1
|
Jelen LA, Young AH, Mehta MA. Opioid Mechanisms and the Treatment of Depression. Curr Top Behav Neurosci 2024; 66:67-99. [PMID: 37923934 DOI: 10.1007/7854_2023_448] [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] [Indexed: 11/06/2023]
Abstract
Opioid receptors are widely expressed in the brain, and the opioid system has a key role in modulating mood, reward processing and stress responsivity. There is mounting evidence that the endogenous opioid system may be dysregulated in depression and that drug treatments targeting mu, delta and kappa opioid receptors may show antidepressant potential. The mechanisms underlying the therapeutic effects of opioid system engagement are complex and likely multi-factorial. This chapter explores various pathways through which the modulation of the opioid system may influence depression. These include impacts on monoaminergic systems, the regulation of stress and the hypothalamic-pituitary-adrenal axis, the immune system and inflammation, brain-derived neurotrophic factors, neurogenesis and neuroplasticity, social pain and social reward, as well as expectancy and placebo effects. A greater understanding of the diverse mechanisms through which opioid system modulation may improve depressive symptoms could ultimately aid in the development of safe and effective alternative treatments for individuals with difficult-to-treat depression.
Collapse
Affiliation(s)
- Luke A Jelen
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
- South London and Maudsley NHS Foundation Trust, London, UK.
| | - Allan H Young
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
| | - Mitul A Mehta
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
2
|
Wu S, Ning K, Wang Y, Zhang L, Liu J. Up-regulation of BDNF/TrkB signaling by δ opioid receptor agonist SNC80 modulates depressive-like behaviors in chronic restraint-stressed mice. Eur J Pharmacol 2023; 942:175532. [PMID: 36708979 DOI: 10.1016/j.ejphar.2023.175532] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023]
Abstract
Depressive disorder is a psychiatric disease characterized by its main symptoms of low mood and anhedonia. Due to its complex etiology, current clinical treatments for depressive disorder are limited. In this study, we assessed the role of the δ opioid receptor (δOR) system in the development of chronic-restraint-stressed (CRS)-induced depressive behaviors. We employed a 21-day CRS model and detected the c-fos activation and protein levels' changes in enkephalin (ENK)/δOR. It was found that the hippocampus and amygdala were involved in CRS-induced depression. The expression of pro-enkephalin (PENK), the precursors of the endogenous ligand for δOR, was significantly decreased in the hippocampus and amygdala following CRS. We then treated the mice with SNC80, a specific δOR agonist, to examine its anti-depressant effects in the tail suspension test (TST), forced swimming test (FST), and sucrose preference test (SPT). SNC80 administration significantly reversed depressive-like behaviors, and this antidepressant effect could be blocked by a TrkB inhibitor: ANA-12. Although ANA-12 treatment had no significant effect on the expression of ENK/δOR, it blocked the promoting effects of brain-derived neurotrophic factor (BDNF)/tyrosine kinase B(TrkB) signaling by SNC80 in the hippocampus and amygdala. Therefore, the present study demonstrates that SNC80 exerts anti-depressant effects by up-regulating the BDNF/TrkB signaling pathway in the hippocampus and amygdala in CRS-induced depression and provides evidence that δOR's agonists may be potential anti-depressant therapeutic agents.
Collapse
Affiliation(s)
- Shuo Wu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Kuan Ning
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yujun Wang
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Lesha Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| | - Jinggen Liu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| |
Collapse
|
3
|
Xu C, Cheng Y, Han M, Tao Y, Liu JG. The Agonist of Adenosine A1 Receptor Induced Desensitization of delta Opioid receptor-mediated Raf-1/MEK/ERK Signaling by Feedback Phosphorylation of Raf-1-Ser289/296/301. Neurochem Res 2022; 48:1531-1542. [PMID: 36525124 DOI: 10.1007/s11064-022-03843-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/22/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
Our previous study found that activation of adenosine A1 receptor (A1R) induced phosphorylation of delta opioid receptor (DOR) and desensitization of its downstream signaling molecules, cAMP and Akt. To further investigate the effect of A1R agonist on DOR signaling and the underlying mechanism, we examined the effect of A1R activation upon binding of its agonist N6-cyclohexyl-adenosine (CHA) on DOR-mediated Raf-1/MEK/ERK activation, and found that prolonged CHA exposure resulted in downregulation of DOR-mediated Raf-1/MEK/ERK signaling pathway. CHA-treatment time dependently attenuated Raf-1-Ser338 phosphorylation induced by [D-Pen2,5] enkephalin (DPDPE), a specific agonist of DOR, and further caused downregulation of the Raf-1/MEK/ERK signaling pathway activated by DOR agonist. Moreover, CHA exposure time-dependently induced the phosphorylation of Raf-1-Ser289/296/301, the inhibitory phosphorylation sites that were regulated by negative feedback, thereby inhibiting activation of the MEK/ERK pathway, and this effect could be blocked by MEK inhibitor U0126. Finally, we proved that the heterologous desensitization of the Raf-1/MEK/ERK cascade was essential in the regulation of anti-nociceptive effect of DOR agonists by confirming that such effect was inhibited by pretreatment of CHA. Therefore, we conclude that the activation of A1R inhibits DOR-mediated MAPK signaling pathway via heterologous desensitization of the Raf-1/MEK/ERK cascade, which is a result of ERK-mediated Raf-1-Ser289/296/301 phosphorylation mediated by activation of A1R.
Collapse
|
4
|
The Opioid System in Depression. Neurosci Biobehav Rev 2022; 140:104800. [PMID: 35914624 PMCID: PMC10166717 DOI: 10.1016/j.neubiorev.2022.104800] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 12/16/2022]
Abstract
Opioid receptors are widely distributed throughout the brain and play an essential role in modulating aspects of human mood, reward, and well-being. Accumulating evidence indicates the endogenous opioid system is dysregulated in depression and that pharmacological modulators of mu, delta, and kappa opioid receptors hold potential for the treatment of depression. Here we review animal and clinical data, highlighting evidence to support: dysregulation of the opioid system in depression, evidence for opioidergic modulation of behavioural processes and brain regions associated with depression, and evidence for opioidergic modulation in antidepressant responses. We evaluate clinical trials that have examined the safety and efficacy of opioidergic agents in depression and consider how the opioid system may be involved in the effects of other treatments, including ketamine, that are currently understood to exert antidepressant effects through non-opioidergic actions. Finally, we explore key neurochemical and molecular mechanisms underlying the potential therapeutic effects of opioid system engagement, that together provides a rationale for further investigation into this relevant target in the treatment of depression.
Collapse
|
5
|
Abstract
After participating in this activity, learners should be better able to:• Identify the effects of dysregulated opioid signalling in depression• Evaluate the use of opioid compounds and ketamine in patients with depression ABSTRACT: Major depressive disorder (MDD) remains one of the leading causes of disability and functional impairment worldwide. Current antidepressant therapeutics require weeks to months of treatment prior to the onset of clinical efficacy on depressed mood but remain ineffective in treating suicidal ideation and cognitive impairment. Moreover, 30%-40% of individuals fail to respond to currently available antidepressant medications. MDD is a heterogeneous disorder with an unknown etiology; novel strategies must be developed to treat MDD more effectively. Emerging evidence suggests that targeting one or more of the four opioid receptors-mu (MOR), kappa (KOR), delta (DOR), and the nociceptin/orphanin FQ receptor (NOP)-may yield effective therapeutics for stress-related psychiatric disorders. Furthermore, the effects of the rapidly acting antidepressant ketamine may involve opioid receptors. This review highlights dysregulated opioid signaling in depression, evaluates clinical trials with opioid compounds, and considers the role of opioid mechanisms in rapidly acting antidepressants.
Collapse
|
6
|
Du Y, Wei J, Yang X, Dou Y, Zhao L, Qi X, Yu X, Guo W, Wang Q, Deng W, Li M, Lin D, Li T, Ma X. Plasma metabolites were associated with spatial working memory in major depressive disorder. Medicine (Baltimore) 2021; 100:e24581. [PMID: 33663067 PMCID: PMC7909221 DOI: 10.1097/md.0000000000024581] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 01/14/2021] [Indexed: 02/05/2023] Open
Abstract
Major depressive disorder (MDD) is a common disease with both affective and cognitive disorders. Alterations in metabolic systems of MDD patients have been reported, but the underlying mechanisms still remains unclear. We sought to identify abnormal metabolites in MDD by metabolomics and to explore the association between differential metabolites and neurocognitive dysfunction.Plasma samples from 53 MDD patients and 83 sex-, gender-, BMI-matched healthy controls (HCs) were collected. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) system was then used to detect metabolites in those samples. Two different algorithms were applied to identify differential metabolites in 2 groups. Of the 136 participants, 35 MDD patients and 48 HCs had completed spatial working memory test. Spearman rank correlation coefficient was applied to explore the relationship between differential metabolites and working memory in these 2 groups.The top 5 metabolites which were found in sparse partial least squares-discriminant analysis (sPLS-DA) model and random forest (RF) model were the same, and significant difference was found in 3 metabolites between MDD and HCs, namely, gamma-glutamyl leucine, leucine-enkephalin, and valeric acid. In addition, MDD patients had higher scores in spatial working memory (SWM) between errors and total errors than HCs. Valeric acid was positively correlated with working memory in MDD group.Gamma-glutamyl leucine, leucine-enkephalin, and valeric acid were preliminarily proven to be decreased in MDD patients. In addition, MDD patients performed worse in working memory than HCs. Dysfunction in working memory of MDD individuals was associated with valeric acid.
Collapse
Affiliation(s)
- Yue Du
- Psychiatric Laboratory and Mental Health Center
| | - Jinxue Wei
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| | - Xiao Yang
- Psychiatric Laboratory and Mental Health Center
| | - Yikai Dou
- Psychiatric Laboratory and Mental Health Center
| | - Liansheng Zhao
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| | - Xueyu Qi
- Psychiatric Laboratory and Mental Health Center
| | - Xueli Yu
- Psychiatric Laboratory and Mental Health Center
| | - Wanjun Guo
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| | - Qiang Wang
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| | - Wei Deng
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| | - Minli Li
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| | - Dongtao Lin
- College of Foreign Languages and Cultures, Sichuan University, PR China
| | - Tao Li
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| | - Xiaohong Ma
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| |
Collapse
|
7
|
Electroacupuncture Ameliorates Cerebral I/R-Induced Inflammation through DOR-BDNF/TrkB Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3495836. [PMID: 32256638 PMCID: PMC7102411 DOI: 10.1155/2020/3495836] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 02/17/2020] [Indexed: 12/01/2022]
Abstract
The beneficial effects of electroacupuncture (EA) at Shuigou (GV26) and Neiguan (PC6) on poststroke rehabilitation are critically related to the activation of the delta-opioid receptor (DOR). The underlying anti-inflammatory mechanisms in DOR activation and EA-mediated neuroprotection in cerebral ischemia/reperfusion (I/R) injury were investigated in the current study. Cell proliferation and apoptosis were detected by morphological changes, cell counting kit-8 (CCK-8) assay, lactate dehydrogenase (LDH) release, and TUNEL staining. The mRNA levels were evaluated by using real-time quantitative polymerase chain reaction (RT-qPCR), and the protein expression was measured by western blot or enzyme-linked immunosorbent assay (ELISA) in vitro. Infarct volume was examined by cresyl violet (CV) staining, neurologic recovery was assessed by neurological deficit scores, and pro- and anti-inflammatory cytokines were determined by immunofluorescence in vivo. DOR activation greatly ameliorated morphological injury, reduced LDH leakage and apoptosis, and increased cell viability. It reversed the oxygen-glucose deprivation/reoxygenation- (OGD/R-) induced downregulation of DOR mRNA and protein, as well as BDNF protein. DOR activation also reduced proinflammatory cytokine gene expression, including TNF-α, IL-1β, and IL-6, and at the same time, increased anti-inflammatory cytokines IL-4 and IL-10 in OGD/R challenged PC12 cells. EA significantly reduced middle cerebral artery occlusion/reperfusion- (MCAO/R-) induced infarct volume and attenuated neurologic deficit scores. It markedly increased the expression of IL-10 and decreased IL-1β, while sham EA did not have any protective effect in MCAO/R-injured rats. DOR activation plays an important role in neuroprotection against OGD/R injury by inhibiting inflammation via the brain-derived neurotrophic factor/tropomyosin-related kinase B (BDNF/TrkB) pathway. The neuroprotective efficacy of EA at Shuigou (GV26) and Neiguan (PC6) on cerebral I/R injury may be also related to the inhibition of inflammatory response through the DOR-BDNF/TrkB pathway.
