1
|
McGregor R, Wu MF, Thannickal TC, Li S, Siegel JM. Opioid-induced neuroanatomical, microglial and behavioral changes are blocked by suvorexant without diminishing opioid analgesia. NATURE. MENTAL HEALTH 2024; 2:1018-1031. [PMID: 39989723 PMCID: PMC11845277 DOI: 10.1038/s44220-024-00278-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 05/31/2024] [Indexed: 02/25/2025]
Abstract
Heroin use disorder in humans and chronic opioid administration to mice result in an increase in the number and a decrease in the size of detected hypocretin (Hcrt, or orexin) neurons. Chronic morphine administration to mice increases Hcrt axonal projections to the ventral tegmental area (VTA), the level of tyrosine hydroxylase (TH) in VTA and the number of detected TH+ cells in VTA, and activates VTA and hypothalamic microglia. Co-administration of morphine with the dual Hcrt receptor antagonist suvorexant prevents morphine-induced changes in the number and size of Hcrt neurons, the increase in Hcrt projections to the VTA and microglial activation in the VTA and hypothalamus. Co-administration of suvorexant with morphine also prevents morphine anticipatory behavior and reduces opioid withdrawal symptoms. However, suvorexant does not diminish morphine analgesia. Here we show that combined administration of opioids and suvorexant may reduce the addiction potential of opioid use for pain relief in humans while maintaining the analgesic effects of opioids.
Collapse
Affiliation(s)
- Ronald McGregor
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- These authors contributed equally: Ronald McGregor, Ming-Fung Wu
| | - Ming-Fung Wu
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- These authors contributed equally: Ronald McGregor, Ming-Fung Wu
| | - Thomas C. Thannickal
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Songlin Li
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Jerome M. Siegel
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
2
|
Kukkonen JP, Jacobson LH, Hoyer D, Rinne MK, Borgland SL. International Union of Basic and Clinical Pharmacology CXIV: Orexin Receptor Function, Nomenclature and Pharmacology. Pharmacol Rev 2024; 76:625-688. [PMID: 38902035 DOI: 10.1124/pharmrev.123.000953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/02/2024] [Accepted: 06/06/2024] [Indexed: 06/22/2024] Open
Abstract
The orexin system consists of the peptide transmitters orexin-A and -B and the G protein-coupled orexin receptors OX1 and OX2 Orexin receptors are capable of coupling to all four families of heterotrimeric G proteins, and there are also other complex features of the orexin receptor signaling. The system was discovered 25 years ago and was immediately identified as a central regulator of sleep and wakefulness; this is exemplified by the symptomatology of the disorder narcolepsy with cataplexy, in which orexinergic neurons degenerate. Subsequent translation of these findings into drug discovery and development has resulted to date in three clinically used orexin receptor antagonists to treat insomnia. In addition to sleep and wakefulness, the orexin system appears to be a central player at least in addiction and reward, and has a role in depression, anxiety and pain gating. Additional antagonists and agonists are in development to treat, for instance, insomnia, narcolepsy with or without cataplexy and other disorders with excessive daytime sleepiness, depression with insomnia, anxiety, schizophrenia, as well as eating and substance use disorders. The orexin system has thus proved an important regulator of numerous neural functions and a valuable drug target. Orexin prepro-peptide and orexin receptors are also expressed outside the central nervous system, but their potential physiological roles there remain unknown. SIGNIFICANCE STATEMENT: The orexin system was discovered 25 years ago and immediately emerged as an essential sleep-wakefulness regulator. This discovery has tremendously increased the understanding of these processes and has thus far resulted in the market approval of three orexin receptor antagonists, which promote more physiological aspects of sleep than previous hypnotics. Further, orexin receptor agonists and antagonists with different pharmacodynamic properties are in development since research has revealed additional potential therapeutic indications. Orexin receptor signaling is complex and may represent novel features.
