- Department of Neurology, University of Miami School of Medicine, FL 33136, USA.
| | | | | | | |
Collapse
|
30
|
Sakurada S, Zadina JE, Kastin AJ, Katsuyama S, Fujimura T, Murayama K, Yuki M, Ueda H, Sakurada T. Differential involvement of mu-opioid receptor subtypes in endomorphin-1- and -2-induced antinociception. Eur J Pharmacol 1999; 372:25-30. [PMID: 10374711 DOI: 10.1016/s0014-2999(99)00181-8] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We investigated the role of mu-opioid receptor subtypes in both endomorphin-1 and endomorphin-2 induced antinociception in mice using supraspinally mediated behavior. With tail pressure as a mechanical noxious stimulus, both intracerebroventricularly (i.c.v.) and intrathecally (i.t.) injected-endomorphins produced potent and significant antinociceptive activity. Antinociception induced by i.t. and i.c.v. injection of endomorphin-1 was not reversed by pretreatment with a selective mu1-opioid receptor antagonist, naloxonazine (35 mg/kg, s.c.). By contrast, antinociception induced by i.t. and i.c.v. endomorphin-2 was significantly decreased by mu1-opioid receptor antagonist. Antinociception of both i.t. and i.c.v. endomorphin-1 and -2 was completely reversed by pretreatment with beta-funaltrexamine (40 mg/kg, s.c.). The results indicate that endomorphins may produce antinociception through the distinct mu1 and mu2 subtypes of mu-opioid receptor.
Collapse
Affiliation(s)
- S Sakurada
- Department of Physiology and Anatomy, Tohoku Pharmaceutical University, Sendai, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Sato T, Sakurada S, Takahashi N, Sakurada T, Tan-No K, Wako K, Kisara K. Contribution of spinal mu1-opioid receptors to morphine-induced antinociception. Eur J Pharmacol 1999; 369:183-7. [PMID: 10206177 DOI: 10.1016/s0014-2999(99)00065-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
To determine the role of mu-opioid receptor subtypes, mu1 and mu2, in antinociception induced by intrathecal (i.t.) or intracerebroventricular (i.c.v.) injection of morphine, we assessed the effect of naloxonazine, a selective antagonist at mu1-opioid receptors. The antinociceptive effects of morphine were measured using four different nociceptive tests. The selective mu1 antagonist, naloxonazine (35 mg/kg, s.c.), 24 h before testing antagonized the antinociceptive effect of morphine on responses to chemical and thermal stimuli to a greater extent than that on responses to mechanical stimuli, as judged from ED50 values. The present results suggest that the antinociceptive activity of both i.t. and i.c.v. morphine on responses to chemical and thermal stimuli may be mediated through the mu1-opioid receptor subtype (naloxonazine-sensitive sites). These findings may be interpreted as indicative of the existence of mu1-receptor subtypes capable of mediating antinociception not only in supraspinal sites but also in spinal sites.
Collapse
Affiliation(s)
- T Sato
- Department of Pharmaceutics, Tohoku College of Pharmacy, Sendai, Japan
| | | | | | | | | | | | | |
Collapse
|
32
|
Rady JJ, Gorny JM, Fujimoto JM. Heroin antinociception changed from mu to delta receptor in streptozotocin-treated mice. JAPANESE JOURNAL OF PHARMACOLOGY 1998; 78:443-54. [PMID: 9920201 DOI: 10.1254/jjp.78.443] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