Collapse
|
8
|
Puryear CB, Brooks J, Tan L, Smith K, Li Y, Cunningham J, Todtenkopf MS, Dean RL, Sanchez C. Opioid receptor modulation of neural circuits in depression: What can be learned from preclinical data? Neurosci Biobehav Rev 2020; 108:658-678. [DOI: 10.1016/j.neubiorev.2019.12.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/02/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
|
9
|
Kibaly C, Xu C, Cahill CM, Evans CJ, Law PY. Non-nociceptive roles of opioids in the CNS: opioids' effects on neurogenesis, learning, memory and affect. Nat Rev Neurosci 2019; 20:5-18. [PMID: 30518959 DOI: 10.1038/s41583-018-0092-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Mortality due to opioid use has grown to the point where, for the first time in history, opioid-related deaths exceed those caused by car accidents in many states in the United States. Changes in the prescribing of opioids for pain and the illicit use of fentanyl (and derivatives) have contributed to the current epidemic. Less known is the impact of opioids on hippocampal neurogenesis, the functional manipulation of which may improve the deleterious effects of opioid use. We provide new insights into how the dysregulation of neurogenesis by opioids can modify learning and affect, mood and emotions, processes that have been well accepted to motivate addictive behaviours.
Collapse
Affiliation(s)
- Cherkaouia Kibaly
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA.
| | - Chi Xu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Catherine M Cahill
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA
| | - Christopher J Evans
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA
| | - Ping-Yee Law
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA
| |
Collapse
|
10
|
Turner CA, Hagenauer MH, Aurbach EL, Maras PM, Fournier CL, Blandino P, Chauhan RB, Panksepp J, Watson SJ, Akil H. Effects of early-life FGF2 on ultrasonic vocalizations (USVs) and the mu-opioid receptor in male Sprague-Dawley rats selectively-bred for differences in their response to novelty. Brain Res 2019; 1715:106-114. [PMID: 30880118 DOI: 10.1016/j.brainres.2019.03.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 01/03/2023]
Abstract
In previous studies, early-life fibroblast growth factor-2 (FGF2) administration conferred resilience to developing anxiety-like behavior in vulnerable animals in adulthood. To follow up on this work, we administered FGF2 the day after birth to animals that differ in emotional behavior and further explored its long-term effects on affective behavior and circuitry. Selectively-bred "high responder" rats (bHRs) exhibit low levels of anxiety-like and depression-like behavior, whereas selectively-bred "low responders" (bLRs) display high levels of anxiety-like and depression-like behavior. We found that early-life administration of FGF2 decreased negative affect in bLRs during the early post-natal period, as indexed by 40 kHz ultrasonic vocalizations (USVs) in response to a brief maternal separation on PND11. FGF2 also increased positive affect during the juvenile period, as measured by 50 kHz USVs in response to heterospecific hand play ("tickling") after weaning. In general, we found that bHRs produced more 50 kHz USVs than bLRs. In adulthood, we measured opioid ligand and receptor expression in brain regions implicated in USV production and affect regulation by mRNA in situ hybridization. Within multiple affective brain regions, bHRs had greater expression of the mu opioid receptor than bLRs. FGF2 increased mu opioid expression in bLRs. The bLRs had more kappa and less delta receptor expression than bHRs, and FGF2 increased prodynorphin in bLRs. Our results provide support for further investigations into the role of growth factors and endogenous opioids in the treatment of disorders characterized by altered affect, such as anxiety and depression.
Collapse
Affiliation(s)
- Cortney A Turner
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA.
| | - Megan H Hagenauer
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Elyse L Aurbach
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Pamela M Maras
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Chelsea L Fournier
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Peter Blandino
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Rikav B Chauhan
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jaak Panksepp
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA, USA
| | - Stanley J Watson
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA; Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Huda Akil
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA; Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
11
|
Peciña M, Karp JF, Mathew S, Todtenkopf MS, Ehrich EW, Zubieta JK. Endogenous opioid system dysregulation in depression: implications for new therapeutic approaches. Mol Psychiatry 2019; 24:576-587. [PMID: 29955162 PMCID: PMC6310672 DOI: 10.1038/s41380-018-0117-2] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/11/2018] [Accepted: 05/25/2018] [Indexed: 12/12/2022]
Abstract
The United States is in the midst of an opioid addiction and overdose crisis precipitated and exacerbated by use of prescription opioid medicines. The majority of opioid prescriptions are dispensed to patients with comorbid mood disorders including major depressive disorder (MDD). A growing body of research indicates that the endogenous opioid system is directly involved in the regulation of mood and is dysregulated in MDD. This involvement of the endogenous opioid system may underlie the disproportionate use of opioids among patients with mood disorders. Emerging approaches to address endogenous opioid dysregulation in MDD may yield novel therapeutics that have a low or absent risk of abuse and addiction relative to µ-opioid agonists. Moreover, agents targeting the endogenous opioid system would be expected to yield clinical benefits qualitatively different from conventional monaminergic antidepressants. The development of safe and effective agents to treat MDD-associated endogenous opioid dysregulation may represent a distinct and currently underappreciated means of addressing treatment resistant depression with the potential to attenuate the on-going opioid crisis.
Collapse
Affiliation(s)
- Marta Peciña
- 0000 0004 1936 9000grid.21925.3dDepartment of Psychiatry, University of Pittsburgh, Pittsburgh, PA USA
| | - Jordan F. Karp
- 0000 0004 1936 9000grid.21925.3dDepartment of Psychiatry, University of Pittsburgh, Pittsburgh, PA USA
| | - Sanjay Mathew
- 0000 0001 2160 926Xgrid.39382.33Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX USA
| | | | | | - Jon-Kar Zubieta
- Department of Psychiatry, University of Utah Health Sciences Center, Salt Lake City, UT, USA.
| |
Collapse
|
12
|
Gendron L, Cahill CM, von Zastrow M, Schiller PW, Pineyro G. Molecular Pharmacology of δ-Opioid Receptors. Pharmacol Rev 2017; 68:631-700. [PMID: 27343248 DOI: 10.1124/pr.114.008979] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Opioids are among the most effective analgesics available and are the first choice in the treatment of acute severe pain. However, partial efficacy, a tendency to produce tolerance, and a host of ill-tolerated side effects make clinically available opioids less effective in the management of chronic pain syndromes. Given that most therapeutic opioids produce their actions via µ-opioid receptors (MOPrs), other targets are constantly being explored, among which δ-opioid receptors (DOPrs) are being increasingly considered as promising alternatives. This review addresses DOPrs from the perspective of cellular and molecular determinants of their pharmacological diversity. Thus, DOPr ligands are examined in terms of structural and functional variety, DOPrs' capacity to engage a multiplicity of canonical and noncanonical G protein-dependent responses is surveyed, and evidence supporting ligand-specific signaling and regulation is analyzed. Pharmacological DOPr subtypes are examined in light of the ability of DOPr to organize into multimeric arrays and to adopt multiple active conformations as well as differences in ligand kinetics. Current knowledge on DOPr targeting to the membrane is examined as a means of understanding how these receptors are especially active in chronic pain management. Insight into cellular and molecular mechanisms of pharmacological diversity should guide the rational design of more effective, longer-lasting, and better-tolerated opioid analgesics for chronic pain management.
Collapse
Affiliation(s)
- Louis Gendron
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Catherine M Cahill
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Mark von Zastrow
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Peter W Schiller
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Graciela Pineyro
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| |
Collapse
|
13
|
Abstract
Depression is a pervasive and debilitating mental disorder that is inadequately treated by current pharmacotherapies in a majority of patients. Although opioids have long been known to regulate mood states, the use of opioids to treat depression is rarely discussed. This chapter explores the preclinical and clinical evidence supporting the antidepressant-like effects of opioid ligands, and in particular, delta opioid receptor (DOR) agonists. DOR agonists have been shown to produce antidepressant-like effects in a number of animal models. Some DOR agonists also produce convulsions which has limited their clinical utility. However, DOR agonists that generate antidepressant-like effects without convulsions have recently been developed and these drugs are beginning to be evaluated in humans. Work investigating potential mechanisms of action for the antidepressant-like effects of DOR agonists is also explored. Understanding mechanisms that give rise to DOR-mediated behaviors is critical for the development of DOR drugs with improved safety and clinical utility, and future work should be devoted to elucidating these pathways.
Collapse
|
14
|
Hoffman JR, Ostfeld I, Kaplan Z, Zohar J, Cohen H. Exercise Enhances the Behavioral Responses to Acute Stress in an Animal Model of PTSD. Med Sci Sports Exerc 2016; 47:2043-52. [PMID: 25699481 DOI: 10.1249/mss.0000000000000642] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION This study examined the effects of endurance exercise on the behavioral response to stress and patterns of brain-derived neurotrophic factor (BDNF), neuropeptide Y (NPY), and δ-opioid receptor (phospho-DOR) expression in the hippocampus. METHODS Animals ran on a treadmill at 15 m·min, 5 min·d gradually increasing to 20 min·d, 5 d·wk for 6 wk. After training, one group of animals was exposed to a predator scent stress (PSS) protocol for 10 min. Outcome measurements included behavior in an elevated plus-maze (EPM) and acoustic startle response (ASR) 7 d after exposure to stress. Immunohistochemical technique was used to detect the expression of the BDNF, NPY, and phospho-DOR in the hippocampus 8 d after exposure. RESULTS Sedentary animals exposed to PSS were observed to have a greater incidence of extreme behavior responses including higher anxiety, less total activity in the EPM, and greater amplitude in the ASR than unexposed and/or trained animals. Exercise-trained animals exposed to PSS developed a resiliency to the stress, reflected by significantly greater total activity in the EPM, reduced anxiety, and reduced ASR compared to the sedentary, exposed animals. Exercise in the absence of stress significantly elevated the expression of BDNF and phospho-DOR, whereas exposure to PSS resulted in a significant decline in the expression of NPY, BDNF, and phospho-DOR. Trained animals that were exposed maintained expression of BDNF, NPY, and phospho-DOR in most subregions of the hippocampus. CONCLUSION Results indicated that endurance training provided a mechanism to promote resilience and/or recovery from stress. In addition, exercise increased expression of BDNF, NPY, and DOR signaling in the hippocampus that was associated with the greater resiliency seen in the trained animals.
Collapse
Affiliation(s)
- Jay R Hoffman
- 1Institute of Exercise Physiology and Wellness, Sport and Exercise Science, University of Central Florida, Orlando, FL; 2Israel Defense Force, Medical Corps, Tel Hashomer, ISRAEL; 3Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Faculty of Health Sciences, Division of Psychiatry, Ben-Gurion University of the Negev, Beer-Sheva, ISRAEL; and 4The State of Israel Ministry of Health, The Chaim Sheba Medical Center, Division of Psychiatry, Ramat-Gan, Israel, Sackler Medical School, Tel-Aviv University, ISRAEL
| | | | | | | | | |
Collapse
|
15
|
Manjoch H, Vainer E, Matar M, Ifergane G, Zohar J, Kaplan Z, Cohen H. Predator-scent stress, ethanol consumption and the opioid system in an animal model of PTSD. Behav Brain Res 2016; 306:91-105. [PMID: 26965572 DOI: 10.1016/j.bbr.2016.03.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/02/2016] [Accepted: 03/04/2016] [Indexed: 12/20/2022]
Abstract
Emerging literature points to stress exposure as a potential contributor to the development of alcohol abuse, but animal models have yielded inconsistent results. Converging experimental data indicate that the endogenous opioid system modulates alcohol consumption and stress regulation. The aim of the present study is to examine the interplay between stress exposure, behavioral stress responses, ethanol (EtOH) consumption and the endogenous opioid system in an animal model of posttraumatic stress disorder. Rats were exposed to stress and then tested in a two-bottle free choice (TBC) assay or in a conditioned place preference paradigm. In some experiments, the endogenous opioid system was pharmacologically manipulated prior to stress exposure. The behavioral outcomes of stress exposure were assessed in an elevated plus-maze, with the acoustic startle response, and by monitoring the freezing response to trauma reminder. Immunoreactivity of phosphorylated opioid receptors in hippocampal subregions was also measured. Stress significantly increased the consumption of EtOH in the TBC assay. The severity of the behavioral response to stress was associated with EtOH consumption, cue-triggered freezing response to a trauma reminder, and endogenous levels of phosphorylated opioid receptors in the hippocampus. Pharmacologically manipulating the endogenous opioid system prior to stress exposure attenuated trauma cue-triggered freezing responses and blocked predator scent stress-induced potentiation of EtOH consumption. These data demonstrate a stress-induced potentiation of EtOH self-administration and reveal a clear association between individual patterns of the behavioral response to stress and alcohol preference, while indicating a role for the endogenous opioid system in the neurobiological response to stress.
