1
|
Gilani M, Abak N, Saberian M. Genetic-epigenetic-neuropeptide associations in mood and anxiety disorders: Toward personalized medicine. Pharmacol Biochem Behav 2024; 245:173897. [PMID: 39424200 DOI: 10.1016/j.pbb.2024.173897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/29/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Mood and anxiety disorders are complex psychiatric conditions shaped by the multifactorial interplay of genetic, epigenetic, and neuropeptide factors. This review aims to elucidate the intricate interactions among these factors and their potential in advancing personalized medicine. We examine the genetic underpinnings, emphasizing key heritability studies and specific gene associations. The role of epigenetics is discussed, focusing on how environmental factors can modify gene expression and contribute to these disorders. Neuropeptides, including substance P, CRF, AVP, NPY, galanin, and kisspeptin, are evaluated for their involvement in mood regulation and their potential as therapeutic targets. Additionally, we address the emerging role of the gut microbiome in modulating neuropeptide activity and its connection to mood disorders. This review integrates findings from genetic, epigenetic, and neuropeptide research, offering a comprehensive overview of their collective impact on mood and anxiety disorders. By highlighting novel insights and potential clinical applications, we underscore the importance of a multi-omics approach in developing personalized treatment strategies. Future research directions are proposed to address existing knowledge gaps and translate these findings into clinical practice. Our review provides a fresh perspective on the pathophysiology of mood and anxiety disorders, paving the way for more effective and individualized therapies.
Collapse
Affiliation(s)
- Maryam Gilani
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloofar Abak
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Saberian
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Hendry E, McCallister B, Elman DJ, Freeman R, Borsook D, Elman I. Validity of mental and physical stress models. Neurosci Biobehav Rev 2024; 158:105566. [PMID: 38307304 PMCID: PMC11082879 DOI: 10.1016/j.neubiorev.2024.105566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/13/2024] [Accepted: 01/27/2024] [Indexed: 02/04/2024]
Abstract
Different stress models are employed to enhance our understanding of the underlying mechanisms and explore potential interventions. However, the utility of these models remains a critical concern, as their validities may be limited by the complexity of stress processes. Literature review revealed that both mental and physical stress models possess reasonable construct and criterion validities, respectively reflected in psychometrically assessed stress ratings and in activation of the sympathoadrenal system and the hypothalamic-pituitary-adrenal axis. The findings are less robust, though, in the pharmacological perturbations' domain, including such agents as adenosine or dobutamine. Likewise, stress models' convergent- and discriminant validity vary depending on the stressors' nature. Stress models share similarities, but also have important differences regarding their validities. Specific traits defined by the nature of the stressor stimulus should be taken into consideration when selecting stress models. Doing so can personalize prevention and treatment of stress-related antecedents, its acute processing, and chronic sequelae. Further work is warranted to refine stress models' validity and customize them so they commensurate diverse populations and circumstances.
Collapse
Affiliation(s)
- Erin Hendry
- Center for Autonomic and Peripheral Nerve Disorders, Harvard Medical School, Boston, MA, USA; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Brady McCallister
- Center for Autonomic and Peripheral Nerve Disorders, Harvard Medical School, Boston, MA, USA
| | - Dan J Elman
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Roy Freeman
- Center for Autonomic and Peripheral Nerve Disorders, Harvard Medical School, Boston, MA, USA; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - David Borsook
- Departments of Psychiatry and Radiology, Massachusetts General Hospital, Harvard Medical School, Department of Anesthesiology, Harvard Medical School, Boston, MA, USA.
| | - Igor Elman
- Department of Psychiatry, Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, USA
| |
Collapse
|
3
|
Ritz NL, Brocka M, Butler MI, Cowan CSM, Barrera-Bugueño C, Turkington CJR, Draper LA, Bastiaanssen TFS, Turpin V, Morales L, Campos D, Gheorghe CE, Ratsika A, Sharma V, Golubeva AV, Aburto MR, Shkoporov AN, Moloney GM, Hill C, Clarke G, Slattery DA, Dinan TG, Cryan JF. Social anxiety disorder-associated gut microbiota increases social fear. Proc Natl Acad Sci U S A 2024; 121:e2308706120. [PMID: 38147649 PMCID: PMC10769841 DOI: 10.1073/pnas.2308706120] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 10/05/2023] [Indexed: 12/28/2023] Open
Abstract
Social anxiety disorder (SAD) is a crippling psychiatric disorder characterized by intense fear or anxiety in social situations and their avoidance. However, the underlying biology of SAD is unclear and better treatments are needed. Recently, the gut microbiota has emerged as a key regulator of both brain and behaviour, especially those related to social function. Moreover, increasing data supports a role for immune function and oxytocin signalling in social responses. To investigate whether the gut microbiota plays a causal role in modulating behaviours relevant to SAD, we transplanted the microbiota from SAD patients, which was identified by 16S rRNA sequencing to be of a differential composition compared to healthy controls, to mice. Although the mice that received the SAD microbiota had normal behaviours across a battery of tests designed to assess depression and general anxiety-like behaviours, they had a specific heightened sensitivity to social fear, a model of SAD. This distinct heightened social fear response was coupled with changes in central and peripheral immune function and oxytocin expression in the bed nucleus of the stria terminalis. This work demonstrates an interkingdom basis for social fear responses and posits the microbiome as a potential therapeutic target for SAD.
Collapse
Affiliation(s)
- Nathaniel L. Ritz
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Anatomy and Neuroscience, University College Cork, CorkT12YT20, Ireland
| | - Marta Brocka
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
| | - Mary I. Butler
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, CorkT12YT20, Ireland
| | - Caitlin S. M. Cowan
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
| | - Camila Barrera-Bugueño
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
| | - Christopher J. R. Turkington
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- School of Microbiology, University College Cork, CorkT12K8AF, Ireland
| | - Lorraine A. Draper
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- School of Microbiology, University College Cork, CorkT12K8AF, Ireland
| | - Thomaz F. S. Bastiaanssen
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Anatomy and Neuroscience, University College Cork, CorkT12YT20, Ireland
| | - Valentine Turpin
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
| | - Lorena Morales
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
| | - David Campos
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
| | - Cassandra E. Gheorghe
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Anatomy and Neuroscience, University College Cork, CorkT12YT20, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, CorkT12YT20, Ireland
| | - Anna Ratsika
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Anatomy and Neuroscience, University College Cork, CorkT12YT20, Ireland
| | - Virat Sharma
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- School of Microbiology, University College Cork, CorkT12K8AF, Ireland
| | - Anna V. Golubeva
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
| | - Maria R. Aburto
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Anatomy and Neuroscience, University College Cork, CorkT12YT20, Ireland
| | - Andrey N. Shkoporov
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- School of Microbiology, University College Cork, CorkT12K8AF, Ireland
| | - Gerard M. Moloney
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Anatomy and Neuroscience, University College Cork, CorkT12YT20, Ireland
| | - Colin Hill
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- School of Microbiology, University College Cork, CorkT12K8AF, Ireland
| | - Gerard Clarke
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, CorkT12YT20, Ireland
| | - David A. Slattery
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, Frankfurt60528, Germany
| | - Timothy G. Dinan
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, CorkT12YT20, Ireland
| | - John F. Cryan
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Anatomy and Neuroscience, University College Cork, CorkT12YT20, Ireland
| |
Collapse
|
4
|
Woodson J, Bergan JF. Uncovering the brain-wide pattern of synaptic input to vasopressin-expressing neurons in the paraventricular nucleus of the hypothalamus. J Comp Neurol 2023; 531:1017-1031. [PMID: 37121600 PMCID: PMC10566340 DOI: 10.1002/cne.25476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 05/02/2023]
Abstract
Arginine vasopressin (AVP) is a neuropeptide critical for the mammalian stress response and social behavior. AVP produced in the hypothalamus regulates water osmolality and vasoconstriction in the body, and in the brain, it regulates social behavior, aggression, and anxiety. However, the circuit mechanisms that link AVP to social behavior, homeostatic function, and disease are not well understood. This study investigates the circuit configurations of AVP-expressing neurons in the rodent hypothalamus and characterizes synaptic input from the entire brain. We targeted the paraventricular nucleus (PVN) using retrograde viral tracing techniques to identify direct afferent synaptic connections made onto AVP-expressing neurons. AVP neurons in the PVN display region-specific anatomical configurations that reflect their unique contributions to homeostatic function, motor behaviors, feeding, and affiliative behavior. The afferent connections identified were similar in both sexes and subsequent molecular investigation of these inputs shows that those local hypothalamic inputs are overwhelmingly nonpeptidergic cells indicating a potential interneuron nexus between hormone cell activation and broader cortical connection. This proposed work reveals new insights into the organization of social behavior circuits in the brain, and how neuropeptides act centrally to modulate social behaviors.
Collapse
Affiliation(s)
- Jonathan Woodson
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Joseph F Bergan
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, Massachusetts, USA
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| |
Collapse
|
5
|
Smith-Osborne L, Duong A, Resendez A, Palme R, Fadok JP. Female dominance hierarchies influence responses to psychosocial stressors. Curr Biol 2023; 33:1535-1549.e5. [PMID: 37003262 PMCID: PMC10321215 DOI: 10.1016/j.cub.2023.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/26/2023] [Accepted: 03/07/2023] [Indexed: 04/03/2023]
Abstract
Social species form dominance hierarchies to ensure survival and promote reproductive success. Traditionally studied in males, rodent hierarchies are considered despotic, and dominant social rank results from a history of winning agonistic encounters. By contrast, female hierarchies are thought to be less despotic, and rank is conferred by intrinsic traits. Both social buffering and elevated social status confer resilience to depression, anxiety, and other consequences of chronic stress. Here, we investigate whether female social hierarchies and individual traits related to social rank likewise influence stress resilience. We observe the formation of dyadic female hierarchies under varying conditions of ambient light and circadian phase and subject mice to two forms of chronic psychosocial stress: social isolation or social instability. We find that stable female hierarchies emerge rapidly in dyads. Individual behavioral and endocrinological traits are characteristic of rank, some of which are circadian phase dependent. Further, female social rank is predicted by behavior and stress status prior to social introduction. Other behavioral characteristics suggest that rank is motivation-based, indicating that female rank identity serves an evolutionarily relevant purpose. Rank is associated with alterations in behavior in response to social instability stress and prolonged social isolation, but the different forms of stress produce disparate rank responses in endocrine status. Histological examination of c-Fos protein expression identified brain regions that respond to social novelty or social reunion following chronic isolation in a rank-specific manner. Collectively, female rank is linked to neurobiology, and hierarchies exert context-specific influence upon stress outcomes.
Collapse
Affiliation(s)
- Lydia Smith-Osborne
- Department of Psychology, Tulane University, New Orleans, LA 70118, USA; Tulane National Primate Research Center, Covington, LA 70433, USA.
| | - Anh Duong
- Department of Psychology, Tulane University, New Orleans, LA 70118, USA; Neuroscience Program, Tulane University, New Orleans, LA 70118, USA
| | - Alexis Resendez
- Department of Psychology, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Rupert Palme
- Department of Biomedical Sciences, University of Veterinary Medicine, Veterinärplatz 1, 1210 Vienna, Austria
| | - Jonathan P Fadok
- Department of Psychology, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA.
| |
Collapse
|
6
|
Kanes SJ, Dennie L, Perera P. Targeting the Arginine Vasopressin V 1b Receptor System and Stress Response in Depression and Other Neuropsychiatric Disorders. Neuropsychiatr Dis Treat 2023; 19:811-828. [PMID: 37077711 PMCID: PMC10106826 DOI: 10.2147/ndt.s402831] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/23/2023] [Indexed: 04/21/2023] Open
Abstract
A healthy stress response is critical for good mental and overall health and promotes neuronal growth and adaptation, but the intricately balanced biological mechanisms that facilitate a stress response can also result in predisposition to disease when that equilibrium is disrupted. The hypothalamic-pituitary-adrenal (HPA) axis neuroendocrine system plays a critical role in the body's response and adaptation to stress, and vasopressinergic regulation of the HPA axis is critical to maintaining system responsiveness during chronic stress. However, exposure to repeated or excessive physical or emotional stress or trauma can shift the body's stress response equilibrium to a "new normal" underpinned by enduring changes in HPA axis function. Exposure to early life stress due to adverse childhood experiences can also lead to lasting neurobiological changes, including in HPA axis function. HPA axis impairment in patients with depression is considered among the most reliable findings in biological psychiatry, and chronic stress has been shown to play a major role in the pathogenesis and onset of depression and other neuropsychiatric disorders. Modulating HPA axis activity, for example via targeted antagonism of the vasopressin V1b receptor, is a promising approach for patients with depression and other neuropsychiatric disorders associated with HPA axis impairment. Despite favorable preclinical indications in animal models, demonstration of clinical efficacy for the treatment of depressive disorders by targeting HPA axis dysfunction has been challenging, possibly due to the heterogeneity and syndromal nature of depressive disorders. Measures of HPA axis function, such as elevated cortisol levels, may be useful biomarkers for identifying patients who may benefit from treatments that modulate HPA axis activity. Utilizing clinical biomarkers to identify subsets of patients with impaired HPA axis function who may benefit is a promising next step in fine-tuning HPA axis activity via targeted antagonism of the V1b receptor.
