Dopamine Partial Agonist Reverses Amphetamine Withdrawal in Rats Cristina Orsini, Ph.D., George F

Total Page:16

File Type:pdf, Size:1020Kb

Dopamine Partial Agonist Reverses Amphetamine Withdrawal in Rats Cristina Orsini, Ph.D., George F BRIEF REPORT Dopamine Partial Agonist Reverses Amphetamine Withdrawal in Rats Cristina Orsini, Ph.D., George F. Koob, Ph.D. and Luigi Pulvirenti, M.D. Decreased motivation to work for a natural reward is a sign reversed the decrease in responding for the sweet solution. of amphetamine withdrawal and is thought to be associated These results suggest that dopamine partial agonists, with hypofunction of the mesolimbic dopamine system. possibly due to their agonistic-like actions under these During withdrawal from repeated amphetamine conditions, are a potential therapeutic approach for the administration, rats showed reduced responding for a sweet acute withdrawal stage of the amphetamine addition cycle. solution in a progressive ratio schedule. Repeated systemic [Neuropsychopharmacology 25:789–792, 2001] treatment with terguride (0.2 and 0.4 mg/kg, i.p.) twice © 2001 American College of Neuropsychopharmacology. daily during the first four days of amphetamine withdrawal Published by Elsevier Science Inc. KEY WORDS: Dopamine; Partial agonist; Amphetamine; limited efficacy has been reported in both preclinical Withdrawal; Terguride; Dependence models and clinical trials with dopamine agonists and antagonists (for review see Withers et al. 1995; Mello Psychostimulant withdrawal in humans is associated and Negus 1996). with fatigue, depression of mood, anhedonia, and psy- Partial dopamine agonists bind to the dopamine re- chomotor retardation and is a critical component of the ceptor with high affinity but low intrinsic activity dependence cycle, but no pharmacotherapeutic treat- (Hoyer and Boddeke 1993), and have been hypothe- ments are currently available for psychostimulant with- sized to act as functional antagonists in conditions of drawal. In rats, psychomotor retardation and decreased high dopaminergic tone and as functional agonists in motivation to work for a natural reward are signs of conditions of DA depletion (Clark et al. 1991; Svensson amphetamine withdrawal (Pulvirenti and Koob 1993; et al. 1991; Pulvirenti and Koob 1994). Within the con- Barr and Phillips 1999). Although dopamine (DA) me- text of drug dependence, the DA D2 receptor partial ag- solimbic neurotransmission appears to be critically in- onists terguride and SDZ 208-911 showed activity as volved in psychostimulant dependence (Wise and functional antagonists in rats trained to self-administer Bozarth 1987; Wise 1996; Koob and Le Moal 1997), only cocaine and amphetamine under different schedules (Pulvirenti and Koob 1994; Pulvirenti et al. 1998; Izzo et al. 2001). However, DA partial agonists might restore From the Department of Neuropharmacology The Scripps the relative functional hypoactivity of the dopamine Research Institute (CO, GFK, LP) and Claude Bernard Neuroscience system during psychostimulant withdrawal (Pulvirenti Institute (LP), La Jolla, CA. and Koob 1994). Address correspondence to: Luigi Pulvirenti, M.D., Department of Neuropharmacology CVN-7, The Scripps Research Institute, The aim of the present study was to investigate the 10550 North Torrey Pines Rd., La Jolla, CA 92037. effects of terguride using a model of amphetamine Received August 26, 2000; revised April 6, 2001; accepted April withdrawal (Barr and Phillips 1999) from sustained, 17, 2001. Online publication: 4/19/01 at www.acnp.org/citations/Npp high doses of amphetamine shown to produce a de- 041901108. crease in operant responding for a sweet solution. NEUROPSYCHOPHARMACOLOGY 2001–VOL. 25, NO. 5 © 2001 American College of Neuropsychopharmacology Published by Elsevier Science Inc. 0893-133X/01/$–see front matter 655 Avenue of the Americas, New York, NY 10010 SSDI S0893-133X(01)00270-6 790 C. Orsini et al. NEUROPSYCHOPHARMACOLOGY 2001–VOL. 25, NO. 5 METHODS RESULTS This study was performed in accordance