Collapse
Affiliation(s)
- Hadar Manjoch
- Ben-Gurion University of the Negev, Department of Psychology, Beer Sheva, Israel; Beer-Sheva Mental Health Center, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, Israel
| | - Ella Vainer
- Beer-Sheva Mental Health Center, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, Israel
| | - Michael Matar
- Beer-Sheva Mental Health Center, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, Israel
| | - Gal Ifergane
- Headache Clinic, Department of Neurology, Soroka Medical Center, Ben-Gurion University of the Negev Beer- Sheva, Israel
| | - Joseph Zohar
- Division of Psychiatry, The State of Israel Ministry of Health, The Chaim Sheba Medical Center, Sackler Medical School, Tel-Aviv University, Tel Hashomer, Israel
| | - Zeev Kaplan
- Beer-Sheva Mental Health Center, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, Israel
| | - Hagit Cohen
- Ben-Gurion University of the Negev, Department of Psychology, Beer Sheva, Israel; Beer-Sheva Mental Health Center, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, Israel.
| |
Collapse
|
16
|
Richards EM, Mathews DC, Luckenbaugh DA, Ionescu DF, Machado-Vieira R, Niciu MJ, Duncan WC, Nolan NM, Franco-Chaves JA, Hudzik T, Maciag C, Li S, Cross A, Smith MA, Zarate CA. A randomized, placebo-controlled pilot trial of the delta opioid receptor agonist AZD2327 in anxious depression. Psychopharmacology (Berl) 2016; 233:1119-30. [PMID: 26728893 PMCID: PMC5103283 DOI: 10.1007/s00213-015-4195-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 12/14/2015] [Indexed: 12/20/2022]
Abstract
RATIONALE Patients with anxious major depressive disorder (AMDD) have more severe symptoms and poorer treatment response than patients with non-AMDD. Increasing evidence implicates the endogenous opioid system in the pathophysiology of depression. AZD2327 is a selective delta opioid receptor (DOR) agonist with anxiolytic and antidepressant activity in animal models. OBJECTIVE This double-blind, parallel group design, placebo-controlled pilot study evaluated the safety and efficacy of AZD2327 in a preclinical model and in patients with AMDD. METHODS We initially tested the effects of AZD2327 in an animal model of AMDD. Subsequently, 22 subjects with AMDD were randomized to receive AZD2327 (3 mg BID) or placebo for 4 weeks. Primary outcome measures included the Hamilton Depression Rating Scale (HAM-D) and the Hamilton Anxiety Rating Scale (HAM-A). We also evaluated neurobiological markers implicated in mood and anxiety disorders, including vascular endothelial growth factor (VEGF) and electroencephalogram (EEG). RESULTS Seven (54 %) patients responded to active drug and three (33 %) responded to placebo. No significant main drug effect was found on either the HAM-D (p = 0.39) or the HAM-A (p = 0.15), but the HAM-A had a larger effect size. Levels of AZ12311418, a major metabolite of AZD2327, were higher in patients with an anti-anxiety response to treatment compared to nonresponders (p = 0.03). AZD2327 treatment decreased VEGF levels (p = 0.02). There was a trend (p < 0.06) for those with an anti-anxiety response to have higher EEG gamma power than nonresponders. CONCLUSION These results suggest that AZD2327 has larger potential anxiolytic than antidepressant efficacy. Additional research with DOR agonists should be considered.
Collapse
Affiliation(s)
- Erica M Richards
- Experimental Therapeutics and Pathophysiology Branch, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, 10 Center Drive CRC, Room 7-5545, Bethesda, MD, 20892, USA.
| | - Daniel C Mathews
- Experimental Therapeutics and Pathophysiology Branch, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, 10 Center Drive CRC, Room 7-5545, Bethesda, MD, 20892, USA
- Lundbeck LLC, Chicago, IL, USA
| | - David A Luckenbaugh
- Experimental Therapeutics and Pathophysiology Branch, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, 10 Center Drive CRC, Room 7-5545, Bethesda, MD, 20892, USA
| | - Dawn F Ionescu
- Experimental Therapeutics and Pathophysiology Branch, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, 10 Center Drive CRC, Room 7-5545, Bethesda, MD, 20892, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Rodrigo Machado-Vieira
- Experimental Therapeutics and Pathophysiology Branch, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, 10 Center Drive CRC, Room 7-5545, Bethesda, MD, 20892, USA
| | - Mark J Niciu
- Experimental Therapeutics and Pathophysiology Branch, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, 10 Center Drive CRC, Room 7-5545, Bethesda, MD, 20892, USA
| | - Wallace C Duncan
- Experimental Therapeutics and Pathophysiology Branch, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, 10 Center Drive CRC, Room 7-5545, Bethesda, MD, 20892, USA
| | - Neal M Nolan
- Experimental Therapeutics and Pathophysiology Branch, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, 10 Center Drive CRC, Room 7-5545, Bethesda, MD, 20892, USA
| | - Jose A Franco-Chaves
- Experimental Therapeutics and Pathophysiology Branch, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, 10 Center Drive CRC, Room 7-5545, Bethesda, MD, 20892, USA
- Veteran Affairs Caribbean Healthcare System, San Juan, Puerto Rico
| | - Thomas Hudzik
- AstraZeneca Neuroscience Innovative Medicines, Cambridge, MA, USA
- AbbVie, Chicago, IL, USA
| | - Carla Maciag
- AstraZeneca Neuroscience Innovative Medicines, Cambridge, MA, USA
- Sage Therapeutics, Cambridge, MA, USA
| | - Shuang Li
- AstraZeneca Neuroscience Innovative Medicines, Cambridge, MA, USA
| | - Alan Cross
- AstraZeneca Neuroscience Innovative Medicines, Cambridge, MA, USA
| | - Mark A Smith
- AstraZeneca Neuroscience Innovative Medicines, Cambridge, MA, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, 10 Center Drive CRC, Room 7-5545, Bethesda, MD, 20892, USA
| |
Collapse
|
17
|
Peppin JF, Raffa RB. Delta opioid agonists: a concise update on potential therapeutic applications. J Clin Pharm Ther 2015; 40:155-66. [PMID: 25726896 DOI: 10.1111/jcpt.12244] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/24/2014] [Indexed: 01/23/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE The endogenous opioid system co-evolved with chemical defences, or at times symbiotic relationships, between plants and other autotrophs and heterotrophic predators - thus, it is not surprising that endogenous opioid ligands and exogenous mimetic ligands produce diverse physiological effects. Among the endogenous opioid peptides (endomorphins, enkephalins, dynorphins and nociception/orphanin FQ) derived from the precursors encoded by four genes (PNOC, PENK, PDYN and POMC) are the pentapeptides Met-enkephalin (Tyr-Gly-Gly-Phe-Met) and Leu-enkephalin (Tyr-Gly-Gly-Phe-Leu). The physiological effects of the enkephalins are mediated via 7-transmembrane G protein-coupled receptors, including delta opioid receptor (DOR). We present a concise update on the status of progress and opportunities of this approach. METHODS A literature search of the PUBMED database and a combination of keywords including delta opioid receptor, analgesia, mood and individual compounds identified therein, from industry and other source, and from www.clinicaltrials.com. RESULTS AND DISCUSSION DOR agonist and antagonist ligands have been developed with ever increasing affinity and selectivity for DOR over other opioid receptor subtypes and studied for therapeutic utility, primarily for pain relief, but also for other clinical endpoints. WHAT IS NEW AND CONCLUSION Selective DOR agonists have been designed with a large increase in therapeutic window for a variety of potential CNS applications including pain, depression, and learning and memory among others.
Collapse
Affiliation(s)
- J F Peppin
- Center for Bioethics, Pain Management and Medicine, University City, MO, USA; Mallinckrodt Pharmaceuticals, Hazelwood, MO, USA
| | | |
Collapse
|
18
|
Szklarczyk K, Korostynski M, Cieslak PE, Wawrzczak-Bargiela A, Przewlocki R. Opioid-dependent regulation of high and low fear responses in two inbred mouse strains. Behav Brain Res 2015; 292:95-101. [PMID: 26051817 DOI: 10.1016/j.bbr.2015.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 05/18/2015] [Accepted: 06/01/2015] [Indexed: 01/16/2023]
Abstract
The molecular mechanisms underlying the susceptibility or resilience to trauma-related disorders remain incompletely understood. Opioids modulate emotional learning, but the roles of specific receptors are unclear. Here, we aimed to analyze the contribution of the opioid system to fear responses in two inbred mouse strains exhibiting distinct behavioral phenotypes. SWR/J and C57BL/6J mice were subjected to five consecutive electric footshocks (1mA each), and the contextual freezing time was measured. Stress-induced alterations in gene expression were analyzed in the amygdala and the hippocampus. In both strains, the fear response was modulated using pharmacological tools. SWR/J mice did not develop conditioned fear but exhibited increased transcriptional expression of Pdyn and Penk in the amygdala region. Blocking opioid receptors prior to the footshocks using naltrexone (2 mg/kg) or naltrindole (5 mg/kg) increased the freezing responses in these animals. The C57BL/6J strain displayed high conditioned fear, although no alteration in the mRNA abundance of genes encoding opioid precursors was observed. Double-injection of morphine (20 mg/kg) following stress and upon context re-exposure prevented the enhancement of freezing. Moreover, selective delta and kappa agonists caused a reduction in conditioned fear responses. To summarize, the increased expression of the Pdyn and Penk genes corresponded to reduced intensity of fear responses. Blockade of the endogenous opioid system restored freezing behavior in stress-resistant animals. The pharmacological stimulation of the kappa and delta opioid receptors in stress-susceptible individuals may alleviate fear. Thus, subtype-selective opioid receptor agonists may protect against the development of trauma-related disorders.
Collapse
Affiliation(s)
- Klaudia Szklarczyk
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Krakow, Poland
| | - Michal Korostynski
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Krakow, Poland
| | - Przemyslaw Eligiusz Cieslak
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Krakow, Poland; Institute of Applied Psychology, Jagiellonian University, Krakow, Poland
| | | | - Ryszard Przewlocki
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Krakow, Poland.
| |
Collapse
|
19
|
Rosenblat C, McIntyre RS, Alves GS, Fountoulakis KN, Carvalho AF. Beyond Monoamines-Novel Targets for Treatment-Resistant Depression: A Comprehensive Review. Curr Neuropharmacol 2015; 13:636-55. [PMID: 26467412 PMCID: PMC4761634 DOI: 10.2174/1570159x13666150630175044] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/06/2014] [Accepted: 11/20/2014] [Indexed: 12/12/2022] Open
Abstract
Major depressive disorder (MDD) is a leading cause of disability worldwide. Current first line therapies target modulation of the monoamine system. A large variety of agents are currently available that effectively alter monoamine levels; however, approximately one third of MDD patients remain treatment refractory after adequate trials of multiple monoamine based therapies. Therefore, patients with treatment-resistant depression (TRD) may require modulation of pathways outside of the classic monoamine system. The purpose of this review was thus to discuss novel targets for TRD, to describe their potential mechanisms of action, the available clinical evidence for these targets, the limitations of available evidence as well as future research directions. Several alternate pathways involved in the patho-etiology of TRD have been uncovered including the following: inflammatory pathways, the oxidative stress pathway, the hypothalamic-pituitary-adrenal (HPA) axis, the metabolic and bioenergetics system, neurotrophic pathways, the glutamate system, the opioid system and the cholinergic system. For each of these systems, several targets have been assessed in preclinical and clinical models. Preclinical models strongly implicate these pathways in the patho-etiology of MDD. Clinical trials for TRD have been conducted for several novel targets; however, most of the trials discussed are small and several are uncontrolled. Therefore, further clinical trials are required to assess the true efficacy of these targets for TRD. As well, several promising novel agents have been clinically tested in MDD populations, but have yet to be assessed specifically for TRD. Thus, their applicability to TRD remains unknown.
