1
|
Liu W, Zhong W, Giusti-Rodríguez P, Jiang Z, Wang GW, Sun H, Hu M, Li Y. SnapHiC-G: identifying long-range enhancer-promoter interactions from single-cell Hi-C data via a global background model. Brief Bioinform 2024; 25:bbae426. [PMID: 39222061 PMCID: PMC11367764 DOI: 10.1093/bib/bbae426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 07/05/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Harnessing the power of single-cell genomics technologies, single-cell Hi-C (scHi-C) and its derived technologies provide powerful tools to measure spatial proximity between regulatory elements and their target genes in individual cells. Using a global background model, we propose SnapHiC-G, a computational method, to identify long-range enhancer-promoter interactions from scHi-C data. We applied SnapHiC-G to scHi-C datasets generated from mouse embryonic stem cells and human brain cortical cells. SnapHiC-G achieved high sensitivity in identifying long-range enhancer-promoter interactions. Moreover, SnapHiC-G can identify putative target genes for noncoding genome-wide association study (GWAS) variants, and the genetic heritability of neuropsychiatric diseases is enriched for single-nucleotide polymorphisms (SNPs) within SnapHiC-G-identified interactions in a cell-type-specific manner. In sum, SnapHiC-G is a powerful tool for characterizing cell-type-specific enhancer-promoter interactions from complex tissues and can facilitate the discovery of chromatin interactions important for gene regulation in biologically relevant cell types.
Collapse
Affiliation(s)
- Weifang Liu
- Department of Biostatistics, University of North Carolina at Chapel Hill, 135 Dauer Drive, Chapel Hill, NC 27599, United States
| | - Wujuan Zhong
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., 126 East Lincoln Ave, Rahway, New Jersey 07065, United States
| | - Paola Giusti-Rodríguez
- Department of Psychiatry, University of Florida, 1149 Newel Dr., Gainesville, FL 32611, United States
| | - Zhiyun Jiang
- Department of Genetics, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC 27599, United States
| | - Geoffery W Wang
- Department of Biostatistics, University of North Carolina at Chapel Hill, 135 Dauer Drive, Chapel Hill, NC 27599, United States
| | - Huaigu Sun
- Department of Genetics, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC 27599, United States
| | - Ming Hu
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44196, United States
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, 135 Dauer Drive, Chapel Hill, NC 27599, United States
- Department of Genetics, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC 27599, United States
- Department of Computer Science, University of North Carolina at Chapel Hill, 201 S. Columbia St, Chapel Hill, NC 27599, United States
| |
Collapse
|
2
|
Weiss N, Zamponi GW. Opioid Receptor Regulation of Neuronal Voltage-Gated Calcium Channels. Cell Mol Neurobiol 2021; 41:839-847. [PMID: 32514826 PMCID: PMC11448596 DOI: 10.1007/s10571-020-00894-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/29/2020] [Indexed: 12/28/2022]
Abstract
Neuronal voltage-gated calcium channels play a pivotal role in the conversion of electrical signals into calcium entry into nerve endings that is required for the release of neurotransmitters. They are under the control of a number of cellular signaling pathways that serve to fine tune synaptic activities, including G-protein coupled receptors (GPCRs) and the opioid system. Besides modulating channel activity via activation of second messengers, GPCRs also physically associate with calcium channels to regulate their function and expression at the plasma membrane. In this mini review, we discuss the mechanisms by which calcium channels are regulated by classical opioid and nociceptin receptors. We highlight the importance of this regulation in the control of neuronal functions and their implication in the development of disease conditions. Finally, we present recent literature concerning the use of novel μ-opioid receptor/nociceptin receptor modulators and discuss their use as potential drug candidates for the treatment of pain.
Collapse
Affiliation(s)
- Norbert Weiss
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| |
Collapse
|
3
|
Parker KE, Sugiarto E, Taylor AMW, Pradhan AA, Al-Hasani R. Pain, Motivation, Migraine, and the Microbiome: New Frontiers for Opioid Systems and Disease. Mol Pharmacol 2020; 98:433-444. [PMID: 32958571 PMCID: PMC7562975 DOI: 10.1124/mol.120.119438] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022] Open
Abstract
For decades the broad role of opioids in addiction, neuropsychiatric disorders, and pain states has been somewhat well established. However, in recent years, with the rise of technological advances, not only is the existing dogma being challenged, but we are identifying new disease areas in which opioids play a critical role. This review highlights four new areas of exploration in the opioid field. The most recent addition to the opioid family, the nociceptin receptor system, shows promise as the missing link in understanding the neurocircuitry of motivation. It is well known that activation of the kappa opioid receptor system modulates negative affect and dysphoria, but recent studies now implicate the kappa opioid system in the modulation of negative affect associated with pain. Opioids are critical in pain management; however, the often-forgotten delta opioid receptor system has been identified as a novel therapeutic target for headache disorders and migraine. Lastly, changes to the gut microbiome have been shown to directly contribute to many of the symptoms of chronic opioid use and opioid related behaviors. This review summarizes the findings from each of these areas with an emphasis on identifying new therapeutic targets. SIGNIFICANCE STATEMENT: The focus of this minireview is to highlight new disease areas or new aspects of disease in which opioids have been implicated; this includes pain, motivation, migraine, and the microbiome. In some cases, this has resulted in the pursuit of a novel therapeutic target and resultant clinical trial. We believe this is very timely and will be a refreshing take on reading about opioids and disease.
Collapse
Affiliation(s)
- Kyle E Parker
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Elizabeth Sugiarto
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Anna M W Taylor
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Amynah A Pradhan
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Ream Al-Hasani
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| |
Collapse
|
4
|
Bellia F, Fernández MS, Fabio MC, Pucci M, Pautassi RM, D'Addario C. Selective alterations in endogenous opioid system genes expression in rats selected for high ethanol intake during adolescence. Drug Alcohol Depend 2020; 212:108025. [PMID: 32442753 DOI: 10.1016/j.drugalcdep.2020.108025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/31/2020] [Accepted: 04/10/2020] [Indexed: 02/07/2023]
Abstract
Historically, the roots of alcoholism have been linked to either environment or heredity. However, the interaction between these factors is still largely unexplored. The evidence supports a link between alcohol consumption and the endogenous opioid system. We here studied the opioid genes expression in male and female Wistar rats derived from a short-term breeding program which selected -- at adolescence -- for high (ADHI line) or low (ADLO line) ethanol drinking. Specifically, in this work we analyzed central opioid gene expression in the rats of the second filial generation (S2-ADLO and S2-ADHI). Selective downregulation of pronociceptin (Pnoc) and its receptor (Oprl1) mRNA levels were observed in the prefrontal cortex of male S2-ADHI rats when compared to S2-ADLO, and for Oprl1 also in the nucleus accumbens. An increase in gene expression was instead observed for pro-opiomelanocortin (Pomc) in the nucleus accumbens of S2-ADHI males when compared to S2-ADLO, as well as for mu opioid receptor (Oprm1) but in females. The differences in mRNA levels may be due to the different alcohol consumption between the two groups of rats or may represent pre-existing differences between them. Moreover, we show a sex-specific modulation of the expression of these genes, thus pointing out the importance of sex on ethanol responses. The results might lead to more specific and effective pharmacological treatments for alcoholism.
Collapse
Affiliation(s)
| | - Macarena Soledad Fernández
- Instituto de Investigación Médica M. y M. Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Carolina Fabio
- Instituto de Investigación Médica M. y M. Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina; Facultad de Psicología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | - Ricardo Marcos Pautassi
- Instituto de Investigación Médica M. y M. Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina; Facultad de Psicología, Universidad Nacional de Córdoba, Córdoba, Argentina.
| | - Claudio D'Addario
- Università degli Studi di Teramo, Teramo, Italy; Karolinska Institutet, Department of Clinical Neuroscience, Stockholm, Sweden.
| |
Collapse
|
5
|
Zhang Y, Luan D, Liu Y, Li H, Dong J, Zhang X, Yuan L, Zhong Z, Jiang L, Li X, Ye M, Tong J. Helicid Reverses Lipopolysaccharide-Induced Inflammation and Promotes GDNF Levels in C6 Glioma Cells through Modulation of Prepronociceptin. Chem Biodivers 2020; 17:e2000063. [PMID: 32329965 DOI: 10.1002/cbdv.202000063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/24/2020] [Indexed: 12/18/2022]
Abstract
Helicid suppresses inflammatory factors and protects nerve cells in the hippocampus of rats with depression, but the mechanisms underlying its protective effects are unclear at present. In this investigation, we conducted gene silencing, Helicid intervention and rescue experiments to explore the protective actions of PNOC, the prepronociceptin gene known to regulate inflammatory processes, and Helicid on a C6 cell model of inflammation induced by LPS. Collective data from Western blots, ELISA, immunofluorescence and flow cytometry experiments showed that PNOC silencing or administration of Helicid led to reduced inflammatory factor levels, oxidative stress and expression of glial fibrillary acidic protein (GFAP), along with increased glial cell lines-derived neurotrophic factor (GDNF) expression. Furthermore, expression of p-Akt in the Akt signaling pathway was increased. Interestingly, overexpression of PNOC in the Helicid treatment group partially reversed the Helicid-induced changes in the above biochemical indexes. Our collective results provide strong evidence of Helicid-mediated regulation of the Akt signaling pathway through PNOC to improve cell inflammation and oxidative stress.
Collapse
Affiliation(s)
| | - Di Luan
- Department of Neurology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, P. R. China
| | - Yanhao Liu
- Wannan Medical College, Wuhu, 241000, P. R. China
| | - Hongjin Li
- Wannan Medical College, Wuhu, 241000, P. R. China
| | - Jian Dong
- Wannan Medical College, Wuhu, 241000, P. R. China
| | | | - Lili Yuan
- Department of Neurology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, P. R. China
| | - Zhengling Zhong
- Department of Clinical Pharmacy, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, P. R. China
| | - Lan Jiang
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, P. R. China
| | - Xuyi Li
- China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Mingquan Ye
- Wannan Medical College, Wuhu, 241000, P. R. China
| | - Jiucui Tong
- Wannan Medical College, Wuhu, 241000, P. R. China.,Department of Clinical Pharmacy, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, P. R. China
| |
Collapse
|
6
|
Piantadosi SC, Pizzagalli DA, Bruchas MR. The Complex Role of Nociceptin Signaling in Stress: Clarity Through Neuroimaging? Biol Psychiatry 2020; 87:489-491. [PMID: 32081252 PMCID: PMC7875195 DOI: 10.1016/j.biopsych.2020.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 01/06/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Sean C Piantadosi
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, Washington; Department of Anesthesiology, University of Washington, Seattle, Washington
| | - Diego A Pizzagalli
- Center for Depression, Anxiety and Stress Research, McLean Hospital, Belmont, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Michael R Bruchas
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, Washington; Department of Anesthesiology, University of Washington, Seattle, Washington.
| |
Collapse
|
7
|
Borruto AM, Fotio Y, Stopponi S, Brunori G, Petrella M, Caputi FF, Romualdi P, Candeletti S, Narendran R, Rorick-Kehn LM, Ubaldi M, Weiss F, Ciccocioppo R. NOP receptor antagonism reduces alcohol drinking in male and female rats through mechanisms involving the central amygdala and ventral tegmental area. Br J Pharmacol 2020; 177:1525-1537. [PMID: 31713848 DOI: 10.1111/bph.14915] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Nociceptin/orphanin FQ (N/OFQ) peptide and its cognate receptor (NOP) are widely expressed in mesolimbic brain regions where they play an important role in modulating reward and motivation. Early evidence suggested that NOP receptor activation attenuates the rewarding effects of drugs of abuse, including alcohol. However, emerging data indicate that NOP receptor blockade also effectively attenuates alcohol drinking and relapse. To advance our understanding of the role of the N/OFQ-NOP receptor system in alcohol abuse, we examined the effect of NOP receptor blockade on voluntary alcohol drinking at the neurocircuitry level. EXPERIMENTAL APPROACH Using male and female genetically selected alcohol-preferring Marchigian Sardinian (msP) rats, we initially evaluated the effects of the selective NOP receptor antagonist LY2817412 (3, 10, and 30 mg·kg-1 , p.o.) on alcohol consumption in a two-bottle free-choice paradigm. We then microinjected LY2817412 (3 and 6 μg·μl-1 per rat) in the central nucleus of the amygdala (CeA), ventral tegmental area (VTA), and nucleus accumbens (NAc). KEY RESULTS Peripheral LY2817412 administration dose-dependently and selectively reduced voluntary alcohol intake in male and female msP rats. Central injections of LY2817412 markedly attenuated voluntary alcohol intake in both sexes following administration in the CeA and VTA but not in the NAc. CONCLUSION AND IMPLICATIONS The present results revealed that the CeA and VTA are neuroanatomical substrates that mediate the effects of NOP receptor antagonism on alcohol consumption. Overall, our findings support the potential of NOP receptor antagonism as a treatment strategy to attenuate alcohol use and addiction.
