1
|
Tomita K, Yamanishi-Taira S, Igarashi K, Oogai Y, Kuwahara Y, Roudkenar MH, Roushandeh AM, Miyawaki S, Kurimasa A, Sato T. Oxytocin ameliorates KCC2 decrease induced by oral bacteria-derived LPS that affect rat primary cultured cells and PC-12 cells. Peptides 2022; 150:170734. [PMID: 34974081 DOI: 10.1016/j.peptides.2021.170734] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/01/2021] [Accepted: 12/28/2021] [Indexed: 01/01/2023]
Abstract
Inflammation, especially neuroinflammation, which is caused by stress, leads to central nervous system (CNS) dysfunction. Because lipopolysaccharides (LPSs) cause neuroinflammation, we investigated the effect of LPSs to CNS. In PC-12 cells, LPSs derived from oral bacteria reduced the expression of KCC2, a Cl- transporter. LPS derived from P. gingivalis (P. g) administered to rat primary cultured cells also reduced the KCC2 expression. However, LPSs derived from E. coli did not reduce the KCC2 expression. LPS treatment activated TLR4, IL-1β, and REST gene expressions, which led to KCC2 inactivation in PC-12 cells. The mechanism of KCC2 has been shown to play an important role in brain maturation, function (such as the GABA switch), and behavioral problems, we investigated the GABA function. We found that the GABA function was changed from inhibitory to excitatory by the LPS derived from P. g treatment. We demonstrated that the GSK3β also involved in the KCC2 reduction by LPS treatment. We show that oxytocin rescued the reduction in KCC2 expression caused by LPSs by inhibiting GSK3β signaling but vasopressin could not. Considered together, our results indicate that the LPSs from oral bacteria but not the LPS from E. coli increase the risk for brain disorders and oxytocin might be a candidate to overcome the abnormal behavior caused by brain disorders such as psychiatric disorders.
Collapse
Affiliation(s)
- Kazuo Tomita
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.
| | - Sayuri Yamanishi-Taira
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan; Department of Orthodontics and Dentofacial Orthopedics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Kento Igarashi
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Yuichi Oogai
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Yoshikazu Kuwahara
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan; Division of Radiation Biology and Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Mehryar Habibi Roudkenar
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan; Burn and Regenerative Medicine Research Center, Velayat Hospital, School of Medicine, Guilan University of Medical Sciences, Parastar St., Rasht, 41887-94755, Iran
| | - Amaneh Mohammadi Roushandeh
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan; Burn and Regenerative Medicine Research Center, Velayat Hospital, School of Medicine, Guilan University of Medical Sciences, Parastar St., Rasht, 41887-94755, Iran
| | - Shouichi Miyawaki
- Department of Orthodontics and Dentofacial Orthopedics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Akihiro Kurimasa
- Division of Radiation Biology and Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Tomoaki Sato
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
2
|
Ibarguen-Vargas Y, Leman S, Palme R, Belzung C, Surget A. CRF-R1 Antagonist Treatment Exacerbates Circadian Corticosterone Secretion under Chronic Stress, but Preserves HPA Feedback Sensitivity. Pharmaceutics 2021; 13:pharmaceutics13122114. [PMID: 34959395 PMCID: PMC8707167 DOI: 10.3390/pharmaceutics13122114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/01/2021] [Accepted: 12/04/2021] [Indexed: 11/16/2022] Open
Abstract
Despite promising initial reports, corticotropin-releasing factor receptor type-1 (CRF-R1) antagonists have mostly failed to display efficacy in clinical trials for anxiety or depression. Rather than broad-spectrum antidepressant/anxiolytic-like drugs, they may represent an ‘antistress’ solution for single stressful situations or for patients with chronic stress conditions. However, the impact of prolonged CRF-R1 antagonist treatments on the hypothalamic–pituitary–adrenal (HPA) axis under chronic stress conditions remained to be characterized. Hence, our study investigated whether a chronic CRF-R1 antagonist (crinecerfont, formerly known as SSR125543, 20 mg·kg−1·day−1 ip, 5 weeks) would alter HPA axis basal circadian activity and negative feedback sensitivity in mice exposed to either control or chronic stress conditions (unpredictable chronic mild stress, UCMS, 7 weeks), through measures of fecal corticosterone metabolites, plasma corticosterone, and dexamethasone suppression test. Despite preserving HPA axis parameters in control non-stressed mice, the 5-week crinercerfont treatment improved the negative feedback sensitivity in chronically stressed mice, but paradoxically exacerbated their basal corticosterone secretion nearly all along the circadian cycle. The capacity of chronic CRF-R1 antagonists to improve the HPA negative feedback in UCMS argues in favor of a potential therapeutic benefit against stress-related conditions. However, the treatment-related overactivation of HPA circadian activity in UCMS raise questions about possible physiological outcomes with long-standing treatments under ongoing chronic stress.
Collapse
Affiliation(s)
- Yadira Ibarguen-Vargas
- UMR1253, iBrain, Université de Tours, Inserm, 37200 Tours, France; (Y.I.-V.); (S.L.)