with the Na- Figure 1 shows the effects of treatment with terguride tional Institutes of Health Guide for the Care and Use of on PR responding for a sweet solution. Overall ANOVA Laboratory Animals. Male Wistar rats (Charles River) revealed that there was a significant difference between were housed on a reversed light/dark cycle (lights on treatment groups: treatment with terguride prevented 10 PM to 10 AM). Testing was performed between 2 PM the reduction of BP (expressed as total reinforcement and 4 PM. Following an initial 48-h exposure to a sweet earned) in rats treated with amphetamine [F(2,162) ϭ solution [3% sucrose (Fisher Scientific) and 0.125% sac- 3.296, p Ͻ .05] and post-hoc analysis revealed that sta- charin (Sigma Chemical Co), all rats were water- tistical significance was reached for the dose of tergu- deprived for 20 h daily before being placed in standard ride 0.4 mg/kg (p Ͻ .05 vs. saline, Tukey’s test). In addi- operant chambers (Coulburn Instruments) (Roberts et tion, comparison at the various levels revealed a al. 2000). Subjects were placed on a progressive-ratio significant reduction of BP (expressed as total reinforce- (PR) schedule for the sweet solution (0.1 ml per rein- ment earned) in rats treated with amphetamine and sa- forcer) for 2 h daily [following initial training on a line [F(4,48) ϭ 3.98, p Ͻ .01] and statistical significance fixed-ratio (Barr and Phillips 1999)] and the response was reached during the first two days of withdrawal (p Ͻ requirement increased according to the following pro- .01, Tukey’s test). gression: 1, 3, 6, 10, 15, 20, 25, 32, 40, 50, 62, 77, 95, 118, ANOVA revealed no significant reduction of break- 145, 175, 205, 235, 265, etc. ing point in rats treated with amphetamine and tergu- The breaking point (BP) was defined as the last ra- ride at the doses of 0.2 mg/kg [F(4,36) ϭ 1.74, NS] and tio attained prior to a 1-h period during which a ratio 0.4 mg/kg [F(4,36) ϭ 2.59, NS]. Finally, Tukey’s test re- was not completed. When stable responding was vealed a significant difference between terguride at the achieved (Ϯ 10% of reinforcements within three con- secutive days and BP Ͼ 16) the rats were injected in- traperitoneally (i.p.) three times per day (9 AM, 5 PM, and 12 AM), starting with a dose of 1 mg/kg of amphetamine and increasing by 1 mg/kg per injec- tion for 10 injections (Barr and Phillips 1999). After the last injection of amphetamine the rats were treated i.p. twice daily (9:30 AM and 9:30 PM) for four days with either saline (n ϭ 13) or terguride at the dose of 0.2 (n ϭ 10) or 0.4 mg/kg (n ϭ 10). PR re- sponding was monitored for the first four days of withdrawal (Pulvirenti and Koob 1993; Barr and Phil- lips 1999). A separate group of rats was trained as described and exposed to three days of saline administration ac- cording to the same amphetamine schedule described above. These rats then were treated with an effective dose of terguride (0.2 mg/kg; n ϭ 7) and PR responding was monitored as in the above experiment. Doses and schedules of terguride administration were chosen ac- cording to previous studies (Pulvirenti and Koob 1993; Pulvirenti et al. 1998). Terguride (Research Biochemicals International) was dissolved in distilled water and 1N HCl, and pH ad- justed to 5.5 with 1M NAOH. d-amphetamine sulfate Figure 1. Effect of repeated treatment with terguride (0.2 and (Sigma) was dissolved in saline and all drugs were in- 0.4 mg/kg, i.p. twice daily for four days) on responding for ϩ jected in a volume of 1.0 ml/kg. Data analyses were a sweet solution (3% sucrose 0.125% saccharin) under a progressive-ratio schedule of reinforcement. Values repre- performed using analysis of variance (ANOVA) as de- sent means Ϯ SEM of breaking points and cumulative total scribed by Pulvirenti and Koob (1993). First an overall responses (right) (saline, n ϭ 13; terguride 0.2 mg/kg, n ϭ analysis was performed to directly explore differences 10; terguride 0.4 mg/kg, n ϭ 10). * p Ͻ .01 Tukey’s test vs. between treatment