Collapse
Affiliation(s)
| | - Yannick Fotio
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | - Serena Stopponi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | - Gloria Brunori
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy.,Department of Biomedical Science, Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| | - Michele Petrella
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | - Francesca Felicia Caputi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Rajesh Narendran
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Linda M Rorick-Kehn
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, USA
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | - Friedbert Weiss
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, California, USA
| | | |
Collapse
|
8
|
Pucci M, Micioni Di Bonaventura MV, Wille-Bille A, Fernández MS, Maccarrone M, Pautassi RM, Cifani C, D’Addario C. Environmental stressors and alcoholism development: Focus on molecular targets and their epigenetic regulation. Neurosci Biobehav Rev 2019; 106:165-181. [DOI: 10.1016/j.neubiorev.2018.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/13/2018] [Accepted: 07/09/2018] [Indexed: 01/17/2023]
|
9
|
Parker KE, Pedersen CE, Gomez AM, Spangler SM, Walicki MC, Feng SY, Stewart SL, Otis JM, Al-Hasani R, McCall JG, Sakers K, Bhatti DL, Copits BA, Gereau RW, Jhou T, Kash TJ, Dougherty JD, Stuber GD, Bruchas MR. A Paranigral VTA Nociceptin Circuit that Constrains Motivation for Reward. Cell 2019; 178:653-671.e19. [PMID: 31348890 PMCID: PMC7001890 DOI: 10.1016/j.cell.2019.06.034] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 08/16/2018] [Accepted: 06/25/2019] [Indexed: 12/26/2022]
Abstract
Nociceptin and its receptor are widely distributed throughout the brain in regions associated with reward behavior, yet how and when they act is unknown. Here, we dissected the role of a nociceptin peptide circuit in reward seeking. We generated a prepronociceptin (Pnoc)-Cre mouse line that revealed a unique subpopulation of paranigral ventral tegmental area (pnVTA) neurons enriched in prepronociceptin. Fiber photometry recordings during progressive ratio operant behavior revealed pnVTAPnoc neurons become most active when mice stop seeking natural rewards. Selective pnVTAPnoc neuron ablation, inhibition, and conditional VTA nociceptin receptor (NOPR) deletion increased operant responding, revealing that the pnVTAPnoc nucleus and VTA NOPR signaling are necessary for regulating reward motivation. Additionally, optogenetic and chemogenetic activation of this pnVTAPnoc nucleus caused avoidance and decreased motivation for rewards. These findings provide insight into neuromodulatory circuits that regulate motivated behaviors through identification of a previously unknown neuropeptide-containing pnVTA nucleus that limits motivation for rewards.
Collapse
Affiliation(s)
- Kyle E Parker
- Departments of Anesthesiology, Division of Basic Research, Anatomy and Neurobiology, and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Christian E Pedersen
- Departments of Anesthesiology, Division of Basic Research, Anatomy and Neurobiology, and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Adrian M Gomez
- Departments of Anesthesiology, Division of Basic Research, Anatomy and Neurobiology, and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Skylar M Spangler
- Departments of Anesthesiology, Division of Basic Research, Anatomy and Neurobiology, and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA; Neuroscience Program (DBBS), Washington University School of Medicine, St. Louis, MO, USA
| | - Marie C Walicki
- Departments of Anesthesiology, Division of Basic Research, Anatomy and Neurobiology, and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Shelley Y Feng
- Departments of Anesthesiology, Division of Basic Research, Anatomy and Neurobiology, and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Sarah L Stewart
- Departments of Anesthesiology, Division of Basic Research, Anatomy and Neurobiology, and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - James M Otis
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Ream Al-Hasani
- Departments of Anesthesiology, Division of Basic Research, Anatomy and Neurobiology, and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, USA; Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University School of Medicine, St. Louis, MO, USA; Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Jordan G McCall
- Departments of Anesthesiology, Division of Basic Research, Anatomy and Neurobiology, and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, USA; Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University School of Medicine, St. Louis, MO, USA; Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Kristina Sakers
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Dionnet L Bhatti
- Departments of Anesthesiology, Division of Basic Research, Anatomy and Neurobiology, and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Bryan A Copits
- Departments of Anesthesiology, Division of Basic Research, Anatomy and Neurobiology, and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Robert W Gereau
- Departments of Anesthesiology, Division of Basic Research, Anatomy and Neurobiology, and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Thomas Jhou
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Thomas J Kash
- Department of Pharmacology and Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Garret D Stuber
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA; Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA; Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Michael R Bruchas
- Departments of Anesthesiology, Division of Basic Research, Anatomy and Neurobiology, and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
10
|
Ciccocioppo R, Borruto AM, Domi A, Teshima K, Cannella N, Weiss F. NOP-Related Mechanisms in Substance Use Disorders. Handb Exp Pharmacol 2019; 254:187-212. [PMID: 30968214 PMCID: PMC6641545 DOI: 10.1007/164_2019_209] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
Nociceptin/orphanin FQ (N/OFQ) is a 17 amino acid peptide that was deorphanized in 1995 and has been widely studied since. The role of the N/OFQ system in drug abuse has attracted researchers' attention since its initial discovery. The first two scientific papers describing the effect of intracranial injection of N/OFQ appeared 20 years ago and reported efficacy of the peptide in attenuating alcohol intake, whereas heroin self-administration was insensitive. Since then more than 100 scientific articles investigating the role of the N/OFQ and N/OFQ receptor (NOP) system in drug abuse have been published. The present article provides an historical overview of the advances in the field with focus on three major elements. First, the most robust data supportive of the efficacy of NOP agonists in treating drug abuse come from studies in the field of alcohol research, followed by psychostimulant and opioid research. In contrast, activation of NOP appears to facilitate nicotine consumption. Second, emerging data challenge the assumption that activation of NOP is the most appropriate strategy to attenuate consumption of substances of abuse. This assumption is based first on the observation that animals carrying an overexpression of NOP system components are more prone to consume substances of abuse, whereas NOP knockout rats are less motivated to self-administer heroin, alcohol, and cocaine. Third, administration of NOP antagonists also reduces alcohol consumption. In addition, NOP blockade reduces nicotine self-administration. Hypothetical mechanisms explaining this apparent paradox are discussed. Finally, we focus on the possibility that co-activation of NOP and mu opioid (MOP) receptors is an alternative strategy, readily testable in the clinic, to reduce the consumption of psychostimulants, opiates, and, possibly, alcohol.
Collapse
Affiliation(s)
- Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy.
| | - Anna Maria Borruto
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Ana Domi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Koji Teshima
- Research Unit/Neuroscience, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Osaka, Japan
| | - Nazzareno Cannella
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Friedbert Weiss
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
11
|
Caputi FF, Romualdi P, Candeletti S. Regulation of the Genes Encoding the ppN/OFQ and NOP Receptor. Handb Exp Pharmacol 2019; 254:141-162. [PMID: 30689088 DOI: 10.1007/164_2018_196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Over the years, the ability of N/OFQ-NOP receptor system in modulating several physiological functions, including the release of neurotransmitters, anxiety-like behavior responses, modulation of the reward circuitry, inflammatory signaling, nociception, and motor function, has been examined in several brain regions and at spinal level. This chapter collects information related to the genes encoding the ppN/OFQ and NOP receptor, their regulation, and relative transcriptional control mechanisms. Furthermore, genetic manipulations, polymorphisms, and epigenetic alterations associated with different pathological conditions are discussed. The evidence here collected indicates that the study of ppN/OFQ and NOP receptor gene expression may offer novel opportunities in the field of personalized therapies and highlights this system as a good "druggable target" for different pathological conditions.
Collapse
Affiliation(s)
- Francesca Felicia Caputi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy.
| | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| |
Collapse
|
12
|
Single-Cell RNA-Seq of Mouse Dopaminergic Neurons Informs Candidate Gene Selection for Sporadic Parkinson Disease. Am J Hum Genet 2018; 102:427-446. [PMID: 29499164 DOI: 10.1016/j.ajhg.2018.02.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/01/2018] [Indexed: 12/22/2022] Open
Abstract
Genetic variation modulating risk of sporadic Parkinson disease (PD) has been primarily explored through genome-wide association studies (GWASs). However, like many other common genetic diseases, the impacted genes remain largely unknown. Here, we used single-cell RNA-seq to characterize dopaminergic (DA) neuron populations in the mouse brain at embryonic and early postnatal time points. These data facilitated unbiased identification of DA neuron subpopulations through their unique transcriptional profiles, including a postnatal neuroblast population and substantia nigra (SN) DA neurons. We use these population-specific data to develop a scoring system to prioritize candidate genes in all 49 GWAS intervals implicated in PD risk, including genes with known PD associations and many with extensive supporting literature. As proof of principle, we confirm that the nigrostriatal pathway is compromised in Cplx1-null mice. Ultimately, this systematic approach establishes biologically pertinent candidates and testable hypotheses for sporadic PD, informing a new era of PD genetic research.
Collapse
|
13
|
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
|
14
|
Adem A, Madjid N, Kahl U, Holst S, Sadek B, Sandin J, Terenius L, Ögren SO. Nociceptin and the NOP receptor in aversive learning in mice. Eur Neuropsychopharmacol 2017; 27:1298-1307. [PMID: 29102248 DOI: 10.1016/j.euroneuro.2017.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 09/13/2017] [Accepted: 09/25/2017] [Indexed: 11/27/2022]
Abstract
The endogenous neuropeptide nociceptin (N/OFQ), which mediates its actions via the nociceptin receptor (NOP), is implicated in multiple behavioural and physiological functions. This study examined the effects of the NOP agonists N/OFQ and the synthetic agonist Ro 64-6198, the antagonists NNN and NalBzoH, as well as deletion of the Pronociceptin gene on emotional memory in mice. The animals were tested in the passive avoidance (PA) task, dependent on hippocampal and amygdala functions. N/OFQ injected intraventricularly (i.c.v.) prior to training produced a biphasic effect on PA retention; facilitation at a low dose and impairment at higher doses. Ro 64-6198 also displayed a biphasic effect with memory facilitation at lower doses and impairment at a high dose. None of the agonists influenced PA training latencies. NNN did not significantly modulate retention in the PA task but antagonized the inhibitory effects of N/OFQ. NalBzoH facilitated memory retention in a dose-dependent manner and blocked the impairing effects of N/OFQ. However, neither NNN nor NalBzoH blocked the memory-impairing effects of Ro 64-6198. Finally, the Pnoc knockout mice exhibited enhanced PA retention latencies compared to the wild type mice. The biphasic effect of the natural ligand and Ro 64-6198 and the failure of the antagonists to block the action of Ro 64-6198 indicate complexity in ligand-receptor interaction. These results indicate that brain nociceptin and its NOP has a subtle role in regulation of mechanisms of relevance for treatment of disorders with processing disturbances of aversive events e.g. Alzheimer's disease, anxiety, depression and PTSD.