- EUK-CVL, Université d’Orléans, 45100 Orléans, France
| | - Samuel Leman
- UMR1253, iBrain, Université de Tours, Inserm, 37200 Tours, France; (Y.I.-V.); (S.L.)
| | - Rupert Palme
- Department of Biomedical Sciences/Biochemistry, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Catherine Belzung
- UMR1253, iBrain, Université de Tours, Inserm, 37200 Tours, France; (Y.I.-V.); (S.L.)
- Correspondence: (C.B.); (A.S.); Tel.: +33-2-47366994 (C.B.); +33-2-47367305 (A.S.)
| | - Alexandre Surget
- UMR1253, iBrain, Université de Tours, Inserm, 37200 Tours, France; (Y.I.-V.); (S.L.)
- Correspondence: (C.B.); (A.S.); Tel.: +33-2-47366994 (C.B.); +33-2-47367305 (A.S.)
| |
Collapse
|
3
|
Komnenov D, Quaal H, Rossi NF. V 1a and V 1b vasopressin receptors within the paraventricular nucleus contribute to hypertension in male rats exposed to chronic mild unpredictable stress. Am J Physiol Regul Integr Comp Physiol 2021; 320:R213-R225. [PMID: 33264070 DOI: 10.1152/ajpregu.00245.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/11/2020] [Accepted: 11/29/2020] [Indexed: 01/06/2023]
Abstract
Depression is an independent nontraditional risk factor for cardiovascular disease and mortality. The chronic unpredictable mild stress (CMS) rat model is a validated model of depression. Within the paraventricular nucleus (PVN), vasopressin (VP) via V1aR and V1bR have been implicated in stress and neurocardiovascular dysregulation. We hypothesized that in conscious, unrestrained CMS rats versus control, unstressed rats, PVN VP results in elevated arterial pressure (MAP), heart rate, and renal sympathetic nerve activity (RSNA) via activation of V1aR and/or V1bR. Male rats underwent 4 wk of CMS or control conditions. They were then equipped with hemodynamic telemetry transmitters, PVN cannula, and left renal nerve electrode. V1aR or V1bR antagonism dose-dependently inhibited MAP after VP injection. V1aR or V1bR blockers at their ED50 doses did not alter baseline parameters in either control or CMS rats but attenuated the pressor response to VP microinjected into PVN by ∼50%. Combined V1aR and V1bR inhibition completely blocked the pressor response to PVN VP in control but not CMS rats. CMS rats required combined maximally inhibitory doses to block either endogenous VP within the PVN or responses to microinjected VP. Compared with unstressed control rats, CMS rats had higher plasma VP levels and greater abundance of V1aR and V1bR transcripts within PVN. Thus, the CMS rat model of depression results in higher resting MAP, heart rate, and RSNA, which can be mitigated by inhibiting vasopressinergic mechanisms involving both V1aR and V1bR within the PVN. Circulating VP may also play a role in the pressor response.
Collapse
Affiliation(s)
- Dragana Komnenov
- Department of Internal Medicine, Wayne State University School of Medicine, Detroit, Michigan
| | - Harrison Quaal
- Department of Internal Medicine, Wayne State University School of Medicine, Detroit, Michigan
| | - Noreen F Rossi
- John D. Dingell Veterans Affairs Medical Center, Detroit, Michigan
- Department of Internal Medicine, Wayne State University School of Medicine, Detroit, Michigan
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
4
|
Abramova O, Zorkina Y, Ushakova V, Zubkov E, Morozova A, Chekhonin V. The role of oxytocin and vasopressin dysfunction in cognitive impairment and mental disorders. Neuropeptides 2020; 83:102079. [PMID: 32839007 DOI: 10.1016/j.npep.2020.102079] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/08/2020] [Accepted: 08/09/2020] [Indexed: 02/06/2023]
Abstract
Oxytocin (OXT) and arginine-vasopressin (AVP) are structurally homologous peptide hormones synthesized in the hypothalamus. Nowadays, the role of OXT and AVP in the regulation of social behaviour and emotions is generally known. However, recent researches indicate that peptides also participate in cognitive functioning. This review presents the evidence that the OXT/AVP systems are involved in the formation of social, working, spatial and episodic memory, mediated by such brain structures as the hippocampal CA2 and CA3 regions, amygdala and prefrontal cortex. Some data have demonstrated that the OXT receptor's polymorphisms are associated with impaired memory in humans, and OXT knockout in mice is connected with memory deficit. Additionally, OXT and AVP are involved in mental disorders' progression. Stress-induced imbalance of the OXT/AVP systems leads to an increased risk of various mental disorders, including depression, schizophrenia, and autism. At the same time, cognitive deficits are observed in stress and mental disorders, and perhaps peptide hormones play a part in this. The final part of the review describes possible therapeutic strategies for the use of OXT and AVP for treatment of various mental disorders.
