151
|
Jordan CJ, Xi ZX. Progress in brain cannabinoid CB 2 receptor research: From genes to behavior. Neurosci Biobehav Rev 2019; 98:208-220. [PMID: 30611802 PMCID: PMC6401261 DOI: 10.1016/j.neubiorev.2018.12.026] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/12/2018] [Accepted: 12/22/2018] [Indexed: 01/01/2023]
Abstract
The type 2 cannabinoid receptor (CB2R) was initially regarded as a peripheral cannabinoid receptor. However, recent technological advances in gene detection, alongside the availability of transgenic mouse lines, indicate that CB2Rs are expressed in both neurons and glial cells in the brain under physiological and pathological conditions, and are involved in multiple functions at cellular and behavioral levels. Brain CB2Rs are inducible and neuroprotective via up-regulation in response to various insults, but display species differences in gene and receptor structures, CB2R expression, and receptor responses to various CB2R ligands. CB2R transcripts also differ between the brain and spleen. In the brain, CB2A is the major transcript isoform, while CB2A and CB2B transcripts are present at higher levels in the spleen. These new findings regarding brain versus spleen CB2R isoforms may in part explain why early studies failed to detect brain CB2R gene expression. Here, we review evidence supporting the expression and function of brain CB2R from gene and receptor levels to cellular functioning, neural circuitry, and animal behavior.
Collapse
Affiliation(s)
- Chloe J Jordan
- Addiction Biology Unit, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Zheng-Xiong Xi
- Addiction Biology Unit, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA.
| |
Collapse
|
152
|
Zhou K, Zhu Y. The paraventricular thalamic nucleus: A key hub of neural circuits underlying drug addiction. Pharmacol Res 2019; 142:70-76. [PMID: 30772461 DOI: 10.1016/j.phrs.2019.02.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 01/19/2019] [Accepted: 02/13/2019] [Indexed: 12/15/2022]
Abstract
Drug addiction is a chronic relapsing brain disease characterized by compulsive, out-of-control drug use and the appearance of negative somatic and emotional consequences when drug access is prevented. The limited efficacy of treatment urges researchers toward a deeper understanding of the neural mechanism of drug addiction. Brain circuits that regulate reward and motivation are considered to be the neural substrate of drug addiction. An increasing body of literature indicates that the paraventricular thalamic nucleus (PVT) could serve as a key node in the neurocircuits that control goal-directed behaviors. In this review, we summarize the anatomical and functional evidence that the PVT regulates drug-related behaviors. The PVT receives extensive inputs from the brainstem and hypothalamus, and is reciprocally connected with the limbic system. Neurons in the PVT are recruited by drug exposure as well as cues and context associated with drug taking. Pathway-specific perturbation studies have begun to decipher the precise role of PVT circuits in drug-related behaviors. We also highlight recent findings about the involvement of neural plasticity of the PVT pathways in drug addiction and provide perspectives on future studies.
Collapse
Affiliation(s)
- Kuikui Zhou
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yingjie Zhu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
153
|
Vidyasagar TR, Levichkina E. An Integrated Neuronal Model of Claustral Function in Timing the Synchrony Between Cortical Areas. Front Neural Circuits 2019; 13:3. [PMID: 30804759 PMCID: PMC6371054 DOI: 10.3389/fncir.2019.00003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/14/2019] [Indexed: 11/22/2022] Open
Abstract
It has been suggested that the function of the claustrum (CL) may be to orchestrate and integrate the activity of the different cortical areas that are involved in a particular function by boosting the synchronized oscillations that occur between these areas. We propose here a model of how this may be done, thanks to the unique synaptic morphology of the CL and its excitatory and inhibitory connections with most cortical areas. Using serial visual search as an example, we describe how the functional anatomy of the claustral connections can potentially execute the sequential activation of the representations of objects that are being processed serially. We also propose that cross-frequency coupling (CFC) between low frequency signals from CL and higher frequency oscillations in the cortical areas will be an efficient means of CL modulating neural activity across multiple brain regions in synchrony. This model is applicable to the wide range of functions one performs, from simple object recognition to reading and writing, listening to or performing music, etc.
Collapse
Affiliation(s)
- Trichur R. Vidyasagar
- Department of Optometry and Vision Science, University of Melbourne, Parkville, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Australian Research Council Centre of Excellence in Integrative Brain Function, University of Melbourne Node, Melbourne, VIC, Australia
| | - Ekaterina Levichkina
- Department of Optometry and Vision Science, University of Melbourne, Parkville, VIC, Australia
- Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
154
|
Smith JB, Alloway KD, Hof PR, Orman R, Reser DH, Watakabe A, Watson GDR. The relationship between the claustrum and endopiriform nucleus: A perspective towards consensus on cross-species homology. J Comp Neurol 2019; 527:476-499. [PMID: 30225888 PMCID: PMC6421118 DOI: 10.1002/cne.24537] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 01/08/2023]
Abstract
With the emergence of interest in studying the claustrum, a recent special issue of the Journal of Comparative Neurology dedicated to the claustrum (Volume 525, Issue 6, pp. 1313-1513) brought to light questions concerning the relationship between the claustrum (CLA) and a region immediately ventral known as the endopiriform nucleus (En). These structures have been identified as separate entities in rodents but appear as a single continuous structure in primates. During the recent Society for Claustrum Research meeting, a panel of experts presented data pertaining to the relationship of these regions and held a discussion on whether the CLA and En should be considered (a) separate unrelated structures, (b) separate nuclei within the same formation, or (c) subregions of a continuous structure. This review article summarizes that discussion, presenting comparisons of the cytoarchitecture, neurochemical profiles, genetic markers, and anatomical connectivity of the CLA and En across several mammalian species. In rodents, we conclude that the CLA and the dorsal endopiriform nucleus (DEn) are subregions of a larger complex, which likely performs analogous computations and exert similar effects on their respective cortical targets (e.g., sensorimotor versus limbic). Moving forward, we recommend that the field retain the nomenclature currently employed for this region but should continue to examine the delineation of these structures across different species. Using thorough descriptions of a variety of anatomical features, this review offers a clear definition of the CLA and En in rodents, which provides a framework for identifying homologous structures in primates.
Collapse
Affiliation(s)
- Jared B. Smith
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Kevin D. Alloway
- Neural and Behavioral Sciences, Center for Neural Engineering, Pennsylvania State University, University Park, PA 16802, USA
| | - Patrick R. Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rena Orman
- Department of Physiology and Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY, 11203 USA
| | - David H. Reser
- Graduate Entry Medicine Program, Monash Rural Health Churchill, Monash University, Churchill, Victoria 3842, Australia
- Department of Physiology, Monash University, Clayton 3800, Victoria, Australia
| | | | - Glenn D. R. Watson
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
| |
Collapse
|
155
|
Klawonn AM, Malenka RC. Nucleus Accumbens Modulation in Reward and Aversion. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2019; 83:119-129. [PMID: 30674650 PMCID: PMC6650377 DOI: 10.1101/sqb.2018.83.037457] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The nucleus accumbens (NAc) is a key node of the brain’s circuitry that is responsible for translating motivation into action. It has been implicated in playing critical roles in virtually all forms of adaptive and pathological motivated behaviors. It is subject to modulation by a broad array of inputs that influence NAc activity in complex ways that are still poorly understood. Here, we briefly review current knowledge about the behavioral consequences of NAc modulation, focusing on recent studies that use novel techniques developed and implemented over the last decade.
Collapse
Affiliation(s)
- Anna M Klawonn
- Department of Psychiatry and Behavioral Sciences, Nancy Pritzker Laboratory, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Robert C Malenka
- Department of Psychiatry and Behavioral Sciences, Nancy Pritzker Laboratory, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
156
|
Li Y, Li CY, Xi W, Jin S, Wu ZH, Jiang P, Dong P, He XB, Xu FQ, Duan S, Zhou YD, Li XM. Rostral and Caudal Ventral Tegmental Area GABAergic Inputs to Different Dorsal Raphe Neurons Participate in Opioid Dependence. Neuron 2019; 101:748-761.e5. [PMID: 30638902 DOI: 10.1016/j.neuron.2018.12.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/26/2018] [Accepted: 12/07/2018] [Indexed: 12/14/2022]
Abstract
Both the ventral tegmental area (VTA) and dorsal raphe nucleus (DRN) are involved in affective control and reward-related behaviors. Moreover, the neuronal activities of the VTA and DRN are modulated by opioids. However, the precise circuits from the VTA to DRN and how opioids modulate these circuits remain unknown. Here, we found that neurons projecting from the VTA to DRN are primarily GABAergic. Rostral VTA (rVTA) GABAergic neurons preferentially innervate DRN GABAergic neurons, thus disinhibiting DRN serotonergic neurons. Optogenetic activation of this circuit induces aversion. In contrast, caudal VTA (cVTA) GABAergic neurons mainly target DRN serotonergic neurons, and activation of this circuit promotes reward. Importantly, μ-opioid receptors (MOPs) are selectively expressed at rVTA→DRN GABAergic synapses, and morphine depresses the synaptic transmission. Chronically elevating the activity of the rVTA→DRN pathway specifically interrupts morphine-induced conditioned place preference. This opioid-modulated inhibitory circuit may yield insights into morphine reward and dependence pathogenesis.