groups. This was followed by com- baseline following significant ANOVA. Tukey’s test also parison at the various levels to explore at what time revealed a significant difference between terguride at the point (day) statistical significance was reached. Post- dose of 0.4 mg/kg and saline at Days 2, 3, and 4 (p Ͻ .05) but hoc Tukey’s test was used where appropriate. not at Day 1 (not indicated). NEUROPSYCHOPHARMACOLOGY 2001–VOL. 25, NO. 5 DA Partial Agonist and Amphetamine Withdrawal 791 Table 1. Effects of Repeated Treatment with Terguride on fects of lisuride on psychomotor retardation were evi- Progressive-Ratio Responding for a Sweet Solution in Rats dent since the first day of treatment. The reason for Not Previously Exposed to Amphetamine these differences is not presently clear, although it is possible that since terguride is a partial agonist its stim- Day ulatory effects on dopamine function may require re- Baseline 1234peated treatment to develop. Terguride 7.4 7.6 7.4 7.0 7.4 Although psychomotor retardation is also a compo- (0.2 mg/kg) Ϯ 0.35 Ϯ 0.81 Ϯ 0.72 Ϯ 0.79 Ϯ 0.65 nent of amphetamine withdrawal (Paulson et al. 1991; Pulvirenti and Koob 1993), it is unlikely that motor im- Ϯ ϭ Values represent means SEM of breaking points (n 7). pairment is solely responsible for the effects reported here and previously (Barr and Phillips 1999). Indeed rats have been shown to maintain high rates of operant dose of 0.4 mg/kg and saline at Days 2, 3, and 4 (p Ͻ responding for intracranial self-stimulation during psy- .05) but not at Day 1. chostimulant withdrawal (Markou and Koob 1992). Ad- Table 1 shows that terguride did not significantly ditionally, the pattern of sucrose consumption reported modify the total reinforcement earned on PR respond- by Barr and Phillips (1999) did not resemble the pattern ing for a sweet solution in animals not previously ex- of consummatory behavior observed after administra- posed to amphetamine [F(4,24)Ͻ1, NS].
Recommended publications
  • INVESTIGATION of NATURAL PRODUCT SCAFFOLDS for the DEVELOPMENT of OPIOID RECEPTOR LIGANDS by Katherine M
    INVESTIGATION OF NATURAL PRODUCT SCAFFOLDS FOR THE DEVELOPMENT OF OPIOID RECEPTOR LIGANDS By Katherine M. Prevatt-Smith Submitted to the graduate degree program in Medicinal Chemistry and the Graduate Faculty of the University of Kansas in partial fulfillment of the requirements for the degree of Doctor of Philosophy. _________________________________ Chairperson: Dr. Thomas E. Prisinzano _________________________________ Dr. Brian S. J. Blagg _________________________________ Dr. Michael F. Rafferty _________________________________ Dr. Paul R. Hanson _________________________________ Dr. Susan M. Lunte Date Defended: July 18, 2012 The Dissertation Committee for Katherine M. Prevatt-Smith certifies that this is the approved version of the following dissertation: INVESTIGATION OF NATURAL PRODUCT SCAFFOLDS FOR THE DEVELOPMENT OF OPIOID RECEPTOR LIGANDS _________________________________ Chairperson: Dr. Thomas E. Prisinzano Date approved: July 18, 2012 ii ABSTRACT Kappa opioid (KOP) receptors have been suggested as an alternative target to the mu opioid (MOP) receptor for the treatment of pain because KOP activation is associated with fewer negative side-effects (respiratory depression, constipation, tolerance, and dependence). The KOP receptor has also been implicated in several abuse-related effects in the central nervous system (CNS). KOP ligands have been investigated as pharmacotherapies for drug abuse; KOP agonists have been shown to modulate dopamine concentrations in the CNS as well as attenuate the self-administration of cocaine in a variety of species, and KOP antagonists have potential in the treatment of relapse. One drawback of current opioid ligand investigation is that many compounds are based on the morphine scaffold and thus have similar properties, both positive and negative, to the parent molecule. Thus there is increasing need to discover new chemical scaffolds with opioid receptor activity.