Collapse
Affiliation(s)
- Jyrki P Kukkonen
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Laura H Jacobson
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Daniel Hoyer
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Maiju K Rinne
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Stephanie L Borgland
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| |
Collapse
|
3
|
Dale NC, Hoyer D, Jacobson LH, Pfleger KDG, Johnstone EKM. Orexin Signaling: A Complex, Multifaceted Process. Front Cell Neurosci 2022; 16:812359. [PMID: 35496914 PMCID: PMC9044999 DOI: 10.3389/fncel.2022.812359] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/07/2022] [Indexed: 11/15/2022] Open
Abstract
The orexin system comprises two G protein-coupled receptors, OX1 and OX2 receptors (OX1R and OX2R, respectively), along with two endogenous agonists cleaved from a common precursor (prepro-orexin), orexin-A (OX-A) and orexin-B (OX-B). For the receptors, a complex array of signaling behaviors has been reported. In particular, it becomes obvious that orexin receptor coupling is very diverse and can be tissue-, cell- and context-dependent. Here, the early signal transduction interactions of the orexin receptors will be discussed in depth, with particular emphasis on the direct G protein interactions of each receptor. In doing so, it is evident that ligands, additional receptor-protein interactions and cellular environment all play important roles in the G protein coupling profiles of the orexin receptors. This has potential implications for our understanding of the orexin system's function in vivo in both central and peripheral environments, as well as the development of novel agonists, antagonists and possibly allosteric modulators targeting the orexin system.
Collapse
Affiliation(s)
- Natasha C. Dale
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, WA, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Perth, WA, Australia
| | - Daniel Hoyer
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Laura H. Jacobson
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Kevin D. G. Pfleger
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, WA, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Perth, WA, Australia
- Dimerix Limited, Nedlands, WA, Australia
| | - Elizabeth K. M. Johnstone
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, WA, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Perth, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
4
|
Gopalakrishnan L, Chatterjee O, Ravishankar N, Suresh S, Raju R, Mahadevan A, Prasad TSK. Opioid receptors signaling network. J Cell Commun Signal 2021; 16:475-483. [PMID: 34724150 DOI: 10.1007/s12079-021-00653-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/12/2021] [Indexed: 10/19/2022] Open
Abstract
Opioid receptors belong to the class A G-protein-coupled receptors and are activated by alkaloid opiates such as morphine, and endogenous ligands such as endorphins and enkephalins. Opioid receptors are widely distributed in the human body and are involved in numerous physiological processes through three major classical opioid receptor subtypes; the mu, delta and kappa along with a lesser characterized subtype, opioid receptor-like (ORL1). Opioids are the most potent analgesics and have been extensively used as a therapeutic drug for the treatment of pain and related disorders. Chronic administration of clinically used opioids is associated with adverse effects such as drug tolerance, addiction and constipation. Several investigations attempted to identify the molecular signaling networks associated with endogenous as well as synthetic opiates, however, there is a paucity of a cumulative depiction of these signaling events. Here, we report a systemic collection of downstream molecules pertaining to four subtypes of opioid receptors (MOR, KOR, DOR and ORL1) in the form of a signaling pathway map. We manually curated reactions induced by the activation of opioid receptors from the literature into five categories- molecular association, activation/inhibition, catalysis, transport, and gene regulation. This led to a dataset of 180 molecules, which is collectively represented in the opioid receptor signaling network following NetPath criteria. We believe that the public availability of an opioid receptor signaling pathway map can accelerate biomedical research in this area because of its high therapeutic significance. The opioid receptors signaling pathway map is uploaded to a freely available web resource, WikiPathways enabling ease of access ( https://www.wikipathways.org/index.php/Pathway:WP5093 ).
Collapse
Affiliation(s)
- Lathika Gopalakrishnan
- Institute of Bioinformatics, International Tech Park, Bangalore, 560 066, India.,Manipal Academy of Higher Education (MAHE), Manipal, 576 104, India.,Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575 018, India
| | - Oishi Chatterjee
- Institute of Bioinformatics, International Tech Park, Bangalore, 560 066, India.,Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575 018, India.,Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, 690 525, India
| | - Namitha Ravishankar
- Institute of Bioinformatics, International Tech Park, Bangalore, 560 066, India
| | - Sneha Suresh
- Institute of Bioinformatics, International Tech Park, Bangalore, 560 066, India
| | - Rajesh Raju
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575 018, India.
