1
|
McCarthy DM, Spencer TJ, Bhide PG. Preclinical Models of Attention Deficit Hyperactivity Disorder: Neurobiology, Drug Discovery, and Beyond. J Atten Disord 2024; 28:880-894. [PMID: 38084074 DOI: 10.1177/10870547231215286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
OBJECTIVE We offer an overview of ADHD research using mouse models of nicotine exposure. METHOD Nicotine exposure of C57BL/6 or Swiss Webster mice occurred during prenatal period only or during the prenatal and the pre-weaning periods. Behavioral, neuroanatomical and neurotransmitter assays were used to investigate neurobiological mechanisms of ADHD and discover candidate ADHD medications. RESULTS Our studies show that norbinaltorphimine, a selective kappa opioid receptor antagonist is a candidate novel non-stimulant ADHD treatment and that a combination of methylphenidate and naltrexone has abuse deterrent potential with therapeutic benefits for ADHD. Other studies showed transgenerational transmission of ADHD-associated behavioral traits and demonstrated that interactions between untreated ADHD and repeated mild traumatic brain injury produced behavioral traits not associated with either condition alone. CONCLUSION Preclinical models contribute to novel insights into ADHD neurobiology and are valuable tools for drug discovery and translation to benefit humans with ADHD.
Collapse
Affiliation(s)
| | - Thomas J Spencer
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Pradeep G Bhide
- Florida State University College of Medicine, Tallahassee, FL, USA
| |
Collapse
|
2
|
Daoud N, Mateos DL, Riley MA, Siegel JB. Computational Drug Design of Novel Agonists of the μ -Opioid Receptor to Inhibit Pain Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.25.554876. [PMID: 37662242 PMCID: PMC10473735 DOI: 10.1101/2023.08.25.554876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Opioids such as Morphine, Codeine, Hydrocodone, and Oxycodone target the μ-opioid receptor, a G-protein-coupled receptor (GPCR), blocking the transmission of nociceptive signals. In this study, four opioids were analyzed for ADMET properties and molecular interactions with a GPCR crystal structure (PDB ID: 8EF6). This aided in the computational design of two novel drug candidates with improved docking scores and ADMET properties when compared to Hydrocodone. Homology analysis indicated that a Mus musculus (house mouse) animal model could be used in the preclinical studies of these drug candidates in the development of safer and more effective opioid drugs for pain management with reduced side effects.
Collapse
Affiliation(s)
- Nancy Daoud
- Department of Chemistry, University of California Davis, Davis, California, United States of America
| | - Diego Lopez Mateos
- Department of Physiology and Membrane Biology, University of California Davis, Davis, California, United States of America
- Biophysics Graduate Group, University of California Davis, Davis, California, United States of America
| | - Mary A Riley
- Microbiology Graduate Group, University of California Davis, Davis, California, United States of America
- Genome Center, University of California Davis, Davis, California, United States of America
| | - Justin B Siegel
- Department of Chemistry, University of California Davis, Davis, California, United States of America
- Microbiology Graduate Group, University of California Davis, Davis, California, United States of America
- Genome Center, University of California Davis, Davis, California, United States of America
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, California, United States of America
| |
Collapse
|
3
|
West AM, Holleran KM, Jones SR. Kappa Opioid Receptors Reduce Serotonin Uptake and Escitalopram Efficacy in the Mouse Substantia Nigra Pars Reticulata. Int J Mol Sci 2023; 24:2080. [PMID: 36768403 PMCID: PMC9916942 DOI: 10.3390/ijms24032080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
The serotonin and kappa opioid receptor (KOR) systems are strongly implicated in disorders of negative affect, such as anxiety and depression. KORs expressed on axon terminals inhibit the release of neurotransmitters, including serotonin. The substantia nigra pars reticulata (SNr) is involved in regulating affective behaviors. It receives the densest serotonergic innervation in the brain and has high KOR expression; however, the influence of KORs on serotonin transmission in this region is yet to be explored. Here, we used ex vivo fast-scan cyclic voltammetry (FSCV) to investigate the effects of a KOR agonist, U50, 488 (U50), and a selective serotonin reuptake inhibitor, escitalopram, on serotonin release and reuptake in the SNr. U50 alone reduced serotonin release and uptake, and escitalopram alone augmented serotonin release and slowed reuptake, while pretreatment with U50 blunted both the release and uptake effects of escitalopram. Here, we show that the KOR influences serotonin signaling in the SNr in multiple ways and short-term activation of the KOR alters serotonin responses to escitalopram. These interactions between KORs and serotonin may contribute to the complexity in the responses to treatments for disorders of negative affect. Ultimately, the KOR system may prove to be a promising pharmacological target, alongside traditional antidepressant treatments.
Collapse
Affiliation(s)
| | | | - Sara R. Jones
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Medical Center Blvd, Winston Salem, NC 27157, USA
| |
Collapse
|
4
|
Kappa Opioid Receptor Mediated Differential Regulation of Serotonin and Dopamine Transporters in Mood and Substance Use Disorder. Handb Exp Pharmacol 2021; 271:97-112. [PMID: 34136961 DOI: 10.1007/164_2021_499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Dynorphin (DYN) is an endogenous neurosecretory peptide which exerts its activity by binding to the family of G protein-coupled receptors, namely the kappa opioid receptor (KOR). Opioids are associated with pain, analgesia, and drug abuse, which play a central role in mood disorders with monoamine neurotransmitter interactions. Growing evidence demonstrates the cellular signaling cascades linked to KOR-mediated monoamine transporters regulation in cell models and native brain tissues. This chapter will review DYN/KOR role in mood and addiction in relevance to dopaminergic and serotonergic neurotransmissions. Also, we discuss the recent findings on KOR-mediated differential regulation of serotonin and dopamine transporters (SERT and DAT). These findings led to a better understanding of the role of DYN/KOR system in aminergic neurotransmission via its modulatory effect on both amine release and clearance. Detailed knowledge of these processes at the molecular level enables designing novel pharmacological reagents to target transporter motifs to treat mood and addiction and reduce unwanted side effects such as aversion, dysphoria, sedation, and psychomimesis.
Collapse
|
5
|
Zhang L, McCarthy DM, Eskow Jaunarajs KL, Biederman J, Spencer TJ, Bhide PG. Frontal Cortical Monoamine Release, Attention, and Working Memory in a Perinatal Nicotine Exposure Mouse Model Following Kappa Opioid Receptor Antagonism. Cereb Cortex 2021; 31:483-496. [PMID: 32869057 DOI: 10.1093/cercor/bhaa238] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 02/27/2024] Open
Abstract
Perinatal nicotine exposure (PNE) produces frontal cortical hypo-dopaminergic state and attention and working memory deficits consistent with neurodevelopmental disorders such as attention deficit hyperactivity disorder (ADHD). Methylphenidate alleviates ADHD symptoms by increasing extracellular dopamine and noradrenaline. Kappa opioid receptor (KOR) antagonism may be another mechanism to achieve the same results because KOR activation inhibits frontal cortical dopamine release. We administered the selective KOR antagonist norbinaltorphimine (norBNI) (20 mg/kg; intraperitoneal) or methylphenidate (0.75 mg/kg; intraperitoneal) to PNE mouse model and examined frontal cortical monoamine release, attention, and working memory. Both compounds increased dopamine and noradrenaline release but neither influenced serotonin release. Both compounds improved object-based attention and working memory in the PNE group, with norBNI's effects evident at 2.5 h and 5.5 h but absent at 24 h. Methylphenidate's effects were evident at 0.5 h but not at 2.5 h. norBNI's effects temporally coincided with frontal cortical c-Jun N-terminal kinase phosphorylation. norBNI did not alter tissue dopamine content in the nucleus accumbens, offering preliminary support for lack of reinforcement.
