1
|
Favoretto CA, Bertagna NB, Miguel TT, Quadros IMH. The CRF/Urocortin systems as therapeutic targets for alcohol use disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 178:97-152. [PMID: 39523064 DOI: 10.1016/bs.irn.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Development and maintenance of alcohol use disorders have been proposed to recruit critical mechanisms involving Corticotropin Releasing Factor and Urocortins (CRF/Ucns). The CRF/Ucns system is comprised of a family of peptides (CRF, Ucn 1, Ucn 2, Ucn 3) which act upon two receptor subtypes, CRFR1 and CRFR2, each with different affinity profiles to the endogenous peptides and differential brain distribution. Activity of CRF/Ucn system is further modulated by CRF binding protein (CRF-BP), which regulates availability of CRF and Ucns to exert their actions. Extensive evidence in preclinical models support the involvement of CRF/Ucn targets in escalated alcohol drinking, as well as point to changes in CRF/Ucn brain function as a result of chronic alcohol exposure and/or withdrawal. It highlights the role of CRF and CRFR1-mediated signaling in conditions of excessive alcohol taking and seeking, including during various stages of withdrawal and relapse to alcohol. Besides its role in the hypothalamic-pituitary-adrenal (HPA) axis, the importance of extra-hypothalamic CRF pathways, especially in the extended amygdala, in the neurobiology of alcohol abuse and dependence is emphasized. Emerging roles for other targets of the CRF/Ucn system, such as CRF2 receptors, CRF-BP and Ucns in escalated alcohol drinking is also discussed. Finally, the limited translational value of CRF/Ucn interventions in stress-related and alcohol use disorders is discussed. So far, CRFR1 antagonists have shown little or no efficacy in human clinical trials, although a range of unexplored conditions and possibilities remain to be explored.
Collapse
Affiliation(s)
- Cristiane Aparecida Favoretto
- Molecular and Behavioral Neuroscience Laboratory, Pharmacology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo (Unifesp), SP, Brazil; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Natalia Bonetti Bertagna
- Molecular and Behavioral Neuroscience Laboratory, Pharmacology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo (Unifesp), SP, Brazil
| | | | - Isabel M H Quadros
- Psychobiology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo (Unifesp), SP, Brazil.
| |
Collapse
|
2
|
Mueller LE, Wexler RS, Lovejoy DA, Stein RB, Slee AM. Teneurin C-terminal associated peptide (TCAP)-1 attenuates the development and expression of naloxone-precipitated morphine withdrawal in male Swiss Webster mice. Psychopharmacology (Berl) 2024; 241:1565-1575. [PMID: 38630316 PMCID: PMC11269454 DOI: 10.1007/s00213-024-06582-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/26/2024] [Indexed: 07/25/2024]
Abstract
RATIONALE Corticotropin-releasing factor (CRF), the apical stress-inducing hormone, exacerbates stress and addictive behaviors. TCAP-1 is a peptide that directly inhibits both CRF-mediated stress and addiction-related behaviors; however, the direct action of TCAP-1 on morphine withdrawal-associated behaviors has not previously been examined. OBJECTIVE To determine whether TCAP-1 administration attenuates behavioral and physiological consequences of morphine withdrawal in mice. METHODS Mice were administered via subcutaneous route TCAP-1 either before or after initial morphine exposure, after which jumping behavior was quantified to assess the effects of TCAP-1 on naloxone-precipitated morphine withdrawal. As a comparison, mice were treated with nonpeptide CRF1 receptor antagonist CP-154,526. In one experiment, plasma corticosterone (CORT) was also measured as a physiological stress indicator. RESULTS Pretreatment with TCAP-1 (10-250 nmol/kg) before morphine treatment significantly inhibited the development of naloxone-precipitated withdrawal. TCAP-1 (250-500 nmol/kg) treatment administered after morphine treatment attenuated the behavioral expression of naloxone-precipitated withdrawal. TCAP-1 (250 nmol/kg) treatment during morphine treatment was more effective than the optimal dosing of CP-154,526 (20 mg/kg) at suppressing the behavioral expression of naloxone-precipitated withdrawal, despite similar reduction of withdrawal-induced plasma CORT level increases. CONCLUSIONS These findings establish TCAP-1 as a potential therapeutic candidate for the prevention and treatment of morphine withdrawal.
Collapse
Affiliation(s)
| | | | - David A Lovejoy
- Protagenic Therapeutics, Inc., New York, NY, USA
- Department of Cell and Systems Biology, University of Toronto, Toronto, CA, Canada
| | | | | |
Collapse
|
3
|
Groenink L, Verdouw PM, Zhao Y, Ter Heegde F, Wever KE, Bijlsma EY. Pharmacological modulation of conditioned fear in the fear-potentiated startle test: a systematic review and meta-analysis of animal studies. Psychopharmacology (Berl) 2023; 240:2361-2401. [PMID: 36651922 PMCID: PMC10593622 DOI: 10.1007/s00213-022-06307-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/21/2022] [Indexed: 01/19/2023]
Abstract
RATIONALE AND OBJECTIVES Fear conditioning is an important aspect in the pathophysiology of anxiety disorders. The fear-potentiated startle test is based on classical fear conditioning and over the years, a broad range of drugs have been tested in this test. Synthesis of the available data may further our understanding of the neurotransmitter systems that are involved in the expression of conditioned fear. METHODS Following a comprehensive search in Medline and Embase, we included 68 research articles that reported on 103 drugs, covering 56 different drug classes. The systematic review was limited to studies using acute, systemic drug administration in naive animals. RESULTS Qualitative data synthesis showed that most clinically active anxiolytics, but not serotonin-reuptake inhibitors, reduced cued fear. Anxiogenic drugs increased fear potentiation in 35% of the experiments, reduced fear potentiation in 29% of the experiments, and were without effect in 29% of the experiments. Meta-analyses could be performed for five drug classes and showed that benzodiazepines, buspirone, 5-HT1A agonists, 5-HT1A antagonists, and mGluR2,3 agonists reduced cued conditioned fear. The non-cued baseline startle response, which may reflect contextual anxiety, was only significantly reduced by benzodiazepines and 5-HT1A antagonists. No associations were found between drug effects and methodological characteristics, except for strain. CONCLUSIONS The fear-potentiated startle test appears to have moderate to high predictive validity and may serve as a valuable tool for the development of novel anxiolytics. Given the limited available data, the generally low study quality and high heterogeneity additional studies are warranted to corroborate the findings of this review.
Collapse
Affiliation(s)
- Lucianne Groenink
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands.
| | - P Monika Verdouw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Yulong Zhao
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Freija Ter Heegde
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Kimberley E Wever
- Department of Anaesthesiology, Pain and Palliative Medicine, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands
| | - Elisabeth Y Bijlsma
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| |
Collapse
|
4
|
van de Poll Y, Cras Y, Ellender TJ. The neurophysiological basis of stress and anxiety - comparing neuronal diversity in the bed nucleus of the stria terminalis (BNST) across species. Front Cell Neurosci 2023; 17:1225758. [PMID: 37711509 PMCID: PMC10499361 DOI: 10.3389/fncel.2023.1225758] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/03/2023] [Indexed: 09/16/2023] Open
Abstract
The bed nucleus of the stria terminalis (BNST), as part of the extended amygdala, has become a region of increasing interest regarding its role in numerous human stress-related psychiatric diseases, including post-traumatic stress disorder and generalized anxiety disorder amongst others. The BNST is a sexually dimorphic and highly complex structure as already evident by its anatomy consisting of 11 to 18 distinct sub-nuclei in rodents. Located in the ventral forebrain, the BNST is anatomically and functionally connected to many other limbic structures, including the amygdala, hypothalamic nuclei, basal ganglia, and hippocampus. Given this extensive connectivity, the BNST is thought to play a central and critical role in the integration of information on hedonic-valence, mood, arousal states, processing emotional information, and in general shape motivated and stress/anxiety-related behavior. Regarding its role in regulating stress and anxiety behavior the anterolateral group of the BNST (BNSTALG) has been extensively studied and contains a wide variety of neurons that differ in their electrophysiological properties, morphology, spatial organization, neuropeptidergic content and input and output synaptic organization which shape their activity and function. In addition to this great diversity, further species-specific differences are evident on multiple levels. For example, classic studies performed in adult rat brain identified three distinct neuron types (Type I-III) based on their electrophysiological properties and ion channel expression. Whilst similar neurons have been identified in other animal species, such as mice and non-human primates such as macaques, cross-species comparisons have revealed intriguing differences such as their comparative prevalence in the BNSTALG as well as their electrophysiological and morphological properties, amongst other differences. Given this tremendous complexity on multiple levels, the comprehensive elucidation of the BNSTALG circuitry and its role in regulating stress/anxiety-related behavior is a major challenge. In the present Review we bring together and highlight the key differences in BNSTALG structure, functional connectivity, the electrophysiological and morphological properties, and neuropeptidergic profiles of BNSTALG neurons between species with the aim to facilitate future studies of this important nucleus in relation to human disease.