Collapse
Affiliation(s)
- Christian Rosenblat
- Mood Disorder Psychopharmacology Unit, University Health Network, Department of Psychiatry,
University of Toronto, ON, Canada
| | - Roger S. McIntyre
- Mood Disorder Psychopharmacology Unit, University Health Network, Department of Psychiatry,
University of Toronto, ON, Canada
- Departments of Psychiatry and Pharmacology, University of
Toronto, Toronto, ON, Canada
| | - Gilberto S. Alves
- Translational Psychiatry Research Group and Department of
Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - André F. Carvalho
- Translational Psychiatry Research Group and Department of
Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
20
|
Steiner JP, Nath A. Neurotrophin strategies for neuroprotection: are they sufficient? J Neuroimmune Pharmacol 2014; 9:182-94. [PMID: 24609976 DOI: 10.1007/s11481-014-9533-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 02/13/2014] [Indexed: 12/30/2022]
Abstract
As people are living longer, the prevalance of neurodegenerative diseases continues to rise resulting in huge socio-economic consequences. Despite major advancements in studying the pathophysiology of these diseases and a large number of clinical trials currently there is no effective treatment for these illnesses. All neuroprotective strategies have either failed or have shown only a minimal effect. There has been a major shift in recent years exploring the potential of neuroregenerative approaches. While the concept of using neurotropins for therapeutic purposes has been in existence for many years, new modes of delivery and expression of this family of molecules makes this approach now feasilble. Further neurotropin mimetics and receptor agonists are also being developed. The use of small molecules to induce the expression of neurotropins including repurposing of FDA approved drugs for this approach is another strategy being pursued. In the review we examine these new developments and discuss the potential for such approaches in the context of the pathophysiology of neurodegenerative diseases.
Collapse
Affiliation(s)
- Joseph P Steiner
- NINDS Translational Neuroscience Center, National Institutes of Health, Room 7C-105; Bldg 10, 10 Center Drive, Bethesda, MD, 20892, USA,
| | | |
Collapse
|
21
|
Sen D, Huchital M, Chen YL. Crosstalk between delta opioid receptor and nerve growth factor signaling modulates neuroprotection and differentiation in rodent cell models. Int J Mol Sci 2013; 14:21114-39. [PMID: 24152443 PMCID: PMC3821661 DOI: 10.3390/ijms141021114] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 09/16/2013] [Accepted: 09/26/2013] [Indexed: 01/19/2023] Open
Abstract
Both opioid signaling and neurotrophic factor signaling have played an important role in neuroprotection and differentiation in the nervous system. Little is known about whether the crosstalk between these two signaling pathways will affect neuroprotection and differentiation. Previously, we found that nerve growth factor (NGF) could induce expression of the delta opioid receptor gene (Oprd1, dor), mainly through PI3K/Akt/NF-κB signaling in PC12h cells. In this study, using two NGF-responsive rodent cell model systems, PC12h cells and F11 cells, we found the delta opioid neuropeptide [d-Ala2, d-Leu5] enkephalin (DADLE)-mediated neuroprotective effect could be blocked by pharmacological reagents: the delta opioid antagonist naltrindole, PI3K inhibitor LY294002, MAPK inhibitor PD98059, and Trk inhibitor K252a, respectively. Western blot analysis revealed that DADLE activated both the PI3K/Akt and MAPK pathways in the two cell lines. siRNA Oprd1 gene knockdown experiment showed that the upregulation of NGF mRNA level was inhibited with concomitant inhibition of the survival effects of DADLE in the both cell models. siRNA Oprd1 gene knockdown also attenuated the DADLE-mediated neurite outgrowth in PC12h cells as well as phosphorylation of MAPK and Akt in PC12h and F11 cells, respectively. These data together strongly suggest that delta opioid peptide DADLE acts through the NGF-induced functional G protein-coupled Oprd1 to provide its neuroprotective and differentiating effects at least in part by regulating survival and differentiating MAPK and PI3K/Akt signaling pathways in NGF-responsive rodent neuronal cells.
Collapse
Affiliation(s)
- Dwaipayan Sen
- Department of Biological Sciences, Binghamton University, the State University of New York at Binghamton, Binghamton, NY 13902, USA; E-Mails: (D.S.); (M.H.)
| | - Michael Huchital
- Department of Biological Sciences, Binghamton University, the State University of New York at Binghamton, Binghamton, NY 13902, USA; E-Mails: (D.S.); (M.H.)
| | - Yulong L. Chen
- Department of Biological Sciences, Binghamton University, the State University of New York at Binghamton, Binghamton, NY 13902, USA; E-Mails: (D.S.); (M.H.)
- The Center for Development and Behavioral Neurosciences, Binghamton University, the State University of New York at Binghamton, Binghamton, NY 13902, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-607-777-5218; Fax: +1-607-777-6521
| |
Collapse
|
22
|
Tian X, Hua F, Sandhu HK, Chao D, Balboni G, Salvadori S, He X, Xia Y. Effect of δ-opioid receptor activation on BDNF-TrkB vs. TNF-α in the mouse cortex exposed to prolonged hypoxia. Int J Mol Sci 2013; 14:15959-76. [PMID: 23912236 PMCID: PMC3759895 DOI: 10.3390/ijms140815959] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 07/25/2013] [Accepted: 07/25/2013] [Indexed: 12/15/2022] Open
Abstract
We investigated whether δ-opioid receptor (DOR)-induced neuroprotection involves the brain-derived neurotrophic factor (BDNF) pathway. We studied the effect of DOR activation on the expression of BDNF and other proteins in the cortex of C57BL/6 mice exposed to hypoxia (10% of oxygen) for 1–10 days. The results showed that: (1) 1-day hypoxia had no appreciable effect on BDNF expression, while 3- and 10-day hypoxia progressively decreased BDNF expression, resulting in 37.3% reduction (p < 0.05) after 10-day exposure; (2) DOR activation with UFP-512 (1 mg/kg, i.p., daily) partially reversed the hypoxia-induced reduction of BDNF expression in the 3- or 10-day exposed cortex; (3) DOR activation partially reversed the hypoxia-induced reduction in functional TrkB (140-kDa) and attenuated hypoxia-induced increase in truncated TrkB (90-kDa) in the 3- or 10-day hypoxic cortex; and (4) prolonged hypoxia (10 days) significantly increased TNF-α level and decreased CD11b expression in the cortex, which was completely reversed following DOR activation; and (5) there was no significant change in pCREB and pATF-1 levels in the hypoxic cortex. We conclude that prolonged hypoxia down-regulates BDNF-TrkB signaling leading to an increase in TNF-α in the cortex, while DOR activation up-regulates BDNF-TrkB signaling thereby decreasing TNF-α levels in the hypoxic cortex.
Collapse
Affiliation(s)
- Xuesong Tian
- The Vivan L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, TX 77030, USA; E-Mails: (X.T.); (H.K.S.); (D.C.)
| | - Fei Hua
- Research Institute of Modern Medicine, The Third Medical College of Soochow University, Changzhou, Jiangsu 213002, China; E-Mail:
| | - Harleen K Sandhu
- The Vivan L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, TX 77030, USA; E-Mails: (X.T.); (H.K.S.); (D.C.)
| | - Dongman Chao
- The Vivan L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, TX 77030, USA; E-Mails: (X.T.); (H.K.S.); (D.C.)
| | - Gianfranco Balboni
- Department of Life and Environment Sciences, University of Cagliari, Cagliari I-09124, Italy; E-Mail:
| | - Severo Salvadori
- Department of Pharmaceutical Sciences, University of Ferrara, Ferrara I-44100, Italy; E-Mail:
| | - Xiaozhou He
- Research Institute of Modern Medicine, The Third Medical College of Soochow University, Changzhou, Jiangsu 213002, China; E-Mail:
- Authors to whom correspondence should be addressed; E-Mails: (X.H.); (Y.X.); Tel./Fax: +1-713-500-6288 (Y.X.)
| | - Ying Xia
- The Vivan L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, TX 77030, USA; E-Mails: (X.T.); (H.K.S.); (D.C.)
- Authors to whom correspondence should be addressed; E-Mails: (X.H.); (Y.X.); Tel./Fax: +1-713-500-6288 (Y.X.)
| |
Collapse
|
23
|
Tian X, Guo J, Zhu M, Li M, Wu G, Xia Y. δ-Opioid receptor activation rescues the functional TrkB receptor and protects the brain from ischemia-reperfusion injury in the rat. PLoS One 2013; 8:e69252. [PMID: 23844255 PMCID: PMC3699518 DOI: 10.1371/journal.pone.0069252] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 06/13/2013] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVES δ-opioid receptor (DOR) activation reduced brain ischemic infarction and attenuated neurological deficits, while DOR inhibition aggravated the ischemic damage. The underlying mechanisms are, however, not well understood yet. In this work, we asked if DOR activation protects the brain against ischemic injury through a brain-derived neurotrophic factor (BDNF) -TrkB pathway. METHODS We exposed adult male Sprague-Dawley rats to focal cerebral ischemia, which was induced by middle cerebral artery occlusion (MCAO). DOR agonist TAN-67 (60 nmol), antagonist Naltrindole (100 nmol) or artificial cerebral spinal fluid was injected into the lateral cerebroventricle 30 min before MCAO. Besides the detection of ischemic injury, the expression of BDNF, full-length and truncated TrkB, total CREB, p-CREB, p-ATF and CD11b was detected by Western blot and fluorescence immunostaining. RESULTS DOR activation with TAN-67 significantly reduced the ischemic volume and largely reversed the decrease in full-length TrkB protein expression in the ischemic cortex and striatum without any appreciable change in cerebral blood flow, while the DOR antagonist Naltrindole aggregated the ischemic injury. However, the level of BDNF remained unchanged in the cortex, striatum and hippocampus at 24 hours after MCAO and did not change in response to DOR activation or inhibition. MCAO decreased both total CREB and pCREB in the striatum, but not in the cortex, while DOR inhibition promoted a further decrease in total and phosphorylated CREB in the striatum and decreased pATF-1 expression in the cortex. In addition, MCAO increased CD11b expression in the cortex, striatum and hippocampus, and DOR activation specifically attenuated the ischemic increase in the cortex but not in the striatum and hippocampus. CONCLUSIONS DOR activation rescues TrkB signaling by reversing ischemia/reperfusion induced decrease in the full-length TrkB receptor and reduces brain injury in ischemia/reperfusion.