Collapse
Affiliation(s)
- Yue Li
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chun-Yue Li
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wang Xi
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Sen Jin
- CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China
| | - Zuo-Hang Wu
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ping Jiang
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ping Dong
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiao-Bin He
- CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China
| | - Fu-Qiang Xu
- CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China
| | - Shumin Duan
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yu-Dong Zhou
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiao-Ming Li
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
157
|
Abstract
Whilst the nociceptin/orphanin FQ (N/OFQ) receptor (NOP) has similar intracellular coupling mechanisms to opioid receptors, it has distinct modulatory effects on physiological functions such as pain. These actions range from agonistic to antagonistic interactions with classical opioids within the spinal cord and brain, respectively. Understanding the electrophysiological actions of N/OFQ has been crucial in ascertaining the mechanisms by which these agonistic and antagonistic interactions occur. These similarities and differences between N/OFQ and opioids are due to the relative location of NOP versus opioid receptors on specific neuronal elements within these CNS regions. These mechanisms result in varied cellular actions including postsynaptic modulation of ion channels and presynaptic regulation of neurotransmitter release.
Collapse
|
158
|
Paretkar T, Dimitrov E. Activation of enkephalinergic (Enk) interneurons in the central amygdala (CeA) buffers the behavioral effects of persistent pain. Neurobiol Dis 2018; 124:364-372. [PMID: 30572023 DOI: 10.1016/j.nbd.2018.12.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/23/2018] [Accepted: 12/10/2018] [Indexed: 12/25/2022] Open
Abstract
Enk neurons in CeA modulate the activity of the amygdala projection neurons and it is very likely that changes of Enk signaling cause the heightened anxiety that accompanies chronic pain. We use chemogenetics and transgenic mice to investigate the effects of acute and continuous activation of the amygdala Enk neurons on persistent pain and anxiodepressive-like behavior in mice. Enk-cre mice were injected bilaterally into the CeA with cre-activated AAV-DREADD/Gq/mCherry, while neuropathic pain was induced by sciatic nerve constriction. A single injection of DREADD's ligand CNO decreased the anxiety-like behavior in both, uninjured mice and in mice with neuropathic pain and produced robust analgesia that lasted for 24 h. Furthermore, the activation of Enk neurons by the DREADD ligand led to increased c-Fos expression in PKC-δ interneurons of the CeA and in non-serotonergic neurons in the ventrolateral periaqueductal gray (vlPAG), a brain structure that is an essential part of the descending pain inhibitory system. Next, we added CNO to the drinking water of the experimental mice for 14 days in order to assess the effects of continuous activation of CeA Enk interneurons on anxiodepressive-like behavior, which is affected by chronic pain. The prolonged activation of the CeA Enk interneurons reduced neohypophagia in the novelty suppressed feeding test and increased ΔFosB (a marker for sustained neuronal activation) in the vlPAG of mice with chronic pain. All together, the results of our experiments point to an important role of the CeA Enk neurons in the control of both nociception and emotion. Activation of Enk neurons resulted in sustained analgesia accompanied by anxiolysis and antidepressant effects. Very likely, these effects of CeA Enk neurons are result of the activation of vlPAG, a brain region that is essential not only for descending inhibition of pain but it is also a core element in the resilience to stress.
Collapse
Affiliation(s)
- Tanvi Paretkar
- Department of Physiology and Biophysics, Chicago Medical School Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd., North Chicago, IL 60064, United States.
| | - Eugene Dimitrov
- Department of Physiology and Biophysics, Chicago Medical School Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd., North Chicago, IL 60064, United States.
| |
Collapse
|
159
|
Szereda-Przestaszewska M, Kaczyńska K. Pharmacologically evoked apnoeas. Receptors and nervous pathways involved. Life Sci 2018; 217:237-242. [PMID: 30553870 DOI: 10.1016/j.lfs.2018.12.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/05/2018] [Accepted: 12/12/2018] [Indexed: 01/07/2023]
Abstract
This review analyses the knowledge about the incidence of transient apnoeic spells, induced by substances which activate vagal chemically sensitive afferents. It considers the specificity and expression of appropriate receptors, and relevant research on pontomedullary circuits contributing to a cessation of respiration. Insight is gained into an excitatory drive of 5-HT1A serotonin receptors in overcoming opioid-induced respiratory inhibition.
Collapse
Affiliation(s)
- Małgorzata Szereda-Przestaszewska
- Department of Respiration Physiology, Mossakowski Medical Research Centre Polish Academy of Sciences, A. Pawińskiego 5, 02-106 Warsaw, Poland
| | - Katarzyna Kaczyńska
- Department of Respiration Physiology, Mossakowski Medical Research Centre Polish Academy of Sciences, A. Pawińskiego 5, 02-106 Warsaw, Poland.
| |
Collapse
|
160
|
Yang L, Brooks AF, Makaravage KJ, Zhang H, Sanford MS, Scott PJH, Shao X. Radiosynthesis of [ 11C]LY2795050 for Preclinical and Clinical PET Imaging Using Cu(II)-Mediated Cyanation. ACS Med Chem Lett 2018; 9:1274-1279. [PMID: 30613339 DOI: 10.1021/acsmedchemlett.8b00460] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/13/2018] [Indexed: 01/05/2023] Open
Abstract
Copper-mediated 11C-cyanation reactions have enabled the synthesis of PET radiotracers from a range of readily available precursors and avoid the need to use more toxic Pd catalysts. In this work we adapt our recently developed 11C-cyanation of arylpinacolboronate (BPin) esters for the cGMP synthesis of [11C]LY2795050, a selective antagonist radiotracer for the kappa opioid receptor (KOR). [11C]LY2795050 was synthesized in 6 ± 1% noncorrected radiochemical yield (based on [11C]HCN, n = 3) using an automated synthesis module. Quality control testing confirmed the suitability of doses for preclinical and clinical PET imaging (radiochemical purity >99%; specific activity >900 mCi/μmol; residual Cu < 0.1 μg/mL). PET imaging was conducted in rodent and nonhuman primates, showing good brain uptake of [11C]LY2795050 and the expected distribution of KOR. Analogous imaging with [11C]carfentanil (a selective mu opioid receptor (MOR) radiotracer) revealed the anticipated regional differences in MOR and KOR distribution in the primate brain.
Collapse
Affiliation(s)
- Lingyun Yang
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Allen F. Brooks
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Katarina J. Makaravage
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Huibin Zhang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Melanie S. Sanford
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Peter J. H. Scott
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Xia Shao
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
161
|
Untangling the complexity of opioid receptor function. Neuropsychopharmacology 2018; 43:2514-2520. [PMID: 30250308 PMCID: PMC6224460 DOI: 10.1038/s41386-018-0225-3] [Citation(s) in RCA: 200] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/29/2018] [Accepted: 09/10/2018] [Indexed: 02/07/2023]
Abstract
Mu opioid receptor agonists are among the most powerful analgesic medications but also among the most addictive. The current opioid crisis has energized a quest to develop opioid analgesics that are devoid of untoward effects. Since their discovery in the 1970's, there have been major advances in our understanding of the endogenous opioid systems that these drugs target. Yet many questions remain and the development of non-addictive opioid analgesics has not been achieved. However, access to new molecular, genetic and computational tools have begun to elucidate the structural dynamics of opioid receptors, the scaffolding that links them to intracellular signaling cascades, their cellular trafficking and the distinct ways that various opioid drugs modify them. This mini-review highlights some of the chemical and pharmacological findings and new perspectives that have arisen from studies using these tools. They reveal multiple layers of complexity of opioid receptor function, including a spatiotemporal specificity in opioid receptor-induced cellular signaling, ligand-directed biased signaling, allosteric modulation of ligand interactions, heterodimerization of different opioid receptors, and the existence of slice variants with different ligand specificity. By untangling these layers, basic research into the chemistry and pharmacology of opioid receptors is guiding the way towards deciphering the mysteries of tolerance and physical dependence that have plagued the field and is providing a platform for the development of more effective and safer opioids.
Collapse
|
162
|
Ryan JD, Zhou Y, Contoreggi NH, Bshesh FK, Gray JD, Kogan JF, Ben KT, McEwen BS, Jeanne Kreek M, Milner TA. Sex Differences in the Rat Hippocampal Opioid System After Oxycodone Conditioned Place Preference. Neuroscience 2018; 393:236-257. [PMID: 30316908 PMCID: PMC6246823 DOI: 10.1016/j.neuroscience.2018.10.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/10/2018] [Accepted: 10/01/2018] [Indexed: 12/12/2022]
Abstract
Although opioid addiction has risen dramatically, the role of gender in addiction has been difficult to elucidate. We previously found sex-dependent differences in the hippocampal opioid system of Sprague-Dawley rats that may promote associative learning relevant to drug abuse. The present studies show that although female and male rats acquired conditioned place preference (CPP) to the mu-opioid receptor (MOR) agonist oxycodone (3 mg/kg, I.P.), hippocampal opioid circuits were differentially altered. In CA3, Leu-Enkephalin-containing mossy fibers had elevated levels in oxycodone CPP (Oxy) males comparable to those in females and sprouted in Oxy-females, suggesting different mechanisms for enhancing opioid sensitivity. Electron microscopy revealed that in Oxy-males delta opioid receptors (DORs) redistributed to mossy fiber-CA3 synapses in a manner resembling females that we previously showed is important for opioid-mediated long-term potentiation. Moreover, in Oxy-females DORs redistributed to CA3 pyramidal cell spines, suggesting the potential for enhanced plasticity processes. In Saline-injected (Sal) females, dentate hilar parvalbumin-containing basket interneuron dendrites had fewer MORs, however plasmalemmal and total MORs increased in Oxy-females. In dentate hilar GABAergic dendrites that contain neuropeptide Y, Sal-females compared to Sal-males had higher plasmalemmal DORs, and near-plasmalemmal DORs increased in Oxy-females. This redistribution of MORs and DORs within hilar interneurons in Oxy-females would potentially enhance disinhibition of granule cells via two different circuits. Together, these results indicate that oxycodone CPP induces sex-dependent redistributions of opioid receptors in hippocampal circuits in a manner facilitating opioid-associative learning processes and may help explain the increased susceptibility of females to opioid addiction acquisition and relapse.