CD-1 mice were treated intravenously with streptozotocin, 200 mg/kg, and tested 2 weeks later or treated with 60 mg/kg and tested 3 days later. Both treatments changed the tail flick response of heroin and 6-monoacetylmorphine (6 MAM) given intracerebroventricularly from a mu- to delta-opioid receptor-mediated action as determined by differential effects of opioid receptor antagonists. The response to morphine remained mu. Heroin and 6 MAM responses involved delta1 (inhibited by 7-benzylidenenaltrexone) and delta2 (inhibited by naltriben) receptors, respectively. These delta-agonist actions did not synergize with the mu-agonist action of morphine in the diabetic mice. The expected synergism between the delta agonist, [D-Pen2-D-Pen5]enkephalin (DPDPE), and morphine was not obtained in diabetic mice. Thus, diabetes disrupted the purported mu/delta-coupled response. In nondiabetic CD-1 mice, heroin and 6 MAM produced a different mu-receptor response (not inhibited by naloxonazine) from that of morphine (inhibited by naloxonazine). Also, these mu actions, unlike that of morphine, did not synergize with DPDPE. The unique receptor actions and changes produced by streptozotocin suggest that extrinsic in addition to genetic factors influence the opioid receptor selectivity of heroin and 6 MAM.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Anti-Bacterial Agents/pharmacology
- Benzylidene Compounds/pharmacology
- Diabetes Mellitus, Experimental/physiopathology
- Dose-Response Relationship, Drug
- Drug Interactions
- Enkephalin, D-Penicillamine (2,5)-
- Enkephalins/pharmacology
- Heroin/pharmacology
- Injections, Intraventricular
- Male
- Mice
- Morphine/pharmacology
- Morphine Derivatives/pharmacology
- Naloxone/analogs & derivatives
- Naloxone/pharmacology
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Nociceptors/drug effects
- Pain/drug therapy
- Pain/physiopathology
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, mu/drug effects
- Streptozocin/pharmacology
- Time Factors
Collapse
Affiliation(s)
- J J Rady
- Research Service, Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI 53295, USA
| | | | | |
Collapse
|
33
|
Thongpradichote S, Matsumoto K, Tohda M, Takayama H, Aimi N, Sakai S, Watanabe H. Identification of opioid receptor subtypes in antinociceptive actions of supraspinally-administered mitragynine in mice. Life Sci 1998; 62:1371-8. [PMID: 9585164 DOI: 10.1016/s0024-3205(98)00075-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mitragynine (MG), a major alkaloidal constituent extracted from the plant Mitragyna speciosa Korth, is known to exert an opioid-like activity. Our previous study showed the involvement of opioid systems in the antinociceptive activity of MG in the tail-pinch and hot-plate tests in mice. In the present study, to clarify the opioid receptor subtypes involved in the antinociceptive action of MG, we investigated the effects of selective antagonists for mu-, delta- and kappa- opioid receptors on antinociception caused by the intracerebroventricular (i.c.v.) injection of MG in the tail-pinch and hot-plate tests in mice. The coadministration of a selective mu-opioid antagonist, cyprodime (1-10 microg, i.c.v.) and the pretreatment with a selective mu1-opioid antagonist naloxonazine (1-3 microg, i.c.v.) significantly antagonized the antinociceptive activities of MG (10 microg, i.c.v.) and morphine (MOR, 3 microg, i.c.v.) in the tail-pinch and hot-plate tests. Naltrindole (1-5 ng, i.c.v.), a selective delta-opioid antagonist, also blocked the effects of MG (10 microg, i.c.v.) without affecting MOR (3 microg, i.c.v.) antinociception. Nor-binaltorphimine, a selective kappa-opioid antagonist, significantly attenuated MG (10 microg, i.c.v.) antinociception in the tail-pinch test but not in the hot-plate test at the dose (1 microg, i.c.v.) that antagonized the antinociceptive effects of the selective kappa-opioid agonist U50,488H in both tests, while it had no effect on MOR antinociception in either tests. These results suggest that antinociception caused by i.c.v. MG is dominantly mediated by mu- and delta-opioid receptor subtypes, and that the selectivity of MG for the supraspinal opioid receptor subtypes differs from that of MOR in mice.