    [Show full text]
  • Emulated Clinical Trials from Longitudinal Real-World Data Efficiently Identify Candidates for Neurological Disease Modification
    medRxiv preprint doi: https://doi.org/10.1101/2020.08.11.20171447; this version posted November 20, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. Emulated Clinical Trials from Longitudinal Real-World Data Efficiently Identify Candidates for Neurological Disease Modification: Examples from Parkinson's Disease Daphna Laifenfeld, PhD1,a,†, Chen Yanover, PhD2,b,†, Michal Ozery-Flato, PhD3,†, Oded Shaham, PhD2,c, Michal Rozen-Zvi, PhD3,4,*, Nirit Lev, MD/PhD1,d,‡ , Yaara Goldschmidt, PhD2,e,‡, Iris Grossman, PhD1,f,‡ 1Formerly Teva Pharmaceuticals Research & Development, Petach Tikva, Israel; 2Formerly IBM Research – Haifa, Israel; 3IBM Research – Haifa, Israel; 4Faculty of Medicine, The HeBrew University, Jerusalem, Israel; aPresent Address: Ibex Medical Analytics, Tel Aviv, Israel; bPresent Address: KI Research Institute, Kfar Malal, Israel; cPresent Address: MeMed Dx, Haifa, Israel; dPresent Address: Meir Medical Center and Sackler Faculty of Medicine Tel Aviv University, Tel Aviv, Israel; ePresent Address: K Health, Tel Aviv, Israel; fPresent Address: Gross Advantage, Brookline, MA, USA; † These authors contriButed equally and share first authorship. ‡ These authors contriButed equally to this work. * Corresponding author: Michal Rosen-Zvi [email protected] Haifa University Campus, Mount Carmel Haifa, 3498825, Israel Tel: (972) 04-8296517 Keywords: Real World Data; Clinical Evidence; Parkinson’s disease; Artificial Intelligence; Causal Inference; Rasagiline; Zolpidem; Disease Modifying Therapeutics; Repurposing. NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice.
    [Show full text]
  • From NMDA Receptor Hypofunction to the Dopamine Hypothesis of Schizophrenia J
    REVIEW The Neuropsychopharmacology of Phencyclidine: From NMDA Receptor Hypofunction to the Dopamine Hypothesis of Schizophrenia J. David Jentsch, Ph.D., and Robert H. Roth, Ph.D. Administration of noncompetitive NMDA/glutamate effects of these drugs are discussed, especially with regard to receptor antagonists, such as phencyclidine (PCP) and differing profiles following single-dose and long-term ketamine, to humans induces a broad range of exposure. The neurochemical effects of NMDA receptor schizophrenic-like symptomatology, findings that have antagonist administration are argued to support a contributed to a hypoglutamatergic hypothesis of neurobiological hypothesis of schizophrenia, which includes schizophrenia. Moreover, a history of experimental pathophysiology within several neurotransmitter systems, investigations of the effects of these drugs in animals manifested in behavioral pathology. Future directions for suggests that NMDA receptor antagonists may model some the application of NMDA receptor antagonist models of behavioral symptoms of schizophrenia in nonhuman schizophrenia to preclinical and pathophysiological research subjects. In this review, the usefulness of PCP are offered. [Neuropsychopharmacology 20:201–225, administration as a potential animal model of schizophrenia 1999] © 1999 American College of is considered. To support the contention that NMDA Neuropsychopharmacology. Published by Elsevier receptor antagonist administration represents a viable Science Inc. model of schizophrenia, the behavioral and neurobiological KEY WORDS: Ketamine; Phencyclidine; Psychotomimetic; widely from the administration of purportedly psychot- Memory; Catecholamine; Schizophrenia; Prefrontal cortex; omimetic drugs (Snyder 1988; Javitt and Zukin 1991; Cognition; Dopamine; Glutamate Jentsch et al. 1998a), to perinatal insults (Lipska et al. Biological psychiatric research has seen the develop- 1993; El-Khodor and Boksa 1997; Moore and Grace ment of many putative animal models of schizophrenia.