Collapse
Affiliation(s)
- Yana van de Poll
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Yasmin Cras
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Tommas J. Ellender
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
5
|
Cain CK. Beyond Fear, Extinction, and Freezing: Strategies for Improving the Translational Value of Animal Conditioning Research. Curr Top Behav Neurosci 2023; 64:19-57. [PMID: 37532965 PMCID: PMC10840073 DOI: 10.1007/7854_2023_434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
Translational neuroscience for anxiety has had limited success despite great progress in understanding the neurobiology of Pavlovian fear conditioning and extinction. This chapter explores the idea that conditioning paradigms have had a modest impact on translation because studies in animals and humans are misaligned in important ways. For instance, animal conditioning studies typically use imminent threats to assess short-duration fear states with single behavioral measures (e.g., freezing), whereas human studies typically assess weaker or more prolonged anxiety states with physiological (e.g., skin conductance) and self-report measures. A path forward may be more animal research on conditioned anxiety phenomena measuring dynamic behavioral and physiological responses in more complex environments. Exploring transitions between defensive brain states during extinction, looming threats, and post-threat recovery may be particularly informative. If care is taken to align paradigms, threat levels, and measures, this strategy may reveal stable patterns of non-conscious defense in animals and humans that correlate better with conscious anxiety. This shift in focus is also warranted because anxiety is a bigger problem than fear, even in disorders defined by dysfunctional fear or panic reactions.
Collapse
Affiliation(s)
- Christopher K Cain
- Department of Child and Adolescent Psychiatry, NYU Langone Health, New York, NY, USA.
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.
| |
Collapse
|
6
|
Schnider M, Jenni R, Ramain J, Camporesi S, Golay P, Alameda L, Conus P, Do KQ, Steullet P. Time of exposure to social defeat stress during childhood and adolescence and redox dysregulation on long-lasting behavioral changes, a translational study. Transl Psychiatry 2022; 12:413. [PMID: 36163247 PMCID: PMC9512907 DOI: 10.1038/s41398-022-02183-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 11/09/2022] Open
Abstract
Traumatic events during childhood/early adolescence can cause long-lasting physiological and behavioral changes with increasing risk for psychiatric conditions including psychosis. Genetic factors and trauma (and their type, degree of repetition, time of occurrence) are believed to influence how traumatic experiences affect an individual. Here, we compared long-lasting behavioral effects of repeated social defeat stress (SD) applied during either peripuberty or late adolescence in adult male WT and Gclm-KO mice, a model of redox dysregulation relevant to schizophrenia. As SD disrupts redox homeostasis and causes oxidative stress, we hypothesized that KO mice would be particularly vulnerable to such stress. We first found that peripubertal and late adolescent SD led to different behavioral outcomes. Peripubertal SD induced anxiety-like behavior in anxiogenic environments, potentiated startle reflex, and increased sensitivity to the NMDA-receptor antagonist, MK-801. In contrast, late adolescent SD led to increased exploration in novel environments. Second, the long-lasting impact of peripubertal but not late adolescent SD differed in KO and WT mice. Peripubertal SD increased anxiety-like behavior in anxiogenic environments and MK-801-sensitivity mostly in KO mice, while it increased startle reflex in WT mice. These suggest that a redox dysregulation during peripuberty interacts with SD to remodel the trajectory of brain maturation, but does not play a significant role during later SD. As peripubertal SD induced persisting anxiety- and fear-related behaviors in male mice, we then investigated anxiety in a cohort of 89 early psychosis male patients for whom we had information about past abuse and clinical assessment during the first year of psychosis. We found that a first exposure to physical/sexual abuse (analogous to SD) before age 12, but not after, was associated with higher anxiety at 6-12 months after psychosis onset. This supports that childhood/peripuberty is a vulnerable period during which physical/sexual abuse in males has wide and long-lasting consequences.
Collapse
Affiliation(s)
- Mirko Schnider
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Raoul Jenni
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Julie Ramain
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Sara Camporesi
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Philippe Golay
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Luis Alameda
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Philippe Conus
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Pascal Steullet
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland.
| |
Collapse
|
7
|
Ruat J, Hartmann A, Heinz DE, Nemcova P, Stoffel R, Deussing JM, Chen A, Wotjak CT. CB1 receptors in corticotropin-releasing factor neurons selectively control the acoustic startle response in male mice. GENES BRAIN AND BEHAVIOR 2021; 20:e12775. [PMID: 34672092 DOI: 10.1111/gbb.12775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 11/30/2022]
Abstract
The endocannabinoid system is an important regulator of the hormonal and behavioral stress responses, which critically involve corticotropin-releasing factor (CRF) and its receptors. While it has been shown that CRF and the cannabinoid type 1 (CB1) receptor are co-localized in several brain regions, the physiological relevance of this co-expression remains unclear. Using double in situ hybridization, we confirmed co-localization in the piriform cortex, the lateral hypothalamic area, the paraventricular nucleus, and the Barrington's nucleus, albeit at low levels. To study the behavioral and physiological implications of this co-expression, we generated a conditional knockout mouse line that selectively lacks the expression of CB1 receptors in CRF neurons. We found no effects on fear and anxiety-related behaviors under basal conditions nor after a traumatic experience. Additionally, plasma corticosterone levels were unaffected at baseline and after restraint stress. Only acoustic startle responses were significantly enhanced in male, but not female, knockout mice. Taken together, the consequences of depleting CB1 in CRF-positive neurons caused a confined hyperarousal phenotype in a sex-dependent manner. The current results suggest that the important interplay between the central endocannabinoid and CRF systems in regulating the organism's stress response is predominantly taking place at the level of CRF receptor-expressing neurons.
Collapse
Affiliation(s)
- Julia Ruat
- Department Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.,International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Max Planck Institute of Psychiatry, Munich, Germany.,Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, Munich, Germany
| | - Alice Hartmann
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, Munich, Germany
| | - Daniel E Heinz
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, Munich, Germany.,Max Planck School of Cognition, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Paulina Nemcova
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, Munich, Germany
| | - Rainer Stoffel
- Department Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jan M Deussing
- Research Group Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.,Scientific Core Unit Genetically Engineered Mouse Models, Max Planck Institute of Psychiatry, Munich, Germany
| | - Alon Chen
- Department Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.,Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Carsten T Wotjak
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, Munich, Germany.,Max Planck School of Cognition, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.,Central Nervous System Diseases Research (CNSDR), Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| |
Collapse
|
8
|
Urien L, Stein N, Ryckman A, Bell L, Bauer EP. Extended amygdala circuits are differentially activated by context fear conditioning in male and female rats. Neurobiol Learn Mem 2021; 180:107401. [PMID: 33581315 PMCID: PMC8076097 DOI: 10.1016/j.nlm.2021.107401] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 02/06/2023]
Abstract
As the incidence of anxiety disorders is more prevalent in females, comparing the neural underpinnings of anxiety in males and females is imperative. The bed nucleus of the stria terminalis (BNST) contributes to long-lasting, anxiety-like states including the expression of context fear conditioning. Currently, there is conflicting evidence as to which nuclei of the BNST contribute to these behaviors. The anterolateral portion of the BNST (BNST-AL) located dorsal to the anterior commissure and lateral to the stria terminalis sends robust projections to the central nucleus of the amygdala (CE). Here we asked whether the BNST-AL is active during the expression of context fear conditioning in both male and female rats. At the cellular level, the expression of context fear produced upregulation of the immediate-early gene ARC in the BNST-AL as well as an upregulation of ARC specifically in neurons projecting to the CE, as labeled by the retrograde tracer Fluorogold infused into the CE. However, this pattern of ARC expression was observed in male rats only. Excitotoxic lesions of the BNST reduced context fear expression in both sexes, suggesting that a different set of BNST subnuclei may be recruited by the expression of fear and anxiety-like behaviors in females. Overall, our data highlight the involvement of the BNST-AL in fear expression in males, and suggest that subnuclei of the BNST may be functionally different in male and female rats.
Collapse
Affiliation(s)
- Louise Urien
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Nicole Stein
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Abigail Ryckman
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Lindsey Bell
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Elizabeth P Bauer
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States.
| |
Collapse
|
9
|
Beyeler A, Dabrowska J. Neuronal diversity of the amygdala and the bed nucleus of the stria terminalis. HANDBOOK OF BEHAVIORAL NEUROSCIENCE 2020; 26:63-100. [PMID: 32792868 DOI: 10.1016/b978-0-12-815134-1.00003-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Anna Beyeler
- Neurocentre Magendie, French National Institutes of Health (INSERM) unit 1215, Neurocampus of Bordeaux University, Bordeaux, France
| | - Joanna Dabrowska
- Center for the Neurobiology of Stress Resilience and Psychiatric Disorders, Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
10
|
Grillon C, Robinson OJ, Cornwell B, Ernst M. Modeling anxiety in healthy humans: a key intermediate bridge between basic and clinical sciences. Neuropsychopharmacology 2019; 44:1999-2010. [PMID: 31226707 PMCID: PMC6897969 DOI: 10.1038/s41386-019-0445-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/06/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022]
Abstract
Animal models of anxiety disorders are important for elucidating neurobiological defense mechanisms. However, animal models are limited when it comes to understanding the more complex processes of anxiety that are unique to humans (e.g., worry) and to screen new treatments. In this review, we outline how the Experimental Psychopathology approach, based on experimental models of anxiety in healthy subjects, can mitigate these limitations and complement research in animals. Experimental psychopathology can bridge basic research in animals and clinical studies, as well as guide and constrain hypotheses about the nature of psychopathology, treatment mechanisms, and treatment targets. This review begins with a brief review of the strengths and limitations of animal models before discussing the need for human models of anxiety, which are especially necessary to probe higher-order cognitive processes. This can be accomplished by combining anxiety-induction procedures with tasks that probe clinically relevant processes to identify neurocircuits that are potentially altered by anxiety. The review then discusses the validity of experimental psychopathology and introduces a methodological approach consisting of five steps: (1) select anxiety-relevant cognitive or behavioral operations and associated tasks, (2) identify the underlying neurocircuits supporting these operations in healthy controls, 3) examine the impact of experimental anxiety on the targeted operations in healthy controls, (4) utilize findings from step 3 to generate hypotheses about neurocircuit dysfunction in anxious patients, and 5) evaluate treatment mechanisms and screen novel treatments. This is followed by two concrete illustrations of this approach and suggestions for future studies.