Collapse
Affiliation(s)
- S Thongpradichote
- Department of Pharmacology, Research Institute for Wakan-Yaka (Oriental Medicines), Toyama Medical and Pharmaceutical University, Sugitani, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Grung M, Skurtveit S, Aasmundstad TA, Handal M, Alkana RL, Mørland J. Morphine-6-glucuronide-induced locomotor stimulation in mice: role of opioid receptors. PHARMACOLOGY & TOXICOLOGY 1998; 82:3-10. [PMID: 9527644 DOI: 10.1111/j.1600-0773.1998.tb01390.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Morphine-6beta-glucuronide is a major metabolite of morphine with potent analgesic actions. To explore the importance of this opiate when administered as a drug by its own or in morphine action, we studied the locomotor activity response to morphine and morphine-6-glucuronide in drug-naive C57 BL/6JBom mice. The effects of administration of the two opiates on a battery of 7 different locomotor activities were studied and compared to saline controls. A dose of 20 micromol/kg morphine-6-glucuronide resulted in more locomotion than the same dose of morphine, while at higher doses (up to 120 micromol/kg), similar increases for most locomotor behaviours were recorded for both drugs. Pretreatment with naltrindole indicated that the delta-receptors play an equivalent but minor role in mediating both morphine-6-glucuronide and morphine hyperlocomotion. Administration of high naltrexone doses (10 mg/kg) completely abolished the locomotor stimulation induced by both opiates. However, at intermediate naltrexone doses of 0.25 and 0.5 mg/kg, morphine-induced behaviours was completely inhibited while morphine-6-glucuronide induced behaviours demonstrated partial resistance to naltrexone inhibition. The mu1-specific receptor antagonist naloxonazine caused 75% reduction of morphine induced behaviours and only 50% inhibition of morphine-6-glucuronide induced behaviors. Taken together our observations indicated general similarity but also marked differences between morphine and morphine-6-glucuronide with respect to opiate receptors mediating the locomotor stimulatory effect.
Collapse
Affiliation(s)
- M Grung
- National Institute of Forensic Toxicology, Oslo, Norway
| | | | | | | | | | | |
Collapse
|
35
|
Backer MM, Schreiber S, Pick CG. Interactions between different antidepressants and morphine alter gastrointestinal transit in mice. Life Sci 1997; 61:PL109-13. [PMID: 9284086 DOI: 10.1016/s0024-3205(97)00595-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To determine different serotoninergic antidepressants' effects on the gastrointestinal (GI) inhibiting effect induced by morphine, mice were pretreated with mianserin (a tetracyclic antidepressant with multiple 5-HT receptor subtypes interactions) and with fluoxetine (a selective 5-HT reuptake inhibitor). Mianserin alone, produced gastrointestinal inhibition in a dose-dependent manner. Naloxone did not reverse this inhibiting effect, indicating that different mechanism of action are involved in morphine- and mianserin-induced inhibition of the gastrointestinal transit. Fluoxetine injected alone produced an increased propulsive motility of the GI transit. This effect was not reversed by naloxone. Fluoxetine did not reduce significantly mianserin-induced inhibition of GI transit. Fluoxetine also mildly reversed morphine-induced gastrointestinal inhibition, suggesting some degree of involvement of the opiates through the serotoninergic system.
Collapse
Affiliation(s)
- M M Backer
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Israel
| | | | | |
Collapse
|
36
|
Gergen KA, Zadina JE, Paul D. Analgesic effects of Tyr-W-MIF-1: a mixed mu2-opioid receptor agonist/mu1-opioid receptor antagonist. Eur J Pharmacol 1996; 316:33-8. [PMID: 8982647 DOI: 10.1016/s0014-2999(96)00656-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Tyr-W-MIF-1 (Tyr-Pro-Trp-Gly-NH2) is a naturally occurring neuropeptide that displays high selectivity for mu-opioid receptors. Recently, intrathecal (i.t.) Tyr-W-MIF-1 was shown to induce potent analgesia mediated through spinal mu2-opioid receptors in mice. In the current study, we investigated the supraspinal analgesic effects of Tyr-W-MIF-1 using intracerebroventricular (i.c.v.) administration in mice. I.c.v. Tyr-W-MIF-1 induced a dose-dependent analgesic response with an ED50 of 31.4 micrograms that was antagonized by i.c.v. naloxone (ED50 = 4.46 nmol) and the mu-opioid receptor antagonist beta-funaltrexamine but not by the mu1-opioid receptor-selective antagonist naloxonazine. I.t. naloxone (ED50 = 0.12 nmol), however, was nearly 40-fold more potent than i.c.v. naloxone at antagonizing i.c.v. Tyr-W-MIF-1-induced analgesia. Tyr-W-MIF-1 also possesses antagonist activity at mu1-opioid receptors in brain. Coadministration of i.c.v. Tyr-W-MIF-1 with i.c.v. morphine or i.c.v. [D-Ala2, MePhe4, Gly(ol)5]enkephalin (DAMGO) significantly decreased the analgesic response to either drug administered alone. Thus, Tyr-W-MIF-1 functions as a mixed mu2-opioid receptor agonist/mu1-opioid receptor antagonist after i.c.v. administration in mice.