Collapse
Affiliation(s)
- Olga Abramova
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia.
| | - Yana Zorkina
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - Valeria Ushakova
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia; Department of Biology, Lomonosov Moscow State University, Russia
| | - Eugene Zubkov
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - Anna Morozova
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - Vladimir Chekhonin
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia; Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
5
|
Dunlop BW, Wong A. The hypothalamic-pituitary-adrenal axis in PTSD: Pathophysiology and treatment interventions. Prog Neuropsychopharmacol Biol Psychiatry 2019; 89:361-379. [PMID: 30342071 DOI: 10.1016/j.pnpbp.2018.10.010] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 12/26/2022]
Abstract
Questions of how altered functioning of the hypothalamic pituitary adrenal (HPA) axis contribute to the development and maintenance of posttraumatic stress disorder (PTSD) have been the focus of extensive animal and human research. As a rule, results have been inconsistent across studies, likely due to a variety of confounding variables that have received inadequate attention. Important confounding factors include the effects of early life stress, biological sex, and the glucocorticoid used for interventions. In this manuscript we review: 1) the literature on identified abnormalities of HPA axis function in PTSD, both in terms of basal functioning and as part of challenge paradigms; 2) the role of HPA axis function pre- and immediately post-trauma as a risk factor for PTSD development; 3) the impact of HPA axis genes' allelic variants and epigenetic modifications on PTSD risk; 4) the contributions of HPA axis components to fear learning and extinction; and 5) therapeutic manipulations of the HPA axis to both prevent and treat PTSD, including the role of glucocorticoids as part of medication enhanced psychotherapy.
Collapse
Affiliation(s)
- Boadie W Dunlop
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA.
| | - Andrea Wong
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
6
|
The selective reversible FAAH inhibitor, SSR411298, restores the development of maladaptive behaviors to acute and chronic stress in rodents. Sci Rep 2018; 8:2416. [PMID: 29403000 PMCID: PMC5799259 DOI: 10.1038/s41598-018-20895-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/26/2018] [Indexed: 12/23/2022] Open
Abstract
Enhancing endogenous cannabinoid (eCB) signaling has been considered as a potential strategy for the treatment of stress-related conditions. Fatty acid amide hydrolase (FAAH) represents the primary degradation enzyme of the eCB anandamide (AEA), oleoylethanolamide (OEA) and palmitoylethanolamide (PEA). This study describes a potent reversible FAAH inhibitor, SSR411298. The drug acts as a selective inhibitor of FAAH, which potently increases hippocampal levels of AEA, OEA and PEA in mice. Despite elevating eCB levels, SSR411298 did not mimic the interoceptive state or produce the behavioral side-effects (memory deficit and motor impairment) evoked by direct-acting cannabinoids. When SSR411298 was tested in models of anxiety, it only exerted clear anxiolytic-like effects under highly aversive conditions following exposure to a traumatic event, such as in the mouse defense test battery and social defeat procedure. Results from experiments in models of depression showed that SSR411298 produced robust antidepressant-like activity in the rat forced-swimming test and in the mouse chronic mild stress model, restoring notably the development of inadequate coping responses to chronic stress. This preclinical profile positions SSR411298 as a promising drug candidate to treat diseases such as post-traumatic stress disorder, which involves the development of maladaptive behaviors.
Collapse
|
7
|
Ramos AT, Tufik S, Troncone LRP. Control of Stress-Induced ACTH Secretion by Vasopressin and CRH: Additional Evidence. Neuropsychobiology 2017; 73:184-90. [PMID: 27221315 DOI: 10.1159/000445480] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 03/08/2016] [Indexed: 11/19/2022]
Abstract
Vasopressin and CRH have complementary roles in the secretion of ACTH following different stress modalities. The concomitant use of V1b and CRF1 receptor antagonists completely inhibits ACTH secretion in response to different stress modalities. The combination of the CRF1 antagonist SSR125543 with the V1b antagonist SSR149415 effectively suppressed plasma ACTH 1.30 h after injection in rats stressed by ether vapor inhalation for 1 min, restraint stress for 1 h or forced swimming for 5 min. The duration of the effect was also studied. The CRF1 antagonist effectively suppressed ACTH secretion in restraint stress, while the V1b antagonist was effective against ether inhalation. Both antagonists were necessary to block the forced swimming stress response. SSR125543 induced a prolonged effect and can be used in a model of prolonged HPA axis blockade.
Collapse
Affiliation(s)
- Adriana T Ramos
- Department of Psychobiology, Universidade Federal de Sx00E3;o Paulo (UNIFESP), Sx00E3;o Paulo, Brazil
| | | | | |
Collapse
|
8
|
Chen NA, Ganella DE, Bathgate RAD, Chen A, Lawrence AJ, Kim JH. Knockdown of corticotropin-releasing factor 1 receptors in the ventral tegmental area enhances conditioned fear. Eur Neuropsychopharmacol 2016; 26:1533-1540. [PMID: 27397862 DOI: 10.1016/j.euroneuro.2016.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/14/2016] [Accepted: 06/06/2016] [Indexed: 12/01/2022]
Abstract
The neuropeptide corticotropin-releasing factor (CRF) coordinates the physiological and behavioural responses to stress. CRF receptors are highly expressed in the ventral tegmental area (VTA), an important region for motivated behaviour. Therefore, we examined the role of CRF receptor type 1 (CRFR1) in the VTA in conditioned fear, using a viral-mediated RNA interference approach. Following stereotaxic injection of a lentivirus that contained either shCRF-R1 or a control sequence, mice received tone-footshock pairings. Intra-VTA shCRF-R1 did not affect tone-elicited freezing during conditioning. Once conditioned fear was acquired, however, shCRF-R1 mice consistently showed stronger freezing to the tone even after extinction and reinstatement. These results implicate a novel role of VTA CRF-R1 in conditioned fear, and suggest how stress may modulate aversive learning and memory.