Collapse
Affiliation(s)
- James D Ryan
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States.
| | - Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Natalina H Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States
| | - Farah K Bshesh
- Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, P.O. Box 24144 Doha, Qatar
| | - Jason D Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Joshua F Kogan
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Konrad T Ben
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States.
| |
Collapse
|
163
|
Endogenous opioid signalling in the brain during pregnancy and lactation. Cell Tissue Res 2018; 375:69-83. [DOI: 10.1007/s00441-018-2948-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/10/2018] [Indexed: 12/24/2022]
|
164
|
Molecular Adaptations in the Rat Dorsal Striatum and Hippocampus Following Abstinence-Induced Incubation of Drug Seeking After Escalated Oxycodone Self-Administration. Mol Neurobiol 2018; 56:3603-3615. [PMID: 30155791 PMCID: PMC6477015 DOI: 10.1007/s12035-018-1318-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022]
Abstract
Repeated exposure to the opioid agonist, oxycodone, can lead to addiction. Here, we sought to identify potential neurobiological consequences of withdrawal from escalated and non-escalated oxycodone self-administration in rats. To reach these goals, we used short-access (ShA) (3 h) and long-access (LgA) (9 h) exposure to oxycodone self-administration followed by protracted forced abstinence. After 31 days of withdrawal, we quantified mRNA and protein levels of opioid receptors in the rat dorsal striatum and hippocampus. Rats in the LgA, but not the ShA, group exhibited escalation of oxycodone SA, with distinction of two behavioral phenotypes of relatively lower (LgA-L) and higher (LgA-H) oxycodone takers. Both LgA, but not ShA, phenotypes showed time-dependent increases in oxycodone seeking during the 31 days of forced abstinence. Rats from both LgA-L and LgA-H groups also exhibited decreased levels of striatal mu opioid receptor protein levels in comparison to saline and ShA rats. In contrast, mu opioid receptor mRNA expression was increased in the dorsal striatum of LgA-H rats. Moreover, hippocampal mu and kappa receptor protein levels were both increased in the LgA-H phenotype. Nevertheless, hippocampal mu receptor mRNA levels were decreased in the two LgA groups whereas kappa receptor mRNA expression was decreased in ShA and LgA oxycodone groups. Decreases in striatal mu opioid receptor protein expression in the LgA rats may serve as substrates for relapse to drug seeking because these changes occur in rats that showed incubation of oxycodone seeking.
Collapse
|
165
|
Abstract
Opioids are the most commonly used and effective analgesic treatments for severe pain, but they have recently come under scrutiny owing to epidemic levels of abuse and overdose. These compounds act on the endogenous opioid system, which comprises four G protein-coupled receptors (mu, delta, kappa, and nociceptin) and four major peptide families (β-endorphin, enkephalins, dynorphins, and nociceptin/orphanin FQ). In this review, we first describe the functional organization and pharmacology of the endogenous opioid system. We then summarize current knowledge on the signaling mechanisms by which opioids regulate neuronal function and neurotransmission. Finally, we discuss the loci of opioid analgesic action along peripheral and central pain pathways, emphasizing the pain-relieving properties of opioids against the affective dimension of the pain experience.
Collapse
Affiliation(s)
- Gregory Corder
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Palo Alto, California 94304, USA; .,Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, California 94304, USA.,Department of Neurosurgery, Stanford University, Palo Alto, California 94304, USA.,Stanford Neurosciences Institute, Palo Alto, California 94304, USA
| | - Daniel C Castro
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri 63130, USA;
| | - Michael R Bruchas
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri 63130, USA; .,Division of Basic Research, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63130, USA.,Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri 63130, USA.,Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri 63130, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, USA
| | - Grégory Scherrer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Palo Alto, California 94304, USA; .,Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, California 94304, USA.,Department of Neurosurgery, Stanford University, Palo Alto, California 94304, USA.,Stanford Neurosciences Institute, Palo Alto, California 94304, USA.,New York Stem Cell Foundation - Robertson Investigator, Stanford University, Palo Alto, California 94304, USA
| |
Collapse
|
166
|
Morphine-potentiated cognitive deficits correlate to suppressed hippocampal iNOS RNA expression and an absent type 1 interferon response in LP-BM5 murine AIDS. J Neuroimmunol 2018. [PMID: 29526406 DOI: 10.1016/j.jneuroim.2018.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Opioid use accelerates neurocognitive impairment in HIV/AIDS patients. We assessed the effect of chronic morphine treatment and LP-BM5/murine AIDS (MAIDS) infection on cognition, cytokine production, and type 1 interferon (IFN) expression in the murine CNS. Morphine treatment decreased expression of pro-inflammatory factors (CCL5, iNOS) and reduced cognitive performance in LP-BM5-infected mice, correlating to increased hippocampal viral load and a blunted type 1 IFN response. In the striatum, morphine reduced viral load while increasing IFN-α RNA expression. Our results suggest that differentially regulated type 1 IFN responses may contribute to distinct regional outcomes in the hippocampus and striatum in LP-BM5/MAIDS.
Collapse
|
167
|
Role of dopamine D1 receptor in 3-fluoromethamphetamine-induced neurotoxicity in mice. Neurochem Int 2018; 113:69-84. [DOI: 10.1016/j.neuint.2017.11.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/14/2017] [Accepted: 11/28/2017] [Indexed: 01/26/2023]
|
168
|
|
169
|
Mallick-Searle T, Fillman M. The pathophysiology, incidence, impact, and treatment of opioid-induced nausea and vomiting. J Am Assoc Nurse Pract 2018; 29:704-710. [PMID: 29131554 DOI: 10.1002/2327-6924.12532] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/02/2017] [Accepted: 10/03/2017] [Indexed: 12/11/2022]
Abstract
PURPOSE Opioid medications are integral in managing acute moderate-to-severe pain. Opioid analgesics bind to μ (mu), κ (kappa), or δ (delta) opioid receptors in the brain, spinal cord, and digestive tract. However, opioids cause adverse effects that may interfere with their therapeutic use. Some adverse effects wane over time, but patients using opioids for acute pain struggle with opioid-induced nausea and vomiting (OINV) the entire time they take the opioid. This article discusses the underlying mechanisms, clinical implications, and treatment strategies of OINV. DATA SOURCES Systematic search and review of Medline, PubMed, and Google Scholar for articles relating to OINV. In addition, package inserts provided pharmacologic data and dose recommendations as needed. CONCLUSIONS Research suggests approximately 40% of patients may experience nausea and 15%-25% of patients may experience vomiting after opioid administration. Nausea often precedes vomiting, although they can occur separately. Many patients receiving opioids rate the nausea and vomiting as worse than their pain. Nausea and vomiting can lead to complications including electrolyte imbalances, malnutrition, and volume depletion, and can also negatively affect quality of life and postoperative recovery. IMPLICATIONS FOR PRACTICE There are several medications that can be used to treat OINV including serotonin receptor antagonists, dopamine receptor antagonists, and neurokinin-1 receptor antagonists. Healthcare providers should be proactive about discussing OINV with patients, as this may improve patient outcomes and pain relief.
Collapse
Affiliation(s)
| | - Mechele Fillman
- Division Pain Medicine, Stanford Health Care, Stanford, California
| |
Collapse
|
170
|
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that compromises multiple neurochemical substrates including dopamine, norepinephrine, serotonin, acetylcholine, and glutamate systems. Loss of these transmitter systems initiates a cascade of neurological deficits beginning with motor function and ending with dementia. Current therapies primarily address the motor symptoms of the disease via dopamine replacement therapy. Exogenous dopamine replacement brings about additional challenges since after years of treatment it almost invariably gives rise to dyskinesia as a side effect. Therefore there is a clear unmet clinical need for improved PD therapeutics. Opioid receptors and their respective peptides are expressed throughout the basal ganglia and cortex where monoaminergic denervation strongly contributes to PD pathology. Delta opioid receptors are of particular interest because of their dense localization in basal ganglia and because activating this system is known to enhance locomotor activity under a variety of conditions. This chapter will outline much of the work that has demonstrated the effectiveness of delta opioid receptor activation in models of PD and its neuroprotective properties. It also discusses some of the challenges that must be addressed before moving delta opioid receptor agonists into a clinical setting.