Collapse
Affiliation(s)
- K A Gergen
- Tulane University School of Medicine, New Orleans, LA 70146, USA
| | | | | |
Collapse
|
37
|
Zadina JE, Paul D, Gergen KA, Ge LJ, Hackler L, Kastin AJ. Binding of Tyr-W-MIF-1 (Tyr-Pro-Trp-Gly-NH2) and related peptides to mu 1 and mu 2 opiate receptors. Neurosci Lett 1996; 215:65-9. [PMID: 8880755 DOI: 10.1016/s0304-3940(96)12928-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Two endogenous brain peptides (Tyr-W-MIF-1 (Tyr-Pro-Trp-Gly-NH2) and Tyr-MIF-1 (Tyr-Pro-Leu-Gly-NH2)), a cyclized analog and two fragments of Tyr-W-MIF-1, and hemorphin (Tyr-Pro-Trp-Thr) were tested for binding to mu 1 and mu 2 opiate receptor. All these peptides bound to both mu 1 and mu 2 sites in assays optimized to discriminate these subtypes of the mu opiate receptor in membranes from bovine thalamus. The cyclized analog of Tyr-W-MIF-1, previously shown to have potency near that of Tyr-D-Ala-Gly-N-MePhe-Gly-ol (DAMGO) and morphine in producing analgesia after intracerebroventricular (i.c.v.) injection, bound to mu 1 and mu 2 sites with affinities similar to those of DAMGO. Tyr-W-MIF-1, previously shown to induce analgesia after i.c.v. injection but with much higher potency after intrathecal (i.t.) injection, also bound to both mu 1 and mu 2 sites with an affinity between that of morphiceptin and hemorphin. Although the highest ratios of Ki's for mu 2/mu 1 were shown by hemorphin, Tyr-W-MIF-1, and Tyr-W-MIF-1, none of the compounds were significantly different in selectivity. The results indicate that the relatively lower potency of Tyr-W-MIF-1 after i.c.v., compared with i.t. injection, is not due to a lack of binding to mu 1 sites. They suggest that it has relatively high efficacy at mu 2, but low efficacy at mu 1 sites, a possibility that might explain some of the novel properties of these peptides.
Collapse
Affiliation(s)
- J E Zadina
- VA Medical Center, New Orleans, LA 70146, USA
| | | | | | | | | | | |
Collapse
|
38
|
Carr DJ, Brockunier LL, Scott M, Bagley JR, France CP. Mirfentanil antagonizes morphine-induced suppression of splenic NK activity in mice. IMMUNOPHARMACOLOGY 1996; 34:9-16. [PMID: 8880221 DOI: 10.1016/0162-3109(95)00051-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Mirfentanil [N-(2-pyrazinyl)-N-(1-phenethyl-4-piperidinyl)-2-furamide] was studied for its antinociceptive and immunomodulatory effects in mice Mirfentanil (1.0-32.0 mg/kg) increased tail-flick latency to a thermal stimulus and this effect was antagonized (94 +/- 2%) by naltrexone (10.0 mg/kg). Unlike naltrexone, the delta opioid selective antagonist naltrindole (20.0 mg/kg) had no effect on mirfentanil-induced analgesia. In a dose-dependent fashion, the mu-selective antagonists beta-funaltrexamine (1.0-40.0 mg/kg) and naloxonazine (1.0-35.0 mg/kg) blocked mirfentanil (10.0 mg/kg)-induced analgesia up to 75% of the maximum analgesic effect. Norbinaltorphimine (10.0 mg/kg) partially blocked (35%) the maximum analgesic effect following mirfentanil (10.0 mg/kg) administration. Single doses of mirfentanil (0.1-32.0 mg/kg) had no effect on splenic NK activity. However, preadministration of mirfentanil (1.0-10.0 mg/kg) blocked morphine-induced suppression of splenic NK activity. Collectively, the results suggest that mirfentanil is a novel opioid that induces antinociception predominately through mu opioid receptors but, unlike morphine or fentanyl, does not suppress splenic NK activity.