Collapse
MESH Headings
- Activating Transcription Factor 1/metabolism
- Animals
- Blotting, Western
- Brain/blood supply
- Brain/drug effects
- Brain/metabolism
- Brain Ischemia/complications
- Brain Ischemia/metabolism
- Brain-Derived Neurotrophic Factor/metabolism
- CD11b Antigen/metabolism
- Cerebrovascular Circulation/drug effects
- Cyclic AMP Response Element-Binding Protein/metabolism
- Infarction, Middle Cerebral Artery/complications
- Infarction, Middle Cerebral Artery/metabolism
- Male
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Quinolines/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptor, trkB/metabolism
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/metabolism
- Reperfusion Injury/etiology
- Reperfusion Injury/metabolism
Collapse
Affiliation(s)
- Xuesong Tian
- State Key Laboratory of Medical Neurobiology, Department of Integrative Medicine and Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurosurgery, University of Texas Medical School at Houston, Houston, Texas, United States of America
| | - Jingchun Guo
- State Key Laboratory of Medical Neurobiology, Department of Integrative Medicine and Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China
- Laboratory of Molecular Neurology, Shanghai Research Center for Acupuncture and Meridians, Shanghai, China
- * E-mail: (JG) (YX)
| | - Min Zhu
- State Key Laboratory of Medical Neurobiology, Department of Integrative Medicine and Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Minwei Li
- State Key Laboratory of Medical Neurobiology, Department of Integrative Medicine and Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Gencheng Wu
- State Key Laboratory of Medical Neurobiology, Department of Integrative Medicine and Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Xia
- Department of Neurosurgery, University of Texas Medical School at Houston, Houston, Texas, United States of America
- * E-mail: (JG) (YX)
| |
Collapse
|
24
|
Burstein SR, Williams TJ, Lane DA, Knudsen MG, Pickel VM, McEwen BS, Waters EM, Milner TA. The influences of reproductive status and acute stress on the levels of phosphorylated delta opioid receptor immunoreactivity in rat hippocampus. Brain Res 2013; 1518:71-81. [PMID: 23583481 DOI: 10.1016/j.brainres.2013.03.051] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 03/07/2013] [Accepted: 03/31/2013] [Indexed: 12/20/2022]
Abstract
In the hippocampus, ovarian hormones and sex can alter the trafficking of delta opioid receptors (DORs) and the proportion of DORs that colocalize with the stress hormone, corticotropin releasing factor. Here, we assessed the effects of acute immobilization stress (AIS) and sex on the phosphorylation of DORs in the rat hippocampus. We first localized an antibody to phosphorylated DOR (pDOR) at the SER363 carboxy-terminal residue, and demonstrated its response to an opioid agonist. By light microscopy, pDOR-immunoreactivity (ir) was located predominantly in CA2/CA3a pyramidal cell apical dendrites and in interneurons in CA1-3 stratum oriens and the dentate hilus. By electron microscopy, pDOR-ir primarily was located in somata and dendrites, associated with endomembranes, or in dendritic spines. pDOR-ir was less frequently found in mossy fibers terminals. Quantitative light microscopy revealed a significant increase in pDOR-ir in the CA2/CA3a region of male rats 1h following an injection of the opioid agonist morphine (20mg/kg, I.P). To look at the effects of stress on pDOR, we compared pDOR-ir in males and cycling females after AIS. The level of pDOR-ir in stratum radiatum of CA2/CA3a was increased in control estrus (elevated estrogen and progesterone) females compared to proestrus and diestrus females and males. However, immediately following 30min of AIS, no significant differences in pDOR levels were seen across estrous cycle phase or sex. These findings suggest that hippocampal levels of phosphorylated DORs vary with estrous cycle phase and that acute stress may dampen the differential effects of hormones on DOR activation in females.
Collapse
Affiliation(s)
- Suzanne R Burstein
- Brain and Mind Research Institute, Weill Cornell Medical College, 407 East 61st Street, New York, NY 10065, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Saitoh A, Yamada M. Antidepressant-like Effects of δ Opioid Receptor Agonists in Animal Models. Curr Neuropharmacol 2013; 10:231-8. [PMID: 23449756 PMCID: PMC3468877 DOI: 10.2174/157015912803217314] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 03/09/2012] [Accepted: 04/05/2012] [Indexed: 12/28/2022] Open
Abstract
Recently, δ opioid receptor agonists have been proposed to be attractive targets for the development of novel antidepressants. Several studies revealed that single treatment of δ opioid receptor agonists produce antidepressant-like effects in the forced swimming test, which is one of the most popular animal models for screening antidepressants. In addition, subchronic treatment with δ opioid receptor agonists has been shown to completely attenuate the hyperemotional responses found in olfactory bulbectomized rats. This animal model exhibits hyperemotional behavior that may mimic the anxiety, aggression, and irritability found in depressed patients, suggesting that δ opioid receptor agonists could be effective in the treatment of these symptoms in depression. On the other hand, prototype δ opioid receptor agonists produce convulsive effects, which limit their therapeutic potential and clinical development. In this review, we presented the current knowledge regarding the antidepressant-like effects of δ opioid receptor agonists, which include some recently developed drugs lacking convulsive effects.
Collapse
Affiliation(s)
- Akiyoshi Saitoh
- Department of Neuropsychopharmacology, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo 187-8553, Japan
| | | |
Collapse
|
26
|
Lutz PE, Kieffer BL. Opioid receptors: distinct roles in mood disorders. Trends Neurosci 2012; 36:195-206. [PMID: 23219016 DOI: 10.1016/j.tins.2012.11.002] [Citation(s) in RCA: 353] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 09/20/2012] [Accepted: 11/05/2012] [Indexed: 12/20/2022]
Abstract
The roles of opioid receptors in pain and addiction have been extensively studied, but their function in mood disorders has received less attention. Accumulating evidence from animal research reveals that mu, delta and kappa opioid receptors (MORs, DORs and KORs, respectively) exert highly distinct controls over mood-related processes. DOR agonists and KOR antagonists have promising antidepressant potential, whereas the risk-benefit ratio of currently available MOR agonists as antidepressants remains difficult to evaluate, in addition to their inherent abuse liability. To date, both human and animal studies have mainly examined MORs in the etiology of depressive disorders, and future studies will address DOR and KOR function in established and emerging neurobiological aspects of depression, including neurogenesis, neurodevelopment, and social behaviors.
Collapse
Affiliation(s)
- Pierre-Eric Lutz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de Recherche Scientifique (CNRS), Strasbourg, France
| | | |
Collapse
|
27
|
Guo J, Gu C, Zhou D, Elmore CS, Bui KH, Grimm SW. In Vitro and In Vivo Metabolism of a Selective δ-Opioid Receptor. Drug Metab Dispos 2011; 39:1883-94. [DOI: 10.1124/dmd.111.040980] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
28
|
Rajkumar R, Mahesh R. Assessing the neuronal serotonergic target-based antidepressant stratagem: impact of in vivo interaction studies and knockout models. Curr Neuropharmacol 2010; 6:215-34. [PMID: 19506722 PMCID: PMC2687932 DOI: 10.2174/157015908785777256] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Revised: 06/05/2008] [Accepted: 06/12/2008] [Indexed: 12/26/2022] Open
Abstract
Depression remains a challenge in the field of affective neuroscience, despite a steady research progress. Six out of nine basic antidepressant mechanisms rely on serotonin neurotransmitter system. Preclinical studies have demonstrated the significance of serotonin receptors (5-HT1-3,6,7), its signal transduction pathways and classical down stream targets (including neurotrophins, neurokinins, other peptides and their receptors) in antidepressant drug action. Serotonergic control of depression embraces the recent molecular requirements such as influence on proliferation, neurogenesis, plasticity, synaptic (re)modeling and transmission in the central nervous system. The present progress report analyses the credibility of each protein as therapeutically relevant target of depression. In vivo interaction studies and knockout models which identified these targets are foreseen to unearth new ligands and help them transform to drug candidates. The importance of the antidepressant assay selection at the preclinical level using salient animal models/assay systems is discussed. Such test batteries would definitely provide antidepressants with faster onset, efficacy in resistant (and co-morbid) types and with least adverse effects. Apart from the selective ligands, only those molecules which bring an overall harmony, by virtue of their affinities to various receptor subtypes, could qualify as effective antidepressants. Synchronised modulation of various serotonergic sub-pathways is the basis for a unique and balanced antidepressant profile, as that of fluoxetine (most exploited antidepressant) and such a profile may be considered as a template for the upcoming antidepressants. In conclusion, 5-HT based multi-targeted antidepressant drug discovery supported by in vivo interaction studies and knockout models is advocated as a strategy to provide classic molecules for clinical trials.
Collapse
Affiliation(s)
- R Rajkumar
- Pharmacy Group, FD-III, Vidya Vihar, Birla Institute of Technology & Science, Pilani, Rajasthan-333031, India.
| | | |
Collapse
|
29
|
Lee HJ, Rao JS, Chang L, Rapoport SI, Kim HW. Chronic imipramine but not bupropion increases arachidonic acid signaling in rat brain: is this related to 'switching' in bipolar disorder? Mol Psychiatry 2010; 15:602-14. [PMID: 18982003 PMCID: PMC2874651 DOI: 10.1038/mp.2008.117] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Agents effective against mania in bipolar disorder are reported to decrease turnover of arachidonic acid (AA) in phospholipids and expression of calcium-dependent AA-selective cytosolic phospholipase A(2) (cPLA(2)) in rat brain. In contrast, fluoxetine, an antidepressant that is reported to switch bipolar depressed patients to mania, increases cPLA(2) expression and AA turnover in rat brain. We therefore hypothesized that antidepressants that increase switching to mania generally increase cPLA(2) and AA turnover in brain. To test this hypothesis, adult male CDF-344 rats were administered imipramine and bupropion, with reported high and low switching rates, respectively, at daily doses of 10 and 30 mg kg(-1) i.p., respectively, or i.p. saline (control) for 21 days. Frontal cortex expression of different PLA(2) enzymes and AA turnover rates in brain when the rats were unanesthetized were measured. Compared with chronic saline, chronic imipramine but not bupropion significantly increased cortex cPLA(2) mRNA activity, protein and phosphorylation, expression of the cPLA(2) transcription factor, activator protein-2alpha (AP-2alpha) and AA turnover in phospholipids. Protein levels of secretory phospholipase A(2), calcium-independent phospholipase A(2), cyclooxygenase (COX)-1 and COX-2 were unchanged, and prostaglandin E(2) was unaffected. These results, taken with prior data on chronic fluoxetine in rats, suggest that antidepressants that increase the switching tendency of bipolar depressed patients to mania do so by increasing AA recycling and metabolism in brain. Mania in bipolar disorder thus may involve upregulated brain AA metabolism.
Collapse
|
30
|
Eyvazzadeh AD, Pennington KP, Pop-Busui R, Sowers M, Zubieta JK, Smith YR. The role of the endogenous opioid system in polycystic ovary syndrome. Fertil Steril 2009; 92:1-12. [PMID: 19560572 DOI: 10.1016/j.fertnstert.2009.05.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Revised: 04/29/2009] [Accepted: 05/07/2009] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To review the complex role of the opioid system in reproduction and carbohydrate metabolism, abnormalities in the opioid system in women with polycystic ovary syndrome (PCOS), and the role of opioid antagonists in the management of PCOS-related infertility. DESIGN Pertinent articles were identified through a computer PubMed search. References of selected articles were hand searched for additional citations. CONCLUSION(S) Endogenous opioids are generally considered inhibitory central neurotransmitters. Peripherally, opioids are involved in the regulation of pancreatic islet function, hepatic insulin clearance, and glucose metabolism, potentially contributing to the pathogenesis of hyperinsulinemia and insulin resistance in PCOS. The presence of sex steroids is required for normal function of the opioid system in both GnRH secretion and carbohydrate metabolism. In women with PCOS, growing evidence suggests dysregulation of the opioid system both centrally and peripherally, with complex interactions. The opioid system effects on carbohydrate metabolism appear to be modulated by obesity. Finally, naltrexone has been demonstrated to successfully augment traditional ovulation induction regimens, but has limited support as a single ovulation induction agent for PCOS.
Collapse
Affiliation(s)
- Aimee D Eyvazzadeh
- Department of Obstetrics and Gynecology, School of Medicine and School of Public Health, University of Michigan, 1500 East Medical Center Drive, Women's Hospital, Ann Arbor, MI 48109-0276, USA
| | | | | | | | | | | |
Collapse
|
31
|
Zhang HN, Ko MC. Seizure activity involved in the up-regulation of BDNF mRNA expression by activation of central mu opioid receptors. Neuroscience 2009; 161:301-10. [PMID: 19303919 DOI: 10.1016/j.neuroscience.2009.03.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Revised: 03/07/2009] [Accepted: 03/10/2009] [Indexed: 01/02/2023]
Abstract
Chemical-induced seizures up-regulated brain-derived neurotrophic factor (BDNF) mRNA expression. Intracerebroventricular (i.c.v.) administration of endogenous opioids preferentially activating mu opioid receptor (MOR) could also increase BDNF mRNA expression. The aim of this study was to determine to what extent i.c.v. administration of synthetic MOR-selective agonists in rats can modulate both seizure activity and up-regulation of BDNF mRNA expression. Effects and potencies of i.c.v. administration of morphine and [D-Ala(2), N-Me-Phe(4), Gly(5)-ol]-enkephalin (DAMGO), were directly investigated by scoring behavioral seizures and measuring BDNF mRNA expression. In addition, effects of the opioid receptor antagonist naloxone and antiepileptic drugs, diazepam, phenobarbital, and valproate, on i.c.v. MOR agonist-induced behavioral seizures and up-regulation of BDNF mRNA expression were determined. A single i.c.v. administration of morphine (10-100 microg) or DAMGO (0.15-1.5 microg) dose-dependently elicited behavioral seizures and increased BDNF mRNA expression in the widespread brain regions. However, s.c. administration of MOR agonists neither produced behavioral seizures nor increased BDNF mRNA expression. Pretreatment with naloxone 1 mg/kg significantly reduced behavioral seizure scores and the up-regulation of BDNF mRNA expression elicited by i.c.v. morphine or DAMGO. Similarly, diazepam 10 mg/kg and phenobarbital 40 mg/kg significantly blocked i.c.v. MOR agonist-induced actions. Pretreatment with valproate 300 mg/kg only attenuated behavioral seizures, but it did not affect morphine-induced increase of BDNF mRNA expression. This study provides supporting evidence that seizure activity plays an important role in the up-regulation of BDNF mRNA expression elicited by central MOR activation and that decreased inhibitory action of GABAergic system through the modulation on GABA receptor synaptic function by central MOR activation is involved in its regulation of BDNF mRNA expression.