Collapse
Affiliation(s)
- Omar S Mabrouk
- Department of Chemistry, University of Michigan, 930 North University, Ann Arbor, MI, 48109, USA.
- Department of Pharmacology, University of Michigan, 930 North University, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
171
|
Abstract
Nowadays, the delta opioid receptor (DOPr) represents a promising target for the treatment of chronic pain and emotional disorders. Despite the fact that they produce limited antinociceptive effects in healthy animals and in most acute pain models, DOPr agonists have shown efficacy in various chronic pain models. In this chapter, we review the progresses that have been made over the last decades in understanding the role played by DOPr in the control of pain. More specifically, the distribution of DOPr within the central nervous system and along pain pathways is presented. We also summarize the literature supporting a role for DOPr in acute, tonic, and chronic pain models, as well as the mechanisms regulating its activity under specific conditions. Finally, novel compounds that have make their way to clinical trials are discussed.
Collapse
Affiliation(s)
- Khaled Abdallah
- Département de pharmacologie-physiologie, Université de Sherbrooke, Sherbrooke, QC, Canada
- Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de recherche du CHUS, Sherbrooke, QC, Canada
| | - Louis Gendron
- Département de pharmacologie-physiologie, Université de Sherbrooke, Sherbrooke, QC, Canada.
- Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
- Centre de recherche du CHUS, Sherbrooke, QC, Canada.
- Département d'anesthésiologie, Université de Sherbrooke, Sherbrooke, QC, Canada.
- Quebec Pain Research Network, Sherbrooke, QC, Canada.
| |
Collapse
|
172
|
Frontal cortex dysfunction as a target for remediation in opiate use disorder: Role in cognitive dysfunction and disordered reward systems. PROGRESS IN BRAIN RESEARCH 2018; 239:179-227. [DOI: 10.1016/bs.pbr.2018.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
173
|
Antidepressant-like effects of 3-carboxamido seco-nalmefene (3CS-nalmefene), a novel opioid receptor modulator, in a rat IFN-α-induced depression model. Brain Behav Immun 2018; 67:152-162. [PMID: 28844812 DOI: 10.1016/j.bbi.2017.08.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/27/2017] [Accepted: 08/22/2017] [Indexed: 01/09/2023] Open
Abstract
Patients receiving the cytokine immunotherapy, interferon-alpha (IFN-α) frequently present with neuropsychiatric consequences and cognitive impairments. Patients (25-80%) report symptoms of depression, including, anhedonia, irritability, fatigue and impaired motivation. Our lab has previously demonstrated treatment (170,000IU/kg sc, 3 times per week for 4weeks) of the pro-inflammatory cytokine, IFN-α, induced a depressive phenotype in rats in the forced swim test (FST). Here, we examine the biological mechanisms underlying behavioral changes induced by IFN-α, which may be reflective of mechanisms underlying inflammation associated depression. We also investigate the potential of 3-carboxamido seco-nalmefene (3CS-nalmefene), a novel opioid modulator (antagonist at mu and partial agonist at kappa and delta opioid receptors in vitro), to reverse IFN-α induced changes. In vitro radioligand receptor binding assays and the [35S] GTPγS were performed to determine the affinity of 3CS-nalmefene for the mu, kappa and delta opioid receptors. IFN-α treatment increased circulating and central markers of inflammation and hypothalamic-pituitaryadrenal (HPA) axis activity (IL-6, IL-1β and corticosterone) while increasing immobility in the FST, impairing of object displacement learning in the object exploration task (OET), and decreasing neuronal proliferation and brain-derived neurotrophic factor (BDNF) in the hippocampus. Treatment with 3CS-nalmefene (0.3mg/kg/sc twice per day, 3 times per week for 4weeks) prevented IFN-α-induced immobility in the FST and impaired object displacement learning. In addition, 3CS-nalmefene prevented IFN-α-induced increases in inflammation and hyperactivity of the HPA-axis, the IFN-α-induced reduction in both neuronal proliferation and BDNF expression in the hippocampus. Overall, these preclinical data would support the hypothesis that opioid receptor modulation is a relevant target for treatment of depression.
Collapse
|
174
|
Dhull DK, Kumar A. Tramadol ameliorates behavioural, biochemical, mitochondrial and histological alterations in ICV-STZ-induced sporadic dementia of Alzheimer's type in rats. Inflammopharmacology 2017; 26:925-938. [PMID: 29249049 DOI: 10.1007/s10787-017-0431-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 12/06/2017] [Indexed: 12/23/2022]
Abstract
Alzheimer disease represents a major public health issue with limited therapeutic interventions. We explored the possibility of therapeutic approach by repurposing of tramadol in a sporadic animal model of Alzheimer's type. Streptozocin (STZ 3 mg/kg; bilaterally) was injected to male SD rats through intracerebroventricular (ICV) route. Drug treatment was started just after streptozocin administration and continued for 3 weeks. The rats were killed on the 21st day following the last behavioral test, and cytoplasmic fractions of the hippocampus and pre-frontal cortex were prepared for the quantification of acetylcholinesterase, oxidative stress parameter, mitochondrial enzymes activity and histological examination. Tramadol (5, 10 and 20 mg/kg, i.p.) was used as a treatment drug, and memantine (10 mg/kg, i.p.) was used as a standard. Tramadol significantly attenuated behavioral, biochemical, mitochondrial and histological alterations at low (5 mg/kg) and intermediate (10 mg/kg) dose, suggesting its neuroprotective potential in ICV-STZ-treated rats. Further, the neuroprotective effect of tramadol (10 mg/kg) was comparable to memantine (10 mg/kg). In conclusion, our results indicate the effectiveness of tramadol in preventing ICV-STZ-induced cognitive impairment as well as mito-oxidative stress. Further, these findings reveal the possibility of MOR agonist as a therapeutic approach for sporadic Alzheimer disease.
Collapse
Affiliation(s)
- Dinesh K Dhull
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
175
|
Melanin-Concentrating Hormone acts through hypothalamic kappa opioid system and p70S6K to stimulate acute food intake. Neuropharmacology 2017; 130:62-70. [PMID: 29191753 DOI: 10.1016/j.neuropharm.2017.11.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/28/2017] [Accepted: 11/24/2017] [Indexed: 12/12/2022]
Abstract
Melanin-Concentrating Hormone (MCH) is one of the most relevant orexigenic factors specifically located in the lateral hypothalamic area (LHA), with its physiological relevance demonstrated in studies using several genetically manipulated mice models. However, the central mechanisms controlling MCH-induced hyperphagia remain largely uncharacterized. Here, we show that central injection of MCH in mice deficient for kappa opoid receptor (k-OR) failed to stimulate feeding. To determine the hypothalamic area responsible for this MCH/k-OR interaction, we performed virogenetic studies and found that downregulation of k-OR by adeno-associated viruses (shOprk1-AAV) in LHA, but not in other hypothalamic nuclei, was sufficient to block MCH-induced food intake. Next, we sought to investigate the molecular signaling pathway within the LHA that mediates acute central MCH stimulation of food intake. We found that MCH activates k-OR and that increased levels of phosphorylated extracellular signal regulated kinase (ERK) are associated with downregulation of phospho-S6 Ribosomal Protein. This effect was prevented when a pharmacological inhibitor of k-OR was co-administered with MCH. Finally, the specific activation of the direct upstream regulator of S6 (p70S6K) in the LHA attenuated MCH-stimulated food consumption. Our results reveal that lateral hypothalamic k-OR system modulates the orexigenic action of MCH via the p70S6K/S6 pathway.