Collapse
Affiliation(s)
- Lin Zhang
- Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Deirdre M McCarthy
- Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | | | - Joseph Biederman
- Pediatric Psychopharmacology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Thomas J Spencer
- Pediatric Psychopharmacology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Pradeep G Bhide
- Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
6
|
Dinoff A, Lynch ST, Sekhri N, Klepacz L. A meta-analysis of the potential antidepressant effects of buprenorphine versus placebo as an adjunctive pharmacotherapy for treatment-resistant depression. J Affect Disord 2020; 271:91-99. [PMID: 32479336 DOI: 10.1016/j.jad.2020.03.089] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/26/2020] [Accepted: 03/25/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND Numerous reports have suggested that buprenorphine may have antidepressant effects. Many individuals with depressive disorders don't respond to first-line treatment and are classified with treatment-resistant depression (TRD). Novel therapies for depression are required to better treat this population. This meta-analysis of randomized placebo-controlled trials sought to evaluate the potential antidepressant effects of buprenorphine as an adjunctive pharmacological treatment for individuals with TRD. METHODS PubMed, Embase, CINAHL, Web of Science, and ClinicalTrials.gov databases were searched until June 2019 for original peer-reviewed reports of buprenorphine used for the treatment of depression. Standardized mean differences (SMD) were generated from random effects models. Risk of publication bias was assessed using a funnel plot. Potential sources of heterogeneity were explored in subgroup analyses. RESULTS In six studies that met inclusion criteria, depression symptom severity in individuals with TRD was not significantly decreased after an adjunctive intervention with buprenorphine when compared to placebo (SMD = -0.07, 95% CI: -0.21-0.06, p = 0.30). Five of the six studies utilized a combination of buprenorphine/samidorphan. In these studies, depression symptom severity was also not significantly reduced after intervention compared to placebo (SMD = -0.08, 95% CI: -0.21 - 0.05, p = 0.23). LIMITATIONS Five included studies were performed by the same research group with significant conflicts of interest. CONCLUSIONS This meta-analysis did not reveal a significant reduction in depression symptom severity in individuals with TRD after an adjunctive intervention with buprenorphine when compared to placebo. However, more optimal doses of buprenorphine (2 mg/day) and longer treatment lengths should be explored.
Collapse
Affiliation(s)
- Adam Dinoff
- New York Medical College School of Medicine, 40 Sunshine Cottage Rd, Valhalla, NY, 10595 USA.
| | - Sean T Lynch
- New York Medical College School of Medicine, 40 Sunshine Cottage Rd, Valhalla, NY, 10595 USA
| | - Nitin Sekhri
- New York Medical College School of Medicine, 40 Sunshine Cottage Rd, Valhalla, NY, 10595 USA; Department of Anesthesiology, Westchester Medical Center, Valhalla, NY, 10595, USA
| | - Lidia Klepacz
- New York Medical College School of Medicine, 40 Sunshine Cottage Rd, Valhalla, NY, 10595 USA; Department of Psychiatry, Westchester Medical Center, Valhalla, NY, 10595, USA
| |
Collapse
|
7
|
Park JY, Chae S, Kim CS, Kim YJ, Yi HJ, Han E, Joo Y, Hong S, Yun JW, Kim H, Shin KH. Role of nociceptin/orphanin FQ and nociceptin opioid peptide receptor in depression and antidepressant effects of nociceptin opioid peptide receptor antagonists. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2019; 23:427-448. [PMID: 31680765 PMCID: PMC6819898 DOI: 10.4196/kjpp.2019.23.6.427] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/11/2019] [Accepted: 08/26/2019] [Indexed: 01/28/2023]
Abstract
Nociceptin/orphanin FQ (N/OFQ) and its receptor, nociceptin opioid peptide (NOP) receptor, are localized in brain areas implicated in depression including the amygdala, bed nucleus of the stria terminalis, habenula, and monoaminergic nuclei in the brain stem. N/OFQ inhibits neuronal excitability of monoaminergic neurons and monoamine release from their terminals by activation of G protein-coupled inwardly rectifying K+ channels and inhibition of voltage sensitive calcium channels, respectively. Therefore, NOP receptor antagonists have been proposed as a potential antidepressant. Indeed, mounting evidence shows that NOP receptor antagonists have antidepressant-like effects in various preclinical animal models of depression, and recent clinical studies again confirmed the idea that blockade of NOP receptor signaling could provide a novel strategy for the treatment of depression. In this review, we describe the pharmacological effects of N/OFQ in relation to depression and explore the possible mechanism of NOP receptor antagonists as potential antidepressants.
Collapse
Affiliation(s)
- Jong Yung Park
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
| | - Suji Chae
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
| | - Chang Seop Kim
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
| | - Yoon Jae Kim
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
| | - Hyun Joo Yi
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
| | - Eunjoo Han
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
| | - Youngshin Joo
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
| | - Surim Hong
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
| | - Jae Won Yun
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
| | - Hyojung Kim
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
| | - Kyung Ho Shin
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
| |
Collapse
|
8
|
Lee DB, Woo YS, Bahk WM. Naltrexone-associated Visual Hallucinations: A Case Report. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2019; 17:329-331. [PMID: 30905136 PMCID: PMC6478085 DOI: 10.9758/cpn.2019.17.2.329] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/26/2017] [Accepted: 12/27/2017] [Indexed: 11/30/2022]
Abstract
Naltrexone is a competitive antagonist of μ, δ, and κ opioid receptors. Naltrexone has been investigated for use an as anti-obesity agent in both the general population and in patients with severe mental illness, including schizophrenia. In patients with schizophrenia, however, potential psychotic symptoms due to adverse effects of naltrexone have not been investigated. Our case study, a relevant case report, and some related articles suggest that naltrexone might be associated with the emergence of visual hallucinations, which clinicians should be aware of.
Collapse
Affiliation(s)
- Dae Bo Lee
- Department of Psychiatry, National Forensic Hospital
| | - Young Sup Woo
- Department of Psychiatry, College of Medicine, The Catholic University of Korea
| | - Won-Myong Bahk
- Department of Psychiatry, College of Medicine, The Catholic University of Korea
| |
Collapse
|
9
|
Miller JM, Zanderigo F, Purushothaman PD, DeLorenzo C, Rubin-Falcone H, Ogden RT, Keilp J, Oquendo MA, Nabulsi N, Huang YH, Parsey RV, Carson RE, Mann JJ. Kappa opioid receptor binding in major depression: A pilot study. Synapse 2018; 72:e22042. [PMID: 29935119 PMCID: PMC7599086 DOI: 10.1002/syn.22042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/31/2018] [Accepted: 06/10/2018] [Indexed: 12/19/2022]
Abstract
Endogenous kappa opioids mediate pathological responses to stress in animal models. However, the relationship of the kappa opioid receptor (KOR) to life stress and to psychopathology in humans is not well described. This pilot study sought, for the first time, to quantify KOR in major depressive disorder (MDD) in vivo in humans using positron emission tomography (PET). KOR binding was quantified in vivo by PET imaging with the [11 C]GR103545 radiotracer in 13 healthy volunteers and 10 participants with current MDD. We examined the relationship between regional [11 C]GR103545 total volume of distribution (VT ) and diagnosis, childhood trauma, recent life stress, and, in a subsample, salivary cortisol levels during a modified Trier Social Stress Test (mTSST), amygdala, hippocampus, ventral striatum and raphe nuclei. Whole-brain voxel-wise analyses were also performed. [11 C]GR103545 VT did not differ significantly between MDD participants and healthy volunteers in the four a priori ROIs (p = 0.50). [11 C]GR103545 VT was unrelated to reported childhood adversity (p = 0.17) or recent life stress (p = 0.56). A trend-level inverse correlation was observed between [11 C]GR103545 VT and cortisol area-under-the curve with respect to ground during the mTSST (p = 0.081). No whole-brain voxel-wise contrasts were significant. Regional [11 C]GR103545 VT , a measure of in vivo KOR binding, does not differentiate MDD from healthy volunteers in this pilot sample. Future studies may examine KOR binding in subgroups of depressed individuals at increased risk for KOR abnormalities, including co-occurring mood and substance use disorders, as well as depression with psychotic features.
Collapse
Affiliation(s)
- Jeffrey M. Miller
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY
- Department of Psychiatry, Columbia University, New York, NY
| | - Francesca Zanderigo
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY
- Department of Psychiatry, Columbia University, New York, NY
| | | | - Christine DeLorenzo
- Department of Psychiatry and Behavioral Science, Stony Brook University School of Medicine
| | - Harry Rubin-Falcone
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY
- Department of Psychiatry, Columbia University, New York, NY
| | - R. Todd Ogden
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY
| | - John Keilp
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY
- Department of Psychiatry, Columbia University, New York, NY
| | - Maria A. Oquendo
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania
| | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine
| | - Yiyun H. Huang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine
| | - Ramin V. Parsey
- Department of Psychiatry and Behavioral Science, Stony Brook University School of Medicine
| | - Richard E. Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine
| | - J. John Mann
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY
- Department of Psychiatry, Columbia University, New York, NY
| |
Collapse
|
10
|
Anderson RI, Moorman DE, Becker HC. Contribution of Dynorphin and Orexin Neuropeptide Systems to the Motivational Effects of Alcohol. Handb Exp Pharmacol 2018. [PMID: 29526023 DOI: 10.1007/164_2018_100] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Understanding the neural systems that drive alcohol motivation and are disrupted in alcohol use disorders is of critical importance in developing novel treatments. The dynorphin and orexin/hypocretin neuropeptide systems are particularly relevant with respect to alcohol use and misuse. Both systems are strongly associated with alcohol-seeking behaviors, particularly in cases of high levels of alcohol use as seen in dependence. Furthermore, both systems also play a role in stress and anxiety, indicating that disruption of these systems may underlie long-term homeostatic dysregulation seen in alcohol use disorders. These systems are also closely interrelated with one another - dynorphin/kappa opioid receptors and orexin/hypocretin receptors are found in similar regions and hypocretin/orexin neurons also express dynorphin - suggesting that these two systems may work together in the regulation of alcohol seeking and may be mutually disrupted in alcohol use disorders. This chapter reviews studies demonstrating a role for each of these systems in motivated behavior, with a focus on their roles in regulating alcohol-seeking and self-administration behaviors. Consideration is also given to evidence indicating that these neuropeptide systems may be viable targets for the development of potential treatments for alcohol use disorders.