Collapse
Affiliation(s)
- Christian Grillon
- Section on the Neurobiology of Fear and Anxiety, National Institute of Mental Health, Bethesda, MD, USA.
| | - Oliver J Robinson
- University College London, Institute of Cognitive Neuroscience, London, UK
| | - Brian Cornwell
- Centre for Mental Health, Faculty of Health, Arts and Design, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - Monique Ernst
- Section on the Neurobiology of Fear and Anxiety, National Institute of Mental Health, Bethesda, MD, USA
| |
Collapse
|
11
|
Martinon D, Lis P, Roman AN, Tornesi P, Applebey SV, Buechner G, Olivera V, Dabrowska J. Oxytocin receptors in the dorsolateral bed nucleus of the stria terminalis (BNST) bias fear learning toward temporally predictable cued fear. Transl Psychiatry 2019; 9:140. [PMID: 31000694 PMCID: PMC6472379 DOI: 10.1038/s41398-019-0474-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/19/2019] [Accepted: 04/01/2019] [Indexed: 11/18/2022] Open
Abstract
The inability to discriminate between threat and safety is a hallmark of stress-induced psychiatric disorders, including post-traumatic stress disorder. Dorsolateral bed nucleus of the stria terminalis (BNSTdl) is critically involved in the modulation of fear and anxiety, and has been proposed to regulate discrimination between signaled (cued, predictable) and unsignaled (unpredictable) threats. We recently showed that oxytocin receptors (OTRs) in the BNSTdl facilitate acquisition of cued fear measured in a fear-potentiated startle (FPS). In the current study, using in vivo microdialysis in awake male Sprague-Dawley rats, a double immunofluorescence approach with confocal microscopy, as well as retrograde tracing of hypothalamic BNST-projecting OT neurons, we investigated whether fear conditioning activates OT system and modulates OT release. To determine the role of OTR in fear memory formation, we also infused OTR antagonist or OT into the BNSTdl before fear conditioning and measured rats' ability to discriminate between cued (signaled) and non-cued (unsignaled) fear using FPS. In contrast to acute stress (exposure to forced swim stress or foot shocks alone), cued fear conditioning increases OT content in BNSTdl microdialysates. In addition, fear conditioning induces moderate activation of OT neurons in the paraventricular nucleus of the hypothalamus and robust activation in the supraoptic and accessory nuclei of the hypothalamus. Application of OT into the BNSTdl facilitates fear learning toward signaled, predictable threats, whereas blocking OTR attenuates this effect. We conclude that OTR neurotransmission in the BNSTdl plays a pivotal role in strengthening fear learning of temporally predictable, signaled threats.
Collapse
Affiliation(s)
- Daisy Martinon
- 0000 0004 0388 7807grid.262641.5Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064 USA
| | - Paulina Lis
- 0000 0004 0388 7807grid.262641.5Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064 USA
| | - Alexandra N. Roman
- 0000 0004 0388 7807grid.262641.5Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064 USA
| | - Patricio Tornesi
- 0000 0004 0388 7807grid.262641.5Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064 USA
| | - Sarah V. Applebey
- 0000 0004 0388 7807grid.262641.5Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064 USA
| | - Garrett Buechner
- 0000 0004 0388 7807grid.262641.5Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064 USA
| | - Valentina Olivera
- 0000 0004 0388 7807grid.262641.5Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064 USA ,0000 0004 0388 7807grid.262641.5Center for the Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064 USA ,0000 0004 0388 7807grid.262641.5School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064 USA
| | - Joanna Dabrowska
- Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA. .,Center for the Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA. .,School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA.
| |
Collapse
|
12
|
Loupy KM, Arnold MR, Hassell JE, Lieb MW, Milton LN, Cler KE, Fox JH, Siebler PH, Schmidt D, Noronha SISR, Day HEW, Lowry CA. Evidence that preimmunization with a heat-killed preparation of Mycobacterium vaccae reduces corticotropin-releasing hormone mRNA expression in the extended amygdala in a fear-potentiated startle paradigm. Brain Behav Immun 2019; 77:127-140. [PMID: 30597198 DOI: 10.1016/j.bbi.2018.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 12/21/2018] [Accepted: 12/22/2018] [Indexed: 01/16/2023] Open
Abstract
Posttraumatic stress disorder (PTSD) is a trauma and stressor-related disorder that is characterized by dysregulation of glucocorticoid signaling, chronic low-grade inflammation, and impairment in the ability to extinguish learned fear. Corticotropin-releasing hormone (Crh) is a stress- and immune-responsive neuropeptide secreted from the paraventricular nucleus of the hypothalamus (PVN) to stimulate the hypothalamic-pituitary-adrenal (HPA) axis; however, extra-hypothalamic sources of Crh from the central nucleus of the amygdala (CeA) and bed nucleus of the stria terminalis (BNST) govern specific fear- and anxiety-related defensive behavioral responses. We previously reported that preimmunization with a heat-killed preparation of the immunoregulatory environmental bacterium Mycobacterium vaccae NCTC 11659 enhances fear extinction in a fear-potentiated startle (FPS) paradigm. In this follow-up study, we utilized an in situ hybridization histochemistry technique to investigate Crh, Crhr1, and Crhr2 mRNA expression in the CeA, BNST, and PVN of the same rats from the original study [Fox et al., 2017, Brain, Behavior, and Immunity, 66: 70-84]. Here, we demonstrate that preimmunization with M. vaccae NCTC 11659 decreases Crh mRNA expression in the CeA and BNST of rats exposed to the FPS paradigm, and, further, that Crh mRNA expression in these regions is correlated with fear behavior during extinction training. These data are consistent with the hypothesis that M. vaccae promotes stress-resilience by attenuating Crh production in fear- and anxiety-related circuits. These data suggest that immunization with M. vaccae may be an effective strategy for prevention of fear- and anxiety-related disorders.
Collapse
Affiliation(s)
- Kelsey M Loupy
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Mathew R Arnold
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - James E Hassell
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Margaret W Lieb
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Lauren N Milton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Kristin E Cler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - James H Fox
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Philip H Siebler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Dominic Schmidt
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Sylvana I S R Noronha
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Heidi E W Day
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Department of Physical Medicine & Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center, Denver Veterans Affairs Medical Center (VAMC), Denver, CO 80045, USA; Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Denver, CO 80045, USA.
| |
Collapse
|
13
|
Kaffman A, White JD, Wei L, Johnson FK, Krystal JH. Enhancing the Utility of Preclinical Research in Neuropsychiatry Drug Development. Methods Mol Biol 2019; 2011:3-22. [PMID: 31273690 DOI: 10.1007/978-1-4939-9554-7_1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Most large pharmaceutical companies have downscaled or closed their clinical neuroscience research programs in response to the low clinical success rate for drugs that showed tremendous promise in animal experiments intended to model psychiatric pathophysiology. These failures have raised serious concerns about the role of preclinical research in the identification and evaluation of new pharmacotherapies for psychiatry. In the absence of a comprehensive understanding of the neurobiology of psychiatric disorders, the task of developing "animal models" seems elusive. The purpose of this review is to highlight emerging strategies to enhance the utility of preclinical research in the drug development process. We address this issue by reviewing how advances in neuroscience, coupled with new conceptual approaches, have recently revolutionized the way we can diagnose and treat common psychiatric conditions. We discuss the implications of these new tools for modeling psychiatric conditions in animals and advocate for the use of systematic reviews of preclinical work as a prerequisite for conducting psychiatric clinical trials. We believe that work in animals is essential for elucidating human psychopathology and that improving the predictive validity of animal models is necessary for developing more effective interventions for mental illness.
Collapse
Affiliation(s)
- Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
| | - Jordon D White
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Lan Wei
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Frances K Johnson
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - John H Krystal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
14
|
Ch'ng S, Fu J, Brown RM, McDougall SJ, Lawrence AJ. The intersection of stress and reward: BNST modulation of aversive and appetitive states. Prog Neuropsychopharmacol Biol Psychiatry 2018; 87:108-125. [PMID: 29330137 DOI: 10.1016/j.pnpbp.2018.01.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 12/27/2017] [Accepted: 01/08/2018] [Indexed: 12/13/2022]
Abstract
The bed nucleus of the stria terminalis (BNST) is widely acknowledged as a brain structure that regulates stress and anxiety states, as well as aversive and appetitive behaviours. The diverse roles of the BNST are afforded by its highly modular organisation, neurochemical heterogeneity, and complex intrinsic and extrinsic circuitry. There has been growing interest in the BNST in relation to psychopathologies such as anxiety and addiction. Although research on the human BNST is still in its infancy, there have been extensive preclinical studies examining the molecular signature and hodology of the BNST and their involvement in stress and reward seeking behaviour. This review examines the neurochemical phenotype and connectivity of the BNST, as well as electrophysiological correlates of plasticity in the BNST mediated by stress and/or drugs of abuse.