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, 560 029, India.,Human Brain Tissue Repository, National Institute of Mental Health and Neurosciences, Neurobiology Research Centre, Bangalore, 560 029, India
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575 018, India.
| |
Collapse
|
5
|
Seoane-Collazo P, Romero-Picó A, Rial-Pensado E, Liñares-Pose L, Estévez-Salguero Á, Fernø J, Nogueiras R, Diéguez C, López M. κ-Opioid Signaling in the Lateral Hypothalamic Area Modulates Nicotine-Induced Negative Energy Balance. Int J Mol Sci 2021; 22:ijms22041515. [PMID: 33546289 PMCID: PMC7913331 DOI: 10.3390/ijms22041515] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/20/2022] Open
Abstract
Several studies have reported that nicotine, the main bioactive component of tobacco, exerts a marked negative energy balance. Apart from its anorectic action, nicotine also modulates energy expenditure, by regulating brown adipose tissue (BAT) thermogenesis and white adipose tissue (WAT) browning. These effects are mainly controlled at the central level by modulation of hypothalamic neuropeptide systems and energy sensors, such as AMP-activated protein kinase (AMPK). In this study, we aimed to investigate the kappa opioid receptor (κOR)/dynorphin signaling in the modulation of nicotine’s effects on energy balance. We found that body weight loss after nicotine treatment is associated with a down-regulation of the κOR endogenous ligand dynorphin precursor and with a marked reduction in κOR signaling and the p70 S6 kinase/ribosomal protein S6 (S6K/rpS6) pathway in the lateral hypothalamic area (LHA). The inhibition of these pathways by nicotine was completely blunted in κOR deficient mice, after central pharmacological blockade of κOR, and in rodents where κOR was genetically knocked down specifically in the LHA. Moreover, κOR-mediated nicotine effects on body weight do not depend on orexin. These data unravel a new central regulatory pathway modulating nicotine’s effects on energy balance.
Collapse
Affiliation(s)
- Patricia Seoane-Collazo
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; (A.R.-P.); (E.R.-P.); (L.L.-P.); (Á.E.-S.); (R.N.); (C.D.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
- Correspondence: (P.S.-C.); (M.L.)
| | - Amparo Romero-Picó
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; (A.R.-P.); (E.R.-P.); (L.L.-P.); (Á.E.-S.); (R.N.); (C.D.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Eva Rial-Pensado
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; (A.R.-P.); (E.R.-P.); (L.L.-P.); (Á.E.-S.); (R.N.); (C.D.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Laura Liñares-Pose
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; (A.R.-P.); (E.R.-P.); (L.L.-P.); (Á.E.-S.); (R.N.); (C.D.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Ánxela Estévez-Salguero
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; (A.R.-P.); (E.R.-P.); (L.L.-P.); (Á.E.-S.); (R.N.); (C.D.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Johan Fernø
- Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway;
| | - Rubén Nogueiras
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; (A.R.-P.); (E.R.-P.); (L.L.-P.); (Á.E.-S.); (R.N.); (C.D.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Carlos Diéguez
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; (A.R.-P.); (E.R.-P.); (L.L.-P.); (Á.E.-S.); (R.N.); (C.D.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Miguel López
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; (A.R.-P.); (E.R.-P.); (L.L.-P.); (Á.E.-S.); (R.N.); (C.D.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
- Correspondence: (P.S.-C.); (M.L.)
| |
Collapse
|
6
|
The role of co-neurotransmitters in sleep and wake regulation. Mol Psychiatry 2019; 24:1284-1295. [PMID: 30377299 PMCID: PMC6491268 DOI: 10.1038/s41380-018-0291-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 09/17/2018] [Accepted: 10/08/2018] [Indexed: 12/11/2022]
Abstract
Sleep and wakefulness control in the mammalian brain requires the coordination of various discrete interconnected neurons. According to the most conventional sleep model, wake-promoting neurons (WPNs) and sleep-promoting neurons (SPNs) compete for network dominance, creating a systematic "switch" that results in either the sleep or awake state. WPNs and SPNs are ubiquitous in the brainstem and diencephalon, areas that together contain <1% of the neurons in the human brain. Interestingly, many of these WPNs and SPNs co-express and co-release various types of the neurotransmitters that often have opposing modulatory effects on the network. Co-transmission is often beneficial to structures with limited numbers of neurons because it provides increasing computational capability and flexibility. Moreover, co-transmission allows subcortical structures to bi-directionally control postsynaptic neurons, thus helping to orchestrate several complex physiological functions such as sleep. Here, we present an in-depth review of co-transmission in hypothalamic WPNs and SPNs and discuss its functional significance in the sleep-wake network.