Collapse
Affiliation(s)
- H N Zhang
- Department of Biomedical Engineering, College of Engineering, 1101 Beal Avenue, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
32
|
Rantamäki T, Castrén E. Targeting TrkB neurotrophin receptor to treat depression. Expert Opin Ther Targets 2008; 12:705-15. [DOI: 10.1517/14728222.12.6.705] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
33
|
Saitoh A, Yamada M, Yamada M, Takahashi K, Yamaguchi K, Murasawa H, Nakatani A, Tatsumi Y, Hirose N, Kamei J. Antidepressant-like effects of the delta-opioid receptor agonist SNC80 ([(+)-4-[(alphaR)-alpha-[(2S,5R)-2,5-dimethyl-4-(2-propenyl)-1-piperazinyl]-(3-methoxyphenyl)methyl]-N,N-diethylbenzamide) in an olfactory bulbectomized rat model. Brain Res 2008; 1208:160-9. [DOI: 10.1016/j.brainres.2007.07.095] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Revised: 07/04/2007] [Accepted: 07/05/2007] [Indexed: 10/22/2022]
|
34
|
Kozisek ME, Middlemas D, Bylund DB. Brain-derived neurotrophic factor and its receptor tropomyosin-related kinase B in the mechanism of action of antidepressant therapies. Pharmacol Ther 2007; 117:30-51. [PMID: 17949819 DOI: 10.1016/j.pharmthera.2007.07.001] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Accepted: 07/24/2007] [Indexed: 12/12/2022]
Abstract
The focus of this review is to critically examine and review the literature on the role of brain-derived neurotrophic factor (BDNF) and its primary receptor, tropomyosin-related kinase B (TrkB), in the actions of pharmacologically diverse antidepressant treatments for depression. This will include a review of the studies on the regulation of BDNF and TrkB by different types of antidepressant drug treatments and animal in models of depression, as well as altered levels of BDNF and TrkB in the blood and postmortem brain of patients with depression. Results from clinical and basic studies have demonstrated that stress and depression decrease BDNF expression and neurogenesis and antidepressant treatment reverses or blocks these effects, leading to the neurotrophic hypothesis of depression. Clinical studies demonstrate an association between BDNF levels and several disorders, including depression, epilepsy, bipolar disorder, Parkinson's and Alzheimer's diseases. Physical activity and diet exert neurotrophic effects and positively modulate BDNF levels. A common single nucleotide polymorphism (SNP) in the BDNF gene, a methionine substitution for valine, is associated with alterations in brain anatomy and memory, but what role it has in clinical disorders is unclear. Findings suggest that early childhood events and adult stress produce neurodegenerative alterations in the brain that can eventually cause breakdown of information processing in the neuronal networks regulating mood. Antidepressant treatments elevate activity-dependent neuronal plasticity by activating BDNF, thereby gradually restoring network function and ultimately mood.
Collapse
Affiliation(s)
- Megan E Kozisek
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, United States.
| | | | | |
Collapse
|
35
|
Zhang H, Shi YG, Woods JH, Watson SJ, Ko MC. Central kappa-opioid receptor-mediated antidepressant-like effects of nor-Binaltorphimine: behavioral and BDNF mRNA expression studies. Eur J Pharmacol 2007; 570:89-96. [PMID: 17601558 PMCID: PMC2031926 DOI: 10.1016/j.ejphar.2007.05.045] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2007] [Revised: 05/17/2007] [Accepted: 05/22/2007] [Indexed: 12/29/2022]
Abstract
kappa-opioid receptor antagonists such as nor-Binaltorphimine (nor-BNI) have been shown to produce antidepressant-like behavioral effects in animal models of depression. The aim of this study was to investigate further the duration of centrally administered nor-BNI-induced antidepressant-like actions measured by both behavior and brain-derived neurotrophic factor (BDNF) gene expression. In addition, antagonist studies were conducted to determine the role of opioid receptor subtypes and the time course of nor-BNI's pharmacological actions. Antidepressant-like behavioral effects were measured by decreased immobility in the rat forced swim test and BDNF mRNA expression was determined by in situ hybridization. Centrally administered nor-BNI (20 microg, i.c.v.) decreased immobility and increased BDNF mRNA expression in the hippocampus on day 1, not on days 3-14, post-administration. Systemic administration of selective mu-, delta- and kappa-opioid receptor antagonists did not block nor-BNI-induced antidepressant-like effects. In contrast, i.c.v. administration of nor-BNI 7 or 14 days earlier significantly blocked subsequent nor-BNI-induced decreased immobility and upregulation of BDNF mRNA expression. Although the duration of nor-BNI's antidepressant-like effects did not synchronize with that of its kappa-opioid receptor antagonist effects, this study is the first to show that centrally administered nor-BNI, like most clinically used antidepressants, can upregulate BDNF mRNA expression in the rat hippocampus. These findings further demonstrate that central kappa-opioid receptor mediates antidepressant-like effects of nor-BNI measured by both behavior and BDNF gene expression.
Collapse
Affiliation(s)
- Huina Zhang
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yong-Gong Shi
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - James H. Woods
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Stanley J. Watson
- Molecular & Behavioral Neuroscience Institute, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mei-Chuan Ko
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Psychology, National Cheng Chi University, Taipei, Taiwan
- * Corresponding author. Department of Pharmacology, University of Michigan Medical School, 1301 MSRB III, Ann Arbor, MI 48109-0632, USA. Tel: +1-734-647-3119; fax: +1-734-764-7118. E-mail address: (M.C. Ko)
| |
Collapse
|
36
|
Onali P, Olianas MC. N-Desmethylclozapine, a major clozapine metabolite, acts as a selective and efficacious delta-opioid agonist at recombinant and native receptors. Neuropsychopharmacology 2007; 32:773-85. [PMID: 16841075 DOI: 10.1038/sj.npp.1301152] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The present study examined the effects of N-desmethylclozapine (NDMC), a biologically active metabolite of the atypical antipsychotic clozapine, at cloned human opioid receptors stably expressed in Chinese hamster ovary (CHO) cells and at native opioid receptors present in NG108-15 cells and rat brain. In CHO cells expressing the delta-opioid receptor (CHO/DOR), NDMC behaved as a full agonist both in stimulating [(35)S]GTPgammaS binding (pEC(50)=7.24) and in inhibiting cyclic AMP formation (pEC(50)=6.40). NDMC inhibited [(3)H]naltrindole binding to CHO/DOR membranes with competition curves that were modulated by guanine nucleotides in an agonist-like manner. Determination of intrinsic efficacies by taking into consideration both the maximal [(35)S]GTPgammaS binding stimulation and the extent of receptor occupancy at which half-maximal effect occurred indicated that NDMC had an efficacy value equal to 82% of that of the full delta-opioid receptor agonist DPDPE, whereas clozapine and the other clozapine metabolite clozapine N-oxide displayed much lower levels of agonist efficacy. NDMC exhibited poor agonist activity and lower affinity at the kappa-opioid receptor and was inactive at mu-opioid and NOP receptors. In NG108-15 cells, NDMC inhibited cyclic AMP formation and stimulated the phosphorylation of extracellular signal-regulated kinase 1/2 by activating the endogenously expressed delta-opioid receptor. Moreover, in membranes of different brain regions, NDMC stimulated [(35)S]GTPgammaS binding and regulated adenylyl cyclase activity and the effects were potently antagonized by naltrindole. These data demonstrate for the first time that NDMC acts as a selective and efficacious delta-opioid receptor agonist and suggest that this unique property may contribute, at least in part, to the clinical actions of the atypical antipsychotic clozapine.
Collapse
Affiliation(s)
- Pierluigi Onali
- Department of Neurosciences, Section of Biochemical Pharmacology, University of Cagliari, Cagliari, Italy.
| | | |
Collapse
|
37
|
Solbrig MV, Adrian R, Wechsler SL, Koob GF. Activators of potassium M currents have anticonvulsant actions in two rat models of encephalitis. Eur J Pharmacol 2006; 555:23-9. [PMID: 17126318 PMCID: PMC2981072 DOI: 10.1016/j.ejphar.2006.10.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2006] [Revised: 09/29/2006] [Accepted: 10/02/2006] [Indexed: 10/24/2022]
Abstract
Opioid systems in hippocampus regulate excitability and kappa opioids have a role in anticonvulsant protection, but their mechanisms of action are incompletely understood. We examined the ability of opioid and nonopioid agents with overlapping ionic mechanisms and actions similar to kappa opioid agonists, to block seizures in rat models of encephalitis due to Borna Disease virus and Herpes Simplex Virus Type-1. Naltrindole, a delta antagonist and thus a kappa opioid sparing agent, (10 mg/kg s.c.) blocked spontaneous and naloxone (opioid antagonist)-induced seizures in the models, but produced somatic signs similar to opioid withdrawal. Given that delta antagonists as well as kappa opioid agonists in hippocampus enhance potassium M currents (I(M)), we tested the effect of the I(M) augmenter flupirtine. Flupirtine (20 mg/kg i.p.) prevented seizures in Borna and herpes infected rats, without signs of withdrawal, hypotonia or sedation. The results support the efficacy of opioid and nonopioid drugs in modulating naloxone-induced seizures in critical illness due to viral encephalitis and by analogy, opioid withdrawal seizures.
Collapse
Affiliation(s)
- Marylou V Solbrig
- Department of Neurology, University of California-Irvine, Irvine, California 92697-4292 USA.
| | | | | | | |
Collapse
|
38
|
Zhang Q, Keenan SM, Peng Y, Nair AC, Yu SJ, Howells RD, Welsh WJ. Discovery of novel triazole-based opioid receptor antagonists. J Med Chem 2006; 49:4044-7. [PMID: 16821764 PMCID: PMC2693423 DOI: 10.1021/jm0601250] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
We report the computer-aided design, chemical synthesis, and biological evaluation of a novel family of delta opioid receptor (DOR) antagonists containing a 1,2,4-triazole core structure that are structurally distinct from other known opioid receptor active ligands. Among those delta antagonists sharing this core structure, 8 exhibited strong binding affinity (K(i) = 50 nM) for the DOR and appreciable selectivity for delta over mu and kappa opioid receptors (delta/mu = 80; delta/kappa > 200).
Collapse
Affiliation(s)
| | | | | | | | | | | | - William J. Welsh
- To whom correspondence should be addressed. Phone: 732−235−3234. Fax: 732−235−3475. E-mail:
| |
Collapse
|
39
|
Narita M, Kuzumaki N, Miyatake M, Sato F, Wachi H, Seyama Y, Suzuki T. Role of delta-opioid receptor function in neurogenesis and neuroprotection. J Neurochem 2006; 97:1494-505. [PMID: 16696856 DOI: 10.1111/j.1471-4159.2006.03849.x] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The present study was undertaken to evaluate the implication of delta-opioid receptor function in neurogenesis and neuroprotection. We found that the stimulation of delta-opioid receptors by the selective delta-opioid receptor agonist SNC80 [(+)-4-[(alphaR)-alpha-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide] (10 nm) promoted neural differentiation from multipotent neural stem cells obtained from embryonic C3H mouse forebrains. In contrast, either a selective micro-opioid receptor agonist, [D-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin (DAMGO), or a specific kappa-opioid receptor agonist, (-)-trans-(1S,2S)-U-50488 hydrochloride (U50,488H), had no such effect. In addition to neural differentiation, the increase in cleaved caspase 3-like immunoreactivity induced by H2O2 (3 microm) was suppressed by treatment with SNC80 in cortical neuron/glia co-cultures. These effects of SNC80 were abolished by a Trk-dependent tyrosine kinase inhibitor: (8R*,9S*,11S*)-(-)-9-hydroxy-9-methoxycarbonyl-8-methyl-2,3,9,10-tetrahydro-8,11-epoxy-1H,8H,11H-2,7b,11a-triazadibenzo(a,g)cycloocta(cde)trinden-1-one (K-252a). The SNC80-induced neural differentiation was also inhibited by treatment with the protein kinase C (PKC) inhibitor, phosphatidylinositol 3-kinase (PI3K) inhibitor, mitogen-activated protein kinase kinase (MEK) inhibitor or Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibitor. These findings raise the possibility that delta-opioid receptors play a crucial role in neurogenesis and neuroprotection, mainly through the activation of Trk-dependent tyrosine kinase, which could be linked to PI3K, PKC, CaMKII and MEK.