Collapse
Affiliation(s)
- Abdu Adem
- Department of Neuroscience, Retzius väg 8, S-171 77 Stockholm, Sweden.
| | - Nather Madjid
- Department of Pharmacology & Therapeutics Faculty of Medicine & Health Sciences UAE University, Al Ain, UAE; Department of Neuroscience, Retzius väg 8, S-171 77 Stockholm, Sweden
| | - Ulrika Kahl
- Department of Pharmacology & Therapeutics Faculty of Medicine & Health Sciences UAE University, Al Ain, UAE
| | - Sarah Holst
- Department of Pharmacology & Therapeutics Faculty of Medicine & Health Sciences UAE University, Al Ain, UAE
| | - Bassem Sadek
- Department of Neuroscience, Retzius väg 8, S-171 77 Stockholm, Sweden
| | - Johan Sandin
- AlzeCure Foundation, Karolinska Institutet Science Park, Hälsovägen 7, S-141 57 Huddinge, Stockholm, Sweden
| | - Lars Terenius
- Department of Clinical Neuroscience, Karolinska Institutet, CMM L8:01, Karolinska University Hospital, S-171 76 Stockholm, Sweden
| | - Sven Ove Ögren
- Department of Pharmacology & Therapeutics Faculty of Medicine & Health Sciences UAE University, Al Ain, UAE.
| |
Collapse
|
15
|
Kallupi M, Scuppa G, de Guglielmo G, Calò G, Weiss F, Statnick MA, Rorick-Kehn LM, Ciccocioppo R. Genetic Deletion of the Nociceptin/Orphanin FQ Receptor in the Rat Confers Resilience to the Development of Drug Addiction. Neuropsychopharmacology 2017; 42:695-706. [PMID: 27562376 PMCID: PMC5240182 DOI: 10.1038/npp.2016.171] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 08/12/2016] [Accepted: 08/13/2016] [Indexed: 12/14/2022]
Abstract
The nociceptin (NOP) receptor is a G-protein-coupled receptor whose natural ligand is the NOP/orphanin FQ (N/OFQ) peptide. Evidence from pharmacological studies suggests that the N/OFQ system is implicated in the regulation of several addiction-related phenomena, such as drug intake, withdrawal, and relapse. Here, to further explore the role of NOP system in addiction, we used NOP (-/-) rats to study the motivation for cocaine, heroin, and alcohol self-administration in the absence of N/OFQ function. Conditioned place preference (CPP) and saccharin (0.2% w/v) self-administration were also investigated. Results showed that NOP (-/-) rats self-administer less cocaine (0.25, 0.125, or 0.5 mg/infusion) both under a fixed ratio 1 and a progressive ratio schedule of reinforcement compared with wild-type (Wt) controls. Consistently, cocaine (10 mg/kg, i.p.) was able to induce CPP in Wt but not in NOP (-/-). When NOP (-/-) rats were tested for heroin (20 μg/infusion) and ethanol (10% v/v) self-administration, they showed significantly lower drug intake compared with Wt. Conversely, saccharin self-administration was not affected by NOP deletion, excluding the possibility of nonspecific learning deficits or generalized disruption of reward mechanisms in NOP (-/-) rats. These findings were confirmed with pharmacological experiments using two selective NOP antagonists, SB-612111 and LY2817412. Both drugs attenuated alcohol self-administration in Wt rats but not in NOP (-/-) rats. In conclusion, our results demonstrate that genetic deletion of NOP receptors confers resilience to drug abuse and support a role for NOP receptor antagonism as a potential treatment option for drug addiction.
Collapse
Affiliation(s)
- Marsida Kallupi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy,Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Giulia Scuppa
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Giordano de Guglielmo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy,Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Girolamo Calò
- Department of Medical Science, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - Friedbert Weiss
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA, USA
| | - Michael A Statnick
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN USA
| | | | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy,School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, 62032 Italy, Tel: +39 07 3740 3313, Fax: +39 07 3740 3325, E-mail:
| |
Collapse
|
16
|
Spahn V, Stein C. Targeting delta opioid receptors for pain treatment: drugs in phase I and II clinical development. Expert Opin Investig Drugs 2017; 26:155-160. [PMID: 28001096 DOI: 10.1080/13543784.2017.1275562] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Opioids are widely used to treat severe pain. Most clinically used opioids activate µ-opioid receptors (MOR). Their ligands induce potent analgesia but also adverse effects. The δ-opioid receptor (DOR) is another member of the opioid receptor family that has been under intense investigation with the aim to avoid MOR-induced side effects. Areas covered: This article reviews DOR ligands which appeared to be promising after preclinical evaluation. A literature search using Pubmed, Cochrane library, ClinicalTrials.gov, EudraCT, AdisInsight database and EBSCO Online Library was conducted. Out of numerous newly synthesized molecules, only few candidates entered phase I and/or II clinical investigation. The publicly accessible results are presented here. Expert opinion: Many compounds showed potent DOR-specific pain inhibition in preclinical studies. ADL5859 and ADL5747 entered clinical trials and successfully passed phase I. However, in phase II studies the primary endpoint (pain reduction) was not met and further investigation was terminated. A third compound, NP2, is in phase II clinical evaluation and results are pending. These findings suggest a potential of DOR ligands according to preclinical studies. Further clinical research and secondary analysis of unpublished data is needed to identify molecules which are useful in humans.
Collapse
Affiliation(s)
- Viola Spahn
- a Klinik für Anästhesiologie und operative Intensivmedizin , Charité Campus Benjamin Franklin, Freie Universität Berlin , Berlin , Germany
| | - Christoph Stein
- a Klinik für Anästhesiologie und operative Intensivmedizin , Charité Campus Benjamin Franklin, Freie Universität Berlin , Berlin , Germany.,b Multifunctional Biomaterials for Medicine , Helmholtz Virtual Institute , Teltow , Germany
| |
Collapse
|
17
|
Post A, Smart TS, Jackson K, Mann J, Mohs R, Rorick-Kehn L, Statnick M, Anton R, O'Malley SS, Wong CJ. Proof-of-Concept Study to Assess the Nociceptin Receptor Antagonist LY2940094 as a New Treatment for Alcohol Dependence. Alcohol Clin Exp Res 2016; 40:1935-44. [DOI: 10.1111/acer.13147] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 06/03/2016] [Indexed: 12/13/2022]
Affiliation(s)
| | | | | | | | | | | | | | - Raymond Anton
- Medical University of South Carolina; Charleston South Carolina
| | | | | |
Collapse
|
18
|
A key role for the N/OFQ-NOP receptor system in modulating nicotine taking in a model of nicotine and alcohol co-administration. Sci Rep 2016; 6:26594. [PMID: 27199205 PMCID: PMC4873733 DOI: 10.1038/srep26594] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/05/2016] [Indexed: 11/08/2022] Open
Abstract
Alcohol and nicotine are often co-abused. Although the N/OFQ-NOP receptor system is considered a potential target for development of drug abuse pharmacotherapies, especially for alcoholism, little is known about the role of this system in nicotine dependence. Furthermore, the effect of prior history of nicotine dependence on subsequent nicotine and alcohol taking is understudied. Using an operant co-administration paradigm, in which rats concurrently self-administer nicotine and alcohol, we found that nicotine dependent rats increased nicotine self-administration over time as compared to non-dependent animals, while patterns of alcohol lever pressing did not change between groups. Pretreatment with the potent NOP receptor agonist AT-202 (0.3–3 mg/kg) increased nicotine lever pressing of both dependent and non-dependent groups, whereas the selective antagonist SB612111 (1–10 mg/kg) elicited a clear reduction of nicotine responses, in both dependent and non-dependent rats. In parallel, AT-202 only produced minor changes on alcohol responses and SB612111 reduced alcohol taking at a dose that also reduced locomotor behavior. Results indicate that a history of nicotine dependence affects subsequent nicotine- but not alcohol-maintained responding, and that NOP receptor antagonism, rather than agonism, blocks nicotine self-administration, which strongly suggests a critical role for the endogenous N/OFQ in the modulation of nicotine reinforcement processes.
Collapse
|
19
|
Abstract
UNLABELLED The nociceptin/orphanin FQ (NOP) receptor, the fourth member of the opioid receptor family, is involved in many processes common to the opioid receptors including pain and drug abuse. To better characterize receptor location and trafficking, knock-in mice were created by inserting the gene encoding enhanced green fluorescent protein (eGFP) into the NOP receptor gene (Oprl1) and producing mice expressing a functional NOP-eGFP C-terminal fusion in place of the native NOP receptor. The NOP-eGFP receptor was present in brain of homozygous knock-in animals in concentrations somewhat higher than in wild-type mice and was functional when tested for stimulation of [(35)S]GTPγS binding in vitro and in patch-clamp electrophysiology in dorsal root ganglia (DRG) neurons and hippocampal slices. Inhibition of morphine analgesia was equivalent when tested in knock-in and wild-type mice. Imaging revealed detailed neuroanatomy in brain, spinal cord, and DRG and was generally consistent with in vitro autoradiographic imaging of receptor location. Multicolor immunohistochemistry identified cells coexpressing various spinal cord and DRG cellular markers, as well as coexpression with μ-opioid receptors in DRG and brain regions. Both in tissue slices and primary cultures, the NOP-eGFP receptors appear throughout the cell body and in processes. These knock-in mice have NOP receptors that function both in vitro and in vivo and appear to be an exceptional tool to study receptor neuroanatomy and correlate with NOP receptor function. SIGNIFICANCE STATEMENT The NOP receptor, the fourth member of the opioid receptor family, is involved in pain, drug abuse, and a number of other CNS processes. The regional and cellular distribution has been difficult to determine due to lack of validated antibodies for immunohistochemical analysis. To provide a new tool for the investigation of receptor localization, we have produced knock-in mice with a fluorescent-tagged NOP receptor in place of the native NOP receptor. These knock-in mice have NOP receptors that function both in vitro and in vivo and have provided a detailed characterization of NOP receptors in brain, spinal cord, and DRG neurons. They appear to be an exceptional tool to study receptor neuroanatomy and correlate with NOP receptor function.