Collapse
Affiliation(s)
- Sarah Ch'ng
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Jingjing Fu
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Robyn M Brown
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Stuart J McDougall
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia.
| |
Collapse
|
15
|
Janeček M, Dabrowska J. Oxytocin facilitates adaptive fear and attenuates anxiety responses in animal models and human studies-potential interaction with the corticotropin-releasing factor (CRF) system in the bed nucleus of the stria terminalis (BNST). Cell Tissue Res 2018; 375:143-172. [PMID: 30054732 DOI: 10.1007/s00441-018-2889-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/04/2018] [Indexed: 01/28/2023]
Abstract
Despite its relatively well-understood role as a reproductive and pro-social peptide, oxytocin (OT) tells a more convoluted story in terms of its modulation of fear and anxiety. This nuanced story has been obscured by a great deal of research into the therapeutic applications of exogenous OT, driving more than 400 ongoing clinical trials. Drawing from animal models and human studies, we review the complex evidence concerning OT's role in fear learning and anxiety, clarifying the existing confusion about modulation of fear versus anxiety. We discuss animal models and human studies demonstrating the prevailing role of OT in strengthening fear memory to a discrete signal or cue, which allows accurate and rapid threat detection that facilitates survival. We also review ostensibly contrasting behavioral studies that nonetheless provide compelling evidence of OT attenuating sustained contextual fear and anxiety-like behavior, arguing that these OT effects on the modulation of fear vs. anxiety are not mutually exclusive. To disambiguate how endogenous OT modulates fear and anxiety, an understudied area compared to exogenous OT, we survey behavioral studies utilizing OT receptor (OTR) antagonists. Based on emerging evidence about the role of OTR in rat dorsolateral bed nucleus of stria terminalis (BNST) and elsewhere, we postulate that OT plays a critical role in facilitating accurate discrimination between stimuli representing threat and safety. Supported by human studies, we demonstrate that OT uniquely facilitates adaptive fear but reduces maladaptive anxiety. Last, we explore the limited literature on endogenous OT and its interaction with corticotropin-releasing factor (CRF) with a special emphasis on the dorsolateral BNST, which may hold the key to the neurobiology of phasic fear and sustained anxiety.
Collapse
Affiliation(s)
- Michael Janeček
- Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Joanna Dabrowska
- Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA. .,Department of Neuroscience, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA.
| |
Collapse
|
16
|
Mazzone CM, Pati D, Michaelides M, DiBerto J, Fox JH, Tipton G, Anderson C, Duffy K, McKlveen JM, Hardaway JA, Magness ST, Falls WA, Hammack SE, McElligott ZA, Hurd YL, Kash TL. Acute engagement of G q-mediated signaling in the bed nucleus of the stria terminalis induces anxiety-like behavior. Mol Psychiatry 2018; 23:143-153. [PMID: 27956747 PMCID: PMC5468515 DOI: 10.1038/mp.2016.218] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 09/21/2016] [Accepted: 10/11/2016] [Indexed: 01/23/2023]
Abstract
The bed nucleus of the stria terminalis (BNST) is a brain region important for regulating anxiety-related behavior in both humans and rodents. Here we used a chemogenetic strategy to investigate how engagement of G protein-coupled receptor (GPCR) signaling cascades in genetically defined GABAergic BNST neurons modulates anxiety-related behavior and downstream circuit function. We saw that stimulation of vesicular γ-aminobutyric acid (GABA) transporter (VGAT)-expressing BNST neurons using hM3Dq, but neither hM4Di nor rM3Ds designer receptors exclusively activated by a designer drug (DREADD), promotes anxiety-like behavior. Further, we identified that activation of hM3Dq receptors in BNST VGAT neurons can induce a long-term depression-like state of glutamatergic synaptic transmission, indicating DREADD-induced changes in synaptic plasticity. Further, we used DREADD-assisted metabolic mapping to profile brain-wide network activity following activation of Gq-mediated signaling in BNST VGAT neurons and saw increased activity within ventral midbrain structures, including the ventral tegmental area and hindbrain structures such as the locus coeruleus and parabrachial nucleus. These results highlight that Gq-mediated signaling in BNST VGAT neurons can drive downstream network activity that correlates with anxiety-like behavior and points to the importance of identifying endogenous GPCRs within genetically defined cell populations. We next used a microfluidics approach to profile the receptorome of single BNST VGAT neurons. This approach yielded multiple Gq-coupled receptors that are associated with anxiety-like behavior and several potential novel candidates for regulation of anxiety-like behavior. From this, we identified that stimulation of the Gq-coupled receptor 5-HT2CR in the BNST is sufficient to elevate anxiety-like behavior in an acoustic startle task. Together, these results provide a novel profile of receptors within genetically defined BNST VGAT neurons that may serve as therapeutic targets for regulating anxiety states and provide a blueprint for examining how G-protein-mediated signaling in a genetically defined cell type can be used to assess behavior and brain-wide circuit function.
Collapse
Affiliation(s)
- Christopher M. Mazzone
- Neurobiology Curriculum, University of North Carolina, Chapel Hill, NC 27599
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Dipanwita Pati
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Michael Michaelides
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Jeffrey DiBerto
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - James H. Fox
- Department of Psychology, University of Vermont, Burlington, VT 05405
| | - Gregory Tipton
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Carlton Anderson
- Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, NC 27599
| | - Kelly Duffy
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Jessica M. McKlveen
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - J. Andrew Hardaway
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Scott T. Magness
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599
- Department of Cell Biology and Physiology, and Biomedical Engineering, University of North Carolina, Chapel Hill, NC 27599
| | - William A. Falls
- Department of Psychology, University of Vermont, Burlington, VT 05405
| | | | - Zoe A. McElligott
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC 27599
| | - Yasmin L. Hurd
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Thomas L. Kash
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, NC, 27599
| |
Collapse
|
17
|
Salvatore M, Wiersielis KR, Luz S, Waxler DE, Bhatnagar S, Bangasser DA. Sex differences in circuits activated by corticotropin releasing factor in rats. Horm Behav 2018; 97:145-153. [PMID: 29037972 PMCID: PMC5831187 DOI: 10.1016/j.yhbeh.2017.10.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 09/01/2017] [Accepted: 10/10/2017] [Indexed: 01/04/2023]
Abstract
Women are more likely than men to suffer from psychiatric disorders characterized by corticotropin releasing factor (CRF) hypersecretion, suggesting sex differences in CRF sensitivity. In rodents, sex differences in the sensitivity of specific brain regions to CRF have been identified. However, regions do not work in isolation, but rather form circuits to coordinate distinct responses to stressful events. Here we examined whether CRF activates different circuits in male and female rats. Following central administration of CRF or artificial cerebrospinal fluid (aCSF), neuronal activation in stress-related areas was assessed using cFOS. Functional connectivity was gauged by correlating the number of cFOS-positive cells between regions and then identifying differences within each sex in correlations for aCSF-treated and CRF-treated groups. This analysis revealed that CRF altered different circuits in males and females. As an example, CRF altered correlations involving the dorsal raphe in males and the bed nucleus of the stria terminalis in females, suggesting sex differences in stress-activated circuits controlling mood and anxiety. Next, plasma estradiol and progesterone levels were correlated with cFOS counts in females. Negative correlations between estradiol and neuronal activation in the regions within the extended amygdala were found in CRF-treated, but not aCSF-treated females. This result suggests that estrogens and CRF together modulate the fear and anxiety responses mediated by these regions. Collectively, these studies reveal sex differences in the way brain regions work together in response to CRF. These differences could drive different stress coping strategies in males and females, perhaps contributing to sex biases in psychopathology.
Collapse
Affiliation(s)
- Madeleine Salvatore
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Kimberly R Wiersielis
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Sandra Luz
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - David E Waxler
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Seema Bhatnagar
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA.
| |
Collapse
|
18
|
Murrough JW, Charney DS. Corticotropin-Releasing Factor Type 1 Receptor Antagonists for Stress-Related Disorders: Time to Call It Quits? Biol Psychiatry 2017; 82:858-860. [PMID: 29129198 DOI: 10.1016/j.biopsych.2017.10.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 10/12/2017] [Indexed: 01/08/2023]
Affiliation(s)
- James W Murrough
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Dennis S Charney
- Office of the Dean, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
19
|
Kaye JT, Bradford DE, Magruder KP, Curtin JJ. Probing for Neuroadaptations to Unpredictable Stressors in Addiction: Translational Methods and Emerging Evidence. J Stud Alcohol Drugs 2017; 78:353-371. [PMID: 28499100 DOI: 10.15288/jsad.2017.78.353] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Stressors clearly contribute to addiction etiology and relapse in humans, but our understanding of specific mechanisms remains limited. Rodent models of addiction offer the power, flexibility, and precision necessary to delineate the causal role and specific mechanisms through which stressors influence alcohol and other drug use. This review describes a program of research using startle potentiation to unpredictable stressors that is well positioned to translate between animal models and clinical research with humans on stress neuroadaptations in addiction. This research rests on a solid foundation provided by three separate pillars of evidence from (a) rodent behavioral neuroscience on stress neuroadaptations in addiction, (b) rodent affective neuroscience on startle potentiation, and (c) human addiction and affective science with startle potentiation. Rodent stress neuroadaptation models implicate adaptations in corticotropin-releasing factor and norepinephrine circuits within the central extended amygdala following chronic alcohol and other drug use that mediate anxious behaviors and stress-induced reinstatement among drug-dependent rodents. Basic affective neuroscience indicates that these same neural mechanisms are involved in startle potentiation to unpredictable stressors in particular (vs. predictable stressors). We believe that synthesis of these evidence bases should focus us on the role of unpredictable stressors in addiction etiology and relapse. Startle potentiation in unpredictable stressor tasks is proposed to provide an attractive and flexible test bed to encourage tight translation and reverse translation between animal models and human clinical research on stress neuroadaptations. Experimental therapeutics approaches focused on unpredictable stressors hold high promise to identify, repurpose, or refine pharmacological and psychosocial interventions for addiction.