Collapse
MESH Headings
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology
- Animals
- Apoptosis/drug effects
- Apoptosis/physiology
- Benzamides/pharmacology
- Brain/cytology
- Brain/embryology
- Brain/growth & development
- Brain-Derived Neurotrophic Factor/metabolism
- Brain-Derived Neurotrophic Factor/pharmacology
- Caspase 3
- Caspase Inhibitors
- Caspases/metabolism
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Cell Line
- Cell Proliferation/drug effects
- Coculture Techniques
- Cytoprotection/drug effects
- Cytoprotection/physiology
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Enzyme Inhibitors/pharmacology
- Mice
- Mice, Inbred C3H
- Narcotics/pharmacology
- Nerve Regeneration/drug effects
- Nerve Regeneration/physiology
- Neurons/cytology
- Neurons/drug effects
- Neurons/metabolism
- Piperazines/pharmacology
- Receptor, trkA/drug effects
- Receptor, trkA/metabolism
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, delta/physiology
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Stem Cells/cytology
- Stem Cells/drug effects
- Stem Cells/metabolism
Collapse
Affiliation(s)
- Minoru Narita
- Department of Toxicology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan.
| | | | | | | | | | | | | |
Collapse
|
40
|
Zhang H, Torregrossa MM, Jutkiewicz EM, Shi YG, Rice KC, Woods JH, Watson SJ, Ko MC. Endogenous opioids upregulate brain-derived neurotrophic factor mRNA through delta- and micro-opioid receptors independent of antidepressant-like effects. Eur J Neurosci 2006; 23:984-94. [PMID: 16519663 PMCID: PMC1462954 DOI: 10.1111/j.1460-9568.2006.04621.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Systemic administration of delta-opioid receptor (DOR) agonists decreases immobility in the forced swim test (FST) and increases brain-derived neurotrophic factor (BDNF) mRNA expression in rats, indicating that DOR agonists may have antidepressant-like effects. The aim of this study was to investigate the effects of central administration of endogenous opioid peptides on behavior in the FST and on brain BDNF mRNA expression in rats. Effects of endogenous opioids were compared with those produced by intracerebroventricular administration of a selective non-peptidic DOR agonist (+)BW373U86. Antidepressant-like effects were measured by decreased immobility in the FST. BDNF mRNA expression was determined by in situ hybridization. Centrally administered (+)BW373U86 decreased immobility and increased BDNF mRNA expression in the frontal cortex through a DOR-mediated mechanism, because these effects were blocked by the DOR antagonist naltrindole, but not by the micro-opioid receptor (MOR) antagonist naltrexone (NTX) or the kappa-opioid receptor antagonist nor-binaltorphimine. Of all the endogenous opioids tested, only leu- and met-enkephalin produced behavioral effects like those of (+)BW373U86 in the FST. Unlike (+)BW373U86, the enkephalins upregulated BDNF mRNA expression in the hippocampus through DOR- and MOR-mediated mechanisms. beta-Endorphin, endomorphin-1 and endomorphin-2 significantly increased BDNF mRNA levels in the frontal cortex, hippocampus and amygdala without reducing immobility; and most of these effects were reversed by NTX. This study is the first to provide evidence that endogenous opioids can upregulate BDNF mRNA expression through the DOR and MOR, and that leu- and met-enkephalin have similar pharmacological profiles to synthetic DOR agonists in producing antidepressant-like effects.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Analysis of Variance
- Animals
- Autoradiography/methods
- Behavior, Animal
- Benzamides/administration & dosage
- Brain-Derived Neurotrophic Factor/genetics
- Brain-Derived Neurotrophic Factor/metabolism
- Dose-Response Relationship, Drug
- Drug Interactions
- Freezing Reaction, Cataleptic/drug effects
- In Situ Hybridization/methods
- Injections, Intraventricular/methods
- Male
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Narcotics/metabolism
- Opioid Peptides/administration & dosage
- Piperazines/administration & dosage
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/physiology
- Receptors, Opioid, mu/physiology
- Swimming
- Up-Regulation/drug effects
- Up-Regulation/physiology
Collapse
Affiliation(s)
- Huina Zhang
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Millan MJ. Multi-target strategies for the improved treatment of depressive states: Conceptual foundations and neuronal substrates, drug discovery and therapeutic application. Pharmacol Ther 2006; 110:135-370. [PMID: 16522330 DOI: 10.1016/j.pharmthera.2005.11.006] [Citation(s) in RCA: 389] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2005] [Accepted: 11/28/2005] [Indexed: 12/20/2022]
Abstract
Major depression is a debilitating and recurrent disorder with a substantial lifetime risk and a high social cost. Depressed patients generally display co-morbid symptoms, and depression frequently accompanies other serious disorders. Currently available drugs display limited efficacy and a pronounced delay to onset of action, and all provoke distressing side effects. Cloning of the human genome has fuelled expectations that symptomatic treatment may soon become more rapid and effective, and that depressive states may ultimately be "prevented" or "cured". In pursuing these objectives, in particular for genome-derived, non-monoaminergic targets, "specificity" of drug actions is often emphasized. That is, priority is afforded to agents that interact exclusively with a single site hypothesized as critically involved in the pathogenesis and/or control of depression. Certain highly selective drugs may prove effective, and they remain indispensable in the experimental (and clinical) evaluation of the significance of novel mechanisms. However, by analogy to other multifactorial disorders, "multi-target" agents may be better adapted to the improved treatment of depressive states. Support for this contention is garnered from a broad palette of observations, ranging from mechanisms of action of adjunctive drug combinations and electroconvulsive therapy to "network theory" analysis of the etiology and management of depressive states. The review also outlines opportunities to be exploited, and challenges to be addressed, in the discovery and characterization of drugs recognizing multiple targets. Finally, a diversity of multi-target strategies is proposed for the more efficacious and rapid control of core and co-morbid symptoms of depression, together with improved tolerance relative to currently available agents.
Collapse
Affiliation(s)
- Mark J Millan
- Institut de Recherches Servier, Centre de Recherches de Croissy, Psychopharmacology Department, 125, Chemin de Ronde, 78290-Croissy/Seine, France.
| |
Collapse
|
42
|
Jutkiewicz EM, Torregrossa MM, Sobczyk-Kojiro K, Mosberg HI, Folk JE, Rice KC, Watson SJ, Woods JH. Behavioral and neurobiological effects of the enkephalinase inhibitor RB101 relative to its antidepressant effects. Eur J Pharmacol 2006; 531:151-9. [PMID: 16442521 PMCID: PMC1828120 DOI: 10.1016/j.ejphar.2005.12.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2005] [Revised: 12/08/2005] [Accepted: 12/12/2005] [Indexed: 10/25/2022]
Abstract
Nonpeptidic delta-opioid receptor agonists produce antidepressant-like effects in rodents, and compounds that inhibit the breakdown of endogenous opioid peptides have antidepressant-like effects in animal models. In this study, the behavioral effects of the enkephalinase inhibitor, RB101 (N-[(R, S)-2-benzyl-3-[(S)(2-amino-4-methyl-thio)-butyldithio]-1-oxopropyl]-l-phenylalanine benzyl ester), were examined. Specifically, the effects of RB101 on convulsive activity, locomotor activity, and antidepressant-like effects in the forced swim test were studied in Sprague-Dawley rats, and the opioid receptor types mediating these effects were examined by antagonist studies. In addition, the effects of RB101 on brain-derived neurotrophic factor (BDNF) mRNA expression were evaluated in relation to its antidepressant effects. RB101 produced delta-opioid receptor-mediated antidepressant effects (32 mg/kg i.v. and 100 mg/kg i.p.) and increased locomotor activity (32 mg/kg i.v.) in rats. RB101 did not produce convulsions or seizures and did not alter BDNF mRNA expression. In conclusion, RB101 has the potential to produce antidepressant effects without convulsions.
Collapse
Affiliation(s)
- Emily M Jutkiewicz
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109-0632, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Torregrossa MM, Jutkiewicz EM, Mosberg HI, Balboni G, Watson SJ, Woods JH. Peptidic delta opioid receptor agonists produce antidepressant-like effects in the forced swim test and regulate BDNF mRNA expression in rats. Brain Res 2005; 1069:172-81. [PMID: 16364263 PMCID: PMC1780167 DOI: 10.1016/j.brainres.2005.11.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2005] [Revised: 11/08/2005] [Accepted: 11/11/2005] [Indexed: 10/25/2022]
Abstract
Systemically active, nonpeptidic delta opioid receptor agonists have been shown to produce antidepressant and anxiolytic effects in animal models in rodents. In addition, delta agonists have been shown to increase expression of brain-derived neurotrophic factor (BDNF) mRNA, an effect of some antidepressants, which may be important for the clinical efficacy of antidepressant drugs. The present study examined whether a variety of peptidic delta agonists, DPDPE, JOM-13, a systemically active derivative of DPDPE, deltorphin II, and H-Dmt-Tic-NH-CH2-Bid could produce convulsions and antidepressant-like effects in the forced swim test. In addition, some of these compounds were examined for their influence on BDNF mRNA expression. All four agonists dose-dependently decreased immobility in the forced swim test, indicating an antidepressant-like effect. Only JOM-13 produced convulsions at doses required for antidepressant-like effects. In addition, DPDPE increased BDNF mRNA expression, as measured by in situ hybridization, in the frontal cortex. The antidepressant-like effect of the agonists in the forced swim test and the increase in BDNF mRNA expression produced by DPDPE were blocked by the delta antagonist naltrindole. Therefore, activation of the delta receptor by centrally administered peptidic agonists and intravenously administered JOM-13 produces behavioral antidepressant-like effects without producing convulsions, and some peptidic agonists can increase BDNF mRNA expression, however, not as consistently as the systemically active nonpeptidic agonists.
Collapse
MESH Headings
- Adamantane/administration & dosage
- Adamantane/analogs & derivatives
- Animals
- Antidepressive Agents/administration & dosage
- Autoradiography/methods
- Behavior, Animal/drug effects
- Brain-Derived Neurotrophic Factor/genetics
- Brain-Derived Neurotrophic Factor/metabolism
- Depression/drug therapy
- Dipeptides/administration & dosage
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Drug Administration Routes
- Drug Interactions
- Enkephalin, D-Penicillamine (2,5)-/administration & dosage
- Enkephalins/administration & dosage
- Gene Expression Regulation/drug effects
- Immobility Response, Tonic/drug effects
- In Situ Hybridization/methods
- Male
- Oligopeptides/administration & dosage
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/antagonists & inhibitors
- Swimming
- Tetrahydroisoquinolines/administration & dosage
Collapse
|
44
|
Urigüen L, Berrendero F, Ledent C, Maldonado R, Manzanares J. Kappa- and delta-opioid receptor functional activities are increased in the caudate putamen of cannabinoid CB1 receptor knockout mice. Eur J Neurosci 2005; 22:2106-10. [PMID: 16262648 DOI: 10.1111/j.1460-9568.2005.04372.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The purpose of this study was to examine the functional interaction between endogenous opioid and cannabinoid receptor systems in the caudate putamen and nucleus accumbens. We therefore examined by autoradiography the functional activity and density of micro-, kappa- and delta-opioid receptors in both brain regions of cannabinoid CB1 receptor knockout mice. Functional activity was estimated by measuring agonist-stimulated [35S]GTPgammaS binding. Results showed that deletion of the CB1 cannabinoid receptor markedly increased kappa-opioid (50%) and delta-opioid (42%) receptor activities whereas no differences were found in micro-opioid receptor in the caudate putamen. In contrast, binding autoradiography showed a similar density of micro-, kappa- and delta-opioid receptors between mutant and wild-type mice. No differences were found in densities or activities of micro-, kappa- and delta-opioid receptors between mutant and wild-type mice in the nucleus accumbens. Taken together, our results revealed that deletion of CB1 cannabinoid receptors produced a pronounced increase in the activity of kappa- and delta-opioid receptors in the caudate putamen. This endogenous interaction between opioid and cannabinoid receptors may be relevant to further understand a variety of neuroadaptative processes involving the participation of opioid receptors, such as motor behaviour, emotional responses and drug dependence.