Collapse
Affiliation(s)
- Rachel I Anderson
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA.,Science and Technology Policy Fellowships, American Association for the Advancement of Science, Washington, DC, USA
| | - David E Moorman
- Department of Psychological and Brain Sciences, Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - Howard C Becker
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA. .,Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, USA. .,Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA. .,Department of Veterans Affairs, Ralph H. Johnson VA Medical Center, Charleston, SC, USA.
| |
Collapse
|
11
|
Abstract
Presynaptic receptors are sites at which transmitters, locally formed mediators or hormones inhibit or facilitate the release of a given transmitter from its axon terminals. The interest in the identification of presynaptic receptors has faded in recent years and it may therefore be justified to give an overview of their occurrence in the autonomic and central nervous system; this review will focus on presynaptic receptors in human tissues. Autoreceptors are presynaptic receptors at which a given transmitter restrains its further release, though in some instances may also increase its release. Inhibitory autoreceptors represent a typical example of a negative feedback; they are tonically activated by the respective endogenous transmitter and/or are constitutively active. Autoreceptors also play a role under pathophysiological conditions, e.g. by limiting the massive noradrenaline release occurring during congestive heart failure. They can be used for therapeutic purposes; e.g., the α2-adrenoceptor antagonist mirtazapine is used as an antidepressant and the inverse histamine H3 receptor agonist pitolisant has been marketed as a new drug for the treatment of narcolepsy in 2016. Heteroreceptors are presynaptic receptors at which transmitters from adjacent neurons, locally formed mediators (e.g. endocannabinoids) or hormones (e.g. adrenaline) can inhibit or facilitate transmitter release; they may be subject to an endogenous tone. The constipating effect of the sympathetic nervous system or of the antihypertensive drug clonidine is related to the activation of inhibitory α2-adrenoceptors on postganglionic parasympathetic neurons. Part of the stimulating effect of adrenaline on the sympathetic nervous system during stress is related to its facilitatory effect on noradrenaline release via β2-adrenoceptors.
Collapse
Affiliation(s)
| | - Thomas Feuerstein
- Sektion Neuroelektronische Systeme, Klinik für Neurochirurgie, Universität Freiburg, Germany
| |
Collapse
|
12
|
Modulation of serotonin transporter function by kappa-opioid receptor ligands. Neuropharmacology 2016; 113:281-292. [PMID: 27743931 DOI: 10.1016/j.neuropharm.2016.10.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 09/16/2016] [Accepted: 10/10/2016] [Indexed: 12/11/2022]
Abstract
Kappa opioid receptor (KOR) agonists produce dysphoria and psychotomimesis. While KOR agonists produce pro-depressant-like effects, KOR antagonists produce anti-depressant-like effects in rodent models. The cellular mechanisms and downstream effector(s) by which KOR ligands produce these effects are not clear. KOR agonists modulate serotonin (5-HT) transmission in the brain regions implicated in mood and motivation regulation. Presynaptic serotonin transporter (SERT) activity is critical in the modulation of synaptic 5-HT and, subsequently, in mood disorders. Detailing the molecular events of KOR-linked SERT regulation is important for examining the postulated role of this protein in mood disorders. In this study, we used heterologous expression systems and native tissue preparations to determine the cellular signaling cascades linked to KOR-mediated SERT regulation. KOR agonists U69,593 and U50,488 produced a time and concentration dependent KOR antagonist-reversible decrease in SERT function. KOR-mediated functional down-regulation of SERT is sensitive to CaMKII and Akt inhibition. The U69,593-evoked decrease in SERT activity is associated with a decreased transport Vmax, reduced SERT cell surface expression, and increased SERT phosphorylation. Furthermore, KOR activation enhanced SERT internalization and decreased SERT delivery to the membrane. These data demonstrate that KOR activation decreases 5-HT uptake by altering SERT trafficking mechanisms and phosphorylation status to reduce the functional availability of surface SERT.
Collapse
|
13
|
Reyes BAS, Kravets JL, Connelly KL, Unterwald EM, Van Bockstaele EJ. Localization of the delta opioid receptor and corticotropin-releasing factor in the amygdalar complex: role in anxiety. Brain Struct Funct 2016; 222:1007-1026. [PMID: 27376372 DOI: 10.1007/s00429-016-1261-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/22/2016] [Indexed: 12/24/2022]
Abstract
It is well established that central nervous system norepinephrine (NE) and corticotropin-releasing factor (CRF) systems are important mediators of behavioral responses to stressors. More recent studies have defined a role for delta opioid receptors (DOPR) in maintaining emotional valence including anxiety. The amygdala plays an important role in processing emotional stimuli, and has been implicated in the development of anxiety disorders. Activation of DOPR or inhibition of CRF in the amygdala reduces baseline and stress-induced anxiety-like responses. It is not known whether CRF- and DOPR-containing amygdalar neurons interact or whether they are regulated by NE afferents. Therefore, this study sought to better define interactions between the CRF, DOPR and NE systems in the basolateral (BLA) and central nucleus of the amygdala (CeA) of the male rat using anatomical and functional approaches. Irrespective of the amygdalar subregion, dual immunofluorescence microscopy showed that DOPR was present in CRF-containing neurons. Immunoelectron microscopy confirmed that DOPR was localized to both dendritic processes and axon terminals in the BLA and CeA. Semi-quantitative dual immunoelectron microscopy analysis of gold-silver labeling for DOPR and immunoperoxidase labeling for CRF revealed that 55 % of the CRF neurons analyzed contained DOPR in the BLA while 67 % of the CRF neurons analyzed contained DOPR in the CeA. Furthermore, approximately 41 % of DOPR-labeled axon terminals targeted BLA neurons that expressed CRF while 29 % of DOPR-labeled axon terminals targeted CeA neurons that expressed CRF. Triple label immunofluorescence microscopy revealed that DOPR and CRF were co-localized in common cellular profiles that were in close proximity to NE-containing fibers in both subregions. These anatomical results indicate significant interactions between DOPR and CRF in this critical limbic region and reveal that NE is poised to regulate these peptidergic systems in the amygdala. Functional studies were performed to determine if activation of DOPR could inhibit the anxiety produced by elevation of NE in the amygdala using the pharmacological stressor yohimbine. Administration of the DOPR agonist, SNC80, significantly attenuated elevated anxiogenic behaviors produced by yohimbine as measured in the rat on the elevated zero maze. Taken together, results from this study demonstrate the convergence of three important systems, NE, CRF, and DOPR, in the amygdala and provide insight into their functional role in modulating stress and anxiety responses.
Collapse
Affiliation(s)
- Beverly A S Reyes
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, 245 S. 15th Street, Philadelphia, PA, 19102, USA.
| | - J L Kravets
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, 245 S. 15th Street, Philadelphia, PA, 19102, USA
| | - K L Connelly
- Center for Substance Abuse Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - E M Unterwald
- Center for Substance Abuse Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - E J Van Bockstaele
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, 245 S. 15th Street, Philadelphia, PA, 19102, USA
| |
Collapse
|
14
|
Toll L, Bruchas MR, Calo' G, Cox BM, Zaveri NT. Nociceptin/Orphanin FQ Receptor Structure, Signaling, Ligands, Functions, and Interactions with Opioid Systems. Pharmacol Rev 2016; 68:419-57. [PMID: 26956246 PMCID: PMC4813427 DOI: 10.1124/pr.114.009209] [Citation(s) in RCA: 222] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The NOP receptor (nociceptin/orphanin FQ opioid peptide receptor) is the most recently discovered member of the opioid receptor family and, together with its endogenous ligand, N/OFQ, make up the fourth members of the opioid receptor and opioid peptide family. Because of its more recent discovery, an understanding of the cellular and behavioral actions induced by NOP receptor activation are less well developed than for the other members of the opioid receptor family. All of these factors are important because NOP receptor activation has a clear modulatory role on mu opioid receptor-mediated actions and thereby affects opioid analgesia, tolerance development, and reward. In addition to opioid modulatory actions, NOP receptor activation has important effects on motor function and other physiologic processes. This review discusses how NOP pharmacology intersects, contrasts, and interacts with the mu opioid receptor in terms of tertiary structure and mechanism of receptor activation; location of receptors in the central nervous system; mechanisms of desensitization and downregulation; cellular actions; intracellular signal transduction pathways; and behavioral actions with respect to analgesia, tolerance, dependence, and reward. This is followed by a discussion of the agonists and antagonists that have most contributed to our current knowledge. Because NOP receptors are highly expressed in brain and spinal cord and NOP receptor activation sometimes synergizes with mu receptor-mediated actions and sometimes opposes them, an understanding of NOP receptor pharmacology in the context of these interactions with the opioid receptors will be crucial to the development of novel therapeutics that engage the NOP receptor.