Collapse
Affiliation(s)
- Stephen J Kanes
- EmbarkNeuro, Oakland, CA, USA
- Correspondence: Stephen J Kanes, EmbarkNeuro, Inc, 1111 Broadway, Suite 1300, Oakland, CA, 94607, USA, Tel +1 610 757 7821, Email
| | | | | |
Collapse
|
7
|
Huang B, Sun B, Yang R, Liang S, Li X, Guo Y, Meng Q, Fu Y, Li W, Zhao P, Gong M, Shi Y, Song L, Wang S, Yuan F, Shi H. Long-lasting effects of postweaning sleep deprivation on cognitive function and social behaviors in adult mice. Neuropharmacology 2022; 215:109164. [PMID: 35716724 DOI: 10.1016/j.neuropharm.2022.109164] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/21/2022] [Accepted: 06/08/2022] [Indexed: 10/18/2022]
Abstract
Sleep deprivation (SD) has adverse effects on physical and mental health. Recently increasing attention has been given to SD in the early-life stage. However, the effects and mechanisms of postweaning SD on cognitive function and social behaviors are still unclear. In this study, SD was conducted in mice from postnatal Day 21 (PND21) to PND42, 6 h a day. Meanwhile, changes in body weight, food and water intake were continuously monitored. Behavioral tests were carried out in adulthood of mice. The levels of serum corticosterone, the proinflammatory cytokines interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α), and the anti-inflammatory cytokines interleukin-10 (IL-10), vasopressin (VP) and oxytocin (OT) were measured by ELISA. Golgi staining was used to calculate neural dendritic spine density in the dorsal hippocampus (dHPC) CA1 region and medial prefrontal cortex (mPFC). We found that postweaning SD increased the food intake and the weight of female mice. Behavioral results showed that postweaning SD caused cognitive impairment and lowered social dominance in adult male mice but not in female mice. ELISA results showed that SD increased the levels of serum corticosterone, VP and OT in male mice and serum OT in female mice. Golgi staining analysis showed that SD decreased neural dendritic spine density in the dHPC in male mice. These results suggest that postweaning SD has a long-term effect on social dominance and cognitive function in male mice, which may provide a new insight into the role of SD in regulating cognitive function and social behaviors.
Collapse
Affiliation(s)
- Boya Huang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Binhuang Sun
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Rui Yang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Shihao Liang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Xinrui Li
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Yi Guo
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Qian Meng
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Yaling Fu
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Wenshuya Li
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China
| | - Penghui Zhao
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China
| | - Miao Gong
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Yun Shi
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medicinal University, Shijiazhuang, 050017, China
| | - Li Song
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, 050017, China; Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medicinal University, Shijiazhuang, 050017, China
| | - Sheng Wang
- Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Fang Yuan
- Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Haishui Shi
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, 050017, China; Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medicinal University, Shijiazhuang, 050017, China.
| |
Collapse
|
8
|
Pomrenze MB, Paliarin F, Maiya R. Friend of the Devil: Negative Social Influences Driving Substance Use Disorders. Front Behav Neurosci 2022; 16:836996. [PMID: 35221948 PMCID: PMC8866771 DOI: 10.3389/fnbeh.2022.836996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/13/2022] [Indexed: 11/20/2022] Open
Abstract
Substance use disorders in humans have significant social influences, both positive and negative. While prosocial behaviors promote group cooperation and are naturally rewarding, distressing social encounters, such as aggression exhibited by a conspecific, are aversive and can enhance the sensitivity to rewarding substances, promote the acquisition of drug-taking, and reinstate drug-seeking. On the other hand, withdrawal and prolonged abstinence from drugs of abuse can promote social avoidance and suppress social motivation, accentuating drug cravings and facilitating relapse. Understanding how complex social states and experiences modulate drug-seeking behaviors as well as the underlying circuit dynamics, such as those interacting with mesolimbic reward systems, will greatly facilitate progress on understanding triggers of drug use, drug relapse and the chronicity of substance use disorders. Here we discuss some of the common circuit mechanisms underlying social and addictive behaviors that may underlie their antagonistic functions. We also highlight key neurochemicals involved in social influences over addiction that are frequently identified in comorbid psychiatric conditions. Finally, we integrate these data with recent findings on (±)3,4-methylenedioxymethamphetamine (MDMA) that suggest functional segregation and convergence of social and reward circuits that may be relevant to substance use disorder treatment through the competitive nature of these two types of reward. More studies focused on the relationship between social behavior and addictive behavior we hope will spur the development of treatment strategies aimed at breaking vicious addiction cycles.
Collapse
Affiliation(s)
- Matthew B. Pomrenze
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
- *Correspondence: Matthew B. Pomrenze Rajani Maiya
| | - Franciely Paliarin
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Rajani Maiya
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- *Correspondence: Matthew B. Pomrenze Rajani Maiya
| |
Collapse
|
9
|
Cid-Jofré V, Moreno M, Reyes-Parada M, Renard GM. Role of Oxytocin and Vasopressin in Neuropsychiatric Disorders: Therapeutic Potential of Agonists and Antagonists. Int J Mol Sci 2021; 22:ijms222112077. [PMID: 34769501 PMCID: PMC8584779 DOI: 10.3390/ijms222112077] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/20/2021] [Accepted: 11/02/2021] [Indexed: 12/27/2022] Open
Abstract
Oxytocin (OT) and vasopressin (AVP) are hypothalamic neuropeptides classically associated with their regulatory role in reproduction, water homeostasis, and social behaviors. Interestingly, this role has expanded in recent years and has positioned these neuropeptides as therapeutic targets for various neuropsychiatric diseases such as autism, addiction, schizophrenia, depression, and anxiety disorders. Due to the chemical-physical characteristics of these neuropeptides including short half-life, poor blood-brain barrier penetration, promiscuity for AVP and OT receptors (AVP-R, OT-R), novel ligands have been developed in recent decades. This review summarizes the role of OT and AVP in neuropsychiatric conditions, as well as the findings of different OT-R and AVP-R agonists and antagonists, used both at the preclinical and clinical level. Furthermore, we discuss their possible therapeutic potential for central nervous system (CNS) disorders.
Collapse
Affiliation(s)
- Valeska Cid-Jofré
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile (USACH), Santiago 9170022, Chile; (V.C.-J.); (M.M.)
| | - Macarena Moreno
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile (USACH), Santiago 9170022, Chile; (V.C.-J.); (M.M.)
- Facultad de Ciencias Sociales, Escuela de Psicología, Universidad Bernardo OHiggins, Santiago 8370993, Chile
| | - Miguel Reyes-Parada
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile (USACH), Santiago 9170022, Chile; (V.C.-J.); (M.M.)
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Providencia 7500912, Chile
- Correspondence: (M.R.-P.); (G.M.R.)
| | - Georgina M. Renard
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile (USACH), Santiago 9170022, Chile; (V.C.-J.); (M.M.)
- Correspondence: (M.R.-P.); (G.M.R.)
| |
Collapse
|
10
|
Newman EL, Covington HE, Leonard MZ, Burk K, Miczek KA. Hypoactive Thalamic Crh+ Cells in a Female Mouse Model of Alcohol Drinking After Social Trauma. Biol Psychiatry 2021; 90:563-574. [PMID: 34281710 PMCID: PMC8463500 DOI: 10.1016/j.biopsych.2021.05.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 05/04/2021] [Accepted: 05/20/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Comorbid stress-induced mood and alcohol use disorders are increasingly prevalent among female patients. Stress exposure can disrupt salience processing and goal-directed decision making, contributing to persistent maladaptive behavioral patterns; these and other stress-sensitive cognitive and behavioral processes rely on dynamic and coordinated signaling by midline and intralaminar thalamic nuclei. Considering the role of social trauma in the trajectory of these debilitating psychopathologies, identifying vulnerable thalamic cells may provide guidance for targeting persistent stress-induced symptoms. METHODS A novel behavioral protocol traced the progression from social trauma to the development of social defensiveness and chronically escalated alcohol consumption in female mice. Recent cell activation-measured as cFos-was quantified in thalamic cells after safe social interactions, revealing stress-sensitive corticotropin-releasing hormone-expressing (Crh+) anterior central medial thalamic (aCMT) cells. These cells were optogenetically stimulated during stress-induced social defensiveness and abstinence-escalated binge drinking. RESULTS Crh+ aCMT neurons exhibited substantial activation after social interactions in stress-naïve but not in stressed female mice. Photoactivating Crh+ aCMT cells dampened stress-induced social deficits, whereas inhibiting these cells increased social defensiveness in stress-naïve mice. Optogenetically activating Crh+ aCMT cells diminished abstinence-escalated binge alcohol drinking in female mice, regardless of stress history. CONCLUSIONS This work uncovers a role for Crh+ aCMT neurons in maladaptive stress-induced social interactions and in binge drinking after forced abstinence in female mice. This molecularly defined thalamic cell population may serve as a critical stress-sensitive hub for social deficits caused by exposure to social trauma and for patterns of excessive alcohol drinking in female populations.
Collapse
Affiliation(s)
- Emily L Newman
- Department of Psychology, Tufts University, Medford, Massachusetts; Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, Massachusetts
| | | | | | - Kelly Burk
- Department of Psychology, Tufts University, Medford, Massachusetts
| | - Klaus A Miczek
- Department of Psychology, Tufts University, Medford, Massachusetts; Department of Neuroscience, Tufts University, Boston, Massachusetts.
| |
Collapse
|
11
|
Yoshimura M, Conway-Campbell B, Ueta Y. Arginine vasopressin: Direct and indirect action on metabolism. Peptides 2021; 142:170555. [PMID: 33905792 PMCID: PMC8270887 DOI: 10.1016/j.peptides.2021.170555] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/12/2021] [Accepted: 04/15/2021] [Indexed: 02/07/2023]
Abstract
From its identification and isolation in 1954, arginine vasopressin (AVP) has attracted attention, not only for its peripheral functions such as vasoconstriction and reabsorption of water from kidney, but also for its central effects. As there is now considerable evidence that AVP plays a crucial role in feeding behavior and energy balance, it has become a promising therapeutic target for treating obesity or other obesity-related metabolic disorders. However, the underlying mechanisms for AVP regulation of these central processes still remain largely unknown. In this review, we will provide a brief overview of the current knowledge concerning how AVP controls energy balance and feeding behavior, focusing on physiological aspects including the relationship between AVP, circadian rhythmicity, and glucocorticoids.