Collapse
MESH Headings
- Adjuvants, Immunologic/pharmacology
- Analgesics/pharmacology
- Analgesics, Opioid/antagonists & inhibitors
- Analgesics, Opioid/pharmacology
- Animals
- Dose-Response Relationship, Drug
- Drug Interactions
- Female
- Fentanyl/analogs & derivatives
- Fentanyl/pharmacology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/physiology
- Mice
- Mice, Inbred ICR
- Morphine/antagonists & inhibitors
- Morphine/pharmacology
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/physiology
- Spleen/cytology
- Spleen/drug effects
- Spleen/physiology
Collapse
Affiliation(s)
- D J Carr
- Department of Microbiology, Immunology, Parasitology, Louisiana State University Medical Center, New Orleans 70112-1393, USA.
| | | | | | | | | |
Collapse
|
39
|
Gergen KA, Zadina JE, Kastin AJ, Paul D. Intrathecal Tyr-W-MIF-1 produces potent, naloxone-reversible analgesia modulated by alpha 2-adrenoceptors. Eur J Pharmacol 1996; 298:235-9. [PMID: 8846821 DOI: 10.1016/0014-2999(95)00823-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Spinal administration of morphine or [D-Ala2,MePhe4,Gly(ol)5)]enkephalin (DAMGO) produces potent, naloxone-reversible analgesia that is modulated by alpha 2-adrenoceptors. Tyr-W-MIF-1 (Tyr-Pro-Trp-Gly-NH2) is a naturally occurring, amidated tetrapeptide that is structurally related to the melanocyte-stimulating hormone release inhibiting factor-1 (MIF-1) family of endogenous peptides. Tyr-W-MIF-1 displays high selectivity for the mu-opioid receptor. We investigated the effect of spinal administration of Tyr-W-MIF-1 on analgesia using the mouse tail-flick assay. Intrathecal (i.t.) administration of Tyr-W-MIF-1 produced a dose-dependent analgesic response, with an ED50 of 0.41 microgram, that was reversed by naloxone. Pretreatment with the mu-opioid receptor-selective antagonist beta-funaltrexamine blocked the effect of i.t. Tyr-W-MIF-1. However, pretreatment with the mu1-opioid receptor-selective antagonist naloxonazine did not antagonize the analgesia, indicating the effect was mediated through spinal mu2-opioid receptors. Pretreatment with desipramine, an inhibitor of norepinephrine reuptake, potentiated the analgesic effect of i.t. Tyr-W-MIF-1, producing a 3.1-fold leftward shift in the dose-response curve. Spinal administration of yohimbine, an alpha 2-adrenoceptor-selective antagonist, significantly attenuated the analgesic effect of Tyr-W-MIF-1. Thus, the potent analgesic effect of i.t. Tyr-W-MIF-1 is mediated through spinal mu2-receptors, and is modulated by norepinephrine and alpha 2-adrenoceptors.
Collapse
|
40
|
Gatch MB, Liguori A, Negus SS, Mello NK, Bergman J, Liguori T. Naloxonazine antagonism of levorphanol-induced antinociception and respiratory depression in rhesus monkeys. Eur J Pharmacol 1996; 298:31-6. [PMID: 8867916 DOI: 10.1016/0014-2999(95)00769-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The mu-opioid receptor antagonist effects of naloxonazine on levorphanol-induced thermal antinociception and respiratory depression were examined in rhesus monkeys. Levorphanol (0.032-3.2 mg/kg) produced dose-dependent increases in tail-withdrawal latencies from 50 degrees C water in a warm-water tail-withdrawal assay and dose-dependent decreases in ventilation in both air and 5% CO2 mixed in air. Naloxonazine (0.1-3.0 mg/kg) antagonized both the antinociceptive and ventilatory effects of levorphanol to a similar degree, and the antagonist effects of naloxonazine were greater after 1 h than after 24 h. Under all conditions, the antagonist effects of naloxonazine were fully surmountable. Schild analysis of the antagonist effects of naloxonazine after 1 h pretreatment in the antinociception assay yielded a pA2 value of 7.6 and a slope of -0.50; by comparison, quadazocine yielded a pA2 value of 7.5 and a slope of -1.05. These results suggest that naloxonazine acts as a potent and fully reversible mu-opioid receptor antagonist with a moderately long duration of action in rhesus monkeys. In addition, these results suggest that the antinociceptive and ventilatory effects of mu-opioid receptor agonists in rhesus monkeys are mediated by pharmacologically similar populations of mu opioid receptors.