Collapse
Affiliation(s)
- Lawrence Toll
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Michael R Bruchas
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Girolamo Calo'
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Brian M Cox
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Nurulain T Zaveri
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| |
Collapse
|
15
|
Shagiakhmetov FS, Proskuryakova TV, Shamakina IY. The dynorphin/kappa-opioid system of the brain as a promising target for therapy for dependence on psychoactive substances. NEUROCHEM J+ 2015. [DOI: 10.1134/s1819712415040157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Finkbeiner S, Frumkin M, Kassner PD. Cell-based screening: extracting meaning from complex data. Neuron 2015; 86:160-74. [PMID: 25856492 PMCID: PMC4457442 DOI: 10.1016/j.neuron.2015.02.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 09/23/2014] [Accepted: 01/22/2015] [Indexed: 01/23/2023]
Abstract
Unbiased discovery approaches have the potential to uncover neurobiological insights into CNS disease and lead to the development of therapies. Here, we review lessons learned from imaging-based screening approaches and recent advances in these areas, including powerful new computational tools to synthesize complex data into more useful knowledge that can reliably guide future research and development.
Collapse
Affiliation(s)
- Steven Finkbeiner
- Director of the Taube/Koret Center for Neurodegenerative Disease and the Hellman Family Foundation Program in Alzheimer's Disease Research, Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Michael Frumkin
- Director of Engineering, Research, Google, Inc., 1600 Amphitheatre Parkway, Mountain View, CA 94043, USA
| | - Paul D Kassner
- Director of Research, Amgen, Inc., 1120 Veterans Boulevard South, San Francisco, CA 94080, USA
| |
Collapse
|
17
|
Low dosage of rimonabant leads to anxiolytic-like behavior via inhibiting expression levels and G-protein activity of kappa opioid receptors in a cannabinoid receptor independent manner. Neuropharmacology 2015; 89:298-307. [DOI: 10.1016/j.neuropharm.2014.10.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/19/2014] [Accepted: 10/04/2014] [Indexed: 12/15/2022]
|
18
|
Kim MJ, Kadayat T, Kim DE, Lee ES, Park PH. TI-I-174, a Synthetic Chalcone Derivative, Suppresses Nitric Oxide Production in Murine Macrophages via Heme Oxygenase-1 Induction and Inhibition of AP-1. Biomol Ther (Seoul) 2014; 22:390-9. [PMID: 25414768 PMCID: PMC4201222 DOI: 10.4062/biomolther.2014.062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 07/30/2014] [Accepted: 07/31/2014] [Indexed: 02/05/2023] Open
Abstract
Chalcones (1,3-diaryl-2-propen-1-ones), a flavonoid subfamily, are widely known for their anti-inflammatory properties. Propenone moiety in chalcones is known to play an important role in generating biological responses by chalcones. In the present study, we synthesized chalcone derivatives structurally modified in propenone moiety and examined inhibitory effect on nitric oxide (NO) production and its potential mechanisms. Among the chalcone derivatives used for this study, TI-I-174 (3-(2-Hydroxyphenyl)-1-(thiophen-3-yl)prop-2-en-1-one) most potently inhibited lipopolysaccharide (LPS)-stimulated nitrite production in RAW 264.7 macrophages. TI-I-174 treatment also markedly inhibited inducible nitric oxide synthase (iNOS) expression. However, TI-I-174 did not significantly affect production of IL-6, cyclooxygenase-2 (COX-2) and tumor necrosis factor-α (TNF-α), implying that TI-I-174 inhibits production of inflammatory mediators in a selective manner. Treatment of macrophages with TI-I-174 significantly inhibited transcriptional activity of activator protein-1 (AP-1) as determined by luciferase reporter gene assay, whereas nuclear factor-κB (NF-κB) activity was not affected by TI-I-1744. In addition, TI-I-174 significantly inhibited activation of c-Jun-N-Terminal kinase (JNK) without affecting ERK1/2 and p38MAPK, indicating that down-regulation of iNOS gene expression by TI-I-174 is mainly attributed by blockade of JNK/AP-1 activation. We also demonstrated that TI-I-174 treatment led to an increase in heme oxygenase-1 (HO-1) expression both at mRNA and protein level. Transfection of siRNA targeting HO-1 reversed TI-I-174-mediated inhibition of nitrite production. Taken together, these results indicate that TI-I-174 suppresses NO production in LPS-stimulated RAW 264.7 macrophages via induction of HO-1 and blockade of AP-1 activation.
Collapse
Affiliation(s)
- Mi Jin Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 712-749, Republic of Korea
| | - Taraman Kadayat
- College of Pharmacy, Yeungnam University, Gyeongsan 712-749, Republic of Korea
| | - Da Eun Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 712-749, Republic of Korea
| | - Eung-Seok Lee
- College of Pharmacy, Yeungnam University, Gyeongsan 712-749, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan 712-749, Republic of Korea
| |
Collapse
|
19
|
Zahiu CDM, Rimbas M. Neuropsychiatric side-effects of interferon-alpha treatment: pathophysiology and therapeutic options. MAEDICA 2014; 9:121-126. [PMID: 25705266 PMCID: PMC4296753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 06/20/2014] [Indexed: 06/04/2023]
Abstract
Interferon alpha (IFN-α) is the approved standard of care for chronic hepatitis C and B. Unfortunately, it has neuropsychiatric side-effects that have a major impact upon the quality of life and the drug adherence. The mechanism of IFN-α-induced behavioral changes is complex, involving interactions between the immune system, the endocrine system, the monoaminergic systems and the opioid receptors. Recent studies support the neurodegeneration hypothesis as a possible mechanism of IFN-α-induced depressive behavior. Although a meta-analysis showed that antidepressant pretreatment effectively reduces the incidence and severity of depressive symptoms, irrespective of pre-existing psychiatric disorders, it is not approved for prophylactic use. The "on demand" treatment strategy is justified as the majority of patients have only mild depressive symptoms. Patients with risk factors for depression undergoing IFN-α therapy need to be regularly screened and followed-up by a psychiatric specialist. Further studies should be conducted to show which therapy is the most appropriate to reduce the neuropsychiatric symptoms that are related to the use of IFN-α and to investigate the clinical significance of IFN-α-induced neurodegeneration.
Collapse
Affiliation(s)
- Carmen Denise Mihaela Zahiu
- Division of Physiology and Neurosciences, Department of Functional Sciences, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania ; Department of Gastroenterology, Colentina Clinical Hospital, Bucharest, Romania
| | - Mihai Rimbas
- Department of Gastroenterology, Colentina Clinical Hospital, Bucharest, Romania ; Department of Internal Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
20
|
Efficient design, synthesis and structure–activity relationship studies of 1-(3′-substituted propyl)-4-arylpiperidines as non-peptide antagonists of nociceptin/orphanin FQ receptor: biological activities, metabolic stabilities and hERG channel bindings. Med Chem Res 2013. [DOI: 10.1007/s00044-013-0595-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
21
|
Tuominen L, Nummenmaa L, Keltikangas-Järvinen L, Raitakari O, Hietala J. Mapping neurotransmitter networks with PET: an example on serotonin and opioid systems. Hum Brain Mapp 2013; 35:1875-84. [PMID: 23671038 DOI: 10.1002/hbm.22298] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2012] [Revised: 01/16/2013] [Accepted: 02/28/2013] [Indexed: 12/31/2022] Open
Abstract
All functions of the human brain are consequences of altered activity of specific neural pathways and neurotransmitter systems. Although the knowledge of "system level" connectivity in the brain is increasing rapidly, we lack "molecular level" information on brain networks and connectivity patterns. We introduce novel voxel-based positron emission tomography (PET) methods for studying internal neurotransmitter network structure and intercorrelations of different neurotransmitter systems in the human brain. We chose serotonin transporter and μ-opioid receptor for this analysis because of their functional interaction at the cellular level and similar regional distribution in the brain. Twenty-one healthy subjects underwent two consecutive PET scans using [(11)C]MADAM, a serotonin transporter tracer, and [(11)C]carfentanil, a μ-opioid receptor tracer. First, voxel-by-voxel "intracorrelations" (hub and seed analyses) were used to study the internal structure of opioid and serotonin systems. Second, voxel-level opioid-serotonin intercorrelations (between neurotransmitters) were computed. Regional μ-opioid receptor binding potentials were uniformly correlated throughout the brain. However, our analyses revealed nonuniformity in the serotonin transporter intracorrelations and identified a highly connected local network (midbrain-striatum-thalamus-amygdala). Regionally specific intercorrelations between the opioid and serotonin tracers were found in anteromedial thalamus, amygdala, anterior cingulate cortex, dorsolateral prefrontal cortex, and left parietal cortex, i.e., in areas relevant for several neuropsychiatric disorders, especially affective disorders. This methodology enables in vivo mapping of connectivity patterns within and between neurotransmitter systems. Quantification of functional neurotransmitter balances may be a useful approach in etiological studies of neuropsychiatric disorders and also in drug development as a biomarker-based rationale for targeted modulation of neurotransmitter networks.