Collapse
Affiliation(s)
- Nicola A Chen
- Behavioural Neuroscience Division, Florey Institute of Neuroscience & Mental Health, Parkville, VIC 3052 Australia; The Florey Department of Neuroscience & Mental Health, Australia
| | - Despina E Ganella
- Behavioural Neuroscience Division, Florey Institute of Neuroscience & Mental Health, Parkville, VIC 3052 Australia; The Florey Department of Neuroscience & Mental Health, Australia
| | - Ross A D Bathgate
- Behavioural Neuroscience Division, Florey Institute of Neuroscience & Mental Health, Parkville, VIC 3052 Australia; The Florey Department of Neuroscience & Mental Health, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, VIC 3010 Australia
| | - Alon Chen
- Department of stress Neurobiology and Neurogenetics, Max-Planck Institute of Psychiatry, Munich 80804, Germany
| | - Andrew J Lawrence
- Behavioural Neuroscience Division, Florey Institute of Neuroscience & Mental Health, Parkville, VIC 3052 Australia; The Florey Department of Neuroscience & Mental Health, Australia
| | - Jee Hyun Kim
- Behavioural Neuroscience Division, Florey Institute of Neuroscience & Mental Health, Parkville, VIC 3052 Australia; The Florey Department of Neuroscience & Mental Health, Australia.
| |
Collapse
|
9
|
Fröhlich EE, Farzi A, Mayerhofer R, Reichmann F, Jačan A, Wagner B, Zinser E, Bordag N, Magnes C, Fröhlich E, Kashofer K, Gorkiewicz G, Holzer P. Cognitive impairment by antibiotic-induced gut dysbiosis: Analysis of gut microbiota-brain communication. Brain Behav Immun 2016; 56:140-55. [PMID: 26923630 PMCID: PMC5014122 DOI: 10.1016/j.bbi.2016.02.020] [Citation(s) in RCA: 480] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/22/2016] [Accepted: 02/22/2016] [Indexed: 12/12/2022] Open
Abstract
Emerging evidence indicates that disruption of the gut microbial community (dysbiosis) impairs mental health. Germ-free mice and antibiotic-induced gut dysbiosis are two approaches to establish causality in gut microbiota-brain relationships. However, both models have limitations, as germ-free mice display alterations in blood-brain barrier and brain ultrastructure and antibiotics may act directly on the brain. We hypothesized that the concerns related to antibiotic-induced gut dysbiosis can only adequately be addressed if the effect of intragastric treatment of adult mice with multiple antibiotics on (i) gut microbial community, (ii) metabolite profile in the colon, (iii) circulating metabolites, (iv) expression of neuronal signaling molecules in distinct brain areas and (v) cognitive behavior is systematically investigated. Of the antibiotics used (ampicillin, bacitracin, meropenem, neomycin, vancomycin), ampicillin had some oral bioavailability but did not enter the brain. 16S rDNA sequencing confirmed antibiotic-induced microbial community disruption, and metabolomics revealed that gut dysbiosis was associated with depletion of bacteria-derived metabolites in the colon and alterations of lipid species and converted microbe-derived molecules in the plasma. Importantly, novel object recognition, but not spatial, memory was impaired in antibiotic-treated mice. This cognitive deficit was associated with brain region-specific changes in the expression of cognition-relevant signaling molecules, notably brain-derived neurotrophic factor, N-methyl-d-aspartate receptor subunit 2B, serotonin transporter and neuropeptide Y system. We conclude that circulating metabolites and the cerebral neuropeptide Y system play an important role in the cognitive impairment and dysregulation of cerebral signaling molecules due to antibiotic-induced gut dysbiosis.
Collapse
Affiliation(s)
- Esther E Fröhlich
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria.
| | - Aitak Farzi
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Raphaela Mayerhofer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Florian Reichmann
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Angela Jačan
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Bernhard Wagner
- Institute of Biomedical Science, FH JOANNEUM University of Applied Sciences, Eggenberger Allee 13, 8020 Graz, Austria
| | - Erwin Zinser
- Institute of Biomedical Science, FH JOANNEUM University of Applied Sciences, Eggenberger Allee 13, 8020 Graz, Austria
| | - Natalie Bordag
- Center for Biomarker Research in Medicine, CBmed GmbH, Stiftingtalstrasse 5, 8010 Graz, Austria
| | - Christoph Magnes
- HEALTH Institute for Biomedicine and Health Sciences, JOANNEUM RESEARCH Forschungsgesellschaft mbH, Neue Stiftingtalstraße 2, Graz, Austria
| | - Eleonore Fröhlich
- Core Facility Microscopy, Center for Medical Research, Medical University of Graz, Stiftingtalstrasse 24/1, 8010 Graz, Austria
| | - Karl Kashofer
- Institute of Pathology, Medical University of Graz, Auenbruggerplatz 25, 8036 Graz, Austria
| | - Gregor Gorkiewicz
- Institute of Pathology, Medical University of Graz, Auenbruggerplatz 25, 8036 Graz, Austria; Theodor Escherich Laboratory for Medical Microbiome Research, Medical University of Graz, Auenbruggerplatz 25, 8036 Graz, Austria; BioTechMed-Graz, Krenngasse 37/1, 8010 Graz, Austria
| | - Peter Holzer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria.