Collapse
Affiliation(s)
- Jesse T Kaye
- University of Wisconsin-Madison, Madison, Wisconsin
| | | | | | | |
Collapse
|
20
|
Liu Y, Liu D, Xu J, Jiang H, Pan F. Early adolescent stress-induced changes in prefrontal cortex miRNA-135a and hippocampal miRNA-16 in male rats. Dev Psychobiol 2017; 59:958-969. [PMID: 28944448 DOI: 10.1002/dev.21558] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 07/19/2017] [Indexed: 01/17/2023]
Abstract
Early-life stress increases susceptibility to post-traumatic stress disorders (PTSD), in which the dysfunction of 5-hydroxytryptamine plays an important role. miRNA-135a in the prefrontal cortex (PFC) and miRNA-16 in the hippocampus (HIP) are closely related to the 5-HT neurotransmitter system. Here, we investigated behavior, miRNA-135a in the PFC, miRNA-16 in the HIP, and 5-HT1AR expression in both brain regions in adolescent and adult rats that were exposed to inescapable stress during their adolescence. Paroxetine hydrochloride and corticotropin-releasing factor antagonist (CP-154,526) were used as intervening measures. Our study demonstrated that early adolescent stress induced anxiety-like behaviors and spatial memory damage, a reduction in miRNA-135a expression was associated with increased 5-HT1AR expression in PFC, and increased miRNA-16 expression in the HIP of stressed rats. Drug treatments alleviated behaviors and reversed the miRNA-135a, miRNA-16, and 5-HT1AR expression in stressed rats.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong, P. R. China
| | - Dexiang Liu
- Department of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong, P. R. China
| | - Jingjing Xu
- Department of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong, P. R. China
| | - Hong Jiang
- Department of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong, P. R. China
| | - Fang Pan
- Department of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong, P. R. China
| |
Collapse
|
21
|
Moaddab M, Dabrowska J. Oxytocin receptor neurotransmission in the dorsolateral bed nucleus of the stria terminalis facilitates the acquisition of cued fear in the fear-potentiated startle paradigm in rats. Neuropharmacology 2017; 121:130-139. [PMID: 28456687 PMCID: PMC5553312 DOI: 10.1016/j.neuropharm.2017.04.039] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/04/2017] [Accepted: 04/25/2017] [Indexed: 01/25/2023]
Abstract
Oxytocin (OT) is a hypothalamic neuropeptide that modulates fear and anxiety-like behaviors. Dorsolateral bed nucleus of the stria terminalis (BNSTdl) plays a critical role in the regulation of fear and anxiety, and expresses high levels of OT receptor (OTR). However, the role of OTR neurotransmission within the BNSTdl in mediating these behaviors is unknown. Here, we used adult male Sprague-Dawley rats to investigate the role of OTR neurotransmission in the BNSTdl in the modulation of the acoustic startle response, as well as in the acquisition and consolidation of conditioned fear using fear potentiated startle (FPS) paradigm. Bilateral intra-BNSTdl administration of OT (100 ng) did not affect the acquisition of conditioned fear response. However, intra-BNSTdl administration of specific OTR antagonist (OTA), (d(CH2)51, Tyr(Me)2, Thr4, Orn8, des-Gly-NH29)-vasotocin, (200 ng), prior to the fear conditioning session, impaired the acquisition of cued fear, without affecting a non-cued fear component of FPS. Neither OTA, nor OT affected baseline startle or shock reactivity during fear conditioning. Therefore, the observed impairment of cued fear after OTA infusion resulted from the specific effect on the formation of cued fear. In contrast to the acquisition, neither OTA nor OT affected the consolidation of FPS, when administered after the completion of fear conditioning session. Taken together, these results reveal the important role of OTR neurotransmission in the BNSTdl in the formation of conditioned fear to a discrete cue. This study also highlights the role of the BNSTdl in learning to discriminate between threatening and safe stimuli.
Collapse
Affiliation(s)
- Mahsa Moaddab
- Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Joanna Dabrowska
- Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA; Department of Neuroscience, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA.
| |
Collapse
|
22
|
Functional Heterogeneity in the Bed Nucleus of the Stria Terminalis. J Neurosci 2017; 36:8038-49. [PMID: 27488624 DOI: 10.1523/jneurosci.0856-16.2016] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/05/2016] [Indexed: 11/21/2022] Open
Abstract
Early work stressed the differing involvement of the central amygdala (CeA) and bed nucleus of the stria terminalis (BNST) in the genesis of fear versus anxiety, respectively. In 2009, Walker, Miles, and Davis proposed a model of amygdala-BNST interactions to explain these functional differences. This model became extremely influential and now guides a new wave of studies on the role of BNST in humans. Here, we consider evidence for and against this model, in the process highlighting central principles of BNST organization. This analysis leads us to conclude that BNST's influence is not limited to the generation of anxiety-like responses to diffuse threats, but that it also shapes the impact of discrete threatening stimuli. It is likely that BNST-CeA interactions are involved in modulating responses to such threats. In addition, whereas current views emphasize the contributions of the anterolateral BNST region in anxiety, accumulating data indicate that the anteromedial and anteroventral regions also play a critical role. The presence of multiple functional subregions within the small volume of BNST raises significant technical obstacles for functional imaging studies in humans.
Collapse
|
23
|
Spierling SR, Zorrilla EP. Don't stress about CRF: assessing the translational failures of CRF 1antagonists. Psychopharmacology (Berl) 2017; 234:1467-1481. [PMID: 28265716 PMCID: PMC5420464 DOI: 10.1007/s00213-017-4556-2] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 01/27/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Dr. Athina Markou sought treatments for a common neural substrate shared by depression and drug dependence. Antagonists of corticotropin-releasing factor (CRF) receptors, a target of interest to her, have not reached the clinic despite strong preclinical rationale and sustained translational efforts. METHODS We explore potential causes for the failure of CRF1 antagonists and review recent findings concerning CRF-CRF1 systems in psychopathology. RESULTS Potential causes for negative outcomes include (1) poor safety and efficacy of initial drug candidates due to bad pharmacokinetic and physicochemical properties, (2) specificity problems with preclinical screens, (3) the acute nature of screens vs. late-presenting patients, (4) positive preclinical results limited to certain models and conditions with dynamic CRF-CRF1 activation not homologous to tested patients, (5) repeated CRF1 activation-induced plasticity that reduces the importance of ongoing CRF1 agonist stimulation, and (6) therapeutic silencing which may need to address CRF2 receptor or CRF-binding protein molecules, constitutive CRF1 activity, or molecules that influence agonist-independent activity or to target structural regions other than the allosteric site bound by all drug candidates. We describe potential markers of activation towards individualized treatment, human genetic, and functional data that still implicate CRF1 systems in emotional disturbance, sex differences, and suggestive clinical findings for CRF1 antagonists in food craving and CRF-driven HPA-axis overactivation. CONCLUSION The therapeutic scope of selective CRF1 antagonists now appears narrower than had been hoped. Yet, much remains to be learned about CRF's role in the neurobiology of dysphoria and addiction and the potential for novel anti-CRF therapies therein.
Collapse
Affiliation(s)
- Samantha R Spierling
- Committee on the Neurobiology of Addictive Disorders, SP30-2400, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA.
| | - Eric P Zorrilla
- Committee on the Neurobiology of Addictive Disorders, SP30-2400, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA.
| |
Collapse
|
24
|
Patriquin MA, Mathew SJ. The Neurobiological Mechanisms of Generalized Anxiety Disorder and Chronic Stress. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2017; 1:2470547017703993. [PMID: 29503978 PMCID: PMC5832062 DOI: 10.1177/2470547017703993] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 03/20/2017] [Indexed: 12/11/2022]
Abstract
Two classification systems are now at the forefront of clinical psychiatric research: (1) Diagnostic and Statistical Manual, Fifth Edition and (2) the National Institutes of Mental Health Research Domain Criteria. Herein, we propose that these two classification systems are complementary rather than mutually exclusive, and when combined provide important information for understanding aspects of the pathophysiology related to Generalized Anxiety Disorder (GAD). The neurobiological literature for GAD and one relevant research domain criteria component, sustained threat, are reviewed from multiple units of analysis (genetic, neuroimaging, neuroendocrine, and psychophysiological). It is hypothesized that generating a comprehensive, biologically based understanding of the relationship between GAD, sustained threat, and the measureable units of analysis will provide information critical to design the most effective treatments.