Collapse
Affiliation(s)
- Lauri Tuominen
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland; Department of Psychiatry, University of Turku, Turku, Finland
| | | | | | | | | |
Collapse
|
22
|
Sirohi S, Bakalkin G, Walker BM. Alcohol-induced plasticity in the dynorphin/kappa-opioid receptor system. Front Mol Neurosci 2012; 5:95. [PMID: 23060746 PMCID: PMC3459013 DOI: 10.3389/fnmol.2012.00095] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 08/29/2012] [Indexed: 01/02/2023] Open
Abstract
Alcoholism is a chronic relapsing disorder characterized by continued alcohol use despite numerous adverse consequences. Alcohol has been shown to interact with numerous neurotransmitter systems to exert its pharmacological effects. The endogenous opioid system (EOS) has been strongly implicated in the positive and negative reinforcing effects of alcohol. Traditionally recognized as dysphoric/anhedonic in nature, the dynorphin/kappa-opioid receptor (DYN/KOR) system has recently received considerable attention due to evidence suggesting that an upregulated DYN/KOR system may be a critical contributor to the complex factors that result in escalated alcohol consumption once dependent. The present review will discuss alcohol-induced plasticity in the DYN/KOR system and how these neuroadaptations could contribute to excessive alcohol seeking and consumption.
Collapse
Affiliation(s)
- Sunil Sirohi
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Washington State University Pullman, WA, USA
| | | | | |
Collapse
|
23
|
Tejeda HA, Shippenberg TS, Henriksson R. The dynorphin/κ-opioid receptor system and its role in psychiatric disorders. Cell Mol Life Sci 2012; 69:857-96. [PMID: 22002579 PMCID: PMC11114766 DOI: 10.1007/s00018-011-0844-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 09/16/2011] [Accepted: 09/19/2011] [Indexed: 10/16/2022]
Abstract
The dynorphin/κ-opioid receptor system has been implicated in the pathogenesis and pathophysiology of several psychiatric disorders. In the present review, we present evidence indicating a key role for this system in modulating neurotransmission in brain circuits that subserve mood, motivation, and cognitive function. We overview the pharmacology, signaling, post-translational, post-transcriptional, transcriptional, epigenetic and cis regulation of the dynorphin/κ-opioid receptor system, and critically review functional neuroanatomical, neurochemical, and pharmacological evidence, suggesting that alterations in this system may contribute to affective disorders, drug addiction, and schizophrenia. We also overview the dynorphin/κ-opioid receptor system in the genetics of psychiatric disorders and discuss implications of the reviewed material for therapeutics development.
Collapse
Affiliation(s)
- H. A. Tejeda
- Integrative Neuroscience Section, Integrative Neuroscience Research Branch, NIDA-IRP, NIH, 333 Cassell Dr., Baltimore, MD 21224 USA
- Department of Anatomy and Neurobiology, University of Maryland, Baltimore, 20 Penn St., Baltimore, MD 21201 USA
| | - T. S. Shippenberg
- Integrative Neuroscience Section, Integrative Neuroscience Research Branch, NIDA-IRP, NIH, 333 Cassell Dr., Baltimore, MD 21224 USA
| | - R. Henriksson
- Integrative Neuroscience Section, Integrative Neuroscience Research Branch, NIDA-IRP, NIH, 333 Cassell Dr., Baltimore, MD 21224 USA
- Department of Clinical Neuroscience, Karolinska Institutet, CMM, L8:04, 17176 Stockholm, Sweden
| |
Collapse
|
24
|
Chieng B, Christie MJ. Somatostatin and nociceptin inhibit neurons in the central nucleus of amygdala that project to the periaqueductal grey. Neuropharmacology 2010; 59:425-30. [PMID: 20541564 DOI: 10.1016/j.neuropharm.2010.06.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 06/01/2010] [Accepted: 06/02/2010] [Indexed: 01/08/2023]
Abstract
The central nucleus of amygdala (CeA) plays an important role in modulation of the descending antinociceptive pathways. Using whole-cell patch clamp recordings from brain slices, we found that CeA neurons responded to the endogenous ligands somatostatin (SST) and nociceptin/orphanin FQ (OFQ) via an increased K-conductance. Co-application with selective antagonists suggested that SST and OFQ act on SSTR2 and ORL1 receptors, respectively. Taking account of anatomical localisation of recorded neurons, the present study showed that many responsive neurons were located within the medial subdivision of CeA and all CeA projection neurons to the midbrain periaqueductal grey invariably responded to these peptides. Randomly selected agonist-responsive neurons in CeA predominantly classified physiologically as low-threshold spiking neurons. The similarity of SST, OFQ and, as previously reported, opioid responsiveness in a sub-population of CeA neurons suggests converging roles of these peptides to inhibit the activity of projections from CeA to vlPAG, and potentially similar antinociceptive actions in this pathway.
Collapse
Affiliation(s)
- Billy Chieng
- Brain & Mind Research Institute, University of Sydney, 100 Mallett St, Camperdown, NSW 2050, Australia.
| | | |
Collapse
|
25
|
Rüedi-Bettschen D, Rowlett JK, Spealman RD, Platt DM. Attenuation of cocaine-induced reinstatement of drug seeking in squirrel monkeys: kappa opioid and serotonergic mechanisms. Psychopharmacology (Berl) 2010; 210:169-77. [PMID: 19859697 PMCID: PMC2866039 DOI: 10.1007/s00213-009-1705-2] [Citation(s) in RCA: 23] [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: 03/12/2009] [Accepted: 10/05/2009] [Indexed: 11/28/2022]
Abstract
RATIONALE Kappa agonists can attenuate reinstatement of cocaine-seeking behavior induced by cocaine priming. The mechanisms underlying this effect have not been characterized fully but may have a serotonergic component as kappa agonists also increase the release of serotonin (5-hydroxytryptamine, 5-HT). OBJECTIVES This study investigated the role of kappa opioid receptor and 5-HT mechanisms in kappa agonist-induced attenuation of cocaine priming in monkeys. METHODS Squirrel monkeys were trained to self-administer cocaine (0.18-0.3 mg/kg/injection) under a second-order schedule in which drug seeking was maintained jointly by cocaine injections and a cocaine-paired visual stimulus. In extinction sessions, saline was substituted for cocaine, and the cocaine-paired stimulus was omitted. During test sessions, only saline was available for self-administration, and response-contingent presentations of the cocaine-paired stimulus were restored. RESULTS Priming injections of cocaine (0.1-1.0 mg/kg) induced reinstatement of drug seeking. Maximal levels of responding were similar to those maintained by active cocaine self-administration. Pretreatment with the kappa agonists enadoline (0.01 mg/kg) and spiradoline (0.3 mg/kg) or the 5-HT transport inhibitors fluoxetine (5.6 mg/kg) and citalopram (10.0 mg/kg) attenuated the priming effects of cocaine, shifting the cocaine dose-response function rightward and downward. Inhibition of cocaine-induced reinstatement of drug seeking by spiradoline and fluoxetine was reversed by R(+)8-hydroxy-2-(di-n-propylamino)tetralin (0.03 mg/kg), a 5HT(1A) agonist that inhibits 5-HT release. The effects of spiradoline also were reversed by the kappa antagonist nor-binaltorphimine (10.0 mg/kg). CONCLUSIONS Results suggest that the capacity of kappa opioid agonists to increase extracellular 5-HT levels may at least partially underlie kappa agonist-induced modulation of cocaine seeking.