Collapse
|
7
|
Santos-Otte P, Leysen H, van Gastel J, Hendrickx JO, Martin B, Maudsley S. G Protein-Coupled Receptor Systems and Their Role in Cellular Senescence. Comput Struct Biotechnol J 2019; 17:1265-1277. [PMID: 31921393 PMCID: PMC6944711 DOI: 10.1016/j.csbj.2019.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 12/17/2022] Open
Abstract
Aging is a complex biological process that is inevitable for nearly all organisms. Aging is the strongest risk factor for development of multiple neurodegenerative disorders, cancer and cardiovascular disorders. Age-related disease conditions are mainly caused by the progressive degradation of the integrity of communication systems within and between organs. This is in part mediated by, i) decreased efficiency of receptor signaling systems and ii) an increasing inability to cope with stress leading to apoptosis and cellular senescence. Cellular senescence is a natural process during embryonic development, more recently it has been shown to be also involved in the development of aging disorders and is now considered one of the major hallmarks of aging. G-protein-coupled receptors (GPCRs) comprise a superfamily of integral membrane receptors that are responsible for cell signaling events involved in nearly every physiological process. Recent advances in the molecular understanding of GPCR signaling complexity have expanded their therapeutic capacity tremendously. Emerging data now suggests the involvement of GPCRs and their associated proteins in the development of cellular senescence. With the proven efficacy of therapeutic GPCR targeting, it is reasonable to now consider GPCRs as potential platforms to control cellular senescence and the consequently, age-related disorders.
Collapse
Key Words
- ADP-ribosylation factor GTPase-activating protein, (Arf-GAP)
- AT1R blockers, (ARB)
- Aging
- Angiotensin II, (Ang II)
- Ataxia telangiectasia mutated, (ATM)
- Cellular senescence
- G protein-coupled receptor kinase interacting protein 2 (GIT2)
- G protein-coupled receptor kinase interacting protein 2, (GIT2)
- G protein-coupled receptor kinase, (GRK)
- G protein-coupled receptors (GPCRs)
- G protein-coupled receptors, (GPCRs)
- Hutchinson–Gilford progeria syndrome, (HGPS)
- Lysophosphatidic acid, (LPA)
- Regulator of G-protein signaling, (RGS)
- Relaxin family receptor 3, (RXFP3)
- active state, (R*)
- angiotensin type 1 receptor, (AT1R)
- angiotensin type 2 receptor, (AT2R)
- beta2-adrenergic receptor, (β2AR)
- cyclin-dependent kinase 2, (CDK2)
- cyclin-dependent kinase inhibitor 1, (cdkn1A/p21)
- endothelial cell differentiation gene, (Edg)
- inactive state, (R)
- latent semantic indexing, (LSI)
- mitogen-activated protein kinase, (MAPK)
- nuclear factor kappa-light-chain-enhancer of activated B cells, (NF- κβ)
- protein kinases, (PK)
- purinergic receptors family, (P2Y)
- renin-angiotensin system, (RAS)
- retinoblastoma, (RB)
- senescence associated secretory phenotype, (SASP)
- stress-induced premature senescence, (SIPS)
- transcription factor E2F3, (E2F3)
- transmembrane, (TM)
- tumor suppressor gene PTEN, (PTEN)
- tumor suppressor protein 53, (p53)
- vascular smooth muscle cells, (VSMC)
- β-Arrestin
Collapse
Affiliation(s)
- Paula Santos-Otte
- Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01062 Dresden, Germany
| | - Hanne Leysen
- Receptor Biology Lab, University of Antwerp, 2610 Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Jaana van Gastel
- Receptor Biology Lab, University of Antwerp, 2610 Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Jhana O. Hendrickx
- Receptor Biology Lab, University of Antwerp, 2610 Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Bronwen Martin
- Receptor Biology Lab, University of Antwerp, 2610 Antwerp, Belgium
| | - Stuart Maudsley
- Receptor Biology Lab, University of Antwerp, 2610 Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
8
|
Effect of orexin-A in the arcuate nucleus on cisplatin-induced gastric side effects in rats. Neurosci Res 2019; 143:53-60. [DOI: 10.1016/j.neures.2018.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 05/10/2018] [Accepted: 06/01/2018] [Indexed: 12/12/2022]
|
9
|
Alizamini MM, Kavianpour M, Karimi-Haghighi S, Fatahi Z, Haghparast A. Intra-hippocampal administration of orexin receptor antagonists dose-dependently attenuates reinstatement of morphine seeking behavior in extinguished rats. Peptides 2018; 110:40-46. [PMID: 30391421 DOI: 10.1016/j.peptides.2018.10.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/27/2018] [Accepted: 10/30/2018] [Indexed: 12/21/2022]
Abstract
It has been shown that the hippocampus plays an essential role in the regulation of reward and memory as indicated by the conditioned place preference (CPP) paradigm. Morphine-induced CPP is a common method to consider motivational properties of morphine in animals. Recently, this model has been used in many laboratories to investigate neuronal mechanisms underlying reinstatement of morphine seeking induced by drug re-exposure. Our previous studies indicate that the hippocampus especially CA1 region is involved in reinstatement of drug-seeking behaviors. Also, several studies have shown that orexin attenuates key functional and behavioral effects of its co-transmitter dynorphin. The present study evaluates the role of orexinergic receptors within the CA1 region of the hippocampus in the reinstatement of morphine-induced CPP. Therefore, after the extinction period, the different doses (SB 334867; 0.3, 3, and 30 nM/0.5 μl DMSO) of either orexin-1 or -2 receptor antagonists were bilaterally microinjected into the CA1, 15 min before receiving an effective priming dose of morphine (1 mg/kg). The results revealed that administration of both SB 334867 and TCS OX2 29 prior to injection of the priming dose of morphine significantly reduced the reinstatement of morphine-induced CPP without altering the animal's locomotor activity. Also, the 50% effective dose value of SB 334867 on the reinstatement of morphine seeking behavior was close three times more than that in TCS OX2 29 treatment group. Therefore, the consequences suggested that both orexin receptors in the CA1 play a considerable role in the reinstatement of morphine-induced CPP.