    [Show full text]
  • Principle of Pharmacodynamics
    Principle of pharmacodynamics Dr. M. Emamghoreishi Full Professor Department of Pharmacology Medical School Shiraz University of Medical Sciences Email:[email protected] Reference: Basic & Clinical Pharmacology: Bertrum G. Katzung and Anthony J. Treveror, 13th edition, 2015, chapter 20, p. 336-351 Learning Objectives: At the end of sessions, students should be able to: 1. Define pharmacology and explain its importance for a clinician. 2. Define ―drug receptor‖. 3. Explain the nature of drug receptors. 4. Describe other sites of drug actions. 5. Explain the drug-receptor interaction. 6. Define the terms ―affinity‖, ―intrinsic activity‖ and ―Kd‖. 7. Explain the terms ―agonist‖ and ―antagonist‖ and their different types. 8. Explain chemical and physiological antagonists. 9. Explain the differences in drug responsiveness. 10. Explain tolerance, tachyphylaxis, and overshoot. 11. Define different dose-response curves. 12. Explain the information that can be obtained from a graded dose-response curve. 13. Describe the potency and efficacy of drugs. 14. Explain shift of dose-response curves in the presence of competitive and irreversible antagonists and its importance in clinical application of antagonists. 15. Explain the information that can be obtained from a quantal dose-response curve. 16. Define the terms ED50, TD50, LD50, therapeutic index and certain safety factor. What is Pharmacology?It is defined as the study of drugs (substances used to prevent, diagnose, and treat disease). Pharmacology is the science that deals with the interactions betweena drug and the bodyor living systems. The interactions between a drug and the body are conveniently divided into two classes. The actions of the drug on the body are termed pharmacodynamicprocesses.These properties determine the group in which the drug is classified, and they play the major role in deciding whether that group is appropriate therapy for a particular symptom or disease.
    [Show full text]
  • Inhibits Dyskinesia Expression and Normalizes Motor Activity in 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine-Treated Primates
    The Journal of Neuroscience, October 8, 2003 • 23(27):9107–9115 • 9107 Behavioral/Systems/Cognitive 3,4-Methylenedioxymethamphetamine (Ecstasy) Inhibits Dyskinesia Expression and Normalizes Motor Activity in 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine-Treated Primates Mahmoud M. Iravani, Michael J. Jackson, Mikko Kuoppama¨ki, Lance A. Smith, and Peter Jenner Neurodegenerative Disease Research Centre, Guy’s, King’s, and St. Thomas’ School of Biomedical Sciences, King’s College, London SE1 1UL, United Kingdom Ecstasy [3,4-methylenedioxymethamphetamine (MDMA)] was shown to prolong the action of L-3,4-dihydroxyphenylalanine (L-DOPA) while suppressing dyskinesia in a single patient with Parkinson’s disease (PD). The clinical basis of this effect of MDMA is unknown but may relate to its actions on either dopaminergic or serotoninergic systems in brain. In normal, drug-naive common marmosets, MDMA administration suppressed motor activity and exploratory behavior. In 1-methyl- 4-phenyl-1,2,3,6-tetrahydropyridine(MPTP)-treated, L-DOPA-primedcommonmarmosets,MDMAtransientlyrelievedmotordisability but over a period of 60 min worsened motor symptoms. When given in conjunction with L-DOPA, however, MDMA markedly decreased dyskinesia by reducing chorea and to a lesser extent dystonia and decreased locomotor activity to the level observed in normal animals. MDMA similarly alleviated dyskinesia induced by the selective dopamine D2/3 agonist pramipexole. The actions of MDMA appeared to be mediated through 5-HT mechanisms because its effects were fully blocked by the selective serotonin reuptake inhibitor fluvoxamine. Furthermore,theeffectofMDMAon L-DOPA-inducedmotoractivityanddyskinesiawaspartiallyinhibitedby5-HT1a/bantagonists.The ability of MDMA to inhibit dyskinesia results from its broad spectrum of action on 5-HT systems.