Collapse
Affiliation(s)
| | - James K. Rowlett
- Harvard Medical School, New England Primate Research Center, Southborough, MA, USA
| | - Roger D. Spealman
- Harvard Medical School, New England Primate Research Center, Southborough, MA, USA
| | - Donna M. Platt
- Harvard Medical School, New England Primate Research Center, Southborough, MA, USA
| |
Collapse
|
26
|
Grilli M, Neri E, Zappettini S, Massa F, Bisio A, Romussi G, Marchi M, Pittaluga A. Salvinorin A exerts opposite presynaptic controls on neurotransmitter exocytosis from mouse brain nerve terminals. Neuropharmacology 2009; 57:523-30. [DOI: 10.1016/j.neuropharm.2009.07.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 07/13/2009] [Accepted: 07/15/2009] [Indexed: 10/20/2022]
|
27
|
Dynorphin, stress, and depression. Brain Res 2009; 1314:56-73. [PMID: 19782055 DOI: 10.1016/j.brainres.2009.09.074] [Citation(s) in RCA: 359] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 09/14/2009] [Accepted: 09/17/2009] [Indexed: 12/29/2022]
Abstract
Stress is most often associated with aversive states. It rapidly induces the release of hormones and neuropeptides including dynorphin, which activates kappa opioid receptors (KORs) in the central and peripheral nervous systems. In animal models, many aversive effects of stress are mimicked or exacerbated by stimulation of KORs in limbic brain regions. Although KOR signaling during acute stress may increase physical ability (by producing analgesia) and motivation to escape a threat (by producing aversion), prolonged KOR signaling in response to chronic or uncontrollable stress can lead to persistent expression of behavioral signs that are characteristic of human depressive disorders (i.e., "prodepressive-like" signs). Accumulating evidence suggests that KORs contribute to the progressive amplification (sensitization) of stress-induced behaviors that occurs with repeated exposure to stress. Many of the aversive effects of stress are blocked by KOR antagonists, suggesting that these agents may have potential as therapeutics for stress-related conditions such as depression and anxiety disorders. This review summarizes current data on how KOR systems contribute to the acute (rapid), delayed, and cumulative molecular and behavioral effects of stress. We focus on behavioral paradigms that provide insight on interactions between stress and KOR function within each of these temporal categories. Using a simplified model, we consider the time course and mechanism of KOR-mediated effects in stress and suggest future directions that may be useful in determining whether KOR antagonists exert their therapeutic effects by preventing the development of stress-induced behaviors, the expression of stress-induced behaviors, or both.
Collapse
|
28
|
Hayashi S, Hirao A, Nakamura H, Yamamura K, Mizuno K, Yamashita H. Discovery of 1-[1-(1-methylcyclooctyl)-4-piperidinyl]-2-[(3R)-3-piperidinyl]-1H-benzimidazole: integrated drug-design and structure-activity relationships for orally potent, metabolically stable and potential-risk reduced novel non-peptide nociceptin/orphanin FQ receptor agonist as antianxiety drug. Chem Biol Drug Des 2009; 74:369-81. [PMID: 19691471 DOI: 10.1111/j.1747-0285.2009.00872.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Anxiety disorders, caused by continuous or acute stress or fear, have been highly prevailing psychiatric disorders. For the acute treatment of the disorders, benzodiazepines have been widely used despite having liabilities that limit their utility. Alternatively, endogenous nociceptin/orphanin FQ and nociceptin/orphanin FQ peptide receptor (or opioid-receptor-like-1 receptor) have important roles in the integration of emotional components, e.g. anxiolytic activity is the key behavioral action of nociceptin/orphanin FQ in brain. In our preceding study, various structurally novel 1,2-disubstituted benzimidazole derivatives were designed and synthesized as highly potent nociceptin/orphanin FQ peptide receptor selective full agonists in vitro with high or moderate nociceptin/orphanin FQ peptide receptor occupancy in the mice brain per os based on appropriate physicochemical properties for the oral brain activity [Hayashi et al. (2009) J Med Chem;52:610-625]. In the present study, drug design and structure-activity relationships for Vogel anticonflict activities in mice per os, metabolic stabilities in human liver microsome, CYP2D6 inhibitions, serum protein bindings, and human ether-a-go-go related gene binding affinities of novel nociceptin/orphanin FQ peptide receptor agonists were investigated. Through the series of coherent drug discovery studies, the strongest nociceptin/orphanin FQ peptide receptor agonist, 1-[1-(1-methylcyclooctyl)-4-piperidinyl]-2-[(3R)-3-piperidinyl]-1H-benzimidazole was designed and identified as a new-class orally potent anxiolytic with little side-effects, as significant findings.
Collapse
Affiliation(s)
- Shigeo Hayashi
- Pfizer Global Research & Development Nagoya Laboratories, Pfizer Japan Inc, 5-2 Taketoyo, Aichi 470-2393, Japan.
| | | | | | | | | | | |
Collapse
|
29
|
Stoll C, Eltze M, Lambrecht G, Zentner J, Feuerstein TJ, Jackisch R. Functional characterization of muscarinic autoreceptors in rat and human neocortex. J Neurochem 2009; 110:837-47. [DOI: 10.1111/j.1471-4159.2009.06193.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
30
|
Horner KA, Noble ES, Lauterbach EC. Differential regulation of prodynophin,c-fos, and serotonin transporter mRNA following withdrawal from a chronic, escalating dose regimen of D-amphetamine. Synapse 2009; 63:257-68. [DOI: 10.1002/syn.20606] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
31
|
Yoshikawa S, Hareyama N, Ikeda K, Kurokawa T, Nakajima M, Nakao K, Mochizuki H, Ichinose H. Effects of TRK-820, a selective kappa opioid receptor agonist, on rat schizophrenia models. Eur J Pharmacol 2009; 606:102-8. [DOI: 10.1016/j.ejphar.2009.01.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Revised: 01/14/2009] [Accepted: 01/27/2009] [Indexed: 10/21/2022]
|
32
|
Do Carmo GP, Folk JE, Rice KC, Chartoff E, Carlezon WA, Negus SS. The selective non-peptidic delta opioid agonist SNC80 does not facilitate intracranial self-stimulation in rats. Eur J Pharmacol 2008; 604:58-65. [PMID: 19133255 DOI: 10.1016/j.ejphar.2008.12.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Revised: 12/09/2008] [Accepted: 12/10/2008] [Indexed: 11/30/2022]
Abstract
Delta opioid receptor agonists are under development for a variety of clinical applications, and some findings in rats raise the possibility that agents with this mechanism have abuse liability. The present study assessed the effects of the non-peptidic delta opioid agonist SNC80 in an assay of intracranial self-stimulation (ICSS) in rats. ICSS was examined at multiple stimulation frequencies to permit generation of frequency-response rate curves and evaluation of curve shifts produced by experimental manipulations. Drug-induced leftward shifts in ICSS frequency-rate curves are often interpreted as evidence of abuse liability. However, SNC80 (1.0-10 mg/kg s.c.; 10-56 mg/kg i.p.) failed to alter ICSS frequency-rate curves at doses up to those that produced convulsions in the present study or other effects (e.g. antidepressant effects) in previous studies. For comparison, the monoamine releaser d-amphetamine (0.1-1.0 mg/kg, i.p.) and the kappa agonist U69,593 (0.1-0.56 mg/kg, i.p.) produced dose-dependent leftward and rightward shifts, respectively, in ICSS frequency-rate curves, confirming the sensitivity of the procedure to drug effects. ICSS frequency-rate curves were also shifted by two non-pharmacological manipulations (reductions in stimulus intensity and increases in response requirement). Thus, SNC80 failed to facilitate or attenuate ICSS-maintained responding under conditions in which other pharmacological and non-pharmacological manipulations were effective. These results suggest that non-peptidic delta opioid receptor agonists have negligible abuse-related effects in rats.
Collapse
Affiliation(s)
- Gail Pereira Do Carmo
- Alcohol and Drug Abuse Research Center, McLean Hospital-Harvard Medical School, Belmont, MA 02478, United States
| | | | | | | | | | | |
Collapse
|
33
|
Negus SS, Schrode K, Stevenson GW. Micro/kappa opioid interactions in rhesus monkeys: implications for analgesia and abuse liability. Exp Clin Psychopharmacol 2008; 16:386-99. [PMID: 18837635 PMCID: PMC2604909 DOI: 10.1037/a0013088] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Micro opioid receptor agonists are clinically valuable as analgesics; however, their use is limited by high abuse liability. Kappa opioid agonists also produce antinociception, but they do not produce micro agonist-like abuse-related effects, suggesting that they may enhance the antinociceptive effects and/or attenuate the abuse-related effects of micro agonists. To evaluate this hypothesis, the present study examined interactions between the micro agonist fentanyl and the kappa agonist U69,593 in three behavioral assays in rhesus monkeys. In an assay of schedule-controlled responding, monkeys responded under a fixed-ratio 30 (FR 30) schedule of food presentation. Fentanyl and U69,593 each produced rate-decreasing effects when administered alone, and mixtures of 0.22:1, 0.65:1, and 1.96:1 U69,593/fentanyl usually produced subadditive effects. In an assay of thermal nociception, tail withdrawal latencies were measured from water heated to 50 degrees C. Fentanyl and U69,593 each produced dose-dependent antinociception, and effects were additive for all mixtures. In an assay of drug self-administration, rhesus monkeys responded for intravenous drug injection, and both dose and FR values were manipulated. Fentanyl maintained self-administration, whereas U69,593 did not. Addition of U69,593 to fentanyl produced a proportion-dependent decrease in rates of fentanyl self-administration. Moreover, addition of U69,593 increased the sensitivity of fentanyl self-administration to increases in the FR value. Taken together, these results suggest that simultaneous activation of mu and kappa receptors, either with a mixture of selective drugs or with a single drug that targets both receptors, may reduce abuse liability without reducing analgesic effects relative to selective micro agonists administered alone.