Collapse
Affiliation(s)
- Mirmohammadali Mirramezani Alizamini
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Mohadeseh Kavianpour
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeideh Karimi-Haghighi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Fatahi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Anderson RI, Moorman DE, Becker HC. Contribution of Dynorphin and Orexin Neuropeptide Systems to the Motivational Effects of Alcohol. Handb Exp Pharmacol 2018. [PMID: 29526023 DOI: 10.1007/164_2018_100] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Understanding the neural systems that drive alcohol motivation and are disrupted in alcohol use disorders is of critical importance in developing novel treatments. The dynorphin and orexin/hypocretin neuropeptide systems are particularly relevant with respect to alcohol use and misuse. Both systems are strongly associated with alcohol-seeking behaviors, particularly in cases of high levels of alcohol use as seen in dependence. Furthermore, both systems also play a role in stress and anxiety, indicating that disruption of these systems may underlie long-term homeostatic dysregulation seen in alcohol use disorders. These systems are also closely interrelated with one another - dynorphin/kappa opioid receptors and orexin/hypocretin receptors are found in similar regions and hypocretin/orexin neurons also express dynorphin - suggesting that these two systems may work together in the regulation of alcohol seeking and may be mutually disrupted in alcohol use disorders. This chapter reviews studies demonstrating a role for each of these systems in motivated behavior, with a focus on their roles in regulating alcohol-seeking and self-administration behaviors. Consideration is also given to evidence indicating that these neuropeptide systems may be viable targets for the development of potential treatments for alcohol use disorders.
Collapse
Affiliation(s)
- Rachel I Anderson
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA.,Science and Technology Policy Fellowships, American Association for the Advancement of Science, Washington, DC, USA
| | - David E Moorman
- Department of Psychological and Brain Sciences, Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - Howard C Becker
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA. .,Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, USA. .,Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA. .,Department of Veterans Affairs, Ralph H. Johnson VA Medical Center, Charleston, SC, USA.
| |
Collapse
|
11
|
Thompson MD, Sakurai T, Rainero I, Maj MC, Kukkonen JP. Orexin Receptor Multimerization versus Functional Interactions: Neuropharmacological Implications for Opioid and Cannabinoid Signalling and Pharmacogenetics. Pharmaceuticals (Basel) 2017; 10:ph10040079. [PMID: 28991183 PMCID: PMC5748636 DOI: 10.3390/ph10040079] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 09/29/2017] [Accepted: 09/29/2017] [Indexed: 12/17/2022] Open
Abstract
Orexins/hypocretins are neuropeptides formed by proteolytic cleavage of a precursor peptide, which are produced by neurons found in the lateral hypothalamus. The G protein-coupled receptors (GPCRs) for these ligands, the OX₁ and OX₂ orexin receptors, are more widely expressed throughout the central nervous system. The orexin/hypocretin system has been implicated in many pathways, and its dysregulation is under investigation in a number of diseases. Disorders in which orexinergic mechanisms are being investigated include narcolepsy, idiopathic sleep disorders, cluster headache and migraine. Human narcolepsy has been associated with orexin deficiency; however, it has only rarely been attributed to mutations in the gene encoding the precursor peptide. While gene variations within the canine OX₂ gene hcrtr2 have been directly linked with narcolepsy, the majority of human orexin receptor variants are weakly associated with diseases (the idiopathic sleep disorders, cluster headache and polydipsia-hyponatremia in schizophrenia) or are of potential pharmacogenetic significance. Evidence for functional interactions and/or heterodimerization between wild-type and variant orexin receptors and opioid and cannabinoid receptors is discussed in the context of its relevance to depression and epilepsy.