| |
Collapse
|
10
|
Javadian N, Rahimi N, Javadi-Paydar M, Doustimotlagh AH, Dehpour AR. The modulatory effect of nitric oxide in pro- and anti-convulsive effects of vasopressin in PTZ-induced seizures threshold in mice. Epilepsy Res 2016; 126:134-40. [PMID: 27497813 DOI: 10.1016/j.eplepsyres.2016.07.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 07/09/2016] [Accepted: 07/25/2016] [Indexed: 11/25/2022]
Abstract
Vasopressin neuropeptides play an important role in the several cognitive, social, and neuroendocrine functions. Also, several studies report the involvement of nitrergic system in the vasopressin functions in central nervous system. This study investigates the effect of Arginine-Vasopressin (AVP) in pentylenetetrazol (PTZ)-induced seizures threshold and the probable role of nitric oxide (NO). AVP is administered intraperitoneally (0.01-20μg/kg, i.p.) 30min before induction of seizures. Administration of AVP (0.1μg/kg) significantly lowered the PTZ-induced seizures threshold. But, administration of AVP (10 and 20μg/kg) increased the seizures threshold, significantly. Pretreatment of SR 49059 (V1a receptor antagonist, 2mg/kg, i.p.) just reversed the pro-convulsant effect of AVP. Meanwhile, SSR 149415 (V1b receptor antagonist, 10mg/kg, i.p.) pretreatment reversed both pro-and anti-convulsant effects of AVP. The nitric oxide precursor, L-arginine (60mg/kg, i.p.) increased pro-convulsant effect of AVP, but did not change anticonvulsant activity. The nitric oxide synthase (NOS) inhibitor L-NAME (10mg/kg, i.p.) reversed both pro- and anti-convulsant effect of AVP. Selective inducible NOS inhibitor, aminoguanidine (100mg/kg, i.p.) just reversed the anti-convulsant effects of AVP. The results of the present study showed nitric oxide system may contribute to the biphasic effects of AVP on PTZ-induced seizures. V1a receptor may modulate only the proconvulsive effect. While, V1b receptors can mediate both the pro- and anti-convulsive effect of AVP.
Collapse
Affiliation(s)
- Nina Javadian
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nastaran Rahimi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrak Javadi-Paydar
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Doustimotlagh
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Xu Y, Cheng X, Cui X, Wang T, Liu G, Yang R, Wang J, Bo X, Wang S, Zhou W, Zhang Y. Effects of 5-h multimodal stress on the molecules and pathways involved in dendritic morphology and cognitive function. Neurobiol Learn Mem 2015; 123:225-38. [DOI: 10.1016/j.nlm.2015.06.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 06/17/2015] [Accepted: 06/23/2015] [Indexed: 11/25/2022]
|
12
|
Vargas-López V, Torres-Berrio A, González-Martínez L, Múnera A, Lamprea MR. Acute restraint stress and corticosterone transiently disrupts novelty preference in an object recognition task. Behav Brain Res 2015; 291:60-66. [DOI: 10.1016/j.bbr.2015.05.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 05/04/2015] [Accepted: 05/08/2015] [Indexed: 12/14/2022]
|
13
|
Barsegyan A, Atsak P, Hornberger WB, Jacobson PB, van Gaalen MM, Roozendaal B. The Vasopressin 1b Receptor Antagonist A-988315 Blocks Stress Effects on the Retrieval of Object-Recognition Memory. Neuropsychopharmacology 2015; 40:1979-89. [PMID: 25669604 PMCID: PMC4839522 DOI: 10.1038/npp.2015.48] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 02/03/2015] [Accepted: 02/03/2015] [Indexed: 12/13/2022]
Abstract
Stress-induced activation of the hypothalamo-pituitary-adrenocortical (HPA) axis and high circulating glucocorticoid levels are well known to impair the retrieval of memory. Vasopressin can activate the HPA axis by stimulating vasopressin 1b (V1b) receptors located on the pituitary. In the present study, we investigated the effect of A-988315, a selective and highly potent non-peptidergic V1b-receptor antagonist with good pharmacokinetic properties, in blocking stress effects on HPA-axis activity and memory retrieval. To study cognitive performance, male Sprague-Dawley rats were trained on an object-discrimination task during which they could freely explore two identical objects. Memory for the objects and their location was tested 24 h later. A-988315 (20 or 60 mg/kg) or water was administered orally 90 min before retention testing, followed 60 min later by stress of footshock exposure. A-988315 dose-dependently dampened stress-induced increases in corticosterone plasma levels, but did not significantly alter HPA-axis activity of non-stressed control rats. Most importantly, A-988315 administration prevented stress-induced impairment of memory retrieval on both the object-recognition and the object-location tasks. A-988315 did not alter the retention of non-stressed rats and did not influence the total time spent exploring the objects or experimental context in either stressed or non-stressed rats. Thus, these findings indicate that direct antagonism of V1b receptors is an effective treatment to block stress-induced activation of the HPA axis and the consequent impairment of retrieval of different aspects of recognition memory.