Collapse
Affiliation(s)
- Michelle A. Patriquin
- Menninger Department of Psychiatry and
Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
- The Menninger Clinic, Houston, TX,
USA
| | - Sanjay J. Mathew
- Menninger Department of Psychiatry and
Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
- Michael E. Debakey VA Medical Center,
Houston, TX, USA
| |
Collapse
|
25
|
Asok A, Schulkin J, Rosen JB. Corticotropin releasing factor type-1 receptor antagonism in the dorsolateral bed nucleus of the stria terminalis disrupts contextually conditioned fear, but not unconditioned fear to a predator odor. Psychoneuroendocrinology 2016; 70:17-24. [PMID: 27153520 PMCID: PMC4907900 DOI: 10.1016/j.psyneuen.2016.04.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/26/2016] [Accepted: 04/27/2016] [Indexed: 10/21/2022]
Abstract
The bed nucleus of the stria terminalis (BNST) plays a critical role in fear and anxiety. The BNST is important for contextual fear learning, but the mechanisms regulating this function remain unclear. One candidate mechanism is corticotropin-releasing-factor (CRF) acting at CRF type 1 receptors (CRFr1s). Yet, there has been little progress in elucidating if CRFr1s in the BNST are involved in different types of fear (conditioned and/or unconditioned). Therefore, the present study investigated the effect of antalarmin, a potent CRFr1 receptor antagonist, injected intracerebroventricularly (ICV) and into the dorsolateral BNST (LBNST) during single trial contextual fear conditioning or exposure to the predator odor 2,5-dihydro-2,4,5-trimethylthiazoline (TMT). Neither ICV nor LBNST antalarmin disrupted unconditioned freezing to TMT. In contrast, ICV and LBNST antalarmin disrupted the retention of contextual fear when tested 24h later. Neither ICV nor LBNST antalarmin affected baseline or post-shock freezing-indicating antalarmin does not interfere with the early phases of contextual fear acquisition. Antalarmin did not (1) permanently affect the ability to learn and express contextual fear, (2) change responsivity to footshocks, or (3) affect the ability to freeze. Our findings highlight an important role for CRFr1s within the LBNST during contextually conditioned fear, but not unconditioned predator odor fear.
Collapse
Affiliation(s)
- Arun Asok
- University of Delaware, Department of Psychological and Brain Sciences, Newark, DE 19716, USA
| | - Jay Schulkin
- Georgetown University, Department of Neuroscience, Washington, DC 20057, USA
| | - Jeffrey B Rosen
- University of Delaware, Department of Psychological and Brain Sciences, Newark, DE 19716, USA.
| |
Collapse
|
26
|
Daldrup T, Lesting J, Meuth P, Seidenbecher T, Pape HC. Neuronal correlates of sustained fear in the anterolateral part of the bed nucleus of stria terminalis. Neurobiol Learn Mem 2016; 131:137-46. [DOI: 10.1016/j.nlm.2016.03.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 03/24/2016] [Accepted: 03/28/2016] [Indexed: 12/23/2022]
|
27
|
Partridge JG, Forcelli PA, Luo R, Cashdan JM, Schulkin J, Valentino RJ, Vicini S. Stress increases GABAergic neurotransmission in CRF neurons of the central amygdala and bed nucleus stria terminalis. Neuropharmacology 2016; 107:239-250. [PMID: 27016019 DOI: 10.1016/j.neuropharm.2016.03.029] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 03/17/2016] [Accepted: 03/21/2016] [Indexed: 11/20/2022]
Abstract
Corticotrophin Releasing Factor (CRF) is a critical stress-related neuropeptide in major output pathways of the amygdala, including the central nucleus (CeA), and in a key projection target of the CeA, the bed nucleus of the stria terminalis (BnST). While progress has been made in understanding the contributions and characteristics of CRF as a neuropeptide in rodent behavior, little attention has been committed to determine the properties and synaptic physiology of specific populations of CRF-expressing (CRF(+)) and non-expressing (CRF(-)) neurons in the CeA and BnST. Here, we fill this gap by electrophysiologically characterizing distinct neuronal subtypes in CeA and BnST. Crossing tdTomato or channelrhodopsin-2 (ChR2-YFP) reporter mice to those expressing Cre-recombinase under the CRF promoter allowed us to identify and manipulate CRF(+) and CRF(-) neurons in CeA and BnST, the two largest areas with fluorescently labeled neurons in these mice. We optogenetically activated CRF(+) neurons to elicit action potentials or synaptic responses in CRF(+) and CRF(-) neurons. We found that GABA is the predominant co-transmitter in CRF(+) neurons within the CeA and BnST. CRF(+) neurons are highly interconnected with CRF(-) neurons and to a lesser extent with CRF(+) neurons. CRF(+) and CRF(-) neurons differentially express tonic GABA currents. Chronic, unpredictable stress increase the amplitude of evoked IPSCs and connectivity between CRF(+) neurons, but not between CRF(+) and CRF(-) neurons in both regions. We propose that reciprocal inhibition of interconnected neurons controls CRF(+) output in these nuclei.
Collapse
Affiliation(s)
- John G Partridge
- Department of Pharmacology & Physiology, Georgetown University School of Medicine, Washington, DC 20007, USA; Interdisciplinary Program in Neuroscience, Georgetown University School of Medicine, Washington, DC 20007, USA.
| | - Patrick A Forcelli
- Department of Pharmacology & Physiology, Georgetown University School of Medicine, Washington, DC 20007, USA; Interdisciplinary Program in Neuroscience, Georgetown University School of Medicine, Washington, DC 20007, USA
| | - Ruixi Luo
- Department of Pharmacology & Physiology, Georgetown University School of Medicine, Washington, DC 20007, USA
| | - Jonah M Cashdan
- Department of Biology, Georgetown University School of Medicine, Washington, DC 20007, USA
| | - Jay Schulkin
- Interdisciplinary Program in Neuroscience, Georgetown University School of Medicine, Washington, DC 20007, USA; Department of Obstetrics & Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Rita J Valentino
- Abramson Pediatric Research Center, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Stefano Vicini
- Department of Pharmacology & Physiology, Georgetown University School of Medicine, Washington, DC 20007, USA; Interdisciplinary Program in Neuroscience, Georgetown University School of Medicine, Washington, DC 20007, USA
| |
Collapse
|
28
|
Hoffman KL. New dimensions in the use of rodent behavioral tests for novel drug discovery and development. Expert Opin Drug Discov 2016; 11:343-53. [DOI: 10.1517/17460441.2016.1153624] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
29
|
Mad men, women and steroid cocktails: a review of the impact of sex and other factors on anabolic androgenic steroids effects on affective behaviors. Psychopharmacology (Berl) 2016; 233:549-69. [PMID: 26758282 PMCID: PMC4751878 DOI: 10.1007/s00213-015-4193-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 12/11/2015] [Indexed: 12/26/2022]
Abstract
RATIONALE For several decades, elite athletes and a growing number of recreational consumers have used anabolic androgenic steroids (AAS) as performance enhancing drugs. Despite mounting evidence that illicit use of these synthetic steroids has detrimental effects on affective states, information available on sex-specific actions of these drugs is lacking. OBJECTIVES The focus of this review is to assess information to date on the importance of sex and its interaction with other environmental factors on affective behaviors, with an emphasis on data derived from non-human studies. METHODS The PubMed database was searched for relevant studies in both sexes. RESULTS Studies examining AAS use in females are limited, reflecting the lower prevalence of use in this sex. Data, however, indicate significant sex-specific differences in AAS effects on anxiety-like and aggressive behaviors, interactions with other drugs of abuse, and the interplay of AAS with other environmental factors such as diet and exercise. CONCLUSIONS Current methods for assessing AAS use have limitations that suggest biases of both under- and over-reporting, which may be amplified for females who are poorly represented in self-report studies of human subjects and are rarely used in animal studies. Data from animal literature suggest that there are significant sex-specific differences in the impact of AAS on aggression, anxiety, and concomitant use of other abused substances. These results have relevance for human females who take these drugs as performance-enhancing substances and for transgender XX individuals who may illicitly self-administer AAS as they transition to a male gender identity.
Collapse
|
30
|
Lifelong disturbance of serotonin transporter functioning results in fear learning deficits: Reversal by blockade of CRF1 receptors. Eur Neuropsychopharmacol 2015; 25:1733-43. [PMID: 26302762 DOI: 10.1016/j.euroneuro.2015.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 05/28/2015] [Accepted: 07/14/2015] [Indexed: 12/28/2022]
Abstract
The inability to associate aversive events with relevant cues (i.e. fear learning) may lead to maladaptive anxiety. To further study the role of the serotonin transporter (SERT) in fear learning, classical fear conditioning was studied in SERT knockout rats (SERT(-/-)) using fear potentiation of the startle reflex. Next, fear acquisition and concomitant development of contextual conditioned fear were monitored during training. To differentiate between developmental and direct effects of reduced SERT functioning, effects of acute and chronic SSRI treatment were studied in adult rats. Considering the known interactions between serotonin and corticotropin-releasing factor (CRF), we studied the effect of the CRFR1 antagonist CP154,526 on behavioral changes observed and determined CRF1 receptor levels in SERT(-/-) rats. SERT(-/-) showed blunted fear potentiation and enhanced contextual fear, which resulted from a deficit in fear acquisition. Paroxetine treatment did not affect acquisition or expression of fear-potentiated startle, suggesting that disturbed fear learning in SERT(-/-) results from developmental changes and not from reduced SERT functioning. Although CRF1 receptor levels did not differ significantly between genotypes, CP154,526 treatment normalized both cue- and contextual fear in SERT(-/-) during acquisition, but not expression of fear-potentiated startle. The disrupted fear acquisition and concomitant increase in contextual conditioned fear-potentiated startle fear in SERT(-/-) resembles the associative learning deficit seen in patients with panic disorder and suggests that normal SERT functioning is crucial for the development of an adequate fear neuro-circuitry. Moreover, the normalization of fear acquisition by CP154,526 suggests a role for central CRF signaling in the generalization of fear.