Collapse
|
20
|
Zamponi GW, Striessnig J, Koschak A, Dolphin AC. The Physiology, Pathology, and Pharmacology of Voltage-Gated Calcium Channels and Their Future Therapeutic Potential. Pharmacol Rev 2015; 67:821-70. [PMID: 26362469 PMCID: PMC4630564 DOI: 10.1124/pr.114.009654] [Citation(s) in RCA: 790] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Voltage-gated calcium channels are required for many key functions in the body. In this review, the different subtypes of voltage-gated calcium channels are described and their physiologic roles and pharmacology are outlined. We describe the current uses of drugs interacting with the different calcium channel subtypes and subunits, as well as specific areas in which there is strong potential for future drug development. Current therapeutic agents include drugs targeting L-type Ca(V)1.2 calcium channels, particularly 1,4-dihydropyridines, which are widely used in the treatment of hypertension. T-type (Ca(V)3) channels are a target of ethosuximide, widely used in absence epilepsy. The auxiliary subunit α2δ-1 is the therapeutic target of the gabapentinoid drugs, which are of value in certain epilepsies and chronic neuropathic pain. The limited use of intrathecal ziconotide, a peptide blocker of N-type (Ca(V)2.2) calcium channels, as a treatment of intractable pain, gives an indication that these channels represent excellent drug targets for various pain conditions. We describe how selectivity for different subtypes of calcium channels (e.g., Ca(V)1.2 and Ca(V)1.3 L-type channels) may be achieved in the future by exploiting differences between channel isoforms in terms of sequence and biophysical properties, variation in splicing in different target tissues, and differences in the properties of the target tissues themselves in terms of membrane potential or firing frequency. Thus, use-dependent blockers of the different isoforms could selectively block calcium channels in particular pathologies, such as nociceptive neurons in pain states or in epileptic brain circuits. Of important future potential are selective Ca(V)1.3 blockers for neuropsychiatric diseases, neuroprotection in Parkinson's disease, and resistant hypertension. In addition, selective or nonselective T-type channel blockers are considered potential therapeutic targets in epilepsy, pain, obesity, sleep, and anxiety. Use-dependent N-type calcium channel blockers are likely to be of therapeutic use in chronic pain conditions. Thus, more selective calcium channel blockers hold promise for therapeutic intervention.
Collapse
Affiliation(s)
- Gerald W Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Joerg Striessnig
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Alexandra Koschak
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Annette C Dolphin
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| |
Collapse
|
21
|
Abstract
In the past decades, a large number of neuropeptides with unknown functions have been identified in the brain. Among the newly discovered peptides, nociceptin or orphanin-FQ (N/OFQ) peptide has attracted considerable attention because of its sequence homology with the opioid peptide family. N/OFQ and its cognate receptor (NOP receptor) are distributed widely in the mammalian central nervous system, though particularly intense expression is found in corticolimbic structures. Such distinctive pattern of expression suggests a key role of N/OFQ system in higher brain functions, such as cognition and emotion. In this chapter, we will outline the findings supporting the role played by N/OFQ and NOP receptors in learning and memory and discuss the underlying mechanisms.
Collapse
|
22
|
Ciccocioppo R, Stopponi S, Economidou D, Kuriyama M, Kinoshita H, Heilig M, Roberto M, Weiss F, Teshima K. Chronic treatment with novel brain-penetrating selective NOP receptor agonist MT-7716 reduces alcohol drinking and seeking in the rat. Neuropsychopharmacology 2014; 39:2601-10. [PMID: 24863033 PMCID: PMC4207340 DOI: 10.1038/npp.2014.113] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 03/31/2014] [Accepted: 04/22/2014] [Indexed: 11/09/2022]
Abstract
Since its discovery, the nociceptin/orphanin FQ (N/OFQ)-NOP receptor system has been extensively investigated as a promising target to treat alcoholism. Encouraging results obtained with the endogenous ligand N/OFQ stimulated research towards the development of novel brain-penetrating NOP receptor agonists with a pharmacological and toxicological profile compatible with clinical development. Here we describe the biochemical and alcohol-related behavioral effects of the novel NOP receptor agonist MT-7716. MT-7716 has high affinity for human NOP receptors expressed in HEK293 cells with a Ki value of 0.21 nM. MT-7716 concentration-dependently stimulated GTPγ(35)S binding with an EC50 value of 0.30 nM and its efficacy was similar to N/OFQ, suggesting that MT7716 is a full agonist at NOP receptors. In the two bottle choice test MT-7716 (0, 0.3, 1, and 3 mg/kg, bid) given orally for 14 days dose-dependently decreased voluntary alcohol intake in Marchigian Sardinian rats. The effect became gradually stronger following repeated administration, and was still significant 1 week after discontinuation of the drug. Oral naltrexone (30 mg/kg, bid) for 14 days also reduced ethanol intake; however, the effect decreased over the treatment period and rapidly disappeared when drug treatment was discontinued. MT-7716 is also effective for preventing reinstatement caused by both ethanol-associated environmental stimuli and stress. Finally, to investigate the effect of MT-7716 on alcohol withdrawal symptoms, Wistar rats were withdrawn from a 7-day alcohol liquid diet. MT-7716 significantly attenuated somatic alcohol withdrawal symptoms. Together these findings indicate that MT-7716 is a promising candidate for alcoholism treatment remaining effective with chronic administration.
Collapse
Affiliation(s)
- Roberto Ciccocioppo
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Serena Stopponi
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Daina Economidou
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Makoto Kuriyama
- Department II (CNS), Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Hiroshi Kinoshita
- Department II (CNS), Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Markus Heilig
- Laboratory of Clinical and Translational Studies, National Institute of Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Friedbert Weiss
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Koji Teshima
- Department II (CNS), Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| |
Collapse
|
23
|
Borgquist A, Rivas VM, Kachani M, Sinchak K, Wagner EJ. Gonadal steroids differentially modulate the actions of orphanin FQ/nociceptin at a physiologically relevant circuit controlling female sexual receptivity. J Neuroendocrinol 2014; 26:329-40. [PMID: 24617903 PMCID: PMC4167875 DOI: 10.1111/jne.12148] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 02/25/2014] [Accepted: 03/06/2014] [Indexed: 11/28/2022]
Abstract
Orphanin FQ/nociceptin (OFQ/N) inhibits the activity of pro-opiomelanocortin (POMC) neurones located in the hypothalamic arcuate nucleus (ARH) that regulate female sexual behaviour and energy balance. We tested the hypothesis that gonadal steroids differentially modulate the ability of OFQ/N to inhibit these cells via presynaptic inhibition of transmitter release and postsynaptic activation of G protein-gated, inwardly-rectifying K(+) (GIRK)-1 channels. Whole-cell patch clamp recordings were performed in hypothalamic slices prepared from ovariectomised rats. OFQ/N (1 μm) decreased the frequency of miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs), and also caused a robust outward current in the presence of tetrodotoxin, in ARH neurones from vehicle-treated animals. A priming dose of oestradiol benzoate (EB; 2 μg) increased basal mEPSC frequency, markedly diminished both the OFQ/N-induced decrease in mEPSC frequency and the activation of GIRK-1 currents, and potentiated the OFQ/N-induced decrease in mIPSC frequency. Steroid treatment regimens that facilitate sexual receptivity reinstate the basal mEPSC frequency, the OFQ/N-induced decrease in mEPSC frequency and the activation of GIRK-1 currents to levels observed in vehicle-treated controls, and largely abolish the ability of OFQ/N to decrease mIPSC frequency. These effects were observed in an appreciable population of identified POMC neurones, almost one-half of which projected to the medial preoptic nucleus. Taken together, these data reveal that gonadal steroids influence the pleiotropic actions of OFQ/N on ARH neurones, including POMC neurones, in a disparate manner. These temporal changes in OFQ/N responsiveness further implicate this neuropeptide system as a critical mediator of the gonadal steroid regulation of reproductive behaviour.
Collapse
Affiliation(s)
- Amanda Borgquist
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766
| | - Virginia Mela Rivas
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766
- Department of Physiology, Complutense University, Madrid, Spain
| | - Malika Kachani
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766
| | - Kevin Sinchak
- Department of Biological Sciences, California State University, Long Beach, Long Beach, CA 90840
| | - Edward J. Wagner
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766
| |
Collapse
|
24
|
Kallupi M, Varodayan FP, Oleata CS, Correia D, Luu G, Roberto M. Nociceptin/orphanin FQ decreases glutamate transmission and blocks ethanol-induced effects in the central amygdala of naive and ethanol-dependent rats. Neuropsychopharmacology 2014; 39:1081-92. [PMID: 24169802 PMCID: PMC3957102 DOI: 10.1038/npp.2013.308] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 10/08/2013] [Accepted: 10/24/2013] [Indexed: 12/11/2022]
Abstract
The central nucleus of the amygdala (CeA) mediates several addiction-related processes and nociceptin/orphanin FQ (nociceptin) regulates ethanol intake and anxiety-like behaviors. Glutamatergic synapses, in the CeA and throughout the brain, are very sensitive to ethanol and contribute to alcohol reinforcement, tolerance, and dependence. Previously, we reported that in the rat CeA, acute and chronic ethanol exposures significantly decrease glutamate transmission by both pre- and postsynaptic actions. In this study, using electrophysiological techniques in an in vitro CeA slice preparation, we investigated the effects of nociceptin on glutamatergic transmission and its interaction with acute ethanol in naive and ethanol-dependent rats. We found that nociceptin (100-1000 nM) diminished basal-evoked compound glutamatergic receptor-mediated excitatory postsynaptic potentials (EPSPs) and spontaneous and miniature EPSCs (s/mEPSCs) by mainly decreasing glutamate release in the CeA of naive rats. Notably, nociceptin blocked the inhibition induced by acute ethanol (44 mM) and ethanol blocked the nociceptin-induced inhibition of evoked EPSPs in CeA neurons of naive rats. In neurons from chronic ethanol-treated (ethanol-dependent) rats, the nociceptin-induced inhibition of evoked EPSP amplitude was not significantly different from that in naive rats. Application of [Nphe1]Nociceptin(1-13)NH2, a nociceptin receptor (NOP) antagonist, revealed tonic inhibitory activity of NOP on evoked CeA glutamatergic transmission only in ethanol-dependent rats. The antagonist also blocked nociceptin-induced decreases in glutamatergic responses, but did not affect ethanol-induced decreases in evoked EPSP amplitude. Taken together, these studies implicate a potential role for the nociceptin system in regulating glutamatergic transmission and a complex interaction with ethanol at CeA glutamatergic synapses.
Collapse
Affiliation(s)
- Marsida Kallupi
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Florence P Varodayan
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Christopher S Oleata
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Diego Correia
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- Department of Pharmacology, Universidade Federal do Paraná, Jardim das Américas, Curitiba, Paraná, Brazil
| | - George Luu
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
25
|
Sobczak M, Mokrowiecka A, Cygankiewicz AI, Zakrzewski PK, Sałaga M, Storr M, Kordek R, Małecka-Panas E, Krajewska WM, Fichna J. Anti-inflammatory and antinociceptive action of an orally available nociceptin receptor agonist SCH 221510 in a mouse model of inflammatory bowel diseases. J Pharmacol Exp Ther 2014; 348:401-9. [PMID: 24345466 DOI: 10.1124/jpet.113.209825] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The nociceptin receptors (NOPs) are expressed in the gastrointestinal (GI) tract on muscle cell membranes and neurons, as well as the immune cells that infiltrate the mucosa. The involvement of NOPs in the pathophysiology of GI inflammation has been suggested, but due to the lack of selective NOP agonists, it never fully elucidated. Our aim was to characterize the anti-inflammatory and antinociceptive effect of the NOP agonist, SCH 221510 [3-endo-8-[bis(2-methylphenyl)methyl]-3-phenyl-8-azabicyclo [3.2.1]octan-3-ol], as a potential therapeutic strategy in the treatment of inflammatory bowel diseases (IBD). The anti-inflammatory action of SCH 221510 was determined after intraperitoneal, oral, and intracolonic administration of SCH 221510 (0.1-3.0 mg/kg once or twice daily) in mice treated with 2,4,6-trinitrobenzenesulfonic acid (TNBS). Antinociceptive action of SCH 221510 was evaluated in the mouse model of mustard oil (MO)-induced abdominal pain. Relative NOP mRNA expression was assessed in patients with IBD using real-time reverse transcriptase-polymerase chain reaction. We found that the expression of NOP mRNA was significantly decreased in patients with IBD. The administration (0.1 and 1.0 mg/kg i.p. twice daily and 3 mg/kg p.o. twice daily) of SCH 221510 attenuated TNBS colitis in mice. This effect was blocked by a selective NOP antagonist [J-113397 [(±)-1-[(3R*,4R*)-1-(cyclooctylmethyl)-3-(hydroxymethyl)-4-piperidinyl]-3-ethyl-1,3-dihydro-2H-benzimidazol-2-one]]. The intracolonic injections of SCH 221510 did not improve colitis in mice. The antinociceptive effect of SCH 221510 was observed after oral administration of SCH 221510 in MO-induced pain tests in mice with acute colitis. In conclusion, our results show a potent anti-inflammatory and antinociceptive effect upon selective activation of NOP receptors and suggest that the NOP agonist SCH 221510 is a promising drug candidate for future treatment of IBD.