Collapse
Affiliation(s)
- Areg Barsegyan
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Nijmegen, The Netherlands,Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Piray Atsak
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Nijmegen, The Netherlands,Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands
| | | | - Peer B Jacobson
- Integrated Sciences and Technology, Imaging Research, AbbVie, North Chicago, IL, USA
| | - Marcel M van Gaalen
- Neuroscience Research, AbbVie Deutschland GmbH & Co KG, Ludwigshafen, Germany
| | - Benno Roozendaal
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Nijmegen, The Netherlands,Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands,Department of Cognitive Neuroscience, Radboud University Medical Centre, Geert Grooteplein Noord 21, PO Box 9101, Nijmegen 6500 HB, The Netherlands, Tel: +31 (0) 24 366 6301, E-mail:
| |
Collapse
|
14
|
Serra-de-Oliveira N, Boilesen SN, Prado de França Carvalho C, LeSueur-Maluf L, Zollner RDL, Spadari RC, Medalha CC, Monteiro de Castro G. Behavioural changes observed in demyelination model shares similarities with white matter abnormalities in humans. Behav Brain Res 2015; 287:265-75. [DOI: 10.1016/j.bbr.2015.03.038] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/15/2015] [Accepted: 03/17/2015] [Indexed: 11/30/2022]
|
15
|
Qureshi S, Galiveeti S, Bichet DG, Roth J. Diabetes insipidus: celebrating a century of vasopressin therapy. Endocrinology 2014; 155:4605-21. [PMID: 25211589 DOI: 10.1210/en.2014-1385] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Diabetes mellitus, widely known to the ancients for polyuria and glycosuria, budded off diabetes insipidus (DI) about 200 years ago, based on the glucose-free polyuria that characterized a subset of patients. In the late 19th century, clinicians identified the posterior pituitary as the site of pathology, and pharmacologists found multiple bioactivities there. Early in the 20th century, the amelioration of the polyuria with extracts of the posterior pituitary inaugurated a new era in therapy and advanced the hypothesis that DI was due to a hormone deficiency. Decades later, a subset of patients with polyuria unresponsive to therapy were recognized, leading to the distinction between central DI and nephrogenic DI, an early example of a hormone-resistant condition. Recognition that the posterior pituitary had 2 hormones was followed by du Vigneaud's Nobel Prize winning isolation, sequencing, and chemical synthesis of oxytocin and vasopressin. The pure hormones accelerated the development of bioassays and immunoassays that confirmed the hormone deficiency in vasopressin-sensitive DI and abundant levels of hormone in patients with the nephrogenic disorder. With both forms of the disease, acquired and inborn defects were recognized. Emerging concepts of receptors and of genetic analysis led to the recognition of patients with mutations in the genes for 1) arginine vasopressin (AVP), 2) the AVP receptor 2 (AVPR2), and 3) the aquaporin 2 water channel (AQP2). We recount here the multiple skeins of clinical and laboratory research that intersected frequently over the centuries since the first recognition of DI.
Collapse
Affiliation(s)
- Sana Qureshi
- Laboratory of Diabetes and Diabetes-Related Disorders (S.Q., S.G., J.R.), Feinstein Institute for Medical Research, North Shore-Long Island Jewish Health System, Manhasset, New York 11030; Albert Einstein College of Medicine (S.Q., J.R.), Yeshiva University, Bronx, New York 10461; James J Peters VA Medical Center (S.G.), Mount Sinai Medical Center Health System, Bronx, New York 10029; Hôpital du Sacré-Coeur de Montréal (D.G.B.), Groupe des Protéines Membranaires, Université de Montréal, Montréal, Québec, Canada H4J IC5; and Hofstra North Shore-Long Island Jewish School of Medicine (J.R.), North Shore-Long Island Jewish Health System, Hempstead, New York 11549
| | | | | | | |
Collapse
|
16
|
CRF1 receptor-deficiency induces anxiety-like vulnerability to cocaine. Psychopharmacology (Berl) 2014; 231:3965-72. [PMID: 24687410 DOI: 10.1007/s00213-014-3534-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 03/03/2014] [Indexed: 01/23/2023]
Abstract
RATIONALE The intake of psychostimulant drugs may induce cognitive dysfunction and negative affective-like states, and is associated with increased activity of stress-responsive systems. The corticotropin-releasing factor (CRF) system mediates neuroendocrine, behavioural and autonomic responses to stressors, and might be implicated in substance-related disorders. CRF signalling is mediated by two receptor types, named CRF1 and CRF2. OBJECTIVES The present study aims to elucidate the role for the CRF1 receptor in cognitive dysfunction and anxiety-like states induced by cocaine. RESULTS The genetic inactivation of the CRF1 receptor (CRF1+/- and CRF1-/-) does not influence recognition memory in drug-naïve mice, as assessed by the novel object recognition (NOR) test. Moreover, the chronic administration of escalating doses of cocaine (5-20 mg/kg, i.p.) induces NOR deficits, which are unaffected by CRF1 receptor-deficiency. However, the same drug regimen reveals an anxiety-like vulnerability to cocaine in CRF1-/- but not in wild-type or CRF1+/- mice, as assessed by the elevated plus maze test. CONCLUSIONS The present findings indicate dissociation of cognitive dysfunction and anxiety-like states induced by cocaine. Moreover, they unravel a novel mechanism of vulnerability to psychostimulant drugs.