Collapse
|
176
|
Ghasemi H, Tamaddonfard E, Soltanalinejad F. Role of thalamic ventral posterolateral nucleus histamine H 2 and opiate receptors in modulation of formalin-induced muscle pain in rats. Pharmacol Rep 2017; 69:1393-1401. [DOI: 10.1016/j.pharep.2017.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/18/2017] [Accepted: 05/05/2017] [Indexed: 02/02/2023]
|
177
|
Heath E, Chieng B, Christie MJ, Balleine BW. Substance P and dopamine interact to modulate the distribution of delta‐opioid receptors on cholinergic interneurons in the striatum. Eur J Neurosci 2017; 47:1159-1173. [DOI: 10.1111/ejn.13750] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 10/11/2017] [Accepted: 10/12/2017] [Indexed: 11/28/2022]
Affiliation(s)
- Emily Heath
- Brain & Mind Centre University of Sydney Sydney NSW Australia
| | - Billy Chieng
- Decision Neuroscience Lab School of Psychology University of New South Wales Kensington NSW 2052 Australia
| | | | - Bernard W. Balleine
- Brain & Mind Centre University of Sydney Sydney NSW Australia
- Decision Neuroscience Lab School of Psychology University of New South Wales Kensington NSW 2052 Australia
| |
Collapse
|
178
|
Role of orexin type-1 receptors in paragiganto-coerulear modulation of opioid withdrawal and tolerance: A site specific focus. Neuropharmacology 2017; 126:25-37. [DOI: 10.1016/j.neuropharm.2017.08.024] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/13/2017] [Accepted: 08/16/2017] [Indexed: 11/21/2022]
|
179
|
Morales I, Currie PJ, Hackenberg TD, Pastor R. Opioidergic and dopaminergic modulation of cost/benefit decision-making in Long Evans Rats. Physiol Behav 2017; 179:442-450. [DOI: 10.1016/j.physbeh.2017.07.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 07/12/2017] [Accepted: 07/12/2017] [Indexed: 10/19/2022]
|
180
|
Tan LA, Vaughan JM, Perrin MH, Rivier JE, Sawchenko PE. Distribution of corticotropin-releasing factor (CRF) receptor binding in the mouse brain using a new, high-affinity radioligand, [125I]-PD-Sauvagine. J Comp Neurol 2017; 525:3840-3864. [DOI: 10.1002/cne.24307] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/15/2017] [Accepted: 08/16/2017] [Indexed: 01/30/2023]
Affiliation(s)
- Laura A. Tan
- Laboratory of Neuronal Structure and Function; The Salk Institute for Biological Studies; La Jolla CA
| | - Joan M. Vaughan
- Laboratory of Neuronal Structure and Function; The Salk Institute for Biological Studies; La Jolla CA
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies; La Jolla CA
| | - Marilyn H. Perrin
- Laboratory of Neuronal Structure and Function; The Salk Institute for Biological Studies; La Jolla CA
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies; La Jolla CA
| | - Jean E. Rivier
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies; La Jolla CA
| | - Paul E. Sawchenko
- Laboratory of Neuronal Structure and Function; The Salk Institute for Biological Studies; La Jolla CA
| |
Collapse
|
181
|
Benedito RB, Alves MF, Pereira WB, de Arruda Torres P, Costa JP, da Rocha Tomé A, de Cássia da Silveira e Sá R, de Sousa DP, Ferreira PMP, de Freitas RM, de Fatima F. Melo Diniz M, de Almeida RN. Perillyl Alcohol: Antinociceptive Effects and Histopathological Analysis in Rodent Brains. Nat Prod Commun 2017. [DOI: 10.1177/1934578x1701200902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Perillyl alcohol (PA) is a natural compound found in essential oils. In this study, the antinociceptive activity of PA was evaluated using acetic acid and formalin tests. The involvement of the opioid system in its mechanism of action was investigated. Potential histological changes in the hippocampus and striatum were also assessed. In the acetic acid induced writhing tests, the mice pretreated with PA exhibited significant reductions in writhing. PA inhibited formalin injected paw licking response, and naloxone partially reversed the antinociceptive activity of perillyl alcohol during the writhing test. And as for the histopathological evaluation, PA did not cause significant tissue changes. This study suggests that PA possesses antinociceptive effects without significant hippocampus or striatum neurotoxicity, and that its activity involves opioid.
Collapse
Affiliation(s)
- Rubens Batista Benedito
- Federal University of Paraíba, Research Institute for Drugs and Medicines, P.O. Box 5009, CEP 58.051–900, João Pessoa, Paraíba, Brazil
| | - Mateus Feitosa Alves
- Federal University of Paraíba, Research Institute for Drugs and Medicines, P.O. Box 5009, CEP 58.051–900, João Pessoa, Paraíba, Brazil
| | - Wendel Batista Pereira
- Federal University of Paraíba, Research Institute for Drugs and Medicines, P.O. Box 5009, CEP 58.051–900, João Pessoa, Paraíba, Brazil
| | - Paula de Arruda Torres
- Federal University of Paraíba, Research Institute for Drugs and Medicines, P.O. Box 5009, CEP 58.051–900, João Pessoa, Paraíba, Brazil
| | - Jéssica Pereira Costa
- Research Laboratory in Experimental Neurochemistry, Post-Graduation Program in Pharmaceutical Science, Federal University of Piauí, CEP 64.049–550, Teresina, Piauí, Brazil
| | - Adriana da Rocha Tomé
- State University of Ceará, Av. Paranjana, 1700, Campus of Itaperi, CEP 60.740-000, Fortaleza, Ceará, Brazil
| | - Rita de Cássia da Silveira e Sá
- Federal University of Paraíba, Research Institute for Drugs and Medicines, P.O. Box 5009, CEP 58.051–900, João Pessoa, Paraíba, Brazil
| | - Damião Pergentino de Sousa
- Federal University of Paraíba, Research Institute for Drugs and Medicines, P.O. Box 5009, CEP 58.051–900, João Pessoa, Paraíba, Brazil
| | - Paulo Michel Pinheiro Ferreira
- Research Laboratory in Experimental Neurochemistry, Post-Graduation Program in Pharmaceutical Science, Federal University of Piauí, CEP 64.049–550, Teresina, Piauí, Brazil
| | - Rivelilson Mendes de Freitas
- Research Laboratory in Experimental Neurochemistry, Post-Graduation Program in Pharmaceutical Science, Federal University of Piauí, CEP 64.049–550, Teresina, Piauí, Brazil
| | - Margareth de Fatima F. Melo Diniz
- Federal University of Paraíba, Research Institute for Drugs and Medicines, P.O. Box 5009, CEP 58.051–900, João Pessoa, Paraíba, Brazil
| | - Reinaldo Nóbrega de Almeida
- Federal University of Paraíba, Research Institute for Drugs and Medicines, P.O. Box 5009, CEP 58.051–900, João Pessoa, Paraíba, Brazil
| |
Collapse
|
182
|
Dopamine D1-like Receptors Regulate Constitutive, μ-Opioid Receptor-Mediated Repression of Use-Dependent Synaptic Plasticity in Dorsal Horn Neurons: More Harm than Good? J Neurosci 2017; 36:5661-73. [PMID: 27194343 DOI: 10.1523/jneurosci.2469-15.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 04/11/2016] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED The current study reports on a synaptic mechanism through which D1-like receptors (D1LRs) modulate spinal nociception and plasticity by regulating activation of the μ-opioid receptor (MOR).D1LR stimulation with agonist SKF 38393 concentration-dependently depressed C-fiber-evoked potentials in rats receiving spinal nerve ligation (SNL), but not in uninjured rats. Depression was prevented by MOR- but not GABA-receptor blockade. Neurons expressing the D1 subtype were immunopositive for met-enkephalin and vesicular glutamate transporter VGLUT2, but not for GABAergic marker vGAT.Nerve ligation was followed by increased immunoreactivity for D1 in synaptic compartment (P3) in dorsal horn homogenates and presynaptic met-enkephalin-containing boutons. SNL led to increased immunoreactivity for met-enkephalin in dorsal horn homogenates, which was dose-dependently attenuated by selective D1LR antagonist SCH 23390. During blockade of either D1R or MOR, low-frequency (0.2 or 3 Hz) stimulation (LFS) to the sciatic nerve induced long-term potentiation (LTP) of C-fiber-evoked potentials, revealing a constituent role of both receptors in repressing afferent-induced synaptic plasticity. LFS consistently induced NMDA receptor-dependent LTP in nerve-injured rats. The ability of MOR both to prevent LTP and to modulate mechanical and thermal pain thresholds in behavioral tests was preserved in nerve-ligated rats that were postoperatively treated with SCH 23390. D1LR priming for 30 min sufficed to disrupt MOR function in otherwise naive rats via a mechanism involving receptor overuse.The current data support that, whereas D1LR-modulated MOR activation is instrumental in antinociception and endogenous repression of synaptic plasticity, this mechanism deteriorates rapidly by sustained use, generating increased vulnerability to afferent input. SIGNIFICANCE STATEMENT The current study shows that dopamine D1-like receptors (D1LRs) and μ-opioid receptors (MOR) in the spinal dorsal horn constitutively repress the expression of synaptic long-term potentiation (LTP) of C-fiber-evoked potentials. Anatomical data are provided supporting that the D1 subtype regulates MOR function by modulating met-enkephalin release. Sustained neuropathic pain induced by spinal nerve ligation is accompanied by D1R and met-enkephalin upregulation, acquired D1LR-mediated antinociception, and a loss of endogenous repression of further synaptic plasticity. We show that the ability of MOR to oppose LTP is rapidly impaired by sustained D1LR activation via a mechanism involving sustained MOR activation.
Collapse
|
183
|
Kappa Opioid Receptors Mediate Heterosynaptic Suppression of Hippocampal Inputs in the Rat Ventral Striatum. J Neurosci 2017. [PMID: 28642282 DOI: 10.1523/jneurosci.0876-17.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Kappa opioid receptors (KORs) are highly enriched within the ventral striatum (VS) and are thought to modulate striatal neurotransmission. This includes presynaptic inhibition of local glutamatergic release from excitatory inputs to the VS. However, it is not known which inputs drive this modulation and what impact they have on the local circuit dynamics within the VS. Individual medium spiny neurons (MSNs) within the VS serve as a site of convergence for glutamatergic inputs arising from the PFC and limbic regions, such as the hippocampus (HP). Recent data suggest that competition can arise between these inputs with robust cortical activation leading to a reduction in ongoing HP-evoked MSN responses. Here, we investigated the contribution of KOR signaling in PFC-driven heterosynaptic suppression of HP inputs onto MSNs using whole-cell patch-clamp recordings in slices from adult rats. Optogenetically evoked HP EPSPs were greatly attenuated after a short latency (50 ms) following burst-like PFC electrical stimulation, and the magnitude of this suppression was partially reversed following blockade of GABAARs (GABA Type A receptors), but not GABABRs (GABA Type B receptors). A similar reduction in suppression was observed in the presence of the KOR antagonist, norBNI. Combined blockade of local GABAARs and KORs resulted in complete blockade of PFC-induced heterosynaptic suppression of less salient HP inputs. These findings highlight a mechanism by which strong, transient PFC activity can take precedence over other excitatory inputs to the VS.SIGNIFICANCE STATEMENT Emerging evidence suggests that kappa opioid receptor (KOR) activation can selectively modulate striatal glutamatergic inputs onto medium spiny neurons (MSNs). In this study, we found that robust cortical stimulation leads to a reduction in ongoing hippocampal-evoked MSNs responses through the combined recruitment of local inhibitory mechanisms and activation of presynaptic KORs in the ventral striatum (VS). These processes are likely to facilitate the efficient transfer of cortical information through the VS during critical decision making by dampening competing information from less salient excitatory inputs. These data provide a novel mechanism through which VS information processing could influence decision making, a function thought to occur primarily in the PFC.