Collapse
MESH Headings
- Abdominal Pain/chemically induced
- Abdominal Pain/drug therapy
- Administration, Oral
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Azabicyclo Compounds/pharmacology
- Azabicyclo Compounds/therapeutic use
- Case-Control Studies
- Colitis, Ulcerative/chemically induced
- Colitis, Ulcerative/drug therapy
- Colitis, Ulcerative/pathology
- Colon/metabolism
- Colon/pathology
- Female
- Humans
- Inflammatory Bowel Diseases/chemically induced
- Inflammatory Bowel Diseases/drug therapy
- Inflammatory Bowel Diseases/pathology
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Male
- Mice
- Mice, Inbred BALB C
- Middle Aged
- Mustard Plant
- Plant Oils
- Receptors, Opioid/agonists
- Receptors, Opioid/metabolism
- Trinitrobenzenesulfonic Acid
- Young Adult
- Nociceptin Receptor
Collapse
Affiliation(s)
- Marta Sobczak
- Department of Biomolecular Chemistry (M.So., M.Sa., J.F.), Department of Digestive Tract Diseases (A.M., E.M.-P.), and Department of Pathology (R.K.), Faculty of Medicine, Medical University of Lodz, Lodz, Poland; Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland (A.I.C., P.K.Z., W.M.K.); and Department of Medicine, Division of Gastroenterology, Ludwig Maximilians University of Munich, Munich, Germany (M.St.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Witkin JM, Statnick MA, Rorick-Kehn LM, Pintar JE, Ansonoff M, Chen Y, Tucker RC, Ciccocioppo R. The biology of Nociceptin/Orphanin FQ (N/OFQ) related to obesity, stress, anxiety, mood, and drug dependence. Pharmacol Ther 2014; 141:283-99. [PMID: 24189487 PMCID: PMC5098338 DOI: 10.1016/j.pharmthera.2013.10.011] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 10/10/2013] [Indexed: 01/16/2023]
Abstract
Nociceptin/Orphanin FQ (N/OFQ) is a 17 amino acid peptide that was deorphanized in 1995. The generation of specific agonists, antagonists and receptor deficient mice and rats has enabled progress in elucidating the biological functions of N/OFQ. Additionally, radio-imaging technologies have been advanced for investigation of this system in animals and humans. Together with traditional neurobehavioral techniques, these tools have been utilized to identify the biological significance of the N/OFQ system and its interacting partners. The present review focuses on the role of N/OFQ in the regulation of feeding, body weight homeostasis, stress, the stress-related psychiatric disorders of depression and anxiety, and in drug and alcohol dependence. Critical evaluation of the current scientific preclinical literature suggests that small molecule modulators of nociceptin opioid peptide receptors (NOP) might be useful in the treatment of diseases related to these biological functions. In particular, the literature data suggest that antagonism of NOP receptors will produce anti-obesity and antidepressant activities in humans. However, there are also contradictory data discussed. The current literature on the role of N/OFQ in anxiety and addiction, on the other hand points primarily to a role of agonist modulation being potentially therapeutic. Some drug-like molecules that function either as agonists or antagonists of NOP receptors have been optimized for human clinical study to test some of these hypotheses. The discovery of PET ligands for NOP receptors, combined with the pharmacological tools and burgeoning preclinical data set discussed here bodes well for a rapid advancement of clinical understanding and potential therapeutic benefit.
Collapse
Key Words
- (1S,3aS)-8- (2,3,3a,4,5,6-hexahydro-1H-phenalen-1-yl)-1-phenyl-1,3,8-triaza-spiro[4.5]decan-4-one, a NOP receptor agonist
- (±)trans-1-[1-cyclooctylmethyl-3-hydroxymethyl-4-piperidyl]-3-ethyl-1,3-dihydro-2H-benzimidazol-2-one, a NOP receptor antagonist
- 2-{3-[1-((1R)-acenaphthen-1-yl)piperidin-4-yl]-2,3-dihydro-2-oxo-benzimidazol-1-yl}-N-methylacetamide, a NOP receptor agonist
- 5-HT
- 5-hydroxytryptamine or serotonin
- 8-[bis(2-methylphenyl)-methyl]-3-phenyl-8-azabicyclo[3.2.1]octan-3-ol
- ACTH
- Alcohol-preferring rats
- Anxiety
- BED
- BNST
- CGRP
- CPP
- CRF
- CTA
- Calcitonin gene related peptide
- CeA
- DA
- Depression
- Drug dependence
- EPSC
- FST
- G-protein activated, inwardly rectifying K(+) channel
- G-protein-coupled receptor
- GIRK
- GPCR
- HPA
- J-113397
- JTC-801
- KO
- MDD
- Marchigian Sardinian Alcohol-Preferring
- N-(4-amino-2-methylquinolin-6-yl)-2-(4-ethylphenoxymethyl)benzamide hydrochloride, a NOP receptor antagonist
- N/OFQ
- NAcc
- NE
- NOP
- NPY
- Nociceptin opioid peptide or Nociceptin opioid peptide receptor
- Nociceptin/Orphanin FQ
- Nociceptin/Orphanin FQ (F: phenylalanine, Q: glutamine, the amino acids that begin and end the peptide sequence)
- ORL
- Obesity
- P rats
- POMC
- Pro-opiomelanocortin
- Ro 64-6198
- SB-612111
- SCH 221510
- SCH 655842
- Stress
- TST
- UFP-101
- VTA
- W212393
- [(–)-cis-1-methyl-7-[[4-(2,6-dichlorophenyl)piperidin-1-yl]methyl]-6,7,8,9-tetrahydro-5H-benzocyclohepten-5-ol, a NOP receptor antagonist
- [Nphe(1),Arg(14),Lys(15)]N/OFQ-NH(2), a NOP receptor antagonist
- adrenocorticotropic hormone
- bed nucleus of stria terminalis
- binge eating disorder
- central nucleus of the amygdala
- conditioned place preference
- conditioned taste aversion
- corticotrophin-releasing factor
- dopamine
- endo-8-[bis(2-chlorophenyl)methyl]-3-phenyl-8-azabicyclo[3.2.1]octane-3-carboxamide, a NOP receptor agonist
- excitatory post-synaptic current
- forced-swim test
- hypothalamic–pituitary axis
- knockout
- mPFC
- major depressive disorder
- medial prefrontal cortex
- msP
- neuropeptide Y
- norepinephrine
- nucleus accumbens
- opioid-receptor-like
- tail-suspension test
- ventral tegmental area
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA.
| | | | | | - John E Pintar
- University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Michael Ansonoff
- University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Yanyun Chen
- Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA
| | - R Craig Tucker
- Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA
| | | |
Collapse
|
27
|
Bourinet E, Altier C, Hildebrand ME, Trang T, Salter MW, Zamponi GW. Calcium-permeable ion channels in pain signaling. Physiol Rev 2014; 94:81-140. [PMID: 24382884 DOI: 10.1152/physrev.00023.2013] [Citation(s) in RCA: 240] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The detection and processing of painful stimuli in afferent sensory neurons is critically dependent on a wide range of different types of voltage- and ligand-gated ion channels, including sodium, calcium, and TRP channels, to name a few. The functions of these channels include the detection of mechanical and chemical insults, the generation of action potentials and regulation of neuronal firing patterns, the initiation of neurotransmitter release at dorsal horn synapses, and the ensuing activation of spinal cord neurons that project to pain centers in the brain. Long-term changes in ion channel expression and function are thought to contribute to chronic pain states. Many of the channels involved in the afferent pain pathway are permeable to calcium ions, suggesting a role in cell signaling beyond the mere generation of electrical activity. In this article, we provide a broad overview of different calcium-permeable ion channels in the afferent pain pathway and their role in pain pathophysiology.
Collapse
|
28
|
Miranda-Morales RS, Nizhnikov ME, Waters DH, Spear NE. Participation of the nociceptin/orphanin FQ receptor in ethanol-mediated locomotor activation and ethanol intake in preweanling rats. Behav Brain Res 2013; 245:137-44. [PMID: 23439216 PMCID: PMC3666860 DOI: 10.1016/j.bbr.2013.02.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 02/12/2013] [Accepted: 02/14/2013] [Indexed: 12/16/2022]
Abstract
Activation of nociceptin/orphanin FQ (NOP) receptors seems to attenuate ethanol-induced reinforcement in adult rodents. Since early ethanol exposure results in later increased responsiveness to ethanol, it is important to analyze NOP receptor modulation of ethanol-related behaviors during early ontogeny. By measuring NOP involvement in ethanol intake and ethanol-induced locomotor activation, we analyzed the specific participation of NOP receptors on these ethanol-related behaviors in two-week-old rats. In each experiment animals were pre-treated with the endogenous ligand for this receptor (nociceptin/orphanin FQ at 0.0, 0.5, 1.0 or 2.0 μg) or a selective NOP antagonist (J-113397 at 0.0, 0.5, 2.0 or 5.0 mg/kg). Results indicated that activation of the nociceptin receptor system had no effect on ethanol or water intake, while blockade of the NOP receptor has an unspecific effect on consummatory behavior: J-113397 increased ethanol (at a dose of 0.5 mg/kg) and water intake (at 0.5 and 5.0 mg/kg). Ethanol-mediated locomotor stimulation was attenuated by activation of the NOP system (nociceptin at 1.0 and 2.0 μg). Nociceptin had no effect on basal locomotor activity. Blockade of NOP receptors did not modify ethanol-induced locomotor activation. Contrary to what has been reported for adult rodents, nociceptin failed to suppress intake of ethanol in infants. Attenuation of ethanol-induced stimulation by activation of NOP receptor system suggests an early role of this receptor in this ethanol-related behavior.