    [Show full text]
  • The Effects of Microdose LSD on Time Perception: a Randomised, Double-Blind, Placebo-Controlled Trial
    Psychopharmacology https://doi.org/10.1007/s00213-018-5119-x ORIGINAL INVESTIGATION The effects of microdose LSD on time perception: a randomised, double-blind, placebo-controlled trial Steliana Yanakieva1 & Naya Polychroni1 & Neiloufar Family2 & Luke T. J. Williams2,3 & David P. Luke4 & Devin B. Terhune1,5 Received: 23 May 2018 /Accepted: 9 November 2018 # The Author(s) 2018 Abstract Rationale Previous research demonstrating that lysergic acid diethylamide (LSD) produces alterations in time perception has implications for its impact on conscious states and a range of psychological functions that necessitate precise interval timing. However, interpretation of this research is hindered by methodological limitations and an inability to dissociate direct neuro- chemical effects on interval timing from indirect effects attributable to altered states of consciousness. Methods We conducted a randomised, double-blind, placebo-controlled study contrasting oral administration of placebo with three microdoses of LSD (5, 10, and 20 μg) in older adults. Subjective drug effects were regularly recorded and interval timing was assessed using a temporal reproduction task spanning subsecond and suprasecond intervals. Results LSD conditions were not associated with any robust changes in self-report indices of perception, mentation, or concen- tration. LSD reliably produced over-reproduction of temporal intervals of 2000 ms and longer with these effects most pronounced in the 10 μg dose condition. Hierarchical regression analyses indicated that LSD-mediated over-reproduction was independent of marginal differences in self-reported drug effects across conditions. Conclusions These results suggest that microdose LSD produces temporal dilation of suprasecond intervals in the absence of subjective alterations of consciousness. Keywords Interval timing .
    [Show full text]
  • Pharmacodynamics Drug Receptor Interactions Part-2
    Pharmacodynamics: (Drug Receptor Interactions, Part 2) ………………………………………………………………………………………………………………………………………………………………………………………………………………… VPT: Unit I; Lecture-22 (Dated 03.12.2020) Dr. Nirbhay Kumar Asstt. Professor & Head Deptt. of Veterinary Pharmacology & Toxicology Bihar Veterinary College, Bihar Animal Sciences University, Patna Drug Receptor Interactions Agonist It is a drug that possesses affinity for a particular receptor and causes a change in the receptor that result in an observable effect. Full agonist: Produces a maximal response by occupying all or a fraction of receptors. (Affinity=1, Efficacy=1) Partial agonist: Produces less than a maximal response even when the drug occupies all of the receptors. (Affinity=1, Efficacy= 0 to 1) Inverse agonist: Activates a receptor to produce an effect in the opposite direction to that of the well recognized agonist. (Affinity=1, Efficacy= –1 to 0). Source: Rang & Dale’s Pharmacology, Elsevier Source: Good & Gilman’s The Pharmacological Basis of Therapeutics, 13th Edn. Antagonist An antagonist is a drug that blocks the response produced by an agonist. Antagonists interact with the receptor or other components of the effector mechanism, but antagonists are devoid of intrinsic activity (Affinity=1, Efficacy=0). Antagonist contd… Competitive Antagonism: It is completely reversible; an increase in the concentration of the agonist in the bio-phase will overcome the effect of the antagonist. Example: Atropine (Antimuscarinic agent) Diphenhydramine (H1 receptor blocker) Non-competitive antagonism: The agonist has no influence upon the degree of antagonism or its reversibility. Example: Platelet inhibiting action of aspirin (The thromboxane synthase enzyme of platelets is irreversibly inhibited by aspirin, a process that is reversed only by production of new platelets).