Collapse
|
31
|
The CRH1 antagonist GSK561679 increases human fear but not anxiety as assessed by startle. Neuropsychopharmacology 2015; 40:1064-71. [PMID: 25430779 PMCID: PMC4367474 DOI: 10.1038/npp.2014.316] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/14/2014] [Accepted: 11/17/2014] [Indexed: 01/27/2023]
Abstract
Fear to predictable threat and anxiety to unpredictable threat reflect distinct processes mediated by different brain structures, the central nucleus of the amygdala and the bed nucleus of the stria terminalis (BNST), respectively. This study tested the hypothesis that the corticotropin-releasing factor (CRF1) antagonist GSK561679 differentially reduces anxiety but increases fear in humans. A total of 31 healthy females received each of four treatments: placebo, 50 mg GSK561679 (low-GSK), 400 mg GSK561679 (high-GSK), and 1 mg alprazolam in a crossover design. Participants were exposed to three conditions during each of the four treatments. The three conditions included one in which predictable aversive shocks were signaled by a cue, a second during which shocks were administered unpredictably, and a third condition without shock. Fear and anxiety were assessed using the acoustic startle reflex. High-GSK had no effect on startle potentiation during unpredictable threat (anxiety) but increased startle potentiation during the predictable condition (fear). Low-GSK did not affect startle potentiation across conditions. Consistent with previous findings, alprazolam reduced startle potentiation during unpredictable threat but not during predictable threat. The increased fear by high-GSK replicates animal findings and suggests a lift of the inhibitory effect of the BNST on the amygdala by the CRF1 antagonist.
Collapse
|
32
|
Flandreau E, Risbrough V, Lu A, Ableitner M, Geyer MA, Holsboer F, Deussing JM. Cell type-specific modifications of corticotropin-releasing factor (CRF) and its type 1 receptor (CRF1) on startle behavior and sensorimotor gating. Psychoneuroendocrinology 2015; 53:16-28. [PMID: 25575243 PMCID: PMC4364548 DOI: 10.1016/j.psyneuen.2014.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 11/25/2014] [Accepted: 12/03/2014] [Indexed: 10/24/2022]
Abstract
The corticotropin-releasing factor (CRF) family of peptides and receptors coordinates the mammalian endocrine, autonomic, and behavioral responses to stress. Excessive CRF production has been implicated in the etiology of stress-sensitive psychiatric disorders such as posttraumatic stress disorder (PTSD), which is associated with alterations in startle plasticity. The CRF family of peptides and receptors mediate acute startle response changes during stress, and chronic CRF activation can induce startle abnormalities. To determine what neural circuits modulate startle in response to chronic CRF activation, transgenic mice overexpressing CRF throughout the central nervous system (CNS; CRF-COE(CNS)) or restricted to inhibitory GABAergic neurons (CRF-COE(GABA)) were compared across multiple domains of startle plasticity. CRF overexpression throughout the CNS increased startle magnitude and reduced ability to inhibit startle (decreased habituation and decreased prepulse inhibition (PPI)), similar to previous reports of exogenous effects of CRF. Conversely, CRF overexpression confined to inhibitory neurons decreased startle magnitude but had no effect on inhibitory measures. Acute CRF receptor 1 (CRF1) antagonist treatment attenuated only the effects on startle induced by CNS-specific CRF overexpression. Specific deletion of CRF1 receptors from forebrain principal neurons failed to alter the effects of exogenous CRF or stress on startle, suggesting that these CRF1 expressing neurons are not required for CRF-induced changes in startle behaviors. These data indicate that the effects of CRF activation on startle behavior utilize an extensive neural circuit that includes both forebrain and non-forebrain regions. Furthermore, these findings suggest that the neural source of increased CRF release determines the startle phenotype elicited. It is conceivable that this may explain why disorders characterized by increased CRF in cerebrospinal fluid (e.g. PTSD and major depressive disorder) have distinct symptom profiles in terms of startle reactivity.
Collapse
Affiliation(s)
| | - Victoria Risbrough
- Veterans Administration Center of Excellence for Stress and Mental Health, 3350 La Jolla Village Drive San Diego, CA 92161, USA.
| | - Ailing Lu
- Unit of Innate Immunity, Key Laboratory of Molecular Virology and Immunology Institut Pasteur of Shanghai, Chinese Academy of Sciences. 320 Yue Yang Road, Shanghai, 200031; China. Phone/Fax: 86-21-54923102/54923101
| | - Martin Ableitner
- Max Planck Institute of Psychiatry, Kraepelinstrasse 2-10 D-80804, Munich Phone: +49 (0)89 / 30622-645 Fax: +49 (0)89 / 30622-610
| | - Mark A Geyer
- Department of Psychiatry University of California San Diego 9500 Gilman Drive MC 0804 La Jolla, CA 92093-0804 ph (619)543-3582 fx (619)543-2493
| | - Florian Holsboer
- Max Planck Institute of Psychiatry Kraepelinstr. 2-10 80804 Munich, Germany Phone: +49-89-30622-220 Fax: +49-89-30622-483
| | - Jan M Deussing
- Department Stress Neurobiology and Neurogenetics Max Planck Institute of Psychiatry Kraepelinstrasse 2-10 D-80804, Munich Phone: +49 (0)89 / 30622-639 Fax: +49 (0)89 / 30622-610
| |
Collapse
|
33
|
Kash TL, Pleil KE, Marcinkiewcz CA, Lowery-Gionta EG, Crowley N, Mazzone C, Sugam J, Hardaway JA, McElligott ZA. Neuropeptide regulation of signaling and behavior in the BNST. Mol Cells 2015; 38:1-13. [PMID: 25475545 PMCID: PMC4314126 DOI: 10.14348/molcells.2015.2261] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 09/29/2014] [Indexed: 12/23/2022] Open
Abstract
Recent technical developments have transformed how neuroscientists can probe brain function. What was once thought to be difficult and perhaps impossible, stimulating a single set of long range inputs among many, is now relatively straight-forward using optogenetic approaches. This has provided an avalanche of data demonstrating causal roles for circuits in a variety of behaviors. However, despite the critical role that neuropeptide signaling plays in the regulation of behavior and physiology of the brain, there have been remarkably few studies demonstrating how peptide release is causally linked to behaviors. This is likely due to both the different time scale by which peptides act on and the modulatory nature of their actions. For example, while glutamate release can effectively transmit information between synapses in milliseconds, peptide release is potentially slower [See the excellent review by Van Den Pol on the time scales and mechanisms of release (van den Pol, 2012)] and it can only tune the existing signals via modulation. And while there have been some studies exploring mechanisms of release, it is still not as clearly known what is required for efficient peptide release. Furthermore, this analysis could be complicated by the fact that there are multiple peptides released, some of which may act in contrast. Despite these limitations, there are a number of groups making progress in this area. The goal of this review is to explore the role of peptide signaling in one specific structure, the bed nucleus of the stria terminalis, that has proven to be a fertile ground for peptide action.
Collapse
Affiliation(s)
- Thomas L. Kash
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - Kristen E. Pleil
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - Catherine A. Marcinkiewcz
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - Emily G. Lowery-Gionta
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - Nicole Crowley
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - Christopher Mazzone
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - Jonathan Sugam
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - J. Andrew Hardaway
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - Zoe A. McElligott
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| |
Collapse
|
34
|
Rainnie DG, Hazra R, Dabrowska J, Guo JD, Li CC, Dewitt S, Muly EC. Distribution and functional expression of Kv4 family α subunits and associated KChIP β subunits in the bed nucleus of the stria terminalis. J Comp Neurol 2014; 522:609-25. [PMID: 24037673 DOI: 10.1002/cne.23435] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 06/28/2013] [Accepted: 07/02/2013] [Indexed: 12/22/2022]
Abstract
Regulation of BNSTALG neuronal firing activity is tightly regulated by the opposing actions of the fast outward potassium current, IA , mediated by α subunits of the Kv4 family of ion channels, and the transient inward calcium current, IT . Together, these channels play a critical role in regulating the latency to action potential onset, duration, and frequency, as well as dendritic back-propagation and synaptic plasticity. Previously we have shown that Type I-III BNSTALG neurons express mRNA transcripts for each of the Kv4 α subunits. However, the biophysical properties of native IA channels are critically dependent on the formation of macromolecular complexes of Kv4 channels with a family of chaperone proteins, the potassium channel-interacting proteins (KChIP1-4). Here we used a multidisciplinary approach to investigate the expression and function of Kv4 channels and KChIPs in neurons of the rat BNSTALG . Using immunofluorescence we demonstrated the pattern of localization of Kv4.2, Kv4.3, and KChIP1-4 proteins in the BNSTALG . Moreover, our single-cell reverse-transcription polymerase chain reaction (scRT-PCR) studies revealed that mRNA transcripts for Kv4.2, Kv4.3, and all four KChIPs were differentially expressed in Type I-III BNSTALG neurons. Furthermore, immunoelectron microscopy revealed that Kv4.2 and Kv4.3 channels were primarily localized to the dendrites and spines of BNSTALG neurons, and are thus ideally situated to modulate synaptic transmission. Consistent with this observation, in vitro patch clamp recordings showed that reducing postsynaptic IA in these neurons lowered the threshold for long-term potentiation (LTP) induction. These results are discussed in relation to potential modulation of IA channels by chronic stress.