Collapse
Affiliation(s)
- Roberto Sebastián Miranda-Morales
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY 13902-6000, USA.
| | | | | | | |
Collapse
|
29
|
Tariq S, Nurulain SM, Tekes K, Adeghate E. Deciphering intracellular localization and physiological role of nociceptin and nocistatin. Peptides 2013; 43:174-83. [PMID: 23454174 DOI: 10.1016/j.peptides.2013.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 02/18/2013] [Accepted: 02/18/2013] [Indexed: 01/16/2023]
Abstract
Nociceptin and nocistatin are endogenous ligands of G protein coupled receptor family. Numerous techniques have been used to study the diverse parameters including, localization, distribution and ultrastructure of these peptides. The majority of the study parameters are based on their physiological roles in different organ systems. The present study presents an overview of the different methods used for the study of nociceptin, nocistatin and their receptors. Nociceptin has been implicated in many physiological functions including, nociception, locomotion, stressed-induced analgesia, learning and memory, neurotransmitter and hormone release, renal function, neuronal differentiation, sexual and reproductive behavior, uterine contraction, feeding, anxiety, gastrointestinal motility, cardiovascular function, micturition, cough, hypoxic-ischemic brain injury, diuresis and sodium balance, temperature regulation, vestibular function, and mucosal transport. It has been noted that the use of light and electron microscopy was less frequent, though it may be one of the most promising tools to study the intracellular localization of these neuropeptides. In addition, more studies on the level of circulating nociceptin and nocistatin are also necessary for investigating their clinical roles in health and disease. A variety of modern tools including physiological, light and electron microscopy (EM) are needed to decipher the extent of intracellular localization, tissue distribution and function of these peptides. The intracellular localization of nociceptin and nocistatin will require a high resolution transmission EM capable of identifying these peptides and other supporting molecules that co-localize with them. A tracing technique could also elucidate a possible migratory ability of nociceptin and nocistatin from one cellular compartment to the other.
Collapse
Affiliation(s)
- Saeed Tariq
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | | | | | | |
Collapse
|
30
|
León-Olea M, Miller-Pérez C, Sánchez-Islas E, Mendoza-Sotelo J, Garduño-Gutiérrez R, de Gortari P, Amaya MI. The nociceptin/orphanin FQ-like opioid peptide in nervous periesophageal ganglia of land snail Helix aspersa. Brain Res 2013; 1505:22-46. [PMID: 23419890 DOI: 10.1016/j.brainres.2013.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Revised: 02/01/2013] [Accepted: 02/02/2013] [Indexed: 01/10/2023]
Abstract
The neuropeptide nociceptin/orphanin FQ (N/OFQ) and its receptor are members of the endogenous opioid peptide family. In mammals N/OFQ modulates a variety of biological functions such as nociception, food intake, endocrine, control of neurotransmitter release, among others. In the molluscs Cepea nemoralis and Helix aspersa the administration of N/OFQ produces a thermopronociceptive effect. However, little is known about its existence and anatomic distribution in invertebrates. The aim of this study was to provide a detailed anatomical distribution of N/OFQ like peptide immunoreactivity (N/OFQ-IL), to quantify the tissue content of this peptide, as well as to demostrate molecular evidence of N/OFQ mRNA in the nervous tissue of periesophageal ganglia of the land snail H. aspersa. Immunohistochemical, immunocytochemical, radioimmunoanalysis (RIA) and reverse transcription-polymerase chain reaction (RT-PCR) techniques were used. With regard to RT-PCR, the primers to detect expression of mRNA transcripts from H. aspersa were derived from the rat N/OFQ opioid peptide. We show a wide distribution of N/OFQ-IL in neurons and fibers in all perioesophageal ganglia, fibers of the neuropile, nerves, periganglionar connective tissue, aortic wall and neurohemal sinuses. The total amount of N/OFQ-IL in the perioesophageal ganglia (7.75 ± 1.75 pmol/g of tissue) quantified by RIA was similar to that found in mouse hypothalamus (10.1 ± 1.6 pmol/g of tissue). In this study, we present molecular evidence of N/OFQ mRNA expression. Some N/OFQ-IL neurons have been identified as neuroendocrine or involved in olfaction, hydro-electrolyte regulation, feeding, and thermonociception. Therefore, we suggest that N/OFQ may participate in these snail functions.
Collapse
Affiliation(s)
- Martha León-Olea
- Departamento de Neuromorfología Funcional, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz. Av. México Xochimilco 101, Col. San Lorenzo Huipulco, México D.F., C.P. 14370, México.
| | | | | | | | | | | | | |
Collapse
|
31
|
Oddi D, Crusio WE, D'Amato FR, Pietropaolo S. Monogenic mouse models of social dysfunction: implications for autism. Behav Brain Res 2013; 251:75-84. [PMID: 23327738 DOI: 10.1016/j.bbr.2013.01.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Revised: 12/14/2012] [Accepted: 01/05/2013] [Indexed: 12/21/2022]
Abstract
Autism is a pervasive disorder characterized by a complex symptomatology, based principally on social dysfunction. The disorder has a highly complex, largely genetic etiology, involving an impressive variety of genes, the precise contributions of which still remain to be determined. For this reason, a reductionist approach to the study of autism has been proposed, employing monogenic animal models of social dysfunction, either by targeting a candidate gene, or by mimicking a single-gene disorder characterized by autistic symptoms. In the present review, we discuss this monogenic approach by comparing examples of each strategy: the mu opioid receptor knock-out (KO) mouse line, which targets the opioid system (known to be involved in the control of social behaviors); and the Fmr1-KO mouse, a model for Fragile X syndrome (a neurodevelopmental syndrome that includes autistic symptoms). The autistic-relevant behavioral phenotypes of the mu-opioid and Fmr1-KO mouse lines are described here, summarizing previous work by our research group and others, but also providing novel experimental evidence. Relevant factors influencing the validity of the two models, such as sex differences and age at testing, are also addressed, permitting an extensive evaluation of the advantages and limits of monogenic mouse models for autism.
Collapse
Affiliation(s)
- D Oddi
- CNR, Cell Biology and Neurobiology Institute, Rome, Italy; IRCCS, Santa Lucia Foundation, Rome, Italy
| | | | | | | |
Collapse
|
32
|
Basic mechanisms of migraine and its acute treatment. Pharmacol Ther 2012; 136:319-33. [DOI: 10.1016/j.pharmthera.2012.08.011] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 08/13/2012] [Indexed: 12/27/2022]
|
33
|
Karakus E, Halici Z, Albayrak A, Bayir Y, Aydin A, Unal D, Cadirci E, Ferah I, Odaci E. Beneficial Pharmacological Effects of Levosimendan on Antioxidant Status of Acute Inflammation Induced in Paw of Rat: Involvement in Inflammatory Mediators. Basic Clin Pharmacol Toxicol 2012; 112:156-63. [DOI: 10.1111/bcpt.12004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 08/21/2012] [Indexed: 12/20/2022]
Affiliation(s)
- Emre Karakus
- Department of Pharmacology and Toxicology; Ataturk University School of Veterinary; Erzurum; Turkey
| | - Zekai Halici
- Department of Pharmacology; Ataturk University School of Medicine; Erzurum; Turkey
| | - Abdulmecit Albayrak
- Department of Pharmacology; Ataturk University School of Medicine; Erzurum; Turkey
| | - Yasin Bayir
- Department of Biochemistry; Ataturk University School of Pharmacy; Erzurum; Turkey
| | - Ali Aydin
- Department of Histology and Embryology; Ataturk University School of Pharmacy; Erzurum; Turkey
| | - Deniz Unal
- Department of Orthopedic and Traumatology; Ataturk University School of Pharmacy; Erzurum; Turkey
| | - Elif Cadirci
- Department of Pharmacology; Ataturk University School of Pharmacy; Erzurum; Turkey
| | - Irmak Ferah
- Department of Pharmacology; Ataturk University School of Medicine; Erzurum; Turkey
| | - Ersan Odaci
- Department of Histology and Embryology; Karadeniz Technical University School of Medicine; Trabzon; Turkey
| |
Collapse
|
34
|
Schank JR, Ryabinin AE, Giardino WJ, Ciccocioppo R, Heilig M. Stress-related neuropeptides and addictive behaviors: beyond the usual suspects. Neuron 2012; 76:192-208. [PMID: 23040815 PMCID: PMC3495179 DOI: 10.1016/j.neuron.2012.09.026] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Addictive disorders are chronic, relapsing conditions that cause extensive disease burden. Genetic factors partly account for susceptibility to addiction, but environmental factors such as stressful experiences and prolonged exposure of the brain to addictive drugs promote its development. Progression to addiction involves neuroadaptations within neurocircuitry that mediates stress responses and is influenced by several peptidergic neuromodulators. While corticotrophin releasing factor is the prototypic member of this class, recent work has identified several additional stress-related neuropeptides that play an important role in regulation of drug intake and relapse, including the urocortins, nociceptin, substance P, and neuropeptide S. Here, we review this emerging literature, discussing to what extent the properties of these neuromodulators are shared or distinct and considering their potential as drug targets.
Collapse
Affiliation(s)
- Jesse R. Schank
- Laboratory of Clinical and Translational Studies, National Inst. on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Andrey E. Ryabinin
- Dept. of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239-3098
| | - William J. Giardino
- Dept. of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239-3098
| | - Roberto Ciccocioppo
- Dept. of Experimental Medicine and Public Health, Camerino University, Italy
| | - Markus Heilig
- Laboratory of Clinical and Translational Studies, National Inst. on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
35
|
Reiss D, Prinssen EP, Wichmann J, Kieffer BL, Ouagazzal AM. The nociceptin orphanin FQ peptide receptor agonist, Ro64-6198, impairs recognition memory formation through interaction with glutamatergic but not cholinergic receptor antagonists. Neurobiol Learn Mem 2012; 98:254-60. [DOI: 10.1016/j.nlm.2012.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 08/14/2012] [Accepted: 09/03/2012] [Indexed: 11/29/2022]
|
36
|
Feng Y, He X, Yang Y, Chao D, Lazarus LH, Xia Y. Current research on opioid receptor function. Curr Drug Targets 2012; 13:230-46. [PMID: 22204322 DOI: 10.2174/138945012799201612] [Citation(s) in RCA: 211] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 08/09/2011] [Accepted: 08/12/2011] [Indexed: 12/11/2022]
Abstract
The use of opioid analgesics has a long history in clinical settings, although the comprehensive action of opioid receptors is still less understood. Nonetheless, recent studies have generated fresh insights into opioid receptor-mediated functions and their underlying mechanisms. Three major opioid receptors (μ-opioid receptor, MOR; δ-opioid receptor, DOR; and κ-opioid receptor, KOR) have been cloned in many species. Each opioid receptor is functionally sub-classified into several pharmacological subtypes, although, specific gene corresponding each of these receptor subtypes is still unidentified as only a single gene has been isolated for each opioid receptor. In addition to pain modulation and addiction, opioid receptors are widely involved in various physiological and pathophysiological activities, including the regulation of membrane ionic homeostasis, cell proliferation, emotional response, epileptic seizures, immune function, feeding, obesity, respiratory and cardiovascular control as well as some neurodegenerative disorders. In some species, they play an essential role in hibernation. One of the most exciting findings of the past decade is the opioid-receptor, especially DOR, mediated neuroprotection and cardioprotection. The upregulation of DOR expression and DOR activation increase the neuronal tolerance to hypoxic/ischemic stress. The DOR signal triggers (depending on stress duration and severity) different mechanisms at multiple levels to preserve neuronal survival, including the stabilization of homeostasis and increased pro-survival signaling (e.g., PKC-ERK-Bcl 2) and antioxidative capacity. In the heart, PKC and KATP channels are involved in the opioid receptor-mediated cardioprotection. The DOR-mediated neuroprotection and cardioprotection have the potential to significantly alter the clinical pharmacology in terms of prevention and treatment of life-threatening conditions like stroke and myocardial infarction. The main purpose of this article is to review the recent work done on opioids and their receptor functions. It shall provide an informative reference for better understanding the opioid system and further elucidation of the opioid receptor function from a physiological and pharmacological point of view.