Collapse
|
17
|
Two weeks of predatory stress induces anxiety-like behavior with co-morbid depressive-like behavior in adult male mice. Behav Brain Res 2014; 275:120-5. [PMID: 25200517 DOI: 10.1016/j.bbr.2014.08.060] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 08/26/2014] [Accepted: 08/30/2014] [Indexed: 02/06/2023]
Abstract
Psychological stress can have devastating and lasting effects on a variety of behaviors, especially those associated with mental illnesses such as anxiety and depression. Animal models of chronic stress are frequently used to elucidate the mechanisms underlying the relationship between stress and mental health disorders and to develop improved treatment options. The current study expands upon a novel chronic stress paradigm for mice: predatory stress. The predatory stress model incorporates the natural predator-prey relationship that exists among rats and mice and allows for greater interaction between the animals, in turn increasing the extent of the stressful experience. In this study, we evaluated the behavioral effects of exposure to 15 days of predatory stress on an array of behavioral indices. Up to 2 weeks after the end of stress, adult male mice showed an increase of anxiety-like behaviors as measured by the open field and social interaction tests. Animals also expressed an increase in depressive-like behavior in the sucrose preference test. Notably, performance on the novel object recognition task, a memory test, improved after predatory stress. Taken as a whole, our results indicate that 15 exposures to this innovative predatory stress paradigm are sufficient to elicit robust anxiety-like behaviors with evidence of co-morbid depressive-like behavior, as well as changes in cognitive behavior in male mice.
Collapse
|
18
|
Beurel E, Nemeroff CB. Interaction of stress, corticotropin-releasing factor, arginine vasopressin and behaviour. Curr Top Behav Neurosci 2014; 18:67-80. [PMID: 24659554 DOI: 10.1007/7854_2014_306] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Stress mediates the activation of a variety of systems ranging from inflammatory to behavioral responses. In this review we focus on two neuropeptide systems, corticotropin-releasing factor (CRF) and arginine vasopressin (AVP), and their roles in regulating stress responses. Both peptides have been demonstrated to be involved in anxiogenic and depressive effects, actions mediated in part through their regulation of the hypothalamic-pituitary-adrenal axis and the release of adrenocorticotropic hormone. Because of the depressive effects of CRF and AVP, drugs modifying the stress-associated detrimental actions of CRF and AVP are under development, particularly drugs antagonizing CRF and AVP receptors for therapy in depression.
Collapse
Affiliation(s)
- Eléonore Beurel
- Department of Psychiatry and Behavioral Sciences, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | | |
Collapse
|
19
|
The CRF₁ receptor antagonist SSR125543 prevents stress-induced cognitive deficit associated with hippocampal dysfunction: comparison with paroxetine and D-cycloserine. Psychopharmacology (Berl) 2013; 228:97-107. [PMID: 23407783 DOI: 10.1007/s00213-013-3020-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 01/31/2013] [Indexed: 12/17/2022]
Abstract
RATIONALE The selective CRF1 (corticotropin releasing factor type 1) receptor antagonist SSR125543 has been previously shown to attenuate the long-term cognitive deficit produced by traumatic stress exposure. Memory disturbances described in post-traumatic stress disorder (PTSD) patients are believed to be associated with changes in neuronal activity, in particular at the level of the hippocampus. OBJECTIVES The present study aims at investigating whether the effects of SSR125543 (10 mg/kg/day for 2 weeks) on cognitive impairment induced by traumatic stress exposure are associated with changes in hippocampal excitability. Effects of SSR125543 were compared to those of the 5-HT reuptake inhibitor, paroxetine (10 mg/kg/day), and the partial N-methyl-D-aspartate (NMDA) receptor agonist, D-cycloserine (10 mg/kg/day), two compounds which have demonstrated clinical efficacy against PTSD. METHODS Mice received two unavoidable electric foot-shocks. Then, 1 or 16 days after stress, they were tested for their memory performance using the object recognition test. Neuronal excitability was recorded during the third week post-stress in the CA1 area of the hippocampus. Drugs were administered from day 1 post-stress to the day preceding the electrophysiological study. RESULTS Application of electric shocks produced cognitive impairment 16, but not 1 day after stress, an effect which was associated with a decrease in hippocampal neuronal excitability. Both stress-induced effects were prevented by repeated administration of SSR125543, paroxetine and D-cycloserine. CONCLUSIONS These findings confirm that the CRF1 receptor antagonist SSR125543 is able to attenuate the behavioral effects of traumatic stress exposure and indicate that these effects are associated with a normalization of hippocampal neuronal excitability impaired by stress.