    [Show full text]
  • Measuring Ligand Efficacy at the Mu- Opioid Receptor Using A
    RESEARCH ARTICLE Measuring ligand efficacy at the mu- opioid receptor using a conformational biosensor Kathryn E Livingston1,2, Jacob P Mahoney1,2, Aashish Manglik3, Roger K Sunahara4, John R Traynor1,2* 1Department of Pharmacology, University of Michigan Medical School, Ann Arbor, United States; 2Edward F Domino Research Center, University of Michigan, Ann Arbor, United States; 3Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, United States; 4Department of Pharmacology, University of California San Diego School of Medicine, La Jolla, United States Abstract The intrinsic efficacy of orthosteric ligands acting at G-protein-coupled receptors (GPCRs) reflects their ability to stabilize active receptor states (R*) and is a major determinant of their physiological effects. Here, we present a direct way to quantify the efficacy of ligands by measuring the binding of a R*-specific biosensor to purified receptor employing interferometry. As an example, we use the mu-opioid receptor (m-OR), a prototypic class A GPCR, and its active state sensor, nanobody-39 (Nb39). We demonstrate that ligands vary in their ability to recruit Nb39 to m- OR and describe methadone, loperamide, and PZM21 as ligands that support unique R* conformation(s) of m-OR. We further show that positive allosteric modulators of m-OR promote formation of R* in addition to enhancing promotion by orthosteric agonists. Finally, we demonstrate that the technique can be utilized with heterotrimeric G protein. The method is cell- free, signal transduction-independent and is generally applicable to GPCRs. DOI: https://doi.org/10.7554/eLife.32499.001 *For correspondence: [email protected] Competing interests: The authors declare that no Introduction competing interests exist.
    [Show full text]
  • Response Vs. Log [L] - Full Agonist
    DavidsonX – D001x – Medicinal Chemistry Chapter 5 – Receptors Part 2 – Ligands Video Clip – Ligand Types Ligands can have different effects on a receptor. Each type of ligand can be readily classified according to its behavior. A type of ligand is the full agonist. The term agonist refers to a compound that binds a receptor and elicits a response (E). Full agonists elicit the same level of full response (E = Emax = 100%) as the endogenous ligand of the receptor. Graphically, a receptor-ligand interaction is plotted as response (E/Emax) vs. log [L]. The relationship is sigmoidal. A full agonist approaches full response (E/Emax = 1.0) as log [L] reaches relatively high levels. response vs. log [L] - full agonist 1 0.9 0.8 0.7 x a 0.6 m E 0.5 / E 0.4 0.3 0.2 0.1 0 log [L] Two ligands can achieve a full response without being equivalent. Ligands can differ with respect to the concentration required to trigger a response. A ligand that affects a response at a lower concentration has a higher potency. Potencies are measured as the effective ligand concentration required to reach a 50% response – EC50 or, in these graphs, log [EC50]. A more potent ligand has a lower EC50 value. full agonist comparison 1 0.9 full agonist 1 0.8 (more potent) full agonist 2 0.7 (less potent) x a 0.6 log EC m 50 E 0.5 / E 0.4 0.3 log EC 0.2 50 0.1 0 log [L] Partial agonists also cause a response, but they cannot reach the same, 100% response level of the endogenous ligand.
    [Show full text]
  • The Use of Stems in the Selection of International Nonproprietary Names (INN) for Pharmaceutical Substances
    WHO/PSM/QSM/2006.3 The use of stems in the selection of International Nonproprietary Names (INN) for pharmaceutical substances 2006 Programme on International Nonproprietary Names (INN) Quality Assurance and Safety: Medicines Medicines Policy and Standards The use of stems in the selection of International Nonproprietary Names (INN) for pharmaceutical substances FORMER DOCUMENT NUMBER: WHO/PHARM S/NOM 15 © World Health Organization 2006 All rights reserved. Publications of the World Health Organization can be obtained from WHO Press, World Health Organization, 20 Avenue Appia, 1211 Geneva 27, Switzerland (tel.: +41 22 791 3264; fax: +41 22 791 4857; e-mail: [email protected]). Requests for permission to reproduce or translate WHO publications – whether for sale or for noncommercial distribution – should be addressed to WHO Press, at the above address (fax: +41 22 791 4806; e-mail: [email protected]). The designations employed and the presentation of the material in this publication do not imply the expression of any opinion whatsoever on the part of the World Health Organization concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted lines on maps represent approximate border lines for which there may not yet be full agreement. The mention of specific companies or of certain manufacturers’ products does not imply that they are endorsed or recommended by the World Health Organization in preference to others of a similar nature that are not mentioned. Errors and omissions excepted, the names of proprietary products are distinguished by initial capital letters.