Collapse
Affiliation(s)
- Mitsuhiro Yoshimura
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Japan; Translational Health Sciences, Bristol Medical School, University of Bristol, UK.
| | | | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Japan
| |
Collapse
|
12
|
Hou W, Ma H, Xun Y, Zhang X, Cai W, Huang S, He Z, Tai F, Jia R. Sex-Dependent Effects of Chronic Social Defeat on Emotional and Social Behaviors, and Parameters of Oxytocin and Vasopressin Systems in Mandarin Voles ( Microtus mandarinus). Front Neurosci 2021; 15:625116. [PMID: 34045941 PMCID: PMC8144301 DOI: 10.3389/fnins.2021.625116] [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: 11/02/2020] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Abstract
In the regulation of emotional and social behaviors, both oxytocin (OT) and vasopressin (AVP) are sex specific. Although significant sex differences have been reported in the context of behavioral and hormonal responses to social stress, such differences in response to chronic social defeat stress (CSDS) and the underlying neural mechanisms remain largely unknown. By investigating monogamous mandarin voles (Microtus mandarinus), CSDS was found to decrease the percentages of time spent in the central area of the open field, in the open arms of the elevated plus maze, as well as in the light area of the light and dark boxes in both male and female voles. CSDS also increased the observed level of social withdrawal in both sex groups. However, CSDS exposure increased the percentages of immobile time in both the tail suspension test and the forced swim test and reduced the locomotor activity in the open field (in females only). Along with these behavioral changes, the oxytocin receptor (OTR) levels in the nucleus accumbens (NAc) were significantly lower in CSDS-exposed voles of both sexes; however, in males, the levels of OTR in the paraventricular nucleus (PVN) were reduced. CSDS-exposed males showed lower levels of V1aR in the NAc than CSDS-exposed females. Furthermore, induced by a single social defeat event, CSDS reduced c-Fos and OT double labeling in the PVN of females but increased c-Fos and AVP double-labeled neurons in the PVN of males exposed to a single social defeat event. Collectively, the present study indicates that OT and AVP systems may play important regulatory roles in the sex differences of behavioral performances in response to CSDS. These findings suggest mandarin voles as a useful animal model for studying sex-specific behavioral performance and the underlying neurobiological mechanisms of stress-related mental disorders in preclinical studies.
Collapse
Affiliation(s)
- Wenjuan Hou
- Laboratory for Brain and Behavioral Science, Shaanxi Normal University, Xi'an, China
| | - Huan Ma
- Laboratory for Brain and Behavioral Science, Shaanxi Normal University, Xi'an, China
| | - Yufeng Xun
- Laboratory for Brain and Behavioral Science, Shaanxi Normal University, Xi'an, China
| | - Xin Zhang
- Laboratory for Brain and Behavioral Science, Shaanxi Normal University, Xi'an, China
| | - Wenqi Cai
- Laboratory for Brain and Behavioral Science, Shaanxi Normal University, Xi'an, China
| | - Shuying Huang
- Laboratory for Brain and Behavioral Science, Shaanxi Normal University, Xi'an, China
| | - Zhixiong He
- Laboratory for Brain and Behavioral Science, Shaanxi Normal University, Xi'an, China
| | - Fadao Tai
- Laboratory for Brain and Behavioral Science, Shaanxi Normal University, Xi'an, China
| | - Rui Jia
- Laboratory for Brain and Behavioral Science, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
13
|
Harshaw C, Lanzkowsky J, Tran AQD, Bradley AR, Jaime M. Oxytocin and 'social hyperthermia': Interaction with β 3-adrenergic receptor-mediated thermogenesis and significance for the expression of social behavior in male and female mice. Horm Behav 2021; 131:104981. [PMID: 33878523 DOI: 10.1016/j.yhbeh.2021.104981] [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: 07/19/2020] [Revised: 04/02/2021] [Accepted: 04/03/2021] [Indexed: 02/07/2023]
Abstract
Oxytocin (OT) is a critical regulator of multiple facets of energy homeostasis, including brown adipose tissue (BAT) thermogenesis. Nevertheless, it is unclear what, if any, consequence the thermoregulatory and metabolic effects of OT have for the display of social behavior in adult rodents. Here, we examine the contribution of the OT receptor (OTR) and β3 adrenergic receptor (β3AR) to the increase in body temperature that typically accompanies social interaction (i.e., social hyperthermia; SH) and whether SH relates to the expression of social behavior in adult mice. Specifically, we examined how OTR antagonism via peripheral injection of L-368,899 (10 mg/kg) affects the expression of social behavior in C57BL/6J mice, in the presence of active/agonized versus antagonized β3AR, the receptor known to mediate stress-induced BAT thermogenesis. After drug treatment and a 30 min delay, mice were provided a 10 min social interaction test with an unfamiliar, same-sex conspecific. We hypothesized that OTR and β3AR/BAT interact to influence behavior during social interaction, with at least some effects of OT on social behavior dependent upon OT's thermal effects via β3AR/BAT. We found that OTR-mediated temperature elevation is largely responsible for SH during social interaction in mice-albeit not substantially via β3AR-dependent BAT thermogenesis. Further, our results reveal a complex relationship between OTR, β3AR, social hyperthermia and the display of specific social behaviors, with SH most closely associated with anxiety and/or vigilance-related behaviors-that is, behaviors that antagonize or interfere with the initiation of close, non-agonistic social behavior.
Collapse
Affiliation(s)
- Christopher Harshaw
- Department of Psychology, University of New Orleans, New Orleans, LA, United States of America.
| | - Jessica Lanzkowsky
- Department of Psychology, University of New Orleans, New Orleans, LA, United States of America
| | | | - Alana Rose Bradley
- Department of Psychology, University of New Orleans, New Orleans, LA, United States of America
| | - Mark Jaime
- Division of Science, Indiana University-Purdue University, Columbus, Columbus, IN, United States of America
| |
Collapse
|
14
|
Chaki S. Vasopressin V1B Receptor Antagonists as Potential Antidepressants. Int J Neuropsychopharmacol 2021; 24:450-463. [PMID: 33733667 PMCID: PMC8278797 DOI: 10.1093/ijnp/pyab013] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/04/2021] [Accepted: 03/16/2021] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence shows that certain populations of depressed patients have impaired hypothalamus-pituitary-adrenal (HPA) axis function. Arginine-vasopressin (AVP) is one of the primary factors in HPA axis regulation under stress situations, and AVP and its receptor subtype (V1B receptor) play a pivotal role in HPA axis abnormalities observed in depression. Based on this hypothesis, several non-peptide V1B receptor antagonists have been synthesized, and the efficacies of some V1B receptor antagonists have been investigated in both animals and humans. V1B receptor antagonists exert antidepressant-like effects in several animal models at doses that attenuate the hyperactivity of the HPA axis, and some of their detailed mechanisms have been delineated. These results obtained in animal models were, at least partly, reproduced in clinical trials. At least 2 V1B receptor antagonists (TS-121 and ABT-436) showed tendencies to reduce the depression scores of patients with major depressive disorder at doses that attenuate HPA axis hyperactivity or block the pituitary V1B receptor. Importantly, TS-121 showed a clearer efficacy for patients with higher basal cortisol levels than for those with lower basal cortisol levels, which was consistent with the hypothesis that V1B receptor antagonists may be more effective for patients with HPA axis hyperactivity. Therefore, V1B receptor antagonists are promising approaches for the treatment of depression involving HPA axis impairment such as depression.
Collapse
Affiliation(s)
- Shigeyuki Chaki
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Kita-ku, Saitama, Saitama, Japan,Correspondence: Shigeyuki Chaki, PhD, Research Headquarters, Taisho Pharmaceutical Co., Ltd., 1–403 Yoshino-cho, Kita-ku, Saitama, Saitama 331–9530, Japan ()
| |
Collapse
|
15
|
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: 2.7] [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
|
16
|
Ferrer-Pérez C, Reguilón MD, Miñarro J, Rodríguez-Arias M. Oxytocin Signaling as a Target to Block Social Defeat-Induced Increases in Drug Abuse Reward. Int J Mol Sci 2021; 22:ijms22052372. [PMID: 33673448 PMCID: PMC7956822 DOI: 10.3390/ijms22052372] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 12/15/2022] Open
Abstract
There is huge scientific interest in the neuropeptide oxytocin (OXT) due to its putative capacity to modulate a wide spectrum of physiological and cognitive processes including motivation, learning, emotion, and the stress response. The present review seeks to increase the understanding of the role of OXT in an individual’s vulnerability or resilience with regard to developing a substance use disorder. It places specific attention on the role of social stress as a risk factor of addiction, and explores the hypothesis that OXT constitutes a homeostatic response to stress that buffers against its negative impact. For this purpose, the review summarizes preclinical and clinical literature regarding the effects of OXT in different stages of the addiction cycle. The current literature affirms that a well-functioning oxytocinergic system has protective effects such as the modulation of the initial response to drugs of abuse, the attenuation of the development of dependence, the blunting of drug reinstatement and a general anti-stress effect. However, this system is dysregulated if there is continuous drug use or chronic exposure to stress. In this context, OXT is emerging as a promising pharmacotherapy to restore its natural beneficial effects in the organism and to help rebalance the functions of the addicted brain.
Collapse
Affiliation(s)
- Carmen Ferrer-Pérez
- Department of Psychology and Sociology, University of Zaragoza, C/Ciudad Escolar s/n, 44003 Teruel, Spain;
| | - Marina D. Reguilón
- Unit of Research Psychobiology of Drug Dependence, Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain; (M.D.R.); (J.M.)
| | - José Miñarro
- Unit of Research Psychobiology of Drug Dependence, Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain; (M.D.R.); (J.M.)
| | - Marta Rodríguez-Arias
- Unit of Research Psychobiology of Drug Dependence, Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain; (M.D.R.); (J.M.)
- Correspondence:
| |
Collapse
|
17
|
Hou W, He Z, Yang Y, Yuan W, Wang L, Zhang J, Zhang X, Cai W, Guo Q, Tai F. The involvement of oxytocin in the effects of chronic social defeat stress on emotional behaviours in adult female mandarin voles. Eur J Neurosci 2020; 52:2853-2872. [PMID: 32011013 DOI: 10.1111/ejn.14691] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/31/2019] [Accepted: 01/23/2020] [Indexed: 12/26/2022]
Abstract
Chronic social defeat stress (CSDS) can induce anxiety and depression in male rodents, but the prevalence of anxiety and depression is much higher in females, and effects of CSDS on adult females and its underlying mechanism remain unclear. Oxytocin is a stress-buffering hormone in the brain that modulates the physiological effects of stress. Strikingly, research regarding the effect of oxytocin on emotional changes caused by CSDS is still lacking in females. Thus, we focused on the involvement of the oxytocin system in changes in emotional regulation induced by CSDS in female voles. Seventy-day-old female mandarin voles (Microtus mandarinus) were exposed to aggressive adult females for 14 days, and the effects of CSDS on emotion and regulation of oxytocin system were characterized. In addition, we injected vehicle, oxytocin and oxytocin receptor antagonist into the nucleus accumbens (Nacc) of female voles to investigate the involvement of Nacc oxytocin in the effect of CSDS on emotion. Herein, we reported that CSDS increased anxiety and depression-like behaviour and the circulating level of corticosterone, but decreased the number of oxytocin projections and the protein and mRNA expression levels of oxytocin receptor in the Nacc. Injection of oxytocin into the Nacc reversed the effects of CSDS on anxiety-like and depressive-like behaviour, whereas combined injections of oxytocin and oxytocin receptor antagonist eliminated these effects. In conclusion, CSDS increases the levels of anxiety and depression possibly via a reduction in oxytocin projections and the oxytocin receptor level in the Nacc. Nacc oxytocin may be involved in the effects of CSDS on emotional behaviours.