Collapse
|
184
|
Mohammad Ahmadi Soleimani S, Azizi H, Pachenari N, Mirnajafi-Zadeh J, Semnanian S. Enhancement of μ-opioid receptor desensitization by orexin-A in rat locus coeruleus neurons. Neuropeptides 2017; 63:28-36. [PMID: 28385341 DOI: 10.1016/j.npep.2017.03.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/05/2017] [Accepted: 03/22/2017] [Indexed: 11/16/2022]
Abstract
Opioids have always been used in clinical practice for pain management. However, development of tolerance to their effects following long term administration, seriously restricts further clinical use of these drugs. In this regard, μ-opioid receptor (MOR) desensitization, as an initial step in development of opioid tolerance, is of particular significance. Previous studies support the involvement of orexinergic system in development of opioid tolerance. Locus coeruleus (LC) nucleus has been shown to modulate pain and development of tolerance. Opioid receptors (particularly μ) are densely expressed within the LC. Moreover, it receives widespread orexinergic inputs and orexin type 1 receptors (OX1Rs) are also highly expressed in this brain region. In the present study, the effect of orexin-A (OXA) on met-enkephalin (ME)-induced MOR desensitization was investigated in locus coeruleus neurons of male Wistar rats (2-3weeks of age). ME (30μM), as a potent MOR agonist, was applied for 10min and the outward K+ current was recorded using whole cell patch clamp recording. The percentage of decrease in ME-induced K+ current was considered as the degree of MOR desensitization. Results indicated that OXA (100nM) enhances ME-induced MOR desensitization via affecting OX1Rs in rat locus coeruleus neurons and this effect is mediated by a protein kinase C dependent mechanism within the LC. The activity of orexinergic system might potentiate the signaling pathways underlying opioid-induced receptor desensitization.
Collapse
Affiliation(s)
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Narges Pachenari
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeed Semnanian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; School of Cognitive Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.
| |
Collapse
|
185
|
Ugur M, Kaya E, Gozen O, Koylu EO, Kanit L, Keser A, Balkan B. Chronic nicotine-induced changes in gene expression of delta and kappa-opioid receptors and their endogenous ligands in the mesocorticolimbic system of the rat. Synapse 2017; 71. [PMID: 28509375 DOI: 10.1002/syn.21985] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/22/2022]
Abstract
Delta and kappa opioid receptors (DOR and KOR, respectively) and their endogenous ligands, proenkephalin (PENK) and prodynorphin (PDYN)-derived opioid peptides are proposed as important mediators of nicotine reward. This study investigated the regulatory effect of chronic nicotine treatment on the gene expression of DOR, KOR, PENK and PDYN in the mesocorticolimbic system. Three groups of rats were injected subcutaneously with nicotine at doses of 0.2, 0.4, or 0.6 mg/kg/day for 6 days. Rats were decapitated 1 hr after the last dose on day six, as this timing coincides with increased dopamine release in the mesocorticolimbic system. mRNA levels in the ventral tegmental area (VTA), lateral hypothalamic area (LHA), amygdala (AMG), dorsal striatum (DST), nucleus accumbens, and medial prefrontal cortex were measured by quantitative real-time PCR. Our results showed that nicotine upregulated DOR mRNA in the VTA at all of the doses employed, in the AMG at the 0.4 and 0.6 mg/kg doses, and in the DST at the 0.4 mg/kg dose. Conversely, PDYN mRNA was reduced in the LHA with 0.6 mg/kg nicotine and in the AMG with 0.4 mg/kg nicotine. KOR mRNA was also decreased in the DST with 0.6 mg/kg nicotine. Nicotine did not regulate PENK mRNA in any brain region studied.
Collapse
Affiliation(s)
- Muzeyyen Ugur
- Department of Physiology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Egemen Kaya
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Oguz Gozen
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Ersin O Koylu
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Lutfiye Kanit
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Aysegul Keser
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Burcu Balkan
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| |
Collapse
|
186
|
Hawes SL, Salinas AG, Lovinger DM, Blackwell KT. Long-term plasticity of corticostriatal synapses is modulated by pathway-specific co-release of opioids through κ-opioid receptors. J Physiol 2017; 595:5637-5652. [PMID: 28449351 DOI: 10.1113/jp274190] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/24/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Both endogenous opioids and opiate drugs of abuse modulate learning of habitual and goal-directed actions, and can also modify long-term plasticity of corticostriatal synapses. Striatal projection neurons of the direct pathway co-release the opioid neuropeptide dynorphin which can inhibit dopamine release via κ-opioid receptors. Theta-burst stimulation of corticostriatal fibres produces long-term potentiation (LTP) in striatal projection neurons when measured using whole-cell patch recording. Optogenetic activation of direct pathway striatal projection neurons inhibits LTP while reducing dopamine release. Because the endogenous release of opioids is activity dependent, this modulation of synaptic plasticity represents a negative feedback mechanism that may limit runaway enhancement of striatal neuron activity in response to drugs of abuse. ABSTRACT Synaptic plasticity in the striatum adjusts behaviour adaptively during skill learning, or maladaptively in the case of addiction. Just as dopamine plays a critical role in synaptic plasticity underlying normal skill learning and addiction, endogenous and exogenous opiates also modulate learning and addiction-related striatal plasticity. Though the role of opioid receptors in long-term depression in striatum has been characterized, their effect on long-term potentiation (LTP) remains unknown. In particular, direct pathway (dopamine D1 receptor-containing; D1R-) spiny projection neurons (SPNs) co-release the opioid neuropeptide dynorphin, which acts at presynaptic κ-opioid receptors (KORs) on dopaminergic afferents and can negatively regulate dopamine release. Therefore, we evaluated the interaction of co-released dynorphin and KOR on striatal LTP. We optogenetically facilitate the release of endogenous dynorphin from D1R-SPNs in brain slice while using whole-cell patch recording to measure changes in the synaptic response of SPNs following theta-burst stimulation (TBS) of cortical afferents. Our results demonstrate that TBS evokes corticostriatal LTP, and that optogenetic activation of D1R-SPNs during induction impairs LTP. Additional experiments demonstrate that optogenetic activation of D1R-SPNs reduces stimulation-evoked dopamine release and that bath application of a KOR antagonist provides full rescue of both LTP induction and dopamine release during optogenetic activation of D1R-SPNs. These results suggest that an increase in the opioid neuropeptide dynorphin is responsible for reduced TBS LTP and illustrate a physiological phenomenon whereby heightened D1R-SPN activity can regulate corticostriatal plasticity. Our findings have important implications for learning in addictive states marked by elevated direct pathway activation.
Collapse
Affiliation(s)
- Sarah L Hawes
- George Mason University, Krasnow Institute for Advanced Study, Fairfax, VA, 22030-4444, USA
| | - Armando G Salinas
- George Mason University, Krasnow Institute for Advanced Study, Fairfax, VA, 22030-4444, USA.,Laboratory for Integrative Neuroscience, Section on Synaptic Pharmacology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20852, USA
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, Section on Synaptic Pharmacology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20852, USA
| | - Kim T Blackwell
- George Mason University, Krasnow Institute for Advanced Study, Fairfax, VA, 22030-4444, USA
| |
Collapse
|
187
|
Zhang HY, Gao M, Shen H, Bi GH, Yang HJ, Liu QR, Wu J, Gardner EL, Bonci A, Xi ZX. Expression of functional cannabinoid CB 2 receptor in VTA dopamine neurons in rats. Addict Biol 2017; 22:752-765. [PMID: 26833913 DOI: 10.1111/adb.12367] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/23/2015] [Accepted: 12/11/2015] [Indexed: 12/23/2022]
Abstract
We have recently reported the expression of functional cannabinoid CB2 receptors (CB2 Rs) in midbrain dopamine (DA) neurons in mice. However, little is known whether CB2 Rs are similarly expressed in rat brain because significant species differences in CB2 R structures and expression are found. In situ hybridization and immunohistochemical assays detected CB2 gene and receptors in DA neurons of the ventral tegmental area (VTA), which was up-regulated in cocaine self-administration rats. Electrophysiological studies demonstrated that activation of CB2 Rs by JWH133 inhibited VTA DA neuronal firing in single dissociated neurons. Systemic administration of JWH133 failed to alter, while local administration of JWH133 into the nucleus accumbens inhibited cocaine-enhanced extracellular DA and i.v. cocaine self-administration. This effect was blocked by AM630, a selective CB2 R antagonist. These data suggest that CB2 Rs are expressed in VTA DA neurons and functionally modulate DA neuronal activities and cocaine self-administration behavior in rats.