    [Show full text]
  • Pharmacokinetics and Pharmacology of Drugs Used in Children
    Drug and Fluid Th erapy SECTION II Pharmacokinetics and Pharmacology of Drugs Used CHAPTER 6 in Children Charles J. Coté, Jerrold Lerman, Robert M. Ward, Ralph A. Lugo, and Nishan Goudsouzian Drug Distribution Propofol Protein Binding Ketamine Body Composition Etomidate Metabolism and Excretion Muscle Relaxants Hepatic Blood Flow Succinylcholine Renal Excretion Intermediate-Acting Nondepolarizing Relaxants Pharmacokinetic Principles and Calculations Atracurium First-Order Kinetics Cisatracurium Half-Life Vecuronium First-Order Single-Compartment Kinetics Rocuronium First-Order Multiple-Compartment Kinetics Clinical Implications When Using Short- and Zero-Order Kinetics Intermediate-Acting Relaxants Apparent Volume of Distribution Long-Acting Nondepolarizing Relaxants Repetitive Dosing and Drug Accumulation Pancuronium Steady State Antagonism of Muscle Relaxants Loading Dose General Principles Central Nervous System Effects Suggamadex The Drug Approval Process, the Package Insert, and Relaxants in Special Situations Drug Labeling Opioids Inhalation Anesthetic Agents Morphine Physicochemical Properties Meperidine Pharmacokinetics of Inhaled Anesthetics Hydromorphone Pharmacodynamics of Inhaled Anesthetics Oxycodone Clinical Effects Methadone Nitrous Oxide Fentanyl Environmental Impact Alfentanil Oxygen Sufentanil Intravenous Anesthetic Agents Remifentanil Barbiturates Butorphanol and Nalbuphine 89 A Practice of Anesthesia for Infants and Children Codeine Antiemetics Tramadol Metoclopramide Nonsteroidal Anti-infl ammatory Agents 5-Hydroxytryptamine
    [Show full text]
  • Anew Drug Design Strategy in the Liht of Molecular Hybridization Concept
    www.ijcrt.org © 2020 IJCRT | Volume 8, Issue 12 December 2020 | ISSN: 2320-2882 “Drug Design strategy and chemical process maximization in the light of Molecular Hybridization Concept.” Subhasis Basu, Ph D Registration No: VB 1198 of 2018-2019. Department Of Chemistry, Visva-Bharati University A Draft Thesis is submitted for the partial fulfilment of PhD in Chemistry Thesis/Degree proceeding. DECLARATION I Certify that a. The Work contained in this thesis is original and has been done by me under the guidance of my supervisor. b. The work has not been submitted to any other Institute for any degree or diploma. c. I have followed the guidelines provided by the Institute in preparing the thesis. d. I have conformed to the norms and guidelines given in the Ethical Code of Conduct of the Institute. e. Whenever I have used materials (data, theoretical analysis, figures and text) from other sources, I have given due credit to them by citing them in the text of the thesis and giving their details in the references. Further, I have taken permission from the copyright owners of the sources, whenever necessary. IJCRT2012039 International Journal of Creative Research Thoughts (IJCRT) www.ijcrt.org 284 www.ijcrt.org © 2020 IJCRT | Volume 8, Issue 12 December 2020 | ISSN: 2320-2882 f. Whenever I have quoted written materials from other sources I have put them under quotation marks and given due credit to the sources by citing them and giving required details in the references. (Subhasis Basu) ACKNOWLEDGEMENT This preface is to extend an appreciation to all those individuals who with their generous co- operation guided us in every aspect to make this design and drawing successful.
    [Show full text]