Collapse
Affiliation(s)
- Yuan Feng
- Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | | | |
Collapse
|
37
|
Todorov PT, Mateeva PI, Zamfirova RN, Pavlov ND, Naydenova ED. Synthesis and biological activity of new series of N-modified analogues of the N/OFQ(1-13)NH2 with aminophosphonate moiety. Amino Acids 2012; 43:1217-23. [PMID: 22143428 DOI: 10.1007/s00726-011-1177-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 11/22/2011] [Indexed: 11/27/2022]
Abstract
New series of N-modified analogues of the N/OFQ(1-13)NH(2) with aminophosphonate moiety have been synthesized and investigated for biological activity. These peptides were prepared by solid-phase peptide synthesis-Fmoc-strategy. The N/OFQ(1-13)NH(2) analogues were tested for agonistic activity in vitro on electrically stimulated rat vas deferens smooth-muscle preparations isolated from Wistar albino rats. Our study has shown that the selectivity of the peptides containing 1-[(methoxyphosphono)methylamino]cycloalkanecarboxylic acids to the N-side of Phe is not changed-they remain selective agonists of NOP receptors. The derivative with the largest ring (NOC-6) demonstrated efficacy similar to that of N/OFQ(1-13)NH(2), but in a 10-fold higher concentration. The agonistic activity of newly synthesized N-modified analogues of N/OFQ(1-13)NH(2) with aminophosphonate moiety was investigated for the first time.
Collapse
Affiliation(s)
- Petar T Todorov
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, Sofia, Bulgaria
| | | | | | | | | |
Collapse
|
38
|
Mandegary A, Pournamdari M, Sharififar F, Pournourmohammadi S, Fardiar R, Shooli S. Alkaloid and flavonoid rich fractions of fenugreek seeds (Trigonella foenum-graecum L.) with antinociceptive and anti-inflammatory effects. Food Chem Toxicol 2012; 50:2503-7. [DOI: 10.1016/j.fct.2012.04.020] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 04/02/2012] [Accepted: 04/11/2012] [Indexed: 12/01/2022]
|
39
|
Opioid Antagonist Naltrexone for the Treatment of Pathological Gambling in Parkinson Disease. Clin Neuropharmacol 2012; 35:118-20. [DOI: 10.1097/wnf.0b013e31824d529b] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
40
|
Hutchinson MR, Shavit Y, Grace PM, Rice KC, Maier SF, Watkins LR. Exploring the neuroimmunopharmacology of opioids: an integrative review of mechanisms of central immune signaling and their implications for opioid analgesia. Pharmacol Rev 2011; 63:772-810. [PMID: 21752874 PMCID: PMC3141878 DOI: 10.1124/pr.110.004135] [Citation(s) in RCA: 291] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Vastly stimulated by the discovery of opioid receptors in the early 1970s, preclinical and clinical research was directed at the study of stereoselective neuronal actions of opioids, especially those played in their crucial analgesic role. However, during the past decade, a new appreciation of the non-neuronal actions of opioids has emerged from preclinical research, with specific appreciation for the nonclassic and nonstereoselective sites of action. Opioid activity at Toll-like receptors, newly recognized innate immune pattern recognition receptors, adds substantially to this unfolding story. It is now apparent from molecular and rodent data that these newly identified signaling events significantly modify the pharmacodynamics of opioids by eliciting proinflammatory reactivity from glia, the immunocompetent cells of the central nervous system. These central immune signaling events, including the release of cytokines and chemokines and the associated disruption of glutamate homeostasis, cause elevated neuronal excitability, which subsequently decreases opioid analgesic efficacy and leads to heightened pain states. This review will examine the current preclinical literature of opioid-induced central immune signaling mediated by classic and nonclassic opioid receptors. A unification of the preclinical pharmacology, neuroscience, and immunology of opioids now provides new insights into common mechanisms of chronic pain, naive tolerance, analgesic tolerance, opioid-induced hyperalgesia, and allodynia. Novel pharmacological targets for future drug development are discussed in the hope that disease-modifying chronic pain treatments arising from the appreciation of opioid-induced central immune signaling may become practical.
Collapse
Affiliation(s)
- Mark R Hutchinson
- Discipline of Pharmacology, School of Medical Science, University of Adelaide, South Australia, Australia, 5005.
| | | | | | | | | | | |
Collapse
|
41
|
Zhou Q, Carlsson A, Hallberg M, Nyberg F. Substance P N-terminal fragment SP(1-7) attenuates chronic morphine tolerance and affects dynorphin B and nociceptin in rats. Peptides 2011; 32:1661-5. [PMID: 21763376 DOI: 10.1016/j.peptides.2011.06.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2011] [Revised: 06/29/2011] [Accepted: 06/30/2011] [Indexed: 01/28/2023]
Abstract
The N-terminal substance P fragment SP(1-7) is known to modulate hyperalgesia and opioid withdrawal in animal models. This study examined the effects of intraperitoneal (i.p.) injections of SP(1-7) on chronic morphine tolerance and on the levels of dynorphin B (DYN B) and nociceptin/orphanin FQ (N/OFQ) in various brain areas of male Sprague-Dawley rats. Morphine tolerance was induced by subcutaneous injections of the opioid (10mg/kg) twice daily for 7 days. SP(1-7) injected i.p. (185 nmol/kg) 30 min prior to morphine reduced the development of morphine tolerance. Immunoreactive (ir) DYN B and N/OFQ peptide levels were measured in several areas of the central nervous system. Levels of ir DYN B in rats treated with SP(1-7) and morphine were decreased in the nucleus accumbens, substantia nigra and ventral tegmental area and increased in the frontal cortex. The ir N/OFQ levels were increased in the periaqueductal gray and decreased in the nucleus accumbens. Since the concentration profiles of the two peptides were altered by SP(1-7) in the areas that are implicated in the modulation of opioid tolerance and analgesia, it is suggested that DYN B and N/OFQ systems may be involved in the effects of SP(1-7) on opioid tolerance.
Collapse
Affiliation(s)
- Qin Zhou
- Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, Uppsala University, P.O. 591, S-751 24 Uppsala, Sweden
| | | | | | | |
Collapse
|
42
|
Mika J, Obara I, Przewlocka B. The role of nociceptin and dynorphin in chronic pain: implications of neuro-glial interaction. Neuropeptides 2011; 45:247-61. [PMID: 21477860 DOI: 10.1016/j.npep.2011.03.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Revised: 03/15/2011] [Accepted: 03/15/2011] [Indexed: 01/08/2023]
Abstract
Nociceptin-opioid peptide (NOP) receptor, also known as opioid receptor like-1 (ORL1), was identified following the cloning of the kappa-opioid peptide (KOP) receptor, and the characterization of these receptors revealed high homology. The endogenous ligand of NOP, nociceptin (NOC), which shares high homology to dynorphin (DYN), was discovered shortly thereafter, and since then, it has been the subject of several investigations. Despite the many advances in our understanding of the involvement of NOC and DYN systems in pain, tolerance and withdrawal, the precise function of these systems has not been fully characterized. Here, we review the recent literature concerning the distribution of the NOC and DYN systems in the central nervous system and the involvement of these systems in nociceptive transmission, especially under chronic pain conditions. We discuss the use of endogenous and exogenous ligands of NOP and KOP receptors in pain perception, as well as the potential utility of NOP ligands in clinical practice for pain management. We also discuss the modulation of opioid effects by NOC and DYN. We emphasize the important role of neuro-glial interactions in the effects of NOC and DYN, focusing on their presence in neuronal and non-neuronal cells and the changes associated with chronic pain conditions. We also present the dynamics of immune and glial regulation of neuronal functions and the importance of this regulation in the roles of NOC and DYN under conditions of neuropathic pain and in the use of drugs that alter these systems for better control of neuropathic pain.
Collapse
Affiliation(s)
- Joanna Mika
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | | | | |
Collapse
|
43
|
Katsuyama S, Mizoguchi H, Komatsu T, Sakurada C, Tsuzuki M, Sakurada S, Sakurada T. Antinociceptive effects of spinally administered nociceptin/orphanin FQ and its N-terminal fragments on capsaicin-induced nociception. Peptides 2011; 32:1530-5. [PMID: 21672568 DOI: 10.1016/j.peptides.2011.05.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 05/28/2011] [Accepted: 05/30/2011] [Indexed: 11/21/2022]
Abstract
Nociceptin/orphanin FQ (N/OFQ), the endogenous ligand for the N/OFQ peptide (NOP) receptors, has been shown to be metabolized into some fragments. We examined to determine whether intrathecal (i.t.) N/OFQ (1-13), (1-11) and (1-7) have antinociceptive activity in the pain-related behavior after intraplantar injection of capsaicin. The i.t. administration of N/OFQ (0.3-1.2 nmol) produced an appreciable and dose-dependent inhibition of capsaicin-induced paw-licking/biting response. The N-terminal fragments of N/OFQ, (1-13) and (1-11), were antinociceptive with a potency lower than N/OFQ. Calculated ID₅₀ values (nmol, i.t.) were 0.83 for N/OFQ, 2.5 for N/OFQ (1-13) and 4.75 for N/OFQ (1-11), respectively. The time-course effect revealed that the antinociceptive effects of these N-terminal fragments lasted longer than those of N/OFQ. Removal of amino acids down to N/OFQ (1-7) led to be less potent than N/OFQ and its fragments, (1-13) and (1-11). Antinociception induced by N/OFQ or N/OFQ (1-13) was reversed significantly by i.t. co-injection of [Nphe¹]N/OFQ (1-13)NH₂, a peptidergic antagonist for NOP receptors, whereas i.t. injection of the antagonist did not interfere with the action of N/OFQ (1-11) and (1-7). Pretreatment with the opioid receptor antagonist naloxone hydrochloride did not affect the antinociception induced by N/OFQ and its N-terminal fragments. These results suggest that N-terminal fragments of N/OFQ are active metabolites and may modulate the antinociceptive effect of N/OFQ in the spinal cord. The results also indicate that N/OFQ (1-13) still possess antinociceptive activity through NOP receptors.
Collapse
Affiliation(s)
- Soh Katsuyama
- Department of Pharmacology, Daiichi College of Pharmaceutical Sciences, 22-1 Tamagawa-cho, Minami-ku, Fukuoka 815-8511, Japan
| | | | | | | | | | | | | |
Collapse
|
44
|
Cannarsa R, Carretta D, Lattanzio F, Candeletti S, Romualdi P. ∆9-Tetrahydrocannabinol Decreases NOP Receptor Density and mRNA Levels in Human SH-SY5Y Cells. J Mol Neurosci 2011; 46:285-92. [DOI: 10.1007/s12031-011-9552-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 05/09/2011] [Indexed: 11/27/2022]
|
45
|
Liao YY, Jiang F, Chiou LC. Quantitative study of the antagonistic effect of (-)-cis-1-Methyl-7-[[4-(2,6-dichlorophenyl)piperidin-1-yl]methyl]-6,7,8,9-tetrahydro-5H-benzocyclohepten-5-ol (SB-612111) on nociceptin/orphanin FQ-mediated potassium channel activation in rat periaqueductal gray slices. Eur J Pharmacol 2011; 657:84-8. [PMID: 21300055 DOI: 10.1016/j.ejphar.2011.01.057] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 01/12/2011] [Accepted: 01/25/2011] [Indexed: 11/22/2022]
Abstract
Nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor, a non-opioid branch of the opioid receptor family, shows structural similarities to traditional opioid receptors but binds opioid with very poor affinity. This receptor has been implicated in many physiological functions including pain regulation. This study quantitatively investigated the effect of (-)-cis-1-Methyl-7-[[4-(2,6-dichlorophenyl)piperidin-1 -yl]methyl]-6,7,8,9-tetrahydro-5H-benzocyclohepten-5-ol (SB-612111), a novel non-peptide ligand of NOP receptor, on the native NOP receptors in the midbrain ventrolateral periaqueductal gray (vlPAG), a crucial region for pain regulation. SB-612111 concentration-dependently antagonized N/OFQ-induced G-protein coupled inwardly rectifying K(+) (GIRK) current in vlPAG neurons. The IC(50) value of SB-612111 estimated from dose-response curves is 87.7±1.2nM. SB-612111 had no intrinsic agonistic activity and did not affect the GIRK current induced by [D-Ala(2), N-Me-Phe(4), Gly(5)-ol]-enkephalin, a mu-opioid receptor agonist, when tested at concentrations of up to 1μM. It is concluded that SB-612111 is a pure, potent and selective antagonist of NOP receptors that mediate GIRK channel activation in the vlPAG neurons.