Collapse
Affiliation(s)
- Donald G Rainnie
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia; Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, Georgia
| | | | | | | | | | | | | |
Collapse
|
35
|
Schulz-Klaus B, Lessmann V, Endres T. BDNF-dependent consolidation of fear memories in the perirhinal cortex. Front Behav Neurosci 2013; 7:205. [PMID: 24381548 PMCID: PMC3865772 DOI: 10.3389/fnbeh.2013.00205] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 12/02/2013] [Indexed: 01/09/2023] Open
Abstract
In the recent years the perirhinal cortex (PRh) has been identified as a crucial brain area in fear learning. Since the neurotrophin brain-derived neurotrophic factor (BDNF) is an important mediator of synaptic plasticity and also crucially involved in memory consolidation of several learning paradigms, we analyzed now whether fear conditioning influences the expression of BDNF protein in the PRh. Here we observed a specific increase of BDNF protein 120 min after fear conditioning training. In order to test whether this increase of BDNF protein level is also required for the consolidation of the fear memory, we locally applied the Trk receptor inhibitor k252a into the PRh during this time window in a second series of experiments. By interfering with Trk-signaling during this critical time window, the formation of a long-term fear memory was completely blocked, indicated by a complete lack of fear potentiated startle 1 day later. In conclusion the present study further emphasizes the important role of the PRh in cued fear learning and identified BDNF as an important mediator for fear memory consolidation in the PRh.
Collapse
Affiliation(s)
| | - Volkmar Lessmann
- Medizinische Fakultät, Institut für Physiologie, Otto-von-Guericke Universität Magdeburg Magdeburg, Germany ; Center for Behavioral Brain Research (CBBS), Otto-von-Guericke Universität Magdeburg Magdeburg, Germany
| | - Thomas Endres
- Medizinische Fakultät, Institut für Physiologie, Otto-von-Guericke Universität Magdeburg Magdeburg, Germany
| |
Collapse
|
36
|
M T, JE G, RL H, AL H, VB R. The role of PKC signaling in CRF-induced modulation of startle. Psychopharmacology (Berl) 2013; 229:579-89. [PMID: 23722830 PMCID: PMC3784645 DOI: 10.1007/s00213-013-3114-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 04/11/2013] [Indexed: 02/06/2023]
Abstract
RATIONALE Hypersignaling of corticotropin releasing factor (CRF) has been implicated in stress disorders; however, many of its downstream mechanisms of action remain unclear. In vitro, CRF1 receptor activation initiates multiple cell signaling cascades, including protein kinase A (PKA), protein kinase C (PKC), and mitogen-activated protein kinase kinase MEK1/2 signaling. It is unclear, however, which of these signaling cascades mediate CRF-induced behaviors during stress. OBJECTIVES We examined the role of PKA, PKC, and MEK1/2 signaling pathways in CRF-induced anxiety as measured by startle hyperreactivity. METHODS Mice treated with intracerbroventricular (ICV) ovine CRF (oCRF) were pretreated with the PKA inhibitor Rp-cAMPS, PKC inhibitor bisindolylmaleimide (BIM), or MEK1/2 inhibitor PD98059 (ICV) and assessed for acoustic startle reactivity. RESULTS The PKC inhibitor BIM significantly attenuated CRF-induced increases in startle. BIM was also able to block startle increases induced by oCRF when both compounds were infused directly into the bed nucleus of stria terminalis (BNST). PKA and MEK1/2 inhibition had no significant effects on CRF-induced changes in startle at the dose ranges tested. CRF-induced disruption of prepulse inhibition was not significantly reversed by any of the three pretreatments at the dose ranges tested. CONCLUSIONS PKC signaling is required for CRF-induced increases in startle, and this effect is mediated at least in part at the BNST. These findings suggest that PKC signaling cascades (1) may be important for the acute effects of CRF to induce startle hyperreactivity and (2) support further research of the role of PKC signaling in startle abnormalities relevant to disorders such as posttraumatic stress disorder.
Collapse
Affiliation(s)
- Toth M
- Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla CA USA,Dept. of Psychiatry, University of California San Diego, La Jolla CA USA
| | - Gresack JE
- Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla CA USA,Dept. of Psychiatry, University of California San Diego, La Jolla CA USA,Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York NY USA
| | - Hauger RL
- Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla CA USA,Dept. of Psychiatry, University of California San Diego, La Jolla CA USA
| | - Halberstadt AL
- Dept. of Psychiatry, University of California San Diego, La Jolla CA USA
| | - Risbrough VB
- Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla CA USA,Dept. of Psychiatry, University of California San Diego, La Jolla CA USA,Corresponding author: Victoria Risbrough, Ph.D., University of California San Diego, 9500 Gilman Dr. MC0804, La Jolla CA 92093-0804, Tel: 16195433582; Fax: 16195432475:
| |
Collapse
|
37
|
Sink KS, Chung A, Ressler KJ, Davis M, Walker DL. Anxiogenic effects of CGRP within the BNST may be mediated by CRF acting at BNST CRFR1 receptors. Behav Brain Res 2013; 243:286-93. [PMID: 23376701 DOI: 10.1016/j.bbr.2013.01.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 01/16/2013] [Accepted: 01/23/2013] [Indexed: 12/17/2022]
Abstract
Calcitonin gene-related peptide (CGRP) acting within the bed nucleus of the stria terminalis (BNST) increases anxiety as well as neural activation in anxiety-related structures, and mediates behavioral stress responses. Similar effects have been described following intra-ventricular as well as intra-BNST infusions of the stress-responsive neuropeptide, corticotropin releasing factor (CRF). Interestingly, CGRP-positive terminals within the lateral division of the BNST form perisomatic baskets around neurons that express CRF, suggesting that BNST CGRP could exert its anxiogenic effects by increasing release of CRF from these neurons. With this in mind, the present set of experiments was designed to examine the role of CRFR1 signaling in the anxiogenic effects of CGRP within the BNST and to determine whether CRF from BNST neurons contributes to these effects. Consistent with previous studies, we found that 400 ng CGRP infused bilaterally into the BNST increased the acoustic startle response and induced anxiety-like behavior in the elevated plus maze compared to vehicle. Both of these effects were attenuated by 10mg/kg PO of the CRFR1 antagonist, GSK876008. GSK876008 alone did not affect startle. An intra-BNST infusion of the CRFR1 antagonist CP376395 (2 μg) also blocked increases in acoustic startle induced by intra-BNST infusion of CGRP, as did virally-mediated siRNA knockdown of CRF expression locally within the BNST. Together, these results suggest that the anxiogenic effects of intra-BNST CGRP may be mediated by CRF from BNST neurons acting at local CRFR1 receptors.
Collapse
Affiliation(s)
- K S Sink
- Department of Psychiatry and Behavioral Sciences, Yerkes National Primate Center, Emory University, Atlanta, GA 30329, USA
| | | | | | | | | |
Collapse
|
38
|
Identification of neurons specifically activated after recall of context fear conditioning. Neurobiol Learn Mem 2012; 98:139-47. [PMID: 22820091 DOI: 10.1016/j.nlm.2012.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Revised: 06/11/2012] [Accepted: 07/05/2012] [Indexed: 11/22/2022]
Abstract
The learning of new information and recall of that information presumably involves modification of and access to shared circuitry in the brain. However, learning and recall may involve the activation of distinct parts of that circuitry, according to the quite distinct functional differences between these two processes. Previously we examined neuronal activation following learning of context fear conditioning. Using the Fos-Tau-LacZ (FTL) transgenic mouse to label activated neurons, we identified a number of distinct populations of neurons in amygdala and hypothalamus which showed learning specific activation. These populations of neurons showed much less activation following recall. Here we ask what populations of neurons might be specifically activated following recall. We trained mice in context fear conditioning, and then looked at FTL activation following recall of context fear. We identified a number of populations of neurons which showed recall specific activation in nucleus accumbens shell, the anterio-medial bed nucleus of stria terminalis, the anterior commissural nucleus and the periventricular hypothalamic nucleus. These were all different populations of neurons compared with those activated following context fear learning. These different functional activation patterns occurring between learning and recall may reflect the different brain functions occurring between these two memory related processes.
Collapse
|
39
|
Oberlander JG, Henderson LP. Corticotropin-releasing factor modulation of forebrain GABAergic transmission has a pivotal role in the expression of anabolic steroid-induced anxiety in the female mouse. Neuropsychopharmacology 2012; 37:1483-99. [PMID: 22298120 