Collapse
Affiliation(s)
- Hai-Ying Zhang
- Neuropsychopharmacology Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse; Intramural Research Program; Baltimore MD 21224 USA
- Synaptic Plasticity Section; National Institute on Drug Abuse, Intramural Research Program; Baltimore MD 21224 USA
| | - Ming Gao
- Divisions of Neurology and Neurobiology; Barrow Neurological Institute, St. Joseph's Hospital and Medical Center; Phoenix AZ 85013 USA
| | - Hui Shen
- Synaptic Plasticity Section; National Institute on Drug Abuse, Intramural Research Program; Baltimore MD 21224 USA
| | - Guo-Hua Bi
- Neuropsychopharmacology Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse; Intramural Research Program; Baltimore MD 21224 USA
| | - Hong-Ju Yang
- Neuropsychopharmacology Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse; Intramural Research Program; Baltimore MD 21224 USA
| | - Qing-Rong Liu
- Neuropsychopharmacology Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse; Intramural Research Program; Baltimore MD 21224 USA
| | - Jie Wu
- Divisions of Neurology and Neurobiology; Barrow Neurological Institute, St. Joseph's Hospital and Medical Center; Phoenix AZ 85013 USA
| | - Eliot L. Gardner
- Neuropsychopharmacology Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse; Intramural Research Program; Baltimore MD 21224 USA
| | - Antonello Bonci
- Synaptic Plasticity Section; National Institute on Drug Abuse, Intramural Research Program; Baltimore MD 21224 USA
- Solomon H. Snyder Neuroscience Institute; Johns Hopkins University School of Medicine; Baltimore MD 21205 USA
- Department of Psychiatry; Johns Hopkins University School of Medicine; Baltimore MD 21205 USA
| | - Zheng-Xiong Xi
- Neuropsychopharmacology Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse; Intramural Research Program; Baltimore MD 21224 USA
| |
Collapse
|
188
|
Affiliation(s)
- Finja Thiermann
- Faculty of Life Sciences Clinical Pharmacy & Diagnostics Vienna Austria
| | - Gerhard Buchbauer
- Faculty of Life Sciences Clinical Pharmacy & Diagnostics Vienna Austria
| |
Collapse
|
189
|
Sharafshah A, Fazel H, Albonaim A, Omarmeli V, Rezaei S, Mirzajani E, Ajamian F, Keshavarz P. Association of OPRD1 Gene Variants with Opioid Dependence in Addicted Male Individuals Undergoing Methadone Treatment in the North of Iran. J Psychoactive Drugs 2017. [DOI: 10.1080/02791072.2017.1290303] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Alireza Sharafshah
- Master’s Student, Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Master’s Student, Genetic Laboratory, Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Hedyeh Fazel
- Master’s Student, Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Albonaim
- Master’s Student, Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Master’s Student, Genetic Laboratory, Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Vahid Omarmeli
- Master’s Student, Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Master’s Student, Genetic Laboratory, Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Sajjad Rezaei
- Assistant Professor, Department of Psychology, University of Guilan, Rasht, Iran
| | - Ebrahim Mirzajani
- Assistant Professor, Department of Biochemistry and Biophysics, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Farzam Ajamian
- Assistant Professor in Molecular Genetics and Engineering, Department of Biology, Faculty of Sciences (FA), University of Guilan, Rasht, Iran
| | - Parvaneh Keshavarz
- Associate Professor, Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
190
|
Samir S, Yllanes AP, Lallemand P, Brewer KL, Clemens S. Morphine responsiveness to thermal pain stimuli is aging-associated and mediated by dopamine D1 and D3 receptor interactions. Neuroscience 2017; 349:87-97. [PMID: 28257894 DOI: 10.1016/j.neuroscience.2017.02.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 01/16/2017] [Accepted: 02/20/2017] [Indexed: 01/09/2023]
Abstract
Morphine actions involve the dopamine (DA) D1 and D3 receptor systems (D1R and D3R), and the responses to morphine change with age. We here explored in differently aged wild-type (WT) and D3R knockout mice (D3KO) the interactions of the D1R/D3R systems with morphine in vivo at three different times of the animals' lifespan (2months, 1year, and 2years). We found that: (1) thermal pain withdrawal reflexes follow an aging-associated phenotype, with relatively longer latencies at 2months and shorter latencies at 1year, (2) over the same age range, a dysfunction of the D3R subtype decreases reflex latencies more than aging alone, (3) morphine altered reflex responses in a dose-dependent manner in WT animals and changed at its higher dose the phenotype of the D3KO animals from a morphine-resistant state to a morphine-responsive state, (4) block of D1R function had an aging-dependent effect on thermal withdrawal latencies in control animals that, in old animals, was stronger than that of low-dose morphine. Lastly, (5) block of D1R function in young D3KO animals mimicked the behavioral phenotype observed in the aged WT. Our proof-of-concept data from the rodent animal model suggest that, with age, block of D1R function may be considered as an alternative to the use of morphine, to modulate the response to painful stimuli.
Collapse
Affiliation(s)
- Sophia Samir
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Alexander P Yllanes
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Perrine Lallemand
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Kori L Brewer
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Stefan Clemens
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States.
| |
Collapse
|
191
|
Abstract
Obesity is a global epidemic that contributes to a number of health complications including cardiovascular disease, type 2 diabetes, cancer and neuropsychiatric disorders. Pharmacotherapeutic strategies to treat obesity are urgently needed. Research over the past two decades has increased substantially our knowledge of central and peripheral mechanisms underlying homeostatic energy balance. Homeostatic mechanisms involve multiple components including neuronal circuits, some originating in hypothalamus and brain stem, as well as peripherally-derived satiety, hunger and adiposity signals that modulate neural activity and regulate eating behavior. Dysregulation of one or more of these homeostatic components results in obesity. Coincident with obesity, reward mechanisms that regulate hedonic aspects of food intake override the homeostatic regulation of eating. In addition to functional interactions between homeostatic and reward systems in the regulation of food intake, homeostatic signals have the ability to alter vulnerability to drug abuse. Regarding the treatment of obesity, pharmacological monotherapies primarily focus on a single protein target. FDA-approved monotherapy options include phentermine (Adipex-P®), orlistat (Xenical®), lorcaserin (Belviq®) and liraglutide (Saxenda®). However, monotherapies have limited efficacy, in part due to the recruitment of alternate and counter-regulatory pathways. Consequently, a multi-target approach may provide greater benefit. Recently, two combination products have been approved by the FDA to treat obesity, including phentermine/topiramate (Qsymia®) and naltrexone/bupropion (Contrave®). The current review provides an overview of homeostatic and reward mechanisms that regulate energy balance, potential therapeutic targets for obesity and current treatment options, including some candidate therapeutics in clinical development. Finally, challenges in anti-obesity drug development are discussed.
Collapse
Affiliation(s)
- Vidya Narayanaswami
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, 40536, USA
| | - Linda P Dwoskin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
192
|
Lichtenberg NT, Wassum KM. Amygdala mu-opioid receptors mediate the motivating influence of cue-triggered reward expectations. Eur J Neurosci 2017; 45:381-387. [PMID: 27862489 PMCID: PMC5293612 DOI: 10.1111/ejn.13477] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/04/2016] [Accepted: 11/07/2016] [Indexed: 01/16/2023]
Abstract
Environmental reward-predictive stimuli can retrieve from memory a specific reward expectation that allows them to motivate action and guide choice. This process requires the basolateral amygdala (BLA), but little is known about the signaling systems necessary within this structure. Here we examined the role of the neuromodulatory opioid receptor system in the BLA in such cue-directed action using the outcome-specific Pavlovian-to-instrumental transfer (PIT) test in rats. Inactivation of BLA mu-, but not delta-opioid receptors was found to dose-dependently attenuate the ability of a reward-predictive cue to selectively invigorate the performance of actions directed at the same unique predicted reward (i.e. to express outcome-specific PIT). BLA mu-opioid receptor inactivation did not affect the ability of a reward itself to similarly motivate action (outcome-specific reinstatement), suggesting a more selective role for the BLA mu-opioid receptor in the motivating influence of currently unobservable rewarding events. These data reveal a new role for BLA mu-opioid receptor activation in the cued recall of precise reward memories and the use of this information to motivate specific action plans.