Collapse
Affiliation(s)
- Miles D Thompson
- Department of Pediatrics, University of California, San Diego 92093, CA, USA.
| | - Takeshi Sakurai
- Department of Molecular Neuroscience and Integrative Physiology, Faculty of Medicine, Kanazawa University, Kanazawa 920-8620, Japan.
| | - Innocenzo Rainero
- Department of Neuroscience, University of Turin, Torino 10124, Italy.
| | - Mary C Maj
- Department of Biochemistry, School of Medicine, Saint George's University, Saint George's 11739, Grenada.
| | - Jyrki P Kukkonen
- Biochemistry and Cell Biology, Department of Veterinary Biosciences, University of Helsinki, Helsinki 11739, Finland.
- Department of Physiology, Institute of Biomedicine, Biomedicum Helsinki, University of Helsinki, Helsinki 00100, Finland.
| |
Collapse
|
12
|
Schattauer SS, Land BB, Reichard KL, Abraham AD, Burgeno LM, Kuhar JR, Phillips PEM, Ong SE, Chavkin C. Peroxiredoxin 6 mediates Gαi protein-coupled receptor inactivation by cJun kinase. Nat Commun 2017; 8:743. [PMID: 28963507 PMCID: PMC5622097 DOI: 10.1038/s41467-017-00791-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 07/27/2017] [Indexed: 12/21/2022] Open
Abstract
Inactivation of opioid receptors limits the therapeutic efficacy of morphine-like analgesics and mediates the long duration of kappa opioid antidepressants by an uncharacterized, arrestin-independent mechanism. Here we use an iterative, discovery-based proteomic approach to show that following opioid administration, peroxiredoxin 6 (PRDX6) is recruited to the opioid receptor complex by c-Jun N-terminal kinase (JNK) phosphorylation. PRDX6 activation generates reactive oxygen species via NADPH oxidase, reducing the palmitoylation of receptor-associated Gαi in a JNK-dependent manner. Selective inhibition of PRDX6 blocks Gαi depalmitoylation, prevents the enhanced receptor G-protein association and blocks acute analgesic tolerance to morphine and kappa opioid receptor inactivation in vivo. Opioid stimulation of JNK also inactivates dopamine D2 receptors in a PRDX6-dependent manner. We show that the loss of this lipid modification distorts the receptor G-protein association, thereby preventing agonist-induced guanine nucleotide exchange. These findings establish JNK-dependent PRDX6 recruitment and oxidation-induced Gαi depalmitoylation as an additional mechanism of Gαi-G-protein-coupled receptor inactivation.Opioid receptors are important modulators of nociceptive pain. Here the authors show that opioid receptor activation recruits peroxiredoxin 6 (PRDX6) to the receptor-Gαi complex by c-Jun N-terminal kinase, resulting in Gαi depalmitoylation and enhanced receptor-Gαi association.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Benzeneacetamides/pharmacology
- Drug Tolerance
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Fentanyl/pharmacology
- GTP-Binding Protein alpha Subunits, Gi-Go/drug effects
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- JNK Mitogen-Activated Protein Kinases/drug effects
- JNK Mitogen-Activated Protein Kinases/metabolism
- Mice
- Morphine/pharmacology
- NADPH Oxidases/drug effects
- NADPH Oxidases/metabolism
- Peroxiredoxin VI/drug effects
- Peroxiredoxin VI/metabolism
- Phosphorylation
- Pyrrolidines/pharmacology
- Reactive Oxygen Species/metabolism
- Receptors, Dopamine D2/drug effects
- Receptors, Dopamine D2/metabolism
- Receptors, Opioid/drug effects
- Receptors, Opioid/metabolism
- Receptors, Opioid, kappa/drug effects
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Selena S Schattauer
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Benjamin B Land
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Kathryn L Reichard
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Antony D Abraham
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Lauren M Burgeno
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, 98195, USA
- Department of Psychiatry, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Jamie R Kuhar
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Paul E M Phillips
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, 98195, USA
- Department of Psychiatry, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Shao En Ong
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Charles Chavkin
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, 98195, USA.