Collapse
Affiliation(s)
- S Stevens Negus
- Alcohol and Drug Abuse Research Center, McLean Hospital-Harvard Medical School, Belmont, MA, USA.
| | | | | |
Collapse
|
34
|
Do Carmo GP, Polt R, Bilsky EJ, Rice KC, Negus SS. Behavioral pharmacology of the mu/delta opioid glycopeptide MMP2200 in rhesus monkeys. J Pharmacol Exp Ther 2008; 326:939-48. [PMID: 18511649 DOI: 10.1124/jpet.108.138180] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
H(2)N-Tyr-D-Thr-Gly-Phe-Leu-Ser-(O-beta-D-lactose)-CONH(2) (MMP2200) is a novel glycopeptide opioid agonist with similar affinities for mu and delta receptors. Glycosylation promoted brain penetration and production of centrally mediated behavioral effects in mice; however, it is unknown whether the magnitude of enhanced brain penetration is sufficient to permit central mediation of drug effects and production of synergistic mu/delta antinociceptive interactions after systemic administration in primates. To address this issue, the present study compared the effects of MMP2200 and the mu-agonist morphine in four behavioral procedures in rhesus monkeys. In an assay of thermal nociception, morphine (1.0-5.6 mg/kg) produced dose-dependent antinociception, whereas MMP2200 (10-56 mg/kg) was ineffective. In an assay of capsaicin-induced thermal allodynia, both morphine (0.01-1.0 mg/kg) and MMP2200 (0.032-3.2 mg/kg) produced dose-dependent antiallodynic effects. MMP2200-induced antiallodynia was blocked by the moderately mu-selective antagonist naltrexone (0.01 mg/kg), the delta-selective antagonist naltrindole (1.0 mg/kg), and the peripherally selective opioid antagonist quaternary naltrexone (0.32 mg/kg). In an assay of schedule-controlled behavior, both morphine (0.01-1.0 mg/kg) and MMP2200 (10-56 mg/kg) decreased response rates. Morphine effects were antagonized by naltrexone (0.001-0.01 mg/kg); however, the effects of MMP2200 were not antagonized by either naltrexone (0.01 mg/kg) or naltrindole (1.0 mg/kg). In an assay of drug self-administration, morphine (0.0032-0.32 mg/kg/injection) produced reinforcing effects, whereas MMP2200 (0.032-0.32 mg/kg/injection) did not. These results suggest that systemically administered MMP2200 acted as a peripheral, mu/delta-opioid agonist with limited distribution to the central nervous system in rhesus monkeys. These results also suggest the existence of species differences in the pharmacokinetics and brain penetration of glycopeptides.
Collapse
Affiliation(s)
- Gail Pereira Do Carmo
- Alcohol and Drug Abuse Research Center, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | | | | | | | | |
Collapse
|
35
|
Abstract
This article concentrates on recent evidence about opioid analgesics for persistent noncancer pain. Evidence confirms that opioids are drugs with a definite risk of adverse events. Therefore, before prescribing opioids, careful consideration must be given to be certain that the intended benefit of a particular opioid merits its use despite the potential side effects and to determine if the co-prescription of other pharmacologic agents could reduce the risk of adverse events. Strong opioids should be reserved for patients who fail to respond to other lower-risk options and only after proper consideration is given to the long-term consequences of strong opioid use. Problems associated with opioids dictate that more efficacious and safer drugs need to be found.
Collapse
Affiliation(s)
- Gary McCleane
- Rampark Pain Centre, 2 Rampark Dromore Road, Lurgan BT66 7JH, Northern Ireland, UK.
| | | |
Collapse
|
36
|
Abstract
Presynaptic receptors for four families of neuropeptides will be discussed: opioids, neuropeptide Y, adrenocorticotropic hormone (ACTH), and orexins. Presynaptic receptors for the opioids (micro, delta, kappa, and ORL(1)) and neuropeptide Y (Y(2)) inhibit transmitter release from a variety of neurones, both in the peripheral and central nervous systems. These receptors, which were also identified in human tissue, are coupled to G(i/o) proteins and block voltage-dependent Ca(2+) channels, activate voltage-dependent K(+) channels, and/or interfere with the vesicle release machinery. Presynaptic receptors for ACTH (MC(2) receptors) have so far been identified almost exclusively in cardiovascular tissues from rabbits, where they facilitate noradrenaline release; they are coupled to G(s) protein and act via stimulation of adenylyl cyclase. Presynaptic receptors for orexins (most probably OX(2) receptors) have so far almost exclusively been identified in the rat and mouse brain, where they facilitate the release of glutamate and gamma-aminobutyric acid (GABA); they are most probably linked to G(q) and directly activate the vesicle release machinery or act via a transduction mechanism upstream of the release process. Agonists and antagonists at opioid receptors owe at least part of their therapeutic effects to actions on presynaptic receptors. Therapeutic drugs targeting neuropeptide Y and orexin receptors and presynaptic ACTH receptors so far are not available.
Collapse
MESH Headings
- Animals
- Humans
- Neuropeptides/metabolism
- Orexin Receptors
- Receptors, Corticotropin/drug effects
- Receptors, Corticotropin/metabolism
- Receptors, G-Protein-Coupled/drug effects
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Neuropeptide/drug effects
- Receptors, Neuropeptide/metabolism
- Receptors, Neuropeptide Y/drug effects
- Receptors, Neuropeptide Y/metabolism
- Receptors, Opioid/drug effects
- Receptors, Opioid/metabolism
- Receptors, Presynaptic/drug effects
- Receptors, Presynaptic/metabolism
Collapse
Affiliation(s)
- E Schlicker
- Institut für Pharmakologie und Toxikologie, Rheinische Friedrich-Wilhelms-Universität, Reuterstrasse 2b, 53113 Bonn, Germany.
| | | |
Collapse
|
37
|
Abstract
This paper is the 29th consecutive installment of the annual review of research concerning the endogenous opioid system, now spanning 30 years of research. It summarizes papers published during 2006 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurological disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, CUNY, 65-30 Kissena Blvd., Flushing, NY 11367, United States.
| |
Collapse
|
38
|
Shannon HE, Eberle EL, Mitch CH, McKinzie DL, Statnick MA. Effects of kappa opioid receptor agonists on attention as assessed by a 5-choice serial reaction time task in rats. Neuropharmacology 2007; 53:930-41. [PMID: 17959202 DOI: 10.1016/j.neuropharm.2007.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2007] [Revised: 08/30/2007] [Accepted: 09/08/2007] [Indexed: 11/28/2022]
Abstract
In humans, kappa opioid receptor agonists produce, among other effects, sedation and difficulty concentrating, suggesting that they may disrupt attention. The purpose of the present studies was therefore to evaluate the effects of kappa opioid receptor agonists on attention as assessed by a 5-choice serial reaction time task in rats. The kappa opioid receptor agonists (+)-U69,593 (0.1-0.56mg/kg), (+/-)-U50,488 (1.0-5.6mg/kg) and racemic GR89,696 (0.0003-0.01mg/kg) all produced dose-related decreases in the percentage of trials terminated by a correct or incorrect response and increases in the percentage of omissions. In contrast, the peripherally restricted opioid agonist ICI-204,448 was ineffective (1.0-10mg/kg). Moreover, the effects of GR89,696 were stereoselective in that (R)-GR89,696 was approximately equipotent to racemic GR89,696 and approximately 100-fold more potent than (S)-GR89,696. The opioid receptor antagonist naltrexone (0.3-3mg/kg) administered alone had no effects on performance. However, naltrexone, over the dose-range of 0.03-1.0mg/kg, produced a dose-related antagonism of the disruption produced by U69,593 (0.56mg/kg). In contrast, naltrexone, over the dose-range of 0.01-0.3mg/kg produced a dose-related antagonism of morphine (5.6mg/kg). Recent evidence has suggested that kappa opioid receptor agonists decrease dopaminergic and noradrenergic neurotransmission in prefrontal cortex and locus coeruleus. Together with previous findings, the present data indicate that kappa opioid receptor agonists disrupt performance of this attention task by decreasing the probability of responding by specific actions at central kappa opioid receptors, perhaps by decreasing dopaminergic and noradrenergic neurotransmission.