Collapse
Affiliation(s)
- Nina T Lichtenberg
- Department of Psychology, UCLA, 1285 Franz Hall, Box 951563, Los Angeles, CA, 90095, USA
| | - Kate M Wassum
- Department of Psychology, UCLA, 1285 Franz Hall, Box 951563, Los Angeles, CA, 90095, USA
- Brain Research Institute, UCLA, Los Angeles, CA, USA
| |
Collapse
|
193
|
Enkephalin and neuropeptide-Y interaction in the intergeniculate leaflet network, a part of the mammalian biological clock. Neuroscience 2017; 343:10-20. [DOI: 10.1016/j.neuroscience.2016.11.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/22/2016] [Accepted: 11/22/2016] [Indexed: 11/20/2022]
|
194
|
Hill EM, Hunt L, Duryea DG. Evolved Vulnerability to Addiction: The Problem of Opiates. EVOLUTIONARY PSYCHOLOGY 2017. [DOI: 10.1007/978-3-319-60576-0_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
195
|
Neuromodulatory effects of the dorsal hippocampal endocannabinoid system in dextromethorphan/morphine-induced amnesia. Eur J Pharmacol 2017; 794:100-105. [DOI: 10.1016/j.ejphar.2016.11.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 10/29/2016] [Accepted: 11/17/2016] [Indexed: 01/15/2023]
|
196
|
Mazid S, Hall BS, Odell SC, Stafford K, Dyer AD, Van Kempen TA, Selegean J, McEwen BS, Waters EM, Milner TA. Sex differences in subcellular distribution of delta opioid receptors in the rat hippocampus in response to acute and chronic stress. Neurobiol Stress 2016; 5:37-53. [PMID: 27981195 PMCID: PMC5145913 DOI: 10.1016/j.ynstr.2016.11.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 10/18/2016] [Accepted: 11/07/2016] [Indexed: 02/06/2023] Open
Abstract
Drug addiction requires associative learning processes that critically involve hippocampal circuits, including the opioid system. We recently found that acute and chronic stress, important regulators of addictive processes, affect hippocampal opioid levels and mu opioid receptor trafficking in a sexually dimorphic manner. Here, we examined whether acute and chronic stress similarly alters the levels and trafficking of hippocampal delta opioid receptors (DORs). Immediately after acute immobilization stress (AIS) or one-day after chronic immobilization stress (CIS), the brains of adult female and male rats were perfusion-fixed with aldehydes. The CA3b region and the dentate hilus of the dorsal hippocampus were quantitatively analyzed by light microscopy using DOR immunoperoxidase or dual label electron microscopy for DOR using silver intensified immunogold particles (SIG) and GABA using immunoperoxidase. At baseline, females compared to males had more DORs near the plasmalemma of pyramidal cell dendrites and about 3 times more DOR-labeled CA3 dendritic spines contacted by mossy fibers. In AIS females, near-plasmalemmal DOR-SIGs decreased in GABAergic hilar dendrites. However, in AIS males, near-plasmalemmal DOR-SIGs increased in CA3 pyramidal cell and hilar GABAergic dendrites and the percentage of CA3 dendritic spines contacted by mossy fibers increased to about half that seen in unstressed females. Conversely, after CIS, near-plasmalemmal DOR-SIGs increased in hilar GABA-labeled dendrites of females whereas in males plasmalemmal DOR-SIGs decreased in CA3 pyramidal cell dendrites and near-plasmalemmal DOR-SIGs decreased hilar GABA-labeled dendrites. As CIS in females, but not males, redistributed DOR-SIGs near the plasmalemmal of hilar GABAergic dendrites, a subsequent experiment examined the acute affect of oxycodone on the redistribution of DOR-SIGs in a separate cohort of CIS females. Plasmalemmal DOR-SIGs were significantly elevated on hilar interneuron dendrites one-hour after oxycodone (3 mg/kg, I.P.) administration compared to saline administration in CIS females. These data indicate that DORs redistribute within CA3 pyramidal cells and dentate hilar GABAergic interneurons in a sexually dimorphic manner that would promote activation and drug related learning in males after AIS and in females after CIS. Females have more near-plasmalemmal DORs in pyramidal CA3 dendrites than males. Acute stress in males relocates DORs in CA3 & GABA dendrites to promote activation. Chronic stress in females relocates DORs in GABA dendrites in females to promote activation. Chronic stress in males relocates DORs in GABA dendrites opposite of females. DOR-stress relocation may contribute to sexually dimorphic effects on drug related learning.
Collapse
Affiliation(s)
- Sanoara Mazid
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States
| | - Baila S Hall
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States
| | - Shannon C Odell
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States
| | - Khalifa Stafford
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States
| | - Andreina D Dyer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States
| | - Tracey A Van Kempen
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States
| | - Jane Selegean
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Elizabeth M Waters
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| |
Collapse
|
197
|
Xie K, Colgan LA, Dao MT, Muntean BS, Sutton LP, Orlandi C, Boye SL, Boye SE, Shih CC, Li Y, Xu B, Smith RG, Yasuda R, Martemyanov KA. NF1 Is a Direct G Protein Effector Essential for Opioid Signaling to Ras in the Striatum. Curr Biol 2016; 26:2992-3003. [PMID: 27773571 DOI: 10.1016/j.cub.2016.09.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 08/24/2016] [Accepted: 09/07/2016] [Indexed: 01/19/2023]
Abstract
It is well recognized that G-protein-coupled receptors (GPCRs) can activate Ras-regulated kinase pathways to produce lasting changes in neuronal function. Mechanisms by which GPCRs transduce these signals and their relevance to brain disorders are not well understood. Here, we identify a major Ras regulator, neurofibromin 1 (NF1), as a direct effector of GPCR signaling via Gβγ subunits in the striatum. We find that binding of Gβγ to NF1 inhibits its ability to inactivate Ras. Deletion of NF1 in striatal neurons prevents the opioid-receptor-induced activation of Ras and eliminates its coupling to Akt-mTOR-signaling pathway. By acting in the striatal medium spiny neurons of the direct pathway, NF1 regulates opioid-induced changes in Ras activity, thereby sensitizing mice to psychomotor and rewarding effects of morphine. These results delineate a novel mechanism of GPCR signaling to Ras pathways and establish a critical role of NF1 in opioid addiction.
Collapse
Affiliation(s)
- Keqiang Xie
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Lesley A Colgan
- Max Planck Florida Institute for Neuroscience, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Maria T Dao
- Department of Metabolism and Aging, The Scripps Research Institute, 120 Scripps Way, Jupiter, FL 33458, USA
| | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Laurie P Sutton
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Sanford L Boye
- Department of Ophthalmology, University of Florida, 1395 Center Drive, Gainesville, FL 32610, USA
| | - Shannon E Boye
- Department of Ophthalmology, University of Florida, 1395 Center Drive, Gainesville, FL 32610, USA
| | - Chien-Cheng Shih
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Yuqing Li
- Department of Neurology, University of Florida, 1600 SW Archer Road, Gainesville, FL 32610, USA
| | - Baoji Xu
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Roy G Smith
- Department of Metabolism and Aging, The Scripps Research Institute, 120 Scripps Way, Jupiter, FL 33458, USA
| | - Ryohei Yasuda
- Max Planck Florida Institute for Neuroscience, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA.
| |
Collapse
|
198
|
Role of Estradiol in the Regulation of Prolactin Secretion During Late Pregnancy. Neurochem Res 2016; 41:3344-3355. [DOI: 10.1007/s11064-016-2067-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 09/06/2016] [Accepted: 09/15/2016] [Indexed: 12/18/2022]
|
199
|
Abstract
The release of the neurotransmitter norepinephrine throughout the mammalian brain is important for modulating attention, arousal, and cognition during many behaviors. Furthermore, disruption of norepinephrine-mediated signaling is strongly associated with several psychiatric and neurodegenerative disorders in humans, emphasizing the clinical importance of this system. Most of the norepinephrine released in the brain is supplied by a very small, bilateral nucleus in the brainstem called the locus coeruleus. The goal of this minireview is to emphasize the complexity of the locus coeruleus beyond its primary definition as a norepinephrine-producing nucleus. Several recent studies utilizing innovative technologies highlight how the locus coeruleus-norepinephrine system can now be targeted with increased accuracy and resolution, in order to better understand its role in modulating diverse behaviors.
Collapse
Affiliation(s)
- Lindsay A Schwarz
- Howard Hughes Medical Institute, Department of Biology, Stanford University, Stanford, CA 94305, USA.
| | - Liqun Luo
- Howard Hughes Medical Institute, Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
200
|
Abstract
The posterior pituitary gland secretes oxytocin and vasopressin (the antidiuretic hormone) into the blood system. Oxytocin is required for normal delivery of the young and for delivery of milk to the young during lactation. Vasopressin increases water reabsorption in the kidney to maintain body fluid balance and causes vasoconstriction to increase blood pressure. Oxytocin and vasopressin secretion occurs from the axon terminals of magnocellular neurons whose cell bodies are principally found in the hypothalamic supraoptic nucleus and paraventricular nucleus. The physiological functions of oxytocin and vasopressin depend on their secretion, which is principally determined by the pattern of action potentials initiated at the cell bodies. Appropriate secretion of oxytocin and vasopressin to meet the challenges of changing physiological conditions relies mainly on integration of afferent information on reproductive, osmotic, and cardiovascular status with local regulation of magnocellular neurons by glia as well as intrinsic regulation by the magnocellular neurons themselves. This review focuses on the control of magnocellular neuron activity with a particular emphasis on their regulation by reproductive function, body fluid balance, and cardiovascular status. © 2016 American Physiological Society. Compr Physiol 6:1701-1741, 2016.
Collapse
Affiliation(s)
- Colin H Brown
- Brain Health Research Centre, Centre for Neuroendocrinology and Department of Physiology, University of Otago, Dunedin, New Zealand
| |
Collapse
|