Collapse
Affiliation(s)
- M B Gatch
- Alcohol and Drug Abuse Research Center, McLean Hospital, Belmont, MA 02178, USA
| | | | | | | | | | | |
Collapse
|
41
|
Opioids and the Control of Pain. Neurotherapeutics 1996. [DOI: 10.1007/978-1-59259-466-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
42
|
Chien CC, Pasternak GW. (-)-Pentazocine analgesia in mice: interactions with a sigma receptor system. Eur J Pharmacol 1995; 294:303-8. [PMID: 8788445 DOI: 10.1016/0014-2999(95)00552-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
(-)-Pentazocine is active in the tailflick assay in CD-1 mice, although it shows a biphasic dose-response curve with a peak effect of only 30%. Co-administration of haloperidol shifts the dose-response curve to the left and elevates the maximal response to 70% through a blockade of sigma 1 receptors, but the curve remains biphasic. (+)-Pentazocine is inactive in all antinociceptive assays, either alone or with haloperidol. The analgesic actions of (-)-pentazocine are readily reversed by nor-binaltorphimine, but not by the mu-selective opioid receptor antagonist beta-funaltrexamine, implying a kappa 1-opioid receptor mechanism of action. This conclusion is supported by the ability of antisense oligodeoxynucleotides directed against the KOR-1 clone, which encodes the kappa 1-opioid receptor, to block (-)-pentazocine analgesia.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacokinetics
- Analgesics, Opioid/pharmacology
- Animals
- Base Sequence
- Dopamine Antagonists/pharmacology
- Dose-Response Relationship, Drug
- Haloperidol/pharmacology
- Male
- Mice
- Oligonucleotides, Antisense/pharmacology
- Pentazocine/pharmacokinetics
- Pentazocine/pharmacology
- Reaction Time/drug effects
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/metabolism
- Receptors, sigma/drug effects
- Receptors, sigma/metabolism
Collapse
Affiliation(s)
- C C Chien
- Department of Neurology and Neuroscience, Cornell II Medical College, New York, NY 10021, USA
| | | |
Collapse
|
43
|
Garzón J, Sánchez-Blázquez P. In vivo injection of antibodies directed against the cloned mu opioid receptor blocked supraspinal analgesia induced by mu-agonists in mice. Life Sci 1995; 56:PL237-42. [PMID: 7475889 DOI: 10.1016/0024-3205(95)00064-d] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The intracerebroventricular (i.c.v.) injection to mice of a polyclonal antibody raised against the peptide sequence 208-216 (TKYRQGSID) of cloned rat mu opioid receptor, reduced the analgesic potency of DAMGO, morphine and beta-endorphin-(1-31) when studied 48 h later in the tail-flick test. Antinociception elicited by delta agonists, DPDPE and [D-Ala2]-Deltorphin II, and by the kappa agonist U-50488H, was fully expressed in mice undergoing this treatment. The specific binding displayed by 0.6 nM [3H]-DAMGO was reduced in membranes preincubated with the antiserum, whereas no change could be detected for 3 nM [3H]-DPDPE or 2 nM [3H]-U-69593 labelling delta and kappa opioid receptors respectively. Naloxonazine, irreversible antagonist of the pharmacologically defined mu 1 opioid receptor, and beta-funaltrexamine (beta-FNA), that also displays irreversible antagonism at mu 1/2 receptors, when injected i.c.v. 24 h before the opioids significantly reduced the activity of DAMGO and morphine. In mice treated with naloxonazine, but not with beta-FNA, the antibody further reduced the remaining analgesic effect of DAMGO and morphine. Thus, both the antibody and beta-FNA blocked a wider population of mu opioid receptors than that tagged by naloxonazine.