Collapse
Affiliation(s)
- Harlan E Shannon
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA.
| | | | | | | | | |
Collapse
|
39
|
Schindler CW, Graczyk Z, Gilman JP, Negus SS, Bergman J, Mello NK, Goldberg SR. Effects of kappa opioid agonists alone and in combination with cocaine on heart rate and blood pressure in conscious squirrel monkeys. Eur J Pharmacol 2007; 576:107-13. [PMID: 17707792 PMCID: PMC2080578 DOI: 10.1016/j.ejphar.2007.07.053] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2007] [Revised: 07/25/2007] [Accepted: 07/26/2007] [Indexed: 11/28/2022]
Abstract
As kappa agonists have been proposed as treatments for cocaine abuse, the cardiovascular effects of the kappa opioid receptor agonists ethylketocyclazocine (EKC) and enadoline were investigated in conscious squirrel monkeys. Both EKC and enadoline increased heart rate with little effect on blood pressure. This effect appeared to be specific for kappa receptors as the mu opioid agonist morphine did not mimic the effects of the kappa agonists. The opioid antagonist naltrexone, at a dose of 1.0 mg/kg, blocked the effect of EKC. An action at both central and peripheral receptors may be responsible for the heart rate increase following kappa agonist treatment. The ganglionic blocker chlorisondamine partially antagonized the effect of EKC on heart rate, suggesting central involvement, while the peripherally-acting agonist ICI 204,448 ((+/-)-1-[2,3- (Dihydro-7-methyl-1H-inden-4-yl)oxy]-3-[(1-methylethyl)amino]-2-butanol hydrochloride) also increased heart rate, supporting a peripheral site of action. When given in combination with cocaine, EKC produced effects that were sub-additive, suggesting that the kappa agonists may be used safely as cocaine abuse treatments.
Collapse
Affiliation(s)
- Charles W Schindler
- Preclinical Pharmacology Section, Behavioral Neuroscience Branch, DHHS/NIH/NIDA Intramural Research Program, Baltimore MD (CWS, ZG, JPG, SRG), United States.
| | | | | | | | | | | | | |
Collapse
|
40
|
Rutz S, Riegert C, Rothmaier AK, Jackisch R. Presynaptic modulation of 5-HT release in the rat septal region. Neuroscience 2007; 146:643-58. [PMID: 17383104 DOI: 10.1016/j.neuroscience.2007.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2006] [Revised: 02/06/2007] [Accepted: 02/07/2007] [Indexed: 10/23/2022]
Abstract
5-HT released from serotonergic axon terminals in the septal nuclei modulates the activity of septal output neurons (e.g. septohippocampal cholinergic neurons) bearing somatodendritic 5-HT receptors. Therefore, we studied the mechanisms involved in the presynaptic modulation of 5-HT release in the lateral (LS) and medial septum (MS), and the diagonal band of Broca (DB). HPLC analysis showed that tissue concentrations of noradrenaline, dopamine and 5-HT were highest in DB (DB>MS>LS). Slices prepared from LS, MS and DB regions were preincubated with [(3)H]5-HT, superfused in the presence of 6-nitro-2-(1-piperazinyl)-quinoline (6-nitroquipazine) and electrically stimulated up to three times (first electrical stimulation period (S(1)), S(2), S(3); 360 pulses, 3 Hz, 2 ms, 26-28 mA). In all septal regions the Ca(2+)-dependent and tetrodotoxin-sensitive electrically-evoked overflow of [(3)H] was inhibited by the 5-HT(1B) agonist CP-93,129 and the alpha(2)-adrenoceptor agonist 5-bromo-6-(2-imidazolin-2-ylamino)-quinoxaline tartrate (UK-14,304). Also the mu- and kappa-opioid receptor agonists (d-Ala(2), N-Me-Phe(4), glycinol(5))-enkephalin (DAMGO) and [trans-(1S,2S(-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl) cyclohexyl]-benzenacetamide hydro-chloride] (U-50,488H), respectively, acted inhibitory (although less potently), whereas the delta-opioid receptor agonist (d-Pen(2), d-Pen(5))-enkephalin (DPDPE), the dopamine D(2) receptor agonist quinpirole and the adenosine A(1) receptor agonist N(6)-cyclopentyladenosine were all ineffective; the GABA(B) receptor agonist baclofen had weak effects. All inhibitory effects of the agonists were antagonized by the corresponding antagonists (3-[3-(dimethylamino)propyl]-4-hydroxy-N-[4-(4-pyridinyl)phenyl]benzamide dihydrochloride (GR-55,562), idazoxan, naloxone, nor-binaltorphimine), which also significantly enhanced the evoked release of 5-HT at S(1). It is concluded that 5-HT release in septal nuclei of the rat is modulated by presynaptic 5-HT(1B) autoreceptors, as well as by alpha(2)-, mu- and kappa-opioid heteroreceptors. All of these receptors seem to be under a tonic inhibitory influence of the corresponding endogenous agonists and show qualitatively comparable modulatory properties along the dorso-ventral distribution of the 5-HT terminals.
Collapse
Affiliation(s)
- S Rutz
- Institute of Experimental and Clinical Pharmacology and Toxicology, Laboratory of Neuropharmacology, University of Freiburg, Hansastrasse 9A, D-79104 Freiburg, Germany
| | | | | | | |
Collapse
|
41
|
Abstract
Opioids are extensively used in the management of all types of pain and their use is underpinned by extensive trial evidence and an abundance of practical experience. We are still amassing insight into how they achieve their pain-relieving effects, however, and this understanding becomes more complex as time progresses and shows that opioids are medications with complex and diverse central and peripheral nervous system effects. Despite the 200 years that have passed since the chemical isolation of morphine, every year brings new understanding of the mode of action, propensity to cause side effects, and appropriate clinical use of opioids. This article concentrates on this "new" evidence as disclosed by recent publications.
Collapse
Affiliation(s)
- Gary McCleane
- Rampark Pain Centre, 2 Rampark Dromore Road, Lurgaqn BT66 7JH, Northern Ireland, UK.
| | | |
Collapse
|
42
|
Butelman ER, Mandau M, Tidgewell K, Prisinzano TE, Yuferov V, Kreek MJ. Effects of salvinorin A, a kappa-opioid hallucinogen, on a neuroendocrine biomarker assay in nonhuman primates with high kappa-receptor homology to humans. J Pharmacol Exp Ther 2006; 320:300-6. [PMID: 17060493 DOI: 10.1124/jpet.106.112417] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study focused on the in vivo effects of the kappa-opioid hallucinogen salvinorin A, derived from the plant Salvia divinorum. The effects of salvinorin A (0.0032-0.056 mg/kg i.v.) were studied in a neuroendocrine biomarker assay of the anterior pituitary hormone prolactin in gonadally intact, adult male and female rhesus monkeys (n = 4 each). Salvinorin A produced dose- and time-dependent neuroendocrine effects, similar to the synthetic high-efficacy kappa-agonist U69,593 ((+)-(5alpha,7 alpha,8beta)-N-methyl-N-[7-(1-pyrrolidiniyl)-1-oxaspiro[4.5]dec-8yl]-benzeneacetamide), but of shorter duration than the latter. Salvinorin A was approximately equipotent to U69,593 in this endpoint (salvinorin A ED50, 0.015 mg/kg; U69,593 ED(50), 0.0098 mg/kg). The effects of i.v. salvinorin A were not prevented by a small dose of the opioid antagonist nalmefene (0.01 mg/kg s.c.) but were prevented by a larger dose of nalmefene (0.1 mg/kg); the latter nalmefene dose is sufficient to produce kappa-antagonist effects in this species. In contrast, the 5HT2 receptor antagonist ketanserin (0.1 mg/kg i.m.) did not prevent the effects of salvinorin A. As expected, the neuroendocrine effects of salvinorin A (0.0032 mg/kg i.v.) were more robust in female than in male subjects. Related studies focused on full-length cloning of the coding region of the rhesus monkey kappa-opioid receptor (OPRK1) gene and revealed a high homology of the nonhuman primate OPRK1 gene compared with the human OPRK1 gene, including particular C-terminal residues thought to be involved in receptor desensitization and internalization. The present studies indicate that the hallucinogen salvinorin A acts as a high-efficacy kappa-agonist in nonhuman primates in a translationally viable neuroendocrine biomarker assay.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, Box 171, 1230 York Avenue, New York NY 10021, USA.
| | | | | | | | | | | |
Collapse
|