Collapse
|
20
|
Koshimizu TA, Nakamura K, Egashira N, Hiroyama M, Nonoguchi H, Tanoue A. Vasopressin V1a and V1b Receptors: From Molecules to Physiological Systems. Physiol Rev 2012; 92:1813-64. [DOI: 10.1152/physrev.00035.2011] [Citation(s) in RCA: 250] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The neurohypophysial hormone arginine vasopressin (AVP) is essential for a wide range of physiological functions, including water reabsorption, cardiovascular homeostasis, hormone secretion, and social behavior. These and other actions of AVP are mediated by at least three distinct receptor subtypes: V1a, V1b, and V2. Although the antidiuretic action of AVP and V2 receptor in renal distal tubules and collecting ducts is relatively well understood, recent years have seen an increasing understanding of the physiological roles of V1a and V1b receptors. The V1a receptor is originally found in the vascular smooth muscle and the V1b receptor in the anterior pituitary. Deletion of V1a or V1b receptor genes in mice revealed that the contributions of these receptors extend far beyond cardiovascular or hormone-secreting functions. Together with extensively developed pharmacological tools, genetically altered rodent models have advanced the understanding of a variety of AVP systems. Our report reviews the findings in this important field by covering a wide range of research, from the molecular physiology of V1a and V1b receptors to studies on whole animals, including gene knockout/knockdown studies.
Collapse
Affiliation(s)
- Taka-aki Koshimizu
- Department of Pharmacology, Division of Molecular Pharmacology, Jichi Medical University, Tochigi, Japan; Department of Pharmacology, National Research Institute for Child Health and Development, Tokyo, Japan; Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan; and Department of Internal Medicine, Kitasato University, Kitasato Institute Medical Center Hospital, Saitama, Japan
| | - Kazuaki Nakamura
- Department of Pharmacology, Division of Molecular Pharmacology, Jichi Medical University, Tochigi, Japan; Department of Pharmacology, National Research Institute for Child Health and Development, Tokyo, Japan; Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan; and Department of Internal Medicine, Kitasato University, Kitasato Institute Medical Center Hospital, Saitama, Japan
| | - Nobuaki Egashira
- Department of Pharmacology, Division of Molecular Pharmacology, Jichi Medical University, Tochigi, Japan; Department of Pharmacology, National Research Institute for Child Health and Development, Tokyo, Japan; Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan; and Department of Internal Medicine, Kitasato University, Kitasato Institute Medical Center Hospital, Saitama, Japan
| | - Masami Hiroyama
- Department of Pharmacology, Division of Molecular Pharmacology, Jichi Medical University, Tochigi, Japan; Department of Pharmacology, National Research Institute for Child Health and Development, Tokyo, Japan; Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan; and Department of Internal Medicine, Kitasato University, Kitasato Institute Medical Center Hospital, Saitama, Japan
| | - Hiroshi Nonoguchi
- Department of Pharmacology, Division of Molecular Pharmacology, Jichi Medical University, Tochigi, Japan; Department of Pharmacology, National Research Institute for Child Health and Development, Tokyo, Japan; Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan; and Department of Internal Medicine, Kitasato University, Kitasato Institute Medical Center Hospital, Saitama, Japan
| | - Akito Tanoue
- Department of Pharmacology, Division of Molecular Pharmacology, Jichi Medical University, Tochigi, Japan; Department of Pharmacology, National Research Institute for Child Health and Development, Tokyo, Japan; Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan; and Department of Internal Medicine, Kitasato University, Kitasato Institute Medical Center Hospital, Saitama, Japan
| |
Collapse
|
21
|
Neumann ID, Landgraf R. Balance of brain oxytocin and vasopressin: implications for anxiety, depression, and social behaviors. Trends Neurosci 2012; 35:649-59. [PMID: 22974560 DOI: 10.1016/j.tins.2012.08.004] [Citation(s) in RCA: 666] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 05/24/2012] [Accepted: 08/13/2012] [Indexed: 02/07/2023]
Abstract
Oxytocin and vasopressin are regulators of anxiety, stress-coping, and sociality. They are released within hypothalamic and limbic areas from dendrites, axons, and perikarya independently of, or coordinated with, secretion from neurohypophysial terminals. Central oxytocin exerts anxiolytic and antidepressive effects, whereas vasopressin tends to show anxiogenic and depressive actions. Evidence from pharmacological and genetic association studies confirms their involvement in individual variation of emotional traits extending to psychopathology. Based on their opposing effects on emotional behaviors, we propose that a balanced activity of both brain neuropeptide systems is important for appropriate emotional behaviors. Shifting the balance between the neuropeptide systems towards oxytocin, by positive social stimuli and/or psychopharmacotherapy, may help to improve emotional behaviors and reinstate mental health.
Collapse
Affiliation(s)
- Inga D Neumann
- Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany.
| | | |
Collapse
|
22
|
The CRF1 receptor antagonist SSR125543 attenuates long-term cognitive deficit induced by acute inescapable stress in mice, independently from the hypothalamic pituitary adrenal axis. Pharmacol Biochem Behav 2012; 102:415-22. [DOI: 10.1016/j.pbb.2012.05.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 05/14/2012] [Accepted: 05/26/2012] [Indexed: 12/22/2022]
|
23
|
Calabrese F, Molteni R, Riva MA. Antistress properties of antidepressant drugs and their clinical implications. Pharmacol Ther 2011; 132:39-56. [DOI: 10.1016/j.pharmthera.2011.05.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 05/09/2011] [Indexed: 02/07/2023]
|