Collapse
MESH Headings
- Amino Acid Sequence
- Analgesia
- Animals
- Antibodies/pharmacology
- Benzeneacetamides
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-
- Enkephalin, D-Penicillamine (2,5)-
- Enkephalins/metabolism
- Immunization, Passive
- Male
- Mice
- Molecular Sequence Data
- Morphine
- Naloxone/analogs & derivatives
- Naloxone/pharmacology
- Peptide Fragments/chemistry
- Peptide Fragments/immunology
- Pyrrolidines/metabolism
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/immunology
- Receptors, Opioid, mu/physiology
- beta-Endorphin
Collapse
Affiliation(s)
- J Garzón
- Neuropharmacology, Cajal Institute, C.S.I.C., Madrid, Spain
| | | |
Collapse
|
44
|
McIntosh C, Yago V, Kwok YN. Beta-funaltrexamine blockade of opioid-induced inhibition of somatostatin secretion from rat stomach. Eur J Pharmacol 1994; 259:331-4. [PMID: 7982463 DOI: 10.1016/0014-2999(94)90663-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Opioid peptides are potent inhibitors of gastric somatostatin secretion. In the current investigation the effect of mu-opioid receptor blockade on responses to [D-Ala2,N-Me-Phe4,Gly5-ol]enkephalin (DAGO) was studied. Gastric inhibitory polypeptide (GIP; 1 nM) -stimulated secretion of immunoreactive somatostatin was almost completely inhibited by DAGO (1 microM). The mu-receptor antagonists, beta-funaltrexamine and naloxonazine, blocked the effect of DAGO. Pretreatment of rats with beta-funaltrexamine, 24 h prior to perfusion, reduced the percentage inhibition by DAGO from 88.6 +/- 5.2% to 50.7 +/- 9.3%. These studies support the involvement of mu-opioid inhibitory receptors in the regulation of gastric somatostatin secretion.
Collapse
Affiliation(s)
- C McIntosh
- Department of Physiology, University of British Columbia, Vancouver, Canada
| | | | | |
Collapse
|
45
|
Abstract
Morphine is a potent analgesic when microinjected into the periaqueductal gray (PAG), the rostral ventral medulla (RVM) which contains the nuclei raphe magnus and reticularis gigantocellularis and the dorsolateral pons (DLP) which includes the locus coeruleus. Coadministration of low morphine doses which are inactive alone into combinations of these three regions elicits dramatic analgesic responses, implying the existence of synergy. The most effective combination is the PAG/RVM, whereas the PAG/DLP and RVM/DLP combinations are much less efficacious. In addition to fixed combinations, inclusion of a low morphine dose in one region shifts the analgesic dose-response curves in the others. The marked synergy between the PAG and the RVM is sensitive to naloxonazine, implying a role for mu 1 receptors. Thus, these studies indicate the presence of intrinsic brainstem mu 1 receptor systems with synergistic interactions which can be pharmacologically distinguished from the brainstem mu 2 receptors mediating supraspinal/spinal synergy.
Collapse
Affiliation(s)
- G C Rossi
- Neuropsychology Doctoral Sub Program, City University of New York, New York 10036
| | | | | |
Collapse
|
46
|
Tamaki T, Hasui K, Aki Y, Kimura S, Abe Y. Effects of NG-nitro-L-arginine on isolated rabbit afferent arterioles. JAPANESE JOURNAL OF PHARMACOLOGY 1993; 62:231-7. [PMID: 8411772 DOI: 10.1254/jjp.62.231] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We examined the effects of NG-nitro-L-arginine (L-NNA) on isolated rabbit afferent arterioles to confirm that nitric oxide is released at the resistance vessel level in the kidney. We microdissected the superficial afferent arterioles from the kidneys of New Zealand White rabbits. Each afferent arteriole was cannulated with a micropipette system, and the intraluminal pressure was set at 80 mmHg. By our methods, we found that norepinephrine (NE) decreased the lumen diameter of the afferent arterioles in a dose-dependent manner, and acetylcholine increased the lumen diameter of NE-constricted afferent arterioles. L-NNA (10(-4) M) gradually decreased the lumen diameter of afferent arterioles from 21.5 +/- 0.9 to 18.6 +/- 0.9 microns in 20 min, but NG-nitro-D-arginine (10(-4) M) did not affect them (from 21.8 +/- 1.3 to 21.8 +/- 1.5 microns). L-Arginine (10(-2) M) restored the lumen diameter of L-NNA-contracted afferent arterioles to the control levels. These findings indicate that the isolated afferent arteriole has the ability to release or to synthesize and release nitric oxide under basal conditions and that this basal release of nitric oxide plays an important role in the basal tone of the afferent arteriole.