Collapse
MESH Headings
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology
- Analgesics, Non-Narcotic/pharmacology
- Analgesics, Opioid/pharmacology
- Animals
- Autoradiography/methods
- Benzamides/pharmacology
- Competitive Bidding/methods
- Drug Interactions
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics
- Male
- Mice
- Mice, Knockout
- Neostriatum/drug effects
- Neostriatum/metabolism
- Piperazines/pharmacology
- Protein Binding/drug effects
- Receptor, Cannabinoid, CB1/deficiency
- Receptors, Opioid, delta/physiology
- Receptors, Opioid, kappa/physiology
- Sulfur Isotopes/pharmacokinetics
Collapse
Affiliation(s)
- Leyre Urigüen
- Servicio de Psiquiatría y Unidad de Investigación, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | | | | |
Collapse
|
45
|
Abstract
This paper is the 27th consecutive installment of the annual review of research concerning the endogenous opioid system, now spanning over 30 years of research. It summarizes papers published during 2004 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; alcohol and drugs of abuse; 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, USA.
| | | |
Collapse
|
46
|
Ribeiro SC, Kennedy SE, Smith YR, Stohler CS, Zubieta JK. Interface of physical and emotional stress regulation through the endogenous opioid system and mu-opioid receptors. Prog Neuropsychopharmacol Biol Psychiatry 2005; 29:1264-80. [PMID: 16256255 DOI: 10.1016/j.pnpbp.2005.08.011] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/26/2005] [Indexed: 12/30/2022]
Abstract
Unraveling the pathways and neurobiological mechanisms that underlie the regulation of physical and emotional stress responses in humans is of critical importance to understand vulnerability and resiliency factors to the development of a number of complex physical and psychopathological states. Dysregulation of central stress response circuits have been implicated in the establishment of conditions as diverse as persistent pain, mood and personality disorders and substance abuse and dependence. The present review examines the contribution of the endogenous opioid system and mu-opioid receptors to the modulation and adaptation of the organism to challenges, such as sustained pain and negative emotional states, which threaten its internal homeostasis. Data accumulated in animal models, and more recently in humans, point to this neurotransmitter system as a critical modulator of the transition from acute (warning signals) to sustained (stressor) environmental adversity. The existence of pathways and regulatory mechanisms common to the regulation of both physical and emotional states transcend classical categorical disease classifications, and point to the need to utilize dimensional, "symptom"-related approximations to their study. Possible future areas of study at the interface of "mind" (cognitive-emotional) and "body" (physical) functions are delineated in this context.
Collapse
Affiliation(s)
- Saulo C Ribeiro
- University of Michigan, Department of Psychiatry and Molecular and Behavioral Neuroscience Institute, MBNI, 205 Zine Pitcher Place, 48109-0720, USA
| | | | | | | | | |
Collapse
|
47
|
Torregrossa MM, Folk JE, Rice KC, Watson SJ, Woods JH. Chronic administration of the delta opioid receptor agonist (+)BW373U86 and antidepressants on behavior in the forced swim test and BDNF mRNA expression in rats. Psychopharmacology (Berl) 2005; 183:31-40. [PMID: 16220339 PMCID: PMC1315298 DOI: 10.1007/s00213-005-0113-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2005] [Accepted: 06/23/2005] [Indexed: 11/26/2022]
Abstract
RATIONALE Selective delta opioid receptor agonists have been shown to produce antidepressant-like behavioral effects and increase brain-derived neurotrophic factor (BDNF) mRNA expression when given acutely, but the chronic effects of delta agonists have been less well characterized. OBJECTIVE The present study examined the effects of chronic exposure to the delta agonist (+)BW373U86 (BW) on antidepressant-like behavior in the forced swim test and on BDNF mRNA expression in comparison to chronic treatment with the antidepressants fluoxetine, desipramine, bupropion, and tranylcypromine. METHODS Sprague-Dawley rats were treated chronic ally with one of the above treatments and were tested for antidepressant effects in the forced swim test, and assayed for BDNF mRNA expression by in situ hybridization. RESULTS Acute administration of 10 mg/kg BW produced a significant antidepressant-like effect in the forced swim test, while chronic (8- or 21-day) BW administration did not produce a significant antidepressant-like effect. When 10 mg/kg BW was administered for 8 days, it produced a significant increase in BDNF mRNA expression in the frontal cortex, while having no effect on BDNF expression when given for 21 days. Chronic bupropion and desipramine significantly decreased BDNF expression in the dentate gyrus of the hippocampus, while fluoxetine had no effect in any brain region. Chronic tranylcypromine produced a significant increase in BDNF expression in the CA1 region of the hippocampus. CONCLUSIONS Chronic exposure to BW produces tolerance to most effects, although at differential rates. In addition, increased BDNF mRNA expression does not appear to be a common effect of chronic administration of various antidepressants.
Collapse
Affiliation(s)
| | | | | | - Stanley J. Watson
- Neuroscience Program, University of Michigan, Ann Arbor, MI, USA
- Mental Health Research Institute, University of Michigan, Ann Arbor, MI, USA
| | - James H. Woods
- Neuroscience Program, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmacology, University of Michigan Medical School, 1301 MSRB III, Ann Arbor, MI 48109-0632, USA
- e-mail:, Tel.: +1-734-7649133, Fax: +1-734-7647118
| |
Collapse
|
48
|
Jutkiewicz EM, Kaminsky ST, Rice KC, Traynor JR, Woods JH. Differential behavioral tolerance to the delta-opioid agonist SNC80 ([(+)-4-[(alphaR)-alpha-[(2S,5R)-2,5-dimethyl-4-(2-propenyl)-1-piperazinyl]-(3-methoxyphenyl)methyl]-N,N-diethylbenzamide) in Sprague-Dawley rats. J Pharmacol Exp Ther 2005; 315:414-22. [PMID: 16014751 PMCID: PMC1307500 DOI: 10.1124/jpet.105.088831] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nonpeptidic delta-opioid agonists produce a number of behaviors, such as antidepressant-like effects, locomotor stimulation, antinociception, and convulsions. To consider this class of compounds as potential therapeutics for humans, the effects of delta-opioid agonists after repeated administration must be evaluated. Therefore, the present study investigated the effects of repeated delta-opioid agonist, SNC80 ([(+)-4-[(alphaR)-alpha-[(2S,5R)-2,5-dimethyl-4-(2-propenyl)-1-piperazinyl]-(3-methoxyphenyl)-methyl]-N,N-diethylbenzamide), administration on its antidepressant-like effects in the forced swim test, locomotor activity, and convulsions in male Sprague-Dawley rats. Tolerance developed rapidly to the convulsive and locomotor-stimulating effects of SNC80 but not to the antidepressant-like effects. In addition, tolerance was evaluated at the level of the receptor-G protein interaction by measuring 5'-O-(3-[35S]thio)triphosphate binding in brains from rats that were pretreated with SNC80. With various exposure durations to SNC80, some brain regions demonstrated tolerance at different times, suggesting that adaptations in the delta-opioid system may occur during agonist exposure. Overall, the lack of observable tolerance to the antidepressant-like effects of SNC80 indicates that this class of compounds has potential as a novel antidepressant therapy.
Collapse
Affiliation(s)
- Emily M Jutkiewicz
- 1301 MSRB III, Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109-0632, USA.
| | | | | | | | | |
Collapse
|
49
|
Holt AG, Asako M, Lomax CA, MacDonald JW, Tong L, Lomax MI, Altschuler RA. Deafness-related plasticity in the inferior colliculus: gene expression profiling following removal of peripheral activity. J Neurochem 2005; 93:1069-86. [PMID: 15934929 DOI: 10.1111/j.1471-4159.2005.03090.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The inferior colliculus (IC) is a major center of integration in the ascending as well as descending auditory pathways, where both excitatory and inhibitory amino acid neurotransmitters play a key role. When normal input to the auditory system is decreased, the balance between excitation and inhibition in the IC is disturbed. We examined global changes in gene expression in the rat IC 3 and 21 days following bilateral deafening, using Affymetrix GeneChip arrays and focused our analysis on changes in expression of neurotransmission-related genes. Over 1400 probe sets in the Affymetrix Rat Genome U34A Array were identified as genes that were differentially expressed. These genes encoded proteins previously reported to change as a consequence of deafness, such as calbindin, as well as proteins not previously reported to be modulated by deafness, such as clathrin. A subset of 19 differentially expressed genes was further examined using quantitative RT-PCR at 3, 21 and 90 days following deafness. These included several GABA, glycine, glutamate receptor and neuropeptide-related genes. Expression of genes for GABA-A receptor subunits beta2, beta3, and gamma2, plus ionotropic glutamate receptor subunits AMPA 2, AMPA 3, and kainate 2, increased at all three times. Expression of glycine receptor alpha1 initially declined and then later increased, while alpha2 increased sharply at 21 days. Glycine receptor alpha3 increased between 3 and 21 days, but decreased at 90 days. Of the neuropeptide-related genes tested with qRT-PCR, tyrosine hydroxylase decreased approximately 50% at all times tested. Serotonin receptor 2C increased at 3, 21, and 90 days. The 5B serotonin receptor decreased at 3 and 21 days and returned to normal by 90 days. Of the genes tested with qRT-PCR, only glycine receptor alpha2 and serotonin receptor 5B returned to normal levels of expression at 90 days. Changes in GABA receptor beta3, GABA receptor gamma2, glutamate receptor 2/3, enkephalin, and tyrosine hydroxylase were further confirmed using immunocytochemistry.
Collapse
Affiliation(s)
- Avril Genene Holt
- Kresge Hearing Research Institute, Department of Otolaryngology/Head Neck Surgery, University of Michigan, Ann Arbor, Michigan, USA.
| | | | | | | | | | | | | |
Collapse
|
50
|
Martínez-Mota L, López-Rubalcava C, Rodríguez-Manzo G. Ejaculation induces long-lasting behavioural changes in male rats in the forced swimming test: evidence for an increased sensitivity to the antidepressant desipramine. Brain Res Bull 2005; 65:323-9. [PMID: 15811598 DOI: 10.1016/j.brainresbull.2004.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2004] [Revised: 11/12/2004] [Accepted: 12/06/2004] [Indexed: 11/21/2022]
Abstract
Copulation to exhaustion induces a sexual inhibitory state featured by a decreased motivation. Since diminished motivation is a key symptom of depression, we analysed if sexually exhausted animals showed increased levels of depressive-like behaviour in the forced swimming test (FST). Besides, sexual activity has been reported to have reinforcing properties. Thus, we analysed whether different levels of sexual activity modified the development of the behavioural despair in the FST. Finally, the effect of a sub-threshold dose of desipramine (DMI, 2.5mg/kg) was evaluated in animals with different sexual conditions. Male adult rats were divided into: (a) naive rats and (b) animals executing one intromission (1-INTR) or (c) one ejaculation (1-EJ) and (d) sexually satiated rats, classified as sexually responsive (R) and non-responsive (NR). No differences were found in immobility behaviour between sexually exhausted and naive rats. In the pre-test sessions of the FST males attaining ejaculation (1-EJ, R and NR) had lower levels of immobility or showed a tendency towards such a diminution, while animals with sexual activity not involving ejaculation did not. Data suggest a "protective" effect of ejaculation against the development of depressive-like behaviour. Finally, the sub-effective dose of DMI produced an antidepressant-like action in all animals that ejaculated. In conclusion, sexual experience involving ejaculation modifies the behavioural expression of rats in the FST and their sensitivity to antidepressant drugs like DMI. Both responses reflect brain plastic changes induced by ejaculation.
Collapse
Affiliation(s)
- Lucía Martínez-Mota
- Subdirección de Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente, Av. México-Xochimilco 101, San Lorenzo Huipulco, C.P. 14370, México D.F., Mexico
| | | | | |
Collapse
|