Collapse
Affiliation(s)
- Yan-Yu Liao
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | |
Collapse
|
46
|
Goeldner C, Reiss D, Kieffer BL, Ouagazzal AM. Endogenous nociceptin/orphanin-FQ in the dorsal hippocampus facilitates despair-related behavior. Hippocampus 2010; 20:911-6. [PMID: 20087887 PMCID: PMC4494786 DOI: 10.1002/hipo.20760] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Nociceptin/orphanin-FQ (N/OFQ) peptide and its receptor (NOP: N/OFQ opioid peptide receptor) are highly expressed in the hippocampus, but their functional role remains poorly understood. We recently showed that hippocampal N/OFQ inhibits learning and memory abilities in mice. Here, we investigated whether the endogenous peptide also regulated emotional responses at the level of the hippocampus. Bilateral infusions of the selective NOP receptor antagonist, UFP-101 (1-3 nmol/side), into the dorsal hippocampus produced antidepressant-like effects in the mouse forced swim and tail suspension tests comparable with those obtained with the prototypical antidepressant, fluoxetine (10-30 mg/kg, intraperitoneal). In the light-dark test, neither UFP-101 (1-3 nmol/side) nor N/OFQ peptide (1-3 nmol/side) modified anxiety measures when injected at behaviorally active doses in the dorsal hippocampus. These findings show a clear dissociation in the involvement of hippocampal N/OFQ system in anxiety- and despair-related behaviors. We conclude that the dorsal hippocampus is a brain region in which there is an important N/OFQ modulation of mnemonic processes and adaptive emotional responses associated to despair states.
Collapse
Affiliation(s)
- Celia Goeldner
- Université de Strasbourg, Strasbourg, F-67000 France
- IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), Département de Neurobiologie et Génétique Illkirch, F-67404 France; Inserm, U596, Illkirch, F-67404 France; CNRS, UMR7104, Illkirch, F-67404 France
| | - David Reiss
- IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), Département de Neurobiologie et Génétique Illkirch, F-67404 France; Inserm, U596, Illkirch, F-67404 France; CNRS, UMR7104, Illkirch, F-67404 France
| | - Brigitte L. Kieffer
- IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), Département de Neurobiologie et Génétique Illkirch, F-67404 France; Inserm, U596, Illkirch, F-67404 France; CNRS, UMR7104, Illkirch, F-67404 France
| | - Abdel-Mouttalib Ouagazzal
- IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), Département de Neurobiologie et Génétique Illkirch, F-67404 France; Inserm, U596, Illkirch, F-67404 France; CNRS, UMR7104, Illkirch, F-67404 France
| |
Collapse
|
47
|
Lü N, Han M, Yang ZL, Wang YQ, Wu GC, Zhang YQ. Nociceptin/Orphanin FQ in PAG modulates the release of amino acids, serotonin and norepinephrine in the rostral ventromedial medulla and spinal cord in rats. Pain 2010; 148:414-425. [PMID: 20036056 DOI: 10.1016/j.pain.2009.11.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Revised: 10/28/2009] [Accepted: 11/30/2009] [Indexed: 11/21/2022]
Abstract
High density Nociceptin/Orphanin FQ (N/OFQ) and its receptor (NOPr) have been found in the ventrolateral periaqueductal gray (vlPAG), a main output pathway involved in the descending pain-control system. Our previous study demonstrated that the microinjection of N/OFQ into the vlPAG markedly facilitated nociceptive responses of spinal dorsal horn neurons. The aim of the present work was to further provide evidence for the supraspinal mechanisms of action for N/OFQ-mediated nociceptive facilitation by examining the effect of N/OFQ in the vlPAG on neurotransmitter release in the descending pain-control system, including the nucleus raphe magnus (NRM), nucleus reticularis gigantocellularis (NGC) and dorsal horn of the spinal cord. The results showed that the microinjection of N/OFQ into the vlPAG produced robust decreases in 5-hydroxytryptamine (5-HT, serotonin), norepinephrine (NE), and gamma-aminobutyric acid (GABA), and increase in glutamate (Glu) release in the spinal dorsal horn. Spinal application of 5-HT, 2-Me-5-HT (5-HT(3) receptor agonist), muscimol (GABA(A) receptor agonist), and baclofen (GABA(B) receptor agonist) significantly blocked intra-vlPAG-induced facilitation on nociceptive responses. However, the extracellular concentrations of these neurotransmitters in the NRM and NGC exhibited diversity following intra-vlPAG of N/OFQ. In the NRM, intra-vlPAG injection of N/OFQ significantly decreased 5-HT, NE, and Glu, but increased GABA release. Differently, in the NGC, both NE and GABA releases were attenuated by intra-vlPAG of N/OFQ, whereas the concentration of 5-HT and Glu exhibited a trend to increase. These findings provide direct support for the hypothesis that intra-PAG of N/OFQ-induced facilitation of nociceptive responses is associated with the release of 5-HT, NE, and amino acids.
Collapse
Affiliation(s)
- Ning Lü
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, 138 Yi Xue Yuan Road, Shanghai 200032, China
| | | | | | | | | | | |
Collapse
|
48
|
Grandi D, Solenghi E, Massi M, Morini G. Peripheral infusion of nociceptin/orphanin FQ influences the response of rat gastric and colonic mucosa to repeated stress. REGULATORY PEPTIDES 2010; 160:49-56. [PMID: 19945488 DOI: 10.1016/j.regpep.2009.11.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 11/18/2009] [Accepted: 11/22/2009] [Indexed: 10/20/2022]
Abstract
The 17-amino acid peptide nociceptin/orphanin FQ (N/OFQ) plays a role in the regulation of stress responses and of emotional disorders. The objective of this study is to evaluate whether long-term peripheral N/OFQ could dose- and time-dependently influence the responses to repeated cold-restraint stress on the rat gastric and colonic mucosa. Rats were exposed to cold-restraint stress for 3h per day for 1, 2 and 3 consecutive days. N/OFQ was administered at doses of 0.1, 1 and 10 microg/kg/h via Alzet osmotic minipumps. In the gastric fundus, N/OFQ exerted dose-dependent beneficial effects against acute and repeated stress but, after prolonged treatment, became damaging in non-stressed rats. In the distal colon, N/OFQ exerted a protective effect against damage by acute and repeated stress with no influence on epithelial integrity in non-stressed rats. In both regions, the peptide itself dose- and time-dependently reduced intraepithelial mucins. The reduction in mucin content caused by stress was effectively counteracted by N/OFQ, 0.1 microg/kg/h, in the distal colon only. N/OFQ did not modify basal mucosal cell proliferation. The peptide at 0.1 and 1 microg/kg/h had no influence while at 10 microg/kg/h abolished stress-induced increase in cell proliferation. The present results provide evidence that N/OFQ is implicated in the regulation of resting and stress-challenged mucosal integrity and activity of mucin-producing cells.
Collapse
Affiliation(s)
- Daniela Grandi
- Department of Human Anatomy, Pharmacology and Forensic Medicine, University of Parma, Parma, Italy
| | | | | | | |
Collapse
|
49
|
Martin-Fardon R, Zorrilla EP, Ciccocioppo R, Weiss F. Role of innate and drug-induced dysregulation of brain stress and arousal systems in addiction: Focus on corticotropin-releasing factor, nociceptin/orphanin FQ, and orexin/hypocretin. Brain Res 2010; 1314:145-61. [PMID: 20026088 PMCID: PMC2819635 DOI: 10.1016/j.brainres.2009.12.027] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2009] [Revised: 12/09/2009] [Accepted: 12/10/2009] [Indexed: 11/25/2022]
Abstract
Stress-like symptoms are an integral part of acute and protracted drug withdrawal, and several lines of evidence have shown that dysregulation of brain stress systems, including the extrahypothalamic corticotropin-releasing factor (CRF) system, following long-term drug use is of major importance in maintaining drug and alcohol addiction. Recently, two other neuropeptide systems have attracted interest, the nociceptin/orphanin FQ (N/OFQ) and orexin/hypocretin (Orx/Hcrt) systems. N/OFQ participates in a wide range of physiological responses, and the hypothalamic Orx/Hcrt system helps regulate several physiological processes, including feeding, energy metabolism, and arousal. Moreover, these two systems have been suggested to participate in psychiatric disorders, including anxiety and drug addiction. Dysregulation of these systems by chronic drug exposure has been hypothesized to play a role in the maintenance of addiction and dependence. Recent evidence demonstrated that interactions between CRF-N/OFQ and CRF-Orx/Hcrt systems may be functionally relevant for the control of stress-related addictive behavior. The present review discusses recent findings that support the hypotheses of the participation and dysregulation of these systems in drug addiction and evaluates the current understanding of interactions among these stress-regulatory peptides.
Collapse
Affiliation(s)
- Rémi Martin-Fardon
- The Scripps Research Institute, Molecular and Integrative Neurosciences Department, SP30-2120, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
50
|
Kuzmin A, Madjid N, Johansson B, Terenius L, Ogren SO. The nociceptin system and hippocampal cognition in mice: a pharmacological and genetic analysis. Brain Res 2009; 1305 Suppl:S7-19. [PMID: 19782658 DOI: 10.1016/j.brainres.2009.09.075] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Revised: 09/10/2009] [Accepted: 09/16/2009] [Indexed: 11/24/2022]
Abstract
This study examines the effects of NOP agonists nociceptin/orphanin FQ (N/OFQ) and Ro 64-6198, NOP antagonists [Nphe(1)]N/OFQ(1-13)-NH(2) Nphe(1) and naloxone benzoylhydrazone (NalBzoH) on spatial memory in NMRI mice and pronociceptin (proNC) knockout (KO) mice using the water maze task. N/OFQ, administered i.c.v. (1, 5 and 10 nmol/mouse) and into hippocampal CA3 (1 nmol/mouse, bilaterally), impaired acquisition and retention in the maze. Impairments were blocked by pre-treatment with Nphe(1) (10 nmol, i.c.v.). Ro 64-6198 (0.1-0.3-1 mg/kg i.p.) also dose-dependently impaired learning. However, pre-treatment with NalBzoH (1 mg/kg, s.c.) failed to modify the effects of Ro 64-6198. Nphe(1) (10 nmol/mouse i.c.v.) and NalBzoH (1 mg/kg, s.c.) by themselves failed to affect maze performance, despite a tendency for enhanced performance. Prepro N/OFQ knockout (ppN/OFQ -/-) showed evidence of improved learning, evident at retention trials and in reversal training. ppN/OFQ -/- mice were not impaired by N/OFQ (10 nmol i.c.v.) in the task, suggesting that changes in postsynaptic NOP receptors may occur in such KO mice. It is concluded that N/OFQ and NOP receptors have an important role in hippocampus-dependent spatial learning and memory, probably by modulation of glutamatergic functions.
Collapse
Affiliation(s)
- Alexander Kuzmin
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | | | | | | |
Collapse
|