| |
Collapse
|
13
|
Baimel C, Borgland SL. Hypocretin/Orexin and Plastic Adaptations Associated with Drug Abuse. Curr Top Behav Neurosci 2017; 33:283-304. [PMID: 28303403 DOI: 10.1007/7854_2016_44] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Dopamine neurons in the ventral tegmental area (VTA) are a critical part of the neural circuits that underlie reward learning and motivation. Dopamine neurons send dense projections throughout the brain and recent observations suggest that both the intrinsic properties and the functional output of dopamine neurons are dependent on projection target and are subject to neuromodulatory influences. Lateral hypothalamic hypocretin (also termed orexin) neurons project to the VTA and contain both hypocretin and dynorphin peptides in the same dense core vesicles suggesting they may be co-released. Hypocretin peptides act at excitatory Gαq protein-coupled receptors and dynorphin acts at inhibitory Gαi/o protein-coupled receptors, which are both expressed on subpopulations of dopamine neurons. This review describes a role for neuromodulation of dopamine neurons and the influence on motivated behaviour in response to natural and drug rewards.
Collapse
Affiliation(s)
- Corey Baimel
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, 2176 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, Canada, T2N 4N1
| | - Stephanie L Borgland
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, Canada, T2N 4N1.
| |
Collapse
|
14
|
Abstract
Orexin/hypocretin peptide (orexin-A and orexin-B) signaling is believed to take place via the two G-protein-coupled receptors (GPCRs), named OX1 and OX2 orexin receptors, as described in the previous chapters. Signaling of orexin peptides has been investigated in diverse endogenously orexin receptor-expressing cells - mainly neurons but also other types of cells - and in recombinant cells expressing the receptors in a heterologous manner. Findings in the different systems are partially convergent but also indicate cellular background-specific signaling. The general picture suggests an inherently high degree of diversity in orexin receptor signaling.In the current chapter, I present orexin signaling on the cellular and molecular levels. Discussion of the connection to (potential) physiological orexin responses is only brief since these are in focus of other chapters in this book. The same goes for the post-synaptic signaling mechanisms, which are dealt with in Burdakov: Postsynaptic actions of orexin. The current chapter is organized according to the tissue type, starting from the central nervous system. Finally, receptor signaling pathways are discussed across tissues, cell types, and even species.
Collapse
Affiliation(s)
- Jyrki P Kukkonen
- Biochemistry and Cell Biology, Department of Veterinary Biosciences, University of Helsinki, POB 66, FIN-00014, Helsinki, Finland.
| |
Collapse
|
15
|
Reiter E, Ayoub MA, Pellissier LP, Landomiel F, Musnier A, Tréfier A, Gandia J, De Pascali F, Tahir S, Yvinec R, Bruneau G, Poupon A, Crépieux P. β-arrestin signalling and bias in hormone-responsive GPCRs. Mol Cell Endocrinol 2017; 449:28-41. [PMID: 28174117 DOI: 10.1016/j.mce.2017.01.052] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 01/31/2017] [Accepted: 01/31/2017] [Indexed: 12/14/2022]
Abstract
G protein-coupled receptors (GPCRs) play crucial roles in the ability of target organs to respond to hormonal cues. GPCRs' activation mechanisms have long been considered as a two-state process connecting the agonist-bound receptor to heterotrimeric G proteins. This view is now challenged as mounting evidence point to GPCRs being connected to large arrays of transduction mechanisms involving heterotrimeric G proteins as well as other players. Amongst the G protein-independent transduction mechanisms, those elicited by β-arrestins upon their recruitment to the active receptors are by far the best characterized and apply to most GPCRs. These concepts, in conjunction with remarkable advances made in the field of GPCR structural biology and biophysics, have supported the notion of ligand-selective signalling also known as pharmacological bias. Interestingly, recent reports have opened intriguing prospects to the way β-arrestins control GPCR-mediated signalling in space and time within the cells. In the present paper, we review the existing evidence linking endocrine-related GPCRs to β-arrestin recruitement, signalling, pathophysiological implications and selective activation by biased ligands and/or receptor modifications. Emerging concepts surrounding β-arrestin-mediated transduction are discussed in the light of the peculiarities of endocrine systems.