Collapse
Affiliation(s)
- T Tamaki
- Department of Pharmacology, Kagawa Medical School, Japan
| | | | | | | | | |
Collapse
|
47
|
|
48
|
Tive LA, Pick CG, Paul D, Roques BP, Gacel GA, Pasternak GW. Analgesic potency of TRIMU-5: a mixed mu 2 opioid receptor agonist/mu 1 opioid receptor antagonist. Eur J Pharmacol 1992; 216:249-55. [PMID: 1327812 DOI: 10.1016/0014-2999(92)90367-d] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
TRIMU-5 (Tyr-D-Ala-Gly-NH-(CH2)2CH(CH3)2) is a potent enkephalin analog with analgesic actions. Detailed studies show high affinity for both mu 1 and mu 2 sites, with poor affinity for delta, kappa 1 and kappa 3 receptors. Of all the mu ligands examined in binding assays, TRIMU-5 was the most mu-selective. In mice, TRIMU-5 administered either intracerebroventricularly (i.c.v.) or intrathecally elicited analgesia which was readily reversed by the mu-selective antagonist beta-funaltrexamine (beta-FNA). However, the analgesia observed following i.c.v. injections differed from traditional mu ligands: (1) the dose of drug required for analgesic activity i.c.v. was 100-fold greater than those following intrathecal administration; (2) although sensitive to beta-FNA, the analgesia was not antagonized by naloxonazine; and (3) the analgesia was reversed by an opioid antagonist given intrathecally (i.t.) but not i.c.v. Thus, TRIMU-5 analgesia appeared to be mediated spinally through mu 2 receptors. TRIMU-5 did have supraspinal actions, inhibiting gastrointestinal transit, another mu 2 action. In binding studies TRIMU-5 had high affinity for mu 1 sites, but pharmacological studies revealed antagonist actions at this receptor. In mice, the analgesia produced by morphine given i.c.v. was antagonized by coinjection of a low TRIMU-5 dose which was inactive alone. Similarly, TRIMU-5 coadministered with morphine into the periaqueductal gray of rats reversed the analgesia seen with morphine alone. Thus, TRIMU-5 is a highly selective mixed mu 2 opioid receptor agonist/mu 1 opioid receptor antagonist.
Collapse
Affiliation(s)
- L A Tive
- Cotzias Laboratory of Neuro-Oncology, Memorial Sloan-Kettering Cancer Center, New York 10021
| | | | | | | | | | | |
Collapse
|
49
|
Pick CG, Paul D, Eison MS, Pasternak GW. Potentiation of opioid analgesia by the antidepressant nefazodone. Eur J Pharmacol 1992; 211:375-81. [PMID: 1319913 DOI: 10.1016/0014-2999(92)90395-k] [Citation(s) in RCA: 56] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Nefazodone is a new antidepressant related structurally to trazodone. In addition to its activity in preclinical assays for antidepressant activity, nefazodone was a potent analgesic in the mouse hotplate assay. At 50 mg/kg s.c. nefazodone doubled baseline latencies in 40% of mice but was inactive in the tailflick test at any dose tested. The hotplate analgesia seen with nefazodone alone was not reversed by naloxone (10 mg/kg s.c.). In the tailflick assay, nefazodone (50 mg/kg s.c.) enhanced morphine's analgesic response, shifting morphine's ED50 from 3.1 mg/kg alone to 0.86 mg/kg in conjunction with nefazodone (P less than 0.05). Two days after implantation of a morphine pellet (75 mg) no mice remained analgesic in the tailflick assay. Administration of nefazodone (50 mg/kg s.c.) restored analgesia to 60% of mice (P less than 0.03). In selective analgesic assays, nefazodone enhanced mu 1, mu 2 and delta analgesia, but not kappa 1 or kappa 3 analgesia. Nefazodone did not affect morphine's LD50 and, in assays of gastrointestinal transit, nefazodone increased morphine's potency only slightly. In conclusion, nefazodone alone is analgesic in certain animal models. In conjunction with morphine, nefazodone potentiated analgesia with no effect on lethality and little effect on gastrointestinal transit, resulting in an increase in morphine's therapeutic index. These results suggest that nefazodone and similar agents may have a significant role in the management of pain.
Collapse
Affiliation(s)
- C G Pick
- Cotzias Laboratory of Neuro-Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021
| | | | | | | |
Collapse
|