Collapse
Affiliation(s)
- Wenjuan Hou
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Zhixiong He
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yang Yang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Wei Yuan
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Limin Wang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Jing Zhang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xueni Zhang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Wenqi Cai
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Qianqian Guo
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Fadao Tai
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
18
|
Wsol A, Wojno O, Puchalska L, Wrzesien R, Szczepanska-Sadowska E, Cudnoch-Jedrzejewska A. Impaired hypotensive effects of centrally acting oxytocin in SHR and WKY rats exposed to chronic mild stress. Am J Physiol Regul Integr Comp Physiol 2020; 318:R160-R172. [DOI: 10.1152/ajpregu.00050.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The present study was designed to determine the role of centrally acting oxytocin (OT) in the regulation of blood pressure during chronic mild stress (CMS) in spontaneously hypertensive (SHR; n = 36) and normotensive Wistar-Kyoto (WKY; n = 38) rats. The rats were implanted with osmotic minipumps for intracerebroventricular infusions of 0.9% NaCl, OT, and oxytocin receptor antagonist (OTANT) and divided into two groups: SHR and WKY 1) exposed to 4-wk CMS and 2) not exposed to stress (controls). After 4 wk, hemodynamic parameters were recorded at rest and after an application of acute stressor [air-jet stress (AJS)]. Resting mean arterial blood pressure (MAP) was significantly lower in CMS-exposed SHR and WKY infused with OT than in the corresponding groups receiving saline. Exposure to CMS exaggerated the AJS-dependent pressor response in WKY receiving saline but not in the corresponding group of SHR. OT infusion reduced the AJS-dependent pressor response in both CMS-exposed and not exposed SHR and in CMS-exposed WKY. Intracerebroventricular infusion of OTANT potentiated the AJS-dependent pressor response in both stressed and not stressed WKY rats but not in SHR. The results show that centrally delivered OT decreases resting MAP during CMS in both SHR and WKY rats and that in SHR it reduces pressor responses to AJS under control and CMS conditions, whereas in WKY this effect is significant only after CMS exposure. The study indicates that endogenous centrally acting OT may play an essential role in buffering pressor responses to AJS in CMS-exposed and not exposed WKY rats and that this function is significantly impaired in SHR.
Collapse
Affiliation(s)
- A. Wsol
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - O. Wojno
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - L. Puchalska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - R. Wrzesien
- Department of Animal Breeding, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - E. Szczepanska-Sadowska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - A. Cudnoch-Jedrzejewska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
19
|
Varlinskaya EI, Hosová D, Towner T, Werner DF, Spear LP. Effects of chronic intermittent ethanol exposure during early and late adolescence on anxiety-like behaviors and behavioral flexibility in adulthood. Behav Brain Res 2019; 378:112292. [PMID: 31626849 DOI: 10.1016/j.bbr.2019.112292] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/19/2019] [Accepted: 10/07/2019] [Indexed: 12/31/2022]
Abstract
Although both humans and laboratory rodents demonstrate cognitive and affective alterations associated with adolescent alcohol exposure, it is still unknown whether the consequences of early initiation of alcohol use differ from those of later binge drinking within the adolescent developmental period. The present study was designed to assess the effects of early and late AIE on (1) anxiety-like behavior under social (modified social interaction test) and non-social test circumstances (modified light/dark box test, elevated plus maze), and (2) behavioral flexibility, indexed via set shifting in males and females. Early-mid adolescent intermittent exposure (early AIE) occurred between postnatal days (P) 25 and 45, whereas late adolescent intermittent exposure (late AIE) was conducted between P45 and P65, with behavioral testing initiated not earlier than 25 days after repeated exposure to ethanol (4.0 g/kg intragastrically, every other day for a total of 11 exposures). Anxiety-like behavior on the EPM was evident in males and females following early AIE, whereas only males demonstrated non-social anxiety on the EPM following late AIE. Social anxiety-like alterations and deficits in behavioral flexibility were evident only in males following early AIE. Taken together, the results of the present study demonstrate a particular vulnerability of young adolescent males to long-lasting detrimental effects of repeated ethanol and an insensitivity of older adolescent females to the intermittent ethanol exposure paradigm.
Collapse
Affiliation(s)
- Elena I Varlinskaya
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Dominika Hosová
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Trevor Towner
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - David F Werner
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States.
| | - Linda P Spear
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| |
Collapse
|
20
|
Lee W, Hiura LC, Yang E, Broekman KA, Ophir AG, Curley JP. Social status in mouse social hierarchies is associated with variation in oxytocin and vasopressin 1a receptor densities. Horm Behav 2019; 114:104551. [PMID: 31279703 DOI: 10.1016/j.yhbeh.2019.06.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 06/03/2019] [Accepted: 06/28/2019] [Indexed: 01/15/2023]
Abstract
The neuropeptides oxytocin and vasopressin and their receptors have established roles in the regulation of mammalian social behavior including parental care, sex, affiliation and pair-bonding, but less is known regarding their relationship to social dominance and subordination within social hierarchies. We have previously demonstrated that male mice can form stable linear dominance hierarchies with individuals occupying one of three classes of social status: alpha, subdominant, subordinate. Alpha males exhibit high levels of aggression and rarely receive aggression. Subdominant males exhibit aggression towards subordinate males but also receive aggression from more dominant individuals. Subordinate males rarely exhibit aggression and receive aggression from more dominant males. Here, we examined whether variation in social status was associated with levels of oxytocin (OTR) and vasopressin 1a (V1aR) receptor binding in socially relevant brain regions. We found that socially dominant males had significantly higher OTR binding in the nucleus accumbens core than subordinate animals. Alpha males also had higher OTR binding in the anterior olfactory nucleus, posterior part of the cortical amygdala and rostral lateral septum compared to more subordinate individuals. Conversely, alpha males had lower V1aR binding in the rostral lateral septum and lateral preoptic area compared to subordinates. These observed relationships have two potential explanations. Preexisting individual differences in the patterns of OTR and V1aR binding may underlie behavioral differences that promote or inhibit the acquisition of social status. More likely, the differential social environments experienced by dominant and subordinate animals may shift receptor expression, potentially facilitating the expression of adaptive social behaviors.
Collapse
Affiliation(s)
- Won Lee
- Department of Psychology, Columbia University, New York, NY, USA
| | - Lisa C Hiura
- Department of Psychology, Cornell University, Ithaca, NY, USA
| | - Eilene Yang
- Department of Psychology, Columbia University, New York, NY, USA
| | - Katherine A Broekman
- Department of Psychology, Columbia University, New York, NY, USA; SUNY Stony Brook University, Stony Brook, NY, USA
| | | | - James P Curley
- Department of Psychology, Columbia University, New York, NY, USA; Center for Integrative Animal Behavior, Columbia University, New York, NY, USA; Department of Psychology, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
21
|
The Impact of Ethologically Relevant Stressors on Adult Mammalian Neurogenesis. Brain Sci 2019; 9:brainsci9070158. [PMID: 31277460 PMCID: PMC6680763 DOI: 10.3390/brainsci9070158] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/30/2019] [Accepted: 07/02/2019] [Indexed: 12/18/2022] Open
Abstract
Adult neurogenesis—the formation and functional integration of adult-generated neurons—remains a hot neuroscience topic. Decades of research have identified numerous endogenous (such as neurotransmitters and hormones) and exogenous (such as environmental enrichment and exercise) factors that regulate the various neurogenic stages. Stress, an exogenous factor, has received a lot of attention. Despite the large number of reviews discussing the impact of stress on adult neurogenesis, no systematic review on ethologically relevant stressors exists to date. The current review details the effects of conspecifically-induced psychosocial stress (specifically looking at the lack or disruption of social interactions and confrontation) as well as non-conspecifically-induced stress on mammalian adult neurogenesis. The underlying mechanisms, as well as the possible functional role of the altered neurogenesis level, are also discussed. The reviewed data suggest that ethologically relevant stressors reduce adult neurogenesis.
Collapse
|
22
|
Neumann ID, Landgraf R. Tracking oxytocin functions in the rodent brain during the last 30 years: From push-pull perfusion to chemogenetic silencing. J Neuroendocrinol 2019; 31:e12695. [PMID: 30748037 DOI: 10.1111/jne.12695] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 12/22/2022]
Abstract
A short overview is provided of the last 30 years of oxytocin (and vasopressin) research performed in our laboratories, starting with attempts to monitor the release of this nonapeptide in the rodent brain during physiological conditions such as suckling in the lactating animal. Using push-pull perfusion and microdialysis approaches, release patterns in hypothalamic and limbic brain regions could be characterised to occur from intact neuronal structures, to be independent of peripheral secretion into blood, and to respond differentially to various stimuli, particularly those related to reproduction and stress. Parallel efforts focused on the functional impact of central oxytocin release, including neuroendocrine and behavioural effects mediated by nonapeptide receptor interactions and subsequent intraneuronal signalling cascades. The use of a variety of sophisticated behavioural paradigms to manipulate central oxytocin release, along with pharmacological, genetic and pharmacogenetic approaches, revealed multiple consequences on social behaviours, particularly social fear.
Collapse
Affiliation(s)
- Inga D Neumann
- Department of Behavioural and Molecular Neurobiology, Regensburg Centre of Neurosciences, University of Regensburg, Regensburg, Germany
| | | |
Collapse
|
23
|
Lee NS, Beery AK. Neural Circuits Underlying Rodent Sociality: A Comparative Approach. Curr Top Behav Neurosci 2019; 43:211-238. [PMID: 30710222 DOI: 10.1007/7854_2018_77] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
All mammals begin life in social groups, but for some species, social relationships persist and develop throughout the course of an individual's life. Research in multiple rodent species provides evidence of relatively conserved circuitry underlying social behaviors and processes such as social recognition and memory, social reward, and social approach/avoidance. Species exhibiting different complex social behaviors and social systems (such as social monogamy or familiarity preferences) can be characterized in part by when and how they display specific social behaviors. Prairie and meadow voles are closely related species that exhibit similarly selective peer preferences but different mating systems, aiding direct comparison of the mechanisms underlying affiliative behavior. This chapter draws on research in voles as well as other rodents to explore the mechanisms involved in individual social behavior processes, as well as specific complex social patterns. Contrasts between vole species exemplify how the laboratory study of diverse species improves our understanding of the mechanisms underlying social behavior. We identify several additional rodent species whose interesting social structures and available ecological and behavioral field data make them good candidates for study. New techniques and integration across laboratory and field settings will provide exciting opportunities for future mechanistic work in non-model species.
Collapse
Affiliation(s)
- Nicole S Lee
- Neuroscience and Behavior Program, University of Massachusetts, Amherst, MA, USA.
| | - Annaliese K Beery
- Neuroscience and Behavior Program, University of Massachusetts, Amherst, MA, USA. .,Department of Psychology, Smith College, Northampton, MA, USA. .,Neuroscience Program, Smith College, Northampton, MA, USA.
| |
Collapse
|
24
|
Harper KM, Knapp DJ, Criswell HE, Breese GR. Vasopressin and alcohol: a multifaceted relationship. Psychopharmacology (Berl) 2018; 235:3363-3379. [PMID: 30392132 PMCID: PMC6286152 DOI: 10.1007/s00213-018-5099-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 10/28/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Arginine vasopressin (VP) has been implicated in a number of neuropsychiatric disorders with an emphasis on situations where stress increased the severity of the disorder. Based on this hypothesized role for VP in neuropsychiatric disorders, much research is currently being undertaken in humans and animals to test VP as a target for treatment of a number of these disorders including alcohol abuse. OBJECTIVES To provide a summary of the literature regarding the role of VP in alcohol- and stress-related behaviors including the use of drugs that target VP in clinical trials. RESULTS Changes in various components of the VP system occur with alcohol and stress. Manipulating VP or its receptors can alter alcohol- and stress-related behaviors including tolerance to alcohol, alcohol drinking, and anxiety-like behavior. Finally, the hypothalamic-pituitary-adrenal axis response to alcohol is also altered by manipulating the VP system. However, clinical trials of VP antagonists have had mixed results. CONCLUSIONS A review of VP's involvement in alcohol's actions demonstrates that there is much to be learned about brain regions involved in VP-mediated effects on behavior. Thus, future work should focus on elucidating relevant brain regions. By using previous knowledge of the actions of VP and determining the brain regions and/or systems involved in its different behavioral effects, it may be possible to identify a specific receptor subtype target, drug treatment combination, or specific clinical contexts that may point toward a more successful treatment.
Collapse
Affiliation(s)
- Kathryn M Harper
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB #7178, Thurston Bowles Building, Chapel Hill, NC, 27599-7178, USA.
| | - Darin J Knapp
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB #7178, Thurston Bowles Building, Chapel Hill, NC, 27599-7178, USA
- Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7178, USA
| | - Hugh E Criswell
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB #7178, Thurston Bowles Building, Chapel Hill, NC, 27599-7178, USA
- Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7178, USA
| | - George R Breese
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB #7178, Thurston Bowles Building, Chapel Hill, NC, 27599-7178, USA
- Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7178, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7178, USA
| |
Collapse
|
25
|
Ferrer-Pérez C, Castro-Zavala A, Luján MÁ, Filarowska J, Ballestín R, Miñarro J, Valverde O, Rodríguez-Arias M. Oxytocin prevents the increase of cocaine-related responses produced by social defeat. Neuropharmacology 2018; 146:50-64. [PMID: 30448423 DOI: 10.1016/j.neuropharm.2018.11.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022]
Abstract
The neuropeptide oxytocin (OXT) plays a critical role in the regulation of social and emotional behaviors. OXT plays a role in stress response and in drug reward, but to date no studies have evaluated its implication in the long-lasting increase of the motivational effects of cocaine induced by repeated social defeat (RSD). During the social defeat procedure, 1 mg/kg of OXT was administered 30 min before each episode of RSD. Three weeks after the last defeat, the effects of cocaine on the conditioned place preference (CPP), locomotor sensitization and the self-administration (SA) paradigms were evaluated. The influence of OXT on the levels of BDNF in the prefrontal cortex (PFC), striatum and hippocampus was also measured. Our results confirm that raising the levels of OXT during social defeat stress can block the long-lasting effects of this type of stress. OXT counteracts the anxiety induced by social defeat and modifies BDNF levels in all the structures we have studied. Moreover, OXT prevents RSD-induced increases in the motivational effects of cocaine. Administration of OXT before each social defeat blocked the social defeat-induced increment in the conditioned rewarding effects of cocaine in the CPP, favored the extinction of cocaine-associated memories in both the CPP and SA, and decreased reinstatement of cocaine-seeking behavior in the SA. In conclusion, the long-lasting effects of RSD are counteracted by administering OXT prior to stress, and changes in BDNF expression may underlie these protective effects.
Collapse
Affiliation(s)
- Carmen Ferrer-Pérez
- Unit of Research on Psychobiology of Drug Dependence, Department of Psychobiology, Faculty of Psychology, Universitat de València, Valencia, Spain
| | - Adriana Castro-Zavala
- Neurobiology of Behavior Research Group (GReNeC-NeuroBio), Department of Health and Experimental Sciences, University Pompeu Fabra, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Miguel Ángel Luján
- Neurobiology of Behavior Research Group (GReNeC-NeuroBio), Department of Health and Experimental Sciences, University Pompeu Fabra, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Joanna Filarowska
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a, 20-093, Lublin, Poland
| | - Raúl Ballestín
- Unit of Research on Psychobiology of Drug Dependence, Department of Psychobiology, Faculty of Psychology, Universitat de València, Valencia, Spain
| | - José Miñarro
- Unit of Research on Psychobiology of Drug Dependence, Department of Psychobiology, Faculty of Psychology, Universitat de València, Valencia, Spain
| | - Olga Valverde
- Neurobiology of Behavior Research Group (GReNeC-NeuroBio), Department of Health and Experimental Sciences, University Pompeu Fabra, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Marta Rodríguez-Arias
- Unit of Research on Psychobiology of Drug Dependence, Department of Psychobiology, Faculty of Psychology, Universitat de València, Valencia, Spain.
| |
Collapse
|
26
|
Oxytocin and vasopressin modulation of social anxiety following adolescent intermittent ethanol exposure. Psychopharmacology (Berl) 2018; 235:3065-3077. [PMID: 30141056 PMCID: PMC6456069 DOI: 10.1007/s00213-018-5003-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 08/16/2018] [Indexed: 02/08/2023]
Abstract
RATIONALE Adolescent intermittent ethanol exposure (AIE) produces lasting, sex-specific social anxiety-like alterations in male, but not female rats. Oxytocin (OXT) and vasopressin (AVP) brain systems play opposite roles in regulating social preference/avoidance, with OXT increasing approach to, and AVP increasing avoidance of social stimuli. OBJECTIVES To test the hypothesis that social anxiety-like alterations seen in adult males after AIE are associated with a shift in the balance between OXT and AVP toward AVP, effectiveness of pharmacological activation of the OXT system and blockade of endogenous activity at AVP receptors for reversing AIE-induced social anxiety-like alterations was assessed, along with examination of the effects of AIE on OXT, vasopressin V1a, and V1b receptor (OXT-R, V1a-R, and V1b-R) surface expression in the hypothalamus. METHODS Sprague-Dawley male and female rats were given 4 g/kg ethanol (AIE) or water intragastrically every 48 h for a total of 11 exposures during postnatal days (P) 25-45. On P70-72, animals were given a social interaction test following administration of a selective OXT-R agonist WAY-267464, selective V1a-R antagonist SR-49059, or V1b-R antagonist SSR-149415, and hypothalamic tissue was collected. RESULTS Social anxiety-like behavior was induced by AIE in males but not females, and was selectively reversed by the selective OXT-R agonist and V1b-R antagonist, but not V1a-R antagonist. AIE was also found to decrease OXT-R, but increase V1b-R neuronal surface expression relative to water-exposed controls in the hypothalamus of males, but not females. CONCLUSIONS These findings demonstrate that AIE induces changes in OXT-R and AVP-R surface expression in the hypothalamus along with social anxiety-like alterations in male rats. These social anxiety-like alterations can be reversed either by activation of the OXT system or by suppression of the AVP system, data that support the hypothesis that social anxiety-like alterations induced by adolescent alcohol exposure in male rats are associated at least in part with an OXT/AVP imbalance.
Collapse
|
27
|
Newman EL, Leonard MZ, Arena DT, de Almeida RMM, Miczek KA. Social defeat stress and escalation of cocaine and alcohol consumption: Focus on CRF. Neurobiol Stress 2018; 9:151-165. [PMID: 30450381 PMCID: PMC6236516 DOI: 10.1016/j.ynstr.2018.09.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/10/2018] [Accepted: 09/17/2018] [Indexed: 12/15/2022] Open
Abstract
Both the ostensibly aversive effects of unpredictable episodes of social stress and the intensely rewarding effects of drugs of abuse activate the mesocorticolimbic dopamine systems. Significant neuroadaptations in interacting stress and reward neurocircuitry may underlie the striking connection between stress and substance use disorders. In rodent models, recurring intermittent exposure to social defeat stress appears to produce a distinct profile of neuroadaptations that translates most readily to the repercussions of social stress in humans. In the present review, preclinical rodent models of social defeat stress and subsequent alcohol, cocaine or opioid consumption are discussed with regard to: (1) the temporal pattern of social defeat stress, (2) male and female protocols of social stress-escalated drug consumption, and (3) the neuroplastic effects of social stress, which may contribute to escalated drug-taking. Neuroadaptations in corticotropin-releasing factor (CRF) and CRF modulation of monoamines in the ventral tegmental area and the bed nucleus of the stria terminalis are highlighted as potential mechanisms underlying stress-escalated drug consumption. However, the specific mechanisms that drive CRF-mediated increases in dopamine require additional investigation as do the stress-induced neuroadaptations that may contribute to the development of compulsive patterns of drug-taking.
Collapse
Affiliation(s)
- Emily L Newman
- Psychology Dept., Tufts University, Medford, MA, 02155, USA
| | | | | | - Rosa M M de Almeida
- Institute of Psychology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Klaus A Miczek
- Psychology Dept., Tufts University, Medford, MA, 02155, USA.,Dept. of Neuroscience, Sackler School of Graduate Biomedical Sciences, Boston, MA, 02111, USA
| |
Collapse
|
28
|
Masis-Calvo M, Schmidtner AK, de Moura Oliveira VE, Grossmann CP, de Jong TR, Neumann ID. Animal models of social stress: the dark side of social interactions. Stress 2018; 21:417-432. [PMID: 29745275 DOI: 10.1080/10253890.2018.1462327] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Social stress occurs in all social species, including humans, and shape both mental health and future interactions with conspecifics. Animal models of social stress are used to unravel the precise role of the main stress system - the HPA axis - on the one hand, and the social behavior network on the other, as these are intricately interwoven. The present review aims to summarize the insights gained from three highly useful and clinically relevant animal models of psychosocial stress: the resident-intruder (RI) test, the chronic subordinate colony housing (CSC), and the social fear conditioning (SFC). Each model brings its own focus: the role of the HPA axis in shaping acute social confrontations (RI test), the physiological and behavioral impairments resulting from chronic exposure to negative social experiences (CSC), and the neurobiology underlying social fear and its effects on future social interactions (SFC). Moreover, these models are discussed with special attention to the HPA axis and the neuropeptides vasopressin and oxytocin, which are important messengers in the stress system, in emotion regulation, as well as in the social behavior network. It appears that both nonapeptides balance the relative strength of the stress response, and simultaneously predispose the animal to positive or negative social interactions.
Collapse
Affiliation(s)
- Marianela Masis-Calvo
- a Department of Behavioral and Molecular Neurobiology , University of Regensburg , Regensburg , Germany
| | - Anna K Schmidtner
- a Department of Behavioral and Molecular Neurobiology , University of Regensburg , Regensburg , Germany
| | | | - Cindy P Grossmann
- a Department of Behavioral and Molecular Neurobiology , University of Regensburg , Regensburg , Germany
| | - Trynke R de Jong
- a Department of Behavioral and Molecular Neurobiology , University of Regensburg , Regensburg , Germany
- b Medische Biobank Noord-Nederland B.V , Groningen , Netherlands
| | - Inga D Neumann
- a Department of Behavioral and Molecular Neurobiology , University of Regensburg , Regensburg , Germany
| |
Collapse
|
29
|
Jurek B, Neumann ID. The Oxytocin Receptor: From Intracellular Signaling to Behavior. Physiol Rev 2018; 98:1805-1908. [DOI: 10.1152/physrev.00031.2017] [Citation(s) in RCA: 408] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The many facets of the oxytocin (OXT) system of the brain and periphery elicited nearly 25,000 publications since 1930 (see FIGURE 1 , as listed in PubMed), which revealed central roles for OXT and its receptor (OXTR) in reproduction, and social and emotional behaviors in animal and human studies focusing on mental and physical health and disease. In this review, we discuss the mechanisms of OXT expression and release, expression and binding of the OXTR in brain and periphery, OXTR-coupled signaling cascades, and their involvement in behavioral outcomes to assemble a comprehensive picture of the central and peripheral OXT system. Traditionally known for its role in milk let-down and uterine contraction during labor, OXT also has implications in physiological, and also behavioral, aspects of reproduction, such as sexual and maternal behaviors and pair bonding, but also anxiety, trust, sociability, food intake, or even drug abuse. The many facets of OXT are, on a molecular basis, brought about by a single receptor. The OXTR, a 7-transmembrane G protein-coupled receptor capable of binding to either Gαior Gαqproteins, activates a set of signaling cascades, such as the MAPK, PKC, PLC, or CaMK pathways, which converge on transcription factors like CREB or MEF-2. The cellular response to OXT includes regulation of neurite outgrowth, cellular viability, and increased survival. OXTergic projections in the brain represent anxiety and stress-regulating circuits connecting the paraventricular nucleus of the hypothalamus, amygdala, bed nucleus of the stria terminalis, or the medial prefrontal cortex. Which OXT-induced patterns finally alter the behavior of an animal or a human being is still poorly understood, and studying those OXTR-coupled signaling cascades is one initial step toward a better understanding of the molecular background of those behavioral effects.
Collapse
Affiliation(s)
- Benjamin Jurek
- Department of Behavioural and Molecular Neurobiology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| | - Inga D. Neumann
- Department of Behavioural and Molecular Neurobiology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
30
|
Borrow AP, Bales NJ, Stover SA, Handa RJ. Chronic Variable Stress Induces Sex-Specific Alterations in Social Behavior and Neuropeptide Expression in the Mouse. Endocrinology 2018; 159:2803-2814. [PMID: 29788320 PMCID: PMC6692887 DOI: 10.1210/en.2018-00217] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/10/2018] [Indexed: 12/11/2022]
Abstract
Chronic exposure to stressors impairs the function of multiple organ systems and has been implicated in increased disease risk. In the rodent, the chronic variable stress (CVS) paradigm has successfully modeled several stress-related illnesses. Despite striking disparities between men and women in the prevalence and etiology of disorders associated with chronic stress, most preclinical research examining chronic stressor exposure has focused on male subjects. One potential mediator of the consequences of CVS is oxytocin (OT), a known regulator of stress neurocircuitry and behavior. To ascertain the sex-specific effects of CVS in the C57BL/6 mouse on OT and the structurally similar neuropeptide arginine vasopressin (AVP), the numbers of immunoreactive and mRNA-containing neurons in the paraventricular nucleus (PVN) and supraoptic nucleus (SON) were determined using immunohistochemistry and in situ hybridization, respectively. In addition, the mice underwent a battery of behavioral tests to determine whether CVS affects social behaviors known to be regulated by OT and AVP. Six weeks of CVS increased sociability in the female mouse and decreased PVN OT immunoreactivity (ir) and AVP mRNA. In the male mice, CVS decreased PVN OT mRNA but had no effect on social behavior, AVP, or OT-ir. CVS also increased the soma volume for PVN OT neurons. In contrast, OT and AVP neurons in the SON were unaffected by CVS treatment. These findings demonstrate clear sex differences in the effects of CVS on neuropeptides in the mouse, suggest a pathway through which CVS alters sociability and stress-coping responses in females and reveals a vulnerability to CVS in the C57BL/6 mouse strain.
Collapse
Affiliation(s)
- Amanda P Borrow
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Natalie J Bales
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Sally A Stover
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Robert J Handa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
- Correspondence: Robert J. Handa, PhD, Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, Colorado 80523. E-mail:
| |
Collapse
|
31
|
Wacker D, Ludwig M. The role of vasopressin in olfactory and visual processing. Cell Tissue Res 2018; 375:201-215. [PMID: 29951699 PMCID: PMC6335376 DOI: 10.1007/s00441-018-2867-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/31/2018] [Indexed: 12/23/2022]
Abstract
Neural vasopressin is a potent modulator of behaviour in vertebrates. It acts at both sensory processing regions and within larger regulatory networks to mediate changes in social recognition, affiliation, aggression, communication and other social behaviours. There are multiple populations of vasopressin neurons within the brain, including groups in olfactory and visual processing regions. Some of these vasopressin neurons, such as those in the main and accessory olfactory bulbs, anterior olfactory nucleus, piriform cortex and retina, were recently identified using an enhanced green fluorescent protein-vasopressin (eGFP-VP) transgenic rat. Based on the interconnectivity of vasopressin-producing and sensitive brain areas and in consideration of autocrine, paracrine and neurohormone-like actions associated with somato-dendritic release, we discuss how these different neuronal populations may interact to impact behaviour.
Collapse
Affiliation(s)
- Douglas Wacker
- School of STEM (Division of Biological Sciences), University of Washington Bothell, Bothell, WA, USA.
| | - Mike Ludwig
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.,Centre for Neuroendocrinology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
32
|
|
33
|
Differential activation of arginine-vasopressin receptor subtypes in the amygdaloid modulation of anxiety in the rat by arginine-vasopressin. Psychopharmacology (Berl) 2018; 235:1015-1027. [PMID: 29306965 DOI: 10.1007/s00213-017-4817-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 12/19/2017] [Indexed: 01/06/2023]
Abstract
RATIONALE The amygdala plays a paramount role in the modulation of anxiety and numerous studies have shown that arginine vasopressin (AVP) elicits anxiogenic effects following either its systemic or septal administration. OBJECTIVES The aim of this paper was to study the involvement of vasopressinergic neurotransmission in the amygdaloid modulation of unconditioned anxiety and to ascertain whether or not AVP receptor subtypes may have a differential role in this modulation. METHODS Anxiety behavior was evaluated both in Shock-Probe Burying Test and Light-Dark Box following the bilateral microinfusion of AVP alone or AVP together with either AVP 1a or AVP 1b receptor antagonists into the central amygdala (CeA). RESULTS AVP microinfusion elicited at low (1 ng/side) but not at high doses (10 ng/side) anxiogenic-like responses in the Shock-Probe Burying Test but not in the Light-Dark Box. SSR149415, an AVP 1b antagonist unlike Manning compound, an AVP 1a antagonist, fully prevented AVP effects in the Shock-Probe Burying Test when it was administered simultaneously with AVP. In addition, oxytocin receptor blockade also failed to affect AVP effects. No effects of any AVP antagonist by itself were observed in both anxiety paradigms. CONCLUSIONS Our results indicate that AVP 1b receptor contribute to the amygdaloid modulation of anxiety at least in the context of the Shock-Probe Burying Test since no effects were noticed in the Light-Dark Box. It remains to the future to ascertain whether AVP receptor subtypes have indeed differential actions either in the modulation of global or specific features of unconditioned anxiety.
Collapse
|
34
|
Lozić M, Šarenac O, Murphy D, Japundžić-Žigon N. Vasopressin, Central Autonomic Control and Blood Pressure Regulation. Curr Hypertens Rep 2018; 20:11. [DOI: 10.1007/s11906-018-0811-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
Nelson BS, Sequeira MK, Schank JR. Bidirectional relationship between alcohol intake and sensitivity to social defeat: association with Tacr1 and Avp expression. Addict Biol 2018; 23:142-153. [PMID: 28150369 DOI: 10.1111/adb.12494] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/01/2017] [Accepted: 01/04/2017] [Indexed: 01/17/2023]
Abstract
While epidemiological studies show that alcohol abuse is often co-morbid with affective disorders, the causal direction of this association is unclear. We examined this relationship using mouse models including social defeat stress (SDS), social interaction (SI) and voluntary alcohol consumption. C57BL6/J mice exposed to SDS segregate into two subpopulations, those that express depressive-like phenotypes ('susceptible') and those that do not ('resilient'). First, we stratified SDS-exposed mice and measured their voluntary alcohol consumption. Next, we determined whether SI behavior in alcohol-naïve mice could predict alcohol intake. Finally, we assessed the effect of binge-like alcohol exposure on sensitivity to SDS. We quantified Tacr1 (neurokinin-1 receptor gene) and Avp (vasopressin peptide gene) mRNA in brain regions involved in depression, addiction and social behavior. We found that susceptible mice consumed more alcohol compared with resilient mice, suggesting that depression-like phenotypes associate with increased alcohol intake. Interestingly, we observed a negative correlation between SI and alcohol intake in stress- and alcohol-naïve mice, suggesting that individual differences in SI associate with alcohol preference. Finally, alcohol pre-treatment increased sensitivity to SDS, indicating that alcohol exposure alters sensitivity to social stress. Quantification of mRNA revealed that increased expression of Tacr1 and Avp generally associated with decreased SI and increased alcohol intake. C57BL6/J mice are an inbred strain; thus, it is likely that individual differences in behavior and gene expression are driven by epigenetic factors. Collectively, these results support a bidirectional relationship between alcohol exposure and susceptibility to stress that is associated with variations in neuropeptide expression.
Collapse
Affiliation(s)
- Britta S. Nelson
- Department of Physiology and Pharmacology, College of Veterinary Medicine; University of Georgia; GA USA
| | - Michelle K. Sequeira
- Department of Physiology and Pharmacology, College of Veterinary Medicine; University of Georgia; GA USA
| | - Jesse R. Schank
- Department of Physiology and Pharmacology, College of Veterinary Medicine; University of Georgia; GA USA
| |
Collapse
|
36
|
Caldwell HK, Aulino EA, Rodriguez KM, Witchey SK, Yaw AM. Social Context, Stress, Neuropsychiatric Disorders, and the Vasopressin 1b Receptor. Front Neurosci 2017; 11:567. [PMID: 29085277 PMCID: PMC5650633 DOI: 10.3389/fnins.2017.00567] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/27/2017] [Indexed: 01/28/2023] Open
Abstract
The arginine vasopressin 1b receptor (Avpr1b) is involved in the modulation of a variety of behaviors and is an important part of the mammalian hormonal stress axis. The Avpr1b is prominent in hippocampal CA2 pyramidal cells and in the anterior pituitary corticotrophs. Decades of research on this receptor has demonstrated its importance to the modulation of social recognition memory, social forms of aggression, and modulation of the hypothalamic-pituitary-adrenal axis, particularly under conditions of acute stress. Further, work in humans suggests that the Avpr1b may play a role in human neuropsychiatric disorders and its modulation may have therapeutic potential. This paper reviews what is known about the role of the Avpr1b in the context of social behaviors, the stress axis, and human neuropsychiatric disorders. Further, possible mechanisms for how Avpr1b activation within the hippocampus vs. Avpr1b activation within anterior pituitary may interact with one another to affect behavioral output are proposed.
Collapse
Affiliation(s)
- Heather K Caldwell
- Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences Kent State University, Kent, OH, United States.,School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Elizabeth A Aulino
- Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences Kent State University, Kent, OH, United States
| | - Karla M Rodriguez
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Shannah K Witchey
- Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences Kent State University, Kent, OH, United States
| | - Alexandra M Yaw
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| |
Collapse
|
37
|
Bodden C, van den Hove D, Lesch KP, Sachser N. Impact of varying social experiences during life history on behaviour, gene expression, and vasopressin receptor gene methylation in mice. Sci Rep 2017; 7:8719. [PMID: 28821809 PMCID: PMC5562890 DOI: 10.1038/s41598-017-09292-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/24/2017] [Indexed: 01/12/2023] Open
Abstract
Both negative and positive social experiences during sensitive life phases profoundly shape brain and behaviour. Current research is therefore increasingly focusing on mechanisms mediating the interaction between varying life experiences and the epigenome. Here, male mice grew up under either adverse or beneficial conditions until adulthood, when they were subdivided into groups exposed to situations that either matched or mismatched previous conditions. It was investigated whether the resulting four life histories were associated with changes in anxiety-like behaviour, gene expression of selected genes involved in anxiety and stress circuits, and arginine vasopressin receptor 1a (Avpr1a) gene methylation. Varying experiences during life significantly modulated (1) anxiety-like behaviour; (2) hippocampal gene expression of Avpr1a, serotonin receptor 1a (Htr1a), monoamine oxidase A (Maoa), myelin basic protein (Mbp), glucocorticoid receptor (Nr3c1), growth hormone (Gh); and (3) hippocampal DNA methylation within the Avpr1a gene. Notably, mice experiencing early beneficial and later adverse conditions showed a most pronounced downregulation of Avpr1a expression, accompanied by low anxiety-like behaviour. This decrease in Avpr1a expression may have been, in part, a consequence of increased methylation in the Avpr1a gene. In summary, this study highlights the impact of interactive social experiences throughout life on the hippocampal epigenotype and associated behaviour.
Collapse
Affiliation(s)
- Carina Bodden
- Department of Behavioural Biology, University of Münster, Münster, Germany. .,Otto Creutzfeldt Center for Cognitive and Behavioral Neuroscience, University of Münster, Münster, Germany.
| | - Daniel van den Hove
- Division of Molecular Psychiatry, Center of Mental Health, Laboratory of Translational Neuroscience, University of Würzburg, Würzburg, Germany.,Department of Translational Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, Laboratory of Translational Neuroscience, University of Würzburg, Würzburg, Germany.,Department of Translational Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Norbert Sachser
- Department of Behavioural Biology, University of Münster, Münster, Germany.,Otto Creutzfeldt Center for Cognitive and Behavioral Neuroscience, University of Münster, Münster, Germany
| |
Collapse
|
38
|
Galbusera A, De Felice A, Girardi S, Bassetto G, Maschietto M, Nishimori K, Chini B, Papaleo F, Vassanelli S, Gozzi A. Intranasal Oxytocin and Vasopressin Modulate Divergent Brainwide Functional Substrates. Neuropsychopharmacology 2017; 42:1420-1434. [PMID: 27995932 PMCID: PMC5436116 DOI: 10.1038/npp.2016.283] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 11/25/2016] [Accepted: 12/13/2016] [Indexed: 12/20/2022]
Abstract
The neuropeptides oxytocin (OXT) and vasopressin (AVP) have been identified as modulators of emotional social behaviors and associated with neuropsychiatric disorders characterized by social dysfunction. Experimental and therapeutic use of OXT and AVP via the intranasal route is the subject of extensive clinical research. However, the large-scale functional substrates directly engaged by these peptides and their functional dynamics remain elusive. By using cerebral blood volume (CBV) weighted fMRI in the mouse, we show that intranasal administration of OXT rapidly elicits the transient activation of cortical regions and a sustained activation of hippocampal and forebrain areas characterized by high oxytocin receptor density. By contrast, intranasal administration of AVP produced a robust and sustained deactivation in cortico-parietal, thalamic and mesolimbic regions. Importantly, intravenous administration of OXT and AVP did not recapitulate the patterns of modulation produced by intranasal dosing, supporting a central origin of the observed functional changes. In keeping with this notion, hippocampal local field potential recordings revealed multi-band power increases upon intranasal OXT administration. We also show that the selective OXT-derivative TGOT reproduced the pattern of activation elicited by OXT and that the deletion of OXT receptors does not affect AVP-mediated deactivation. Collectively, our data document divergent modulation of brainwide neural systems by intranasal administration of OXT and AVP, an effect that involves key substrates of social and emotional behavior. The observed divergence calls for a deeper investigation of the systems-level mechanisms by which exogenous OXT and AVP modulate brain function and exert their putative therapeutic effects.
Collapse
Affiliation(s)
- Alberto Galbusera
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto (TN), Italy
| | - Alessia De Felice
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto (TN), Italy
| | - Stefano Girardi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Giacomo Bassetto
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marta Maschietto
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Katsuhiko Nishimori
- Laboratory of Molecular Biology, Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan
| | - Bice Chini
- CNR, Institute of Neuroscience, Milan, Italy,Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Francesco Papaleo
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova, Italy
| | | | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto (TN), Italy,Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto (TN) 38068, Italy, Tel: +39 04648028701, E-mail:
| |
Collapse
|
39
|
Florez Acevedo S, Cardenas Parra LF. Rol Modulador de la Oxitocina en la Interacción Social y el Estrés Social. UNIVERSITAS PSYCHOLOGICA 2017. [DOI: 10.11144/javeriana.upsy15-5.rmoi] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
La Oxitocina es un neuropéptido conocido por facilitar funciones del sistema nervioso periférico, relacionadas específicamente con el sistema reproductivo. Sin embargo, en las últimas décadas se ha reconocido la función moduladora de la Oxitocina en el comportamiento social, a través de su liberación en el sistema nervioso central. Así mismo, estudios han mencionado que la Oxitocina es un potencial ansiolítico cuando un individuo ha sido sometido a estrés social. Por lo tanto, el objetivo de esta revisión es presentar una caracterización de la Oxitocina y su relación con distintas formas de interacción social y el estrés social; a través de los resultados presentados en distintos estudios, tanto en modelos animales como en humanos. Además, se intenta mostrar la importancia de continuar con el estudio de la Oxitocina, dados los posibles vacíos teóricos y experimentales existentes, teniendo en cuenta las potenciales cualidades ansiolíticas de esta hormona.
Collapse
|
40
|
Müller L, Weinert D. Individual recognition of social rank and social memory performance depends on a functional circadian system. Behav Processes 2016; 132:85-93. [DOI: 10.1016/j.beproc.2016.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/22/2016] [Accepted: 10/11/2016] [Indexed: 10/20/2022]
|
41
|
Baracz SJ, Cornish JL. The neurocircuitry involved in oxytocin modulation of methamphetamine addiction. Front Neuroendocrinol 2016; 43:1-18. [PMID: 27546878 DOI: 10.1016/j.yfrne.2016.08.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 07/14/2016] [Accepted: 08/11/2016] [Indexed: 12/13/2022]
Abstract
The role of oxytocin in attenuating the abuse of licit and illicit drugs, including the psychostimulant methamphetamine, has been examined with increased ferocity in recent years. This is largely driven by the potential application of oxytocin as a pharmacotherapy. However, the neural mechanisms by which oxytocin modulates methamphetamine abuse are not well understood. Recent research identified an important role for the accumbens core and subthalamic nucleus in this process, which likely involves an interaction with dopamine, glutamate, GABA, and vasopressin. In addition to providing an overview of methamphetamine, the endogenous oxytocin system, and the effects of exogenous oxytocin on drug abuse, we propose a neural circuit through which exogenous oxytocin modulates methamphetamine abuse, focusing on its interaction with neurochemicals within the accumbens core and subthalamic nucleus. A growing understanding of exogenous oxytocin effects at a neurochemical and neurobiological level will assist in its evaluation as a pharmacotherapy for drug addiction.
Collapse
Affiliation(s)
- Sarah J Baracz
- School of Psychology, University of Sydney, Sydney, NSW 2109, Australia; Department of Psychology, Macquarie University, North Ryde, NSW 2109, Australia.
| | - Jennifer L Cornish
- Department of Psychology, Macquarie University, North Ryde, NSW 2109, Australia.
| |
Collapse
|
42
|
Steinman MQ, Duque-Wilckens N, Greenberg GD, Hao R, Campi KL, Laredo SA, Laman-Maharg A, Manning CE, Doig IE, Lopez EM, Walch K, Bales KL, Trainor BC. Sex-Specific Effects of Stress on Oxytocin Neurons Correspond With Responses to Intranasal Oxytocin. Biol Psychiatry 2016; 80:406-14. [PMID: 26620251 PMCID: PMC4837091 DOI: 10.1016/j.biopsych.2015.10.007] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 09/18/2015] [Accepted: 10/02/2015] [Indexed: 01/13/2023]
Abstract
BACKGROUND Oxytocin (OT) is considered to be a stress-buffering hormone, dampening the physiologic effects of stress. However, OT can also be anxiogenic. We examined acute and long-lasting effects of social defeat on OT neurons in male and female California mice. METHODS We used immunohistochemistry for OT and c-fos cells to examine OT neuron activity immediately after defeat (n = 6-9) and 2 weeks (n = 6-9) and 10 weeks (n = 4-5) later. We quantified Oxt messenger RNA with quantitative polymerase chain reaction (n = 5-9). Intranasal OT was administered to naïve and stressed mice tested in social interaction and resident-intruder tests (n = 8-14). RESULTS Acute exposure to a third episode of defeat increased OT/c-fos colocalizations in the paraventricular nucleus of both sexes. In the medioventral bed nucleus of the stria terminalis, defeat increased Oxt messenger RNA, total OT neurons, and OT/c-fos colocalizations in female mice but not male mice. Intranasal OT failed to reverse stress-induced social withdrawal in female mice and reduced social interaction behavior in female mice naïve to defeat. In contrast, intranasal OT increased social interaction in stressed male mice and reduced freezing in the resident-intruder test. CONCLUSIONS Social defeat induces long-lasting increases in OT production and OT/c-fos cells in the medioventral bed nucleus of the stria terminalis of female mice but not male mice. Intranasal OT largely reversed the effects of stress on behavior in male mice, but effects were mixed in female mice. These results suggest that changes in OT-sensitive networks contribute to sex differences in behavioral responses to stress.
Collapse
Affiliation(s)
- Michael Q. Steinman
- Molecular, Cellular, Integrative Physiology Graduate Group, University of California Davis, CA 95616,Department of Psychology, University of California, Davis, CA 95616
| | - Natalia Duque-Wilckens
- Department of Psychology, University of California, Davis, CA 95616,Animal Behavior Graduate Group, University of California, Davis, CA 95616
| | - Gian D. Greenberg
- Department of Psychology, University of California, Davis, CA 95616,Neuroscience Graduate Group, University of California, Davis, CA 95616
| | - Rebecca Hao
- Department of Psychology, University of California, Davis, CA 95616
| | | | - Sarah A. Laredo
- Department of Psychology, University of California, Davis, CA 95616,Animal Behavior Graduate Group, University of California, Davis, CA 95616
| | - Abigail Laman-Maharg
- Department of Psychology, University of California, Davis, CA 95616,Neuroscience Graduate Group, University of California, Davis, CA 95616
| | | | - Ian E. Doig
- Department of Psychology, University of California, Davis, CA 95616
| | - Eduardo M. Lopez
- Department of Psychology, University of California, Davis, CA 95616
| | - Keenan Walch
- Department of Psychology, University of California, Davis, CA 95616
| | - Karen L. Bales
- Department of Psychology, University of California, Davis, CA 95616
| | - Brian C. Trainor
- Department of Psychology, University of California, Davis, CA 95616,Animal Behavior Graduate Group, University of California, Davis, CA 95616,Neuroscience Graduate Group, University of California, Davis, CA 95616
| |
Collapse
|
43
|
Steinman MQ, Trainor BC. Sex differences in the effects of social defeat on brain and behavior in the California mouse: Insights from a monogamous rodent. Semin Cell Dev Biol 2016; 61:92-98. [PMID: 27375045 DOI: 10.1016/j.semcdb.2016.06.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 06/28/2016] [Accepted: 06/29/2016] [Indexed: 11/20/2022]
Abstract
Women are nearly twice as likely as men to be diagnosed with major depressive disorder, yet the use of female animal models in studying the biological basis of depression lags behind that of males. The social defeat model uses social stress to generate depression-like symptoms in order to study the neurobiological mechanisms. In general, social defeat is difficult to apply in female rodents. However, male and female California mice (Peromyscus californicus) are territorial. This allows defeat to be studied in both sexes. Males exposed to defeat tend to exhibit proactive coping mechanisms and demonstrate aggression and reduced cognitive flexibility. Females exposed to defeat engage more in reactive coping mechanisms which is highlighted by social avoidance and low aggression. Importantly, effects of defeat on social interaction behavior in females is independent of adult gonadal steroids. These behavioral phenotypes are associated with sex-specific changes in arginine vasopressin (AVP) and oxytocin (OT), closely related peptides that regulate social behavior and stress reactivity. In brain regions associated with stress responses and social behavior, defeat induced long term decreases in AVP activity and increases in OT activity in males and females respectively. Intranasal OT administration was shown to mimic the effects of defeat-induced increases in endogenous OT activity, causing social withdrawal in undefeated females. This suggests that inhibition of OT activity could reduce the impact of stress on behavior in females. These results highlight the value of maintaining diverse rodent models in the search for sex-specific pharmacological approaches to treating mood disorders.
Collapse
Affiliation(s)
- Michael Q Steinman
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA 92037, U.S.A
| | - Brian C Trainor
- Department of Psychology and Center for Neuroscience, University of California, Davis, CA 95616, U.S.A..
| |
Collapse
|
44
|
Ng E, Browne CJ, Samsom JN, Wong AHC. Depression and substance use comorbidity: What we have learned from animal studies. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2016; 43:456-474. [PMID: 27315335 DOI: 10.1080/00952990.2016.1183020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Depression and substance use disorders are often comorbid, but the reasons for this are unclear. In human studies, it is difficult to determine how one disorder may affect predisposition to the other and what the underlying mechanisms might be. Instead, animal studies allow experimental induction of behaviors relevant to depression and drug-taking, and permit direct interrogation of changes to neural circuits and molecular pathways. While this field is still new, here we review animal studies that investigate whether depression-like states increase vulnerability to drug-taking behaviors. Since chronic psychosocial stress can precipitate or predispose to depression in humans, we review studies that use psychosocial stressors to produce depression-like phenotypes in animals. Specifically, we describe how postweaning isolation stress, repeated social defeat stress, and chronic mild (or unpredictable) stress affect behaviors relevant to substance abuse, especially operant self-administration. Potential brain changes mediating these effects are also discussed where available, with an emphasis on mesocorticolimbic dopamine circuits. Postweaning isolation stress and repeated social defeat generally increase acquisition or maintenance of drug self-administration, and alter dopamine sensitivity in various brain regions. However, the effects of chronic mild stress on drug-taking have been much less studied. Future studies should consider standardizing stress-induction protocols, including female subjects, and using multi-hit models (e.g. genetic vulnerabilities and environmental stress).
Collapse
Affiliation(s)
- Enoch Ng
- a Lunenfeld-Tanenbaum Research Institute , Mount Sinai Hospital , Toronto , Canada.,b Institute of Medical Science, University of Toronto , Toronto , Canada
| | - Caleb J Browne
- c Department of Psychology , University of Toronto , Toronto , Canada.,d Campbell Family Health Institute , Centre for Addiction and Mental Health , Toronto , Canada
| | - James N Samsom
- d Campbell Family Health Institute , Centre for Addiction and Mental Health , Toronto , Canada.,e Department of Pharmacology , University of Toronto , Toronto , Canada
| | - Albert H C Wong
- b Institute of Medical Science, University of Toronto , Toronto , Canada.,d Campbell Family Health Institute , Centre for Addiction and Mental Health , Toronto , Canada.,e Department of Pharmacology , University of Toronto , Toronto , Canada.,f Department of Psychiatry , University of Toronto , Toronto , Canada
| |
Collapse
|
45
|
Murakami G. Distinct Effects of Estrogen on Mouse Maternal Behavior: The Contribution of Estrogen Synthesis in the Brain. PLoS One 2016; 11:e0150728. [PMID: 27007402 PMCID: PMC4805179 DOI: 10.1371/journal.pone.0150728] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 02/17/2016] [Indexed: 11/18/2022] Open
Abstract
Estrogen surge following progesterone withdrawal at parturition plays an important role in initiating maternal behavior in various rodent species. Systemic estrogen treatment shortens the latency to onset of maternal behavior in nulliparous female rats that have not experienced parturition. In contrast, nulliparous laboratory mice show rapid onset of maternal behavior without estrogen treatment, and the role of estrogen still remains unclear. Here the effect of systemic estrogen treatment (for 2 h, 1 day, 3 days, and 7 days) after progesterone withdrawal was examined on maternal behavior of C57BL/6 mice. This estrogen regimen led to different effects on nursing, pup retrieval, and nest building behaviors. Latency to nursing was shortened by estrogen treatment within 2 h. Moreover, pup retrieval and nest building were decreased. mRNA expression was also investigated for estrogen receptor α (ERα) and for genes involved in regulating maternal behavior, specifically, the oxytocin receptor (OTR) and vasopressin receptor in the medial amygdala (MeA) and medial preoptic area (MPOA). Estrogen treatment led to decreased ERα mRNA in both regions. Although OTR mRNA was increased in the MeA, OTR and vasopressin receptor mRNA were reduced in the MPOA, showing region-dependent transcription regulation. To determine the mechanisms for the actions of estrogen treatment, the contribution of estrogen synthesis in the brain was examined. Blockade of estrogen synthesis in the brain by systemic letrozole treatment in ovariectomized mice interfered with pup retrieval and nest building but not nursing behavior, indicating different contributions of estrogen synthesis to maternal behavior. Furthermore, letrozole treatment led to an increase in ERα mRNA in the MeA but not in the MPOA, suggesting that involvement of estrogen synthesis is brain region dependent. Altogether, these results suggest that region-dependent estrogen synthesis leads to differential transcriptional activation due to exogenous estrogen treatment, and thereby results in different effects on maternal behavior.
Collapse
Affiliation(s)
- Gen Murakami
- Laboratory of Neurobiology and Behavior, The Rockefeller University, 1230 York Avenue, Box 275, New York, New York, 10065, United States of America
- Department of Psychology and Behavioral Neuroscience, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431–3192, Japan
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431–3192, Japan
- * E-mail:
| |
Collapse
|
46
|
Hehar H, Ma I, Mychasiuk R. Effects of Metabolic Programming on Juvenile Play Behavior and Gene Expression in the Prefrontal Cortex of Rats. Dev Neurosci 2016; 38:96-104. [PMID: 26967530 DOI: 10.1159/000444015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 01/14/2016] [Indexed: 11/19/2022] Open
Abstract
Early developmental processes, such as metabolic programming, can provide cues to an organism, which allow it to make modifications that are predicted to be beneficial for survival. Similarly, social play has a multifaceted role in promoting survival and fitness of animals. Play is a complex behavior that is greatly influenced by motivational and reward circuits, as well as the energy reserves and metabolism of an organism. This study examined the association between metabolic programming and juvenile play behavior in an effort to further elucidate insight into the consequences that early adaptions have on developmental trajectories. The study also examined changes in expression of four genes (Drd2, IGF1, Opa1, and OxyR) in the prefrontal cortex known to play significant roles in reward, bioenergetics, and social-emotional functioning. Using four distinct variations in developmental programming (high-fat diet, caloric restriction, exercise, or high-fat diet combined with exercise), we found that dietary programming (high-fat diet vs. caloric restriction) had the greatest impact on play behavior and gene expression. However, exercise also induced changes in both measures. This study demonstrates that metabolic programming can alter neural circuits and bioenergetics involved in play behavior, thus providing new insights into mechanisms that allow programming to influence the evolutionary success of an organism.
Collapse
Affiliation(s)
- Harleen Hehar
- Alberta Children's Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alta., Canada
| | | | | |
Collapse
|
47
|
Abstract
The neuropeptide oxytocin (OXT) has been revealed as a profound anxiolytic and antistress factor of the brain, besides its many prosocial and reproductive effects. Therefore, there is substantial scientific and medical interest in its potential therapeutic use for the treatment of psychopathologies associated with anxiety, fear, and social dysfunctions, such as generalized anxiety disorder, posttraumatic stress disorder, and social anxiety disorder, as well as autism and schizophrenia, among others. Focusing on preclinical studies, we review the existing evidence for the regulatory capacity of OXT to fine-tune general and social anxiety-related behaviors, as well as cued and social fear conditioning from a translational perspective. The available evidence from animal and human studies substantiates the hypothesis of an imbalance of the endogenous brain OXT system in the etiology of anxiety disorders, particularly those with a social component such as social anxiety disorder. In addition, such an imbalance of the OXT system is also likely to be the consequence of chronic OXT treatment resulting in a dose-dependent reduction in OXT receptor availability and increased anxiety.
Collapse
|
48
|
Hypothalamic vasopressin systems are more sensitive to the long term effects of social defeat in males versus females. Psychoneuroendocrinology 2015; 51:122-34. [PMID: 25306217 PMCID: PMC4268083 DOI: 10.1016/j.psyneuen.2014.09.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 09/09/2014] [Accepted: 09/10/2014] [Indexed: 12/17/2022]
Abstract
Vasopressin signaling has important effects on the regulation of social behaviors and stress responses, and is considered a promising pathway to target for new therapeutics of stress-induced psychiatric disorders. Although there is evidence for sex differences in the behavioral effects of arginine vasopressin (AVP), few data have directly compared the effects of stress on endogenous AVP signaling in males and females. We used California mice (Peromyscus californicus) to study the short and long term effects of social defeat stress on AVP immunoreactive cells in the paraventricular nucleus (PVN) and the posteromedial bed nucleus of the stria terminalis (BNSTmp). Acute exposure to defeat increased AVP/c-fos cells in the PVN and SON of both males and females. In contrast, there were sex differences in the long term effects of defeat. Males but not females exposed to defeat had less avp mRNA in the PVN, and in two experiments defeat reduced the number of AVP positive cells in the caudal PVN of males but not females. Interestingly, during relatively benign social encounters with a target mouse, there was a rapid decrease in AVP percent staining (including cell bodies and fibers) in the PVN of males but not females. Defeat reduced AVP percent staining in males, but did not block the socially induced decrease in percent staining. When mice were tested in resident-intruder tests, males exposed to defeat were no less aggressive than control males whereas aggression was abolished in females. However, bouts of aggression were positively correlated with the number of AVP neurons in the BNSTmp of control males but not stressed males, suggesting that different mechanisms mediate aggression in control and stressed males. These data show that while acute AVP responses to defeat are similar in males and females, the long term effects of defeat on AVP are stronger in males.
Collapse
|
49
|
Brain oxytocin in social fear conditioning and its extinction: involvement of the lateral septum. Neuropsychopharmacology 2014; 39:3027-35. [PMID: 24964815 PMCID: PMC4229574 DOI: 10.1038/npp.2014.156] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 06/15/2014] [Accepted: 06/19/2014] [Indexed: 01/02/2023]
Abstract
Central oxytocin (OXT) has anxiolytic and pro-social properties both in humans and rodents, and has been proposed as a therapeutic option for anxiety and social dysfunctions. Here, we utilized a mouse model of social fear conditioning (SFC) to study the effects of OXT on social fear, and to determine whether SFC causes alterations in central OXT receptor (OXTR) binding and local OXT release. Central infusion of OXT, but not arginine vasopressin, prior to social fear extinction training completely abolished social fear expression in an OXTR-mediated fashion without affecting general anxiety or locomotion. SFC caused increased OXTR binding in the dorso-lateral septum (DLS), central amygdala, dentate gyrus, and cornu ammunis 1, which normalized after social fear extinction, suggesting that these areas form part of a brain network involved in the development and neural support of social fear. Microdialysis revealed that the increase in OXT release observed in unconditioned mice within the DLS during social fear extinction training was attenuated in conditioned mice. Consequently, increasing the availability of local OXT by infusion of OXT into the DLS reversed social fear. Thus, alterations in the brain OXT system, including altered OXTR binding and OXT release within the DLS, play an important role in SFC and social fear extinction. Thus, we suggest that the OXT system is adversely affected in disorders associated with social fear, such as social anxiety disorder and reinstalling an appropriate balance of the OXT system may alleviate some of the symptoms.
Collapse
|
50
|
Bayerl DS, Klampfl SM, Bosch OJ. Central V1b receptor antagonism in lactating rats: impairment of maternal care but not of maternal aggression. J Neuroendocrinol 2014; 26:918-26. [PMID: 25283607 DOI: 10.1111/jne.12226] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 09/30/2014] [Accepted: 09/30/2014] [Indexed: 01/22/2023]
Abstract
Maternal behaviour in rodents is mediated by the central oxytocin and vasopressin systems, amongst others. The role of vasopressin, acting via the V1a receptor (V1aR), on maternal care and maternal aggression has recently been described. However, a potential involvement of the V1b receptor (V1bR) in maternal behaviour has only been demonstrated in knockout mice. The present study aimed to examine the effects of central pharmacological manipulation of the V1bR on maternal behaviour in lactating Wistar rats. On pregnancy day 18, female rats were implanted with a guide cannula targeting the lateral ventricle. After parturition, dams received an acute central infusion of a specific V1bR agonist (d[Leu4,Lys8]VP) or V1bR antagonist (SSR149415) once daily, followed by observations of maternal care [lactation day (LD) 1], maternal motivation in the pup retrieval test (LD 2), anxiety-related behaviour on the elevated plus-maze (LD 3) and maternal aggression in the maternal defence test followed by maternal care monitoring (LD 4). Our data demonstrate that, under nonstress conditions, the V1bR antagonist decreased the occurrence of both nursing and mother-pup interaction, whereas the V1bR agonist did not affect either parameter. Under stress conditions (i.e. after the maternal defence test), mother-pup interaction was decreased by infusion of the V1bR antagonist. During the maternal defence test, neither treatment affected aggressive or non-aggressive behaviour. Finally, neither treatment altered maternal motivation or anxiety. In conclusion, central V1bR antagonism modulates aspects of maternal care but not of maternal aggression or maternal motivation in lactating rats. These findings further extend our knowledge on the vasopressin system as a vital mediator of maternal behaviour.
Collapse
Affiliation(s)
- D S Bayerl
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | | | | |
Collapse
|