Collapse
Affiliation(s)
- Eric Reiter
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France.
| | - Mohammed Akli Ayoub
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France; LE STUDIUM(®) Loire Valley Institute for Advanced Studies, 45000, Orléans, France; Biology Department, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | | | - Flavie Landomiel
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Astrid Musnier
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Aurélie Tréfier
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Jorge Gandia
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | | | - Shifa Tahir
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Romain Yvinec
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Gilles Bruneau
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Anne Poupon
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Pascale Crépieux
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| |
Collapse
|
16
|
Polter AM, Barcomb K, Chen RW, Dingess PM, Graziane NM, Brown TE, Kauer JA. Constitutive activation of kappa opioid receptors at ventral tegmental area inhibitory synapses following acute stress. eLife 2017; 6. [PMID: 28402252 PMCID: PMC5389861 DOI: 10.7554/elife.23785] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 03/13/2017] [Indexed: 12/14/2022] Open
Abstract
Stressful experiences potently activate kappa opioid receptors (κORs). κORs in the ventral tegmental area regulate multiple aspects of dopaminergic and non-dopaminergic cell function. Here we show that at GABAergic synapses on rat VTA dopamine neurons, a single exposure to a brief cold-water swim stress induces prolonged activation of κORs. This is mediated by activation of the receptor during the stressor followed by a persistent, ligand-independent constitutive activation of the κOR itself. This lasting change in function is not seen at κORs at neighboring excitatory synapses, suggesting distinct time courses and mechanisms of regulation of different subsets of κORs. We also provide evidence that constitutive activity of κORs governs the prolonged reinstatement to cocaine-seeking observed after cold water swim stress. Together, our studies indicate that stress-induced constitutive activation is a novel mechanism of κOR regulation that plays a critical role in reinstatement of drug seeking. DOI:http://dx.doi.org/10.7554/eLife.23785.001 People who are recovering from drug addiction are more vulnerable to cravings and relapse when under stress. This ability of stress to boost drug relapse can also be shown in animals previously exposed to addictive drugs. Rats can learn to press a lever to administer themselves a dose of cocaine and, during withdrawal, rats previously exposed to the drug will press the lever more often if they are stressed. Indeed, just a few minutes of stress is enough to increase lever pressing for several days. Stress and addictive drugs both act on a region of the brain called the ventral tegmental area, or VTA, which is part of the brain’s reward system. Stress indirectly increases the activity of the VTA. It does so by activating a protein on the surface of VTA neurons called the kappa opioid receptor (κOR for short). Previous studies revealed that five minutes of stress increases the activity of κORs in the VTA of rats for five days. Conversely, blocking κORs stopped stressed rats from pressing the lever more often for cocaine. Together, these findings suggested that activating κORs in the VTA contributes to stress-induced drug relapse. Polter et al. have now discovered how stress activates κORs. It turns out that stressful or unpleasant experiences cause the brain to produce a protein called dynorphin, which binds to and activates the κORs. After a stressful event, the receptors are said to have become constitutively active, and blocking this constitutive activity prevents stress from inducing drug-seeking behavior. Polter et al. show that binding of dynorphin is needed to change the shape of the receptors so that they remain active even after dynorphin has detached, but it is likely that other molecules are also involved. This is the first study to show a link between stress, constitutive activation of κORs, and drug relapse. The next step is to work out why this process occurs on only some and not all occasions when the brain releases dynorphin, and why only certain κORs in the VTA respond in this way. Whether constitutive kOR activity drives stress-related craving in people with a history of drug abuse and how to halt these cravings also remain to be determined. DOI:http://dx.doi.org/10.7554/eLife.23785.002
Collapse
Affiliation(s)
- Abigail M Polter
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, United States
| | - Kelsey Barcomb
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, United States
| | - Rudy W Chen
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, United States
| | - Paige M Dingess
- Neuroscience Program, University of Wyoming, Laramie, United States.,University of Wyoming, School of Pharmacy, Laramie, United States
| | - Nicholas M Graziane
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, United States
| | - Travis E Brown
- Neuroscience Program, University of Wyoming, Laramie, United States.,University of Wyoming, School of Pharmacy, Laramie, United States
| | - Julie A Kauer
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, United States
| |
Collapse
|
17
|
Abstract
This paper is the thirty-eighth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2015 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia, stress and social status, tolerance and dependence, learning and memory, eating and drinking, drug abuse and alcohol, sexual activity and hormones, pregnancy, development and endocrinology, mental illness and mood, seizures and neurologic disorders, electrical-related activity and neurophysiology, general activity and locomotion, gastrointestinal, renal and hepatic functions, cardiovascular responses, respiration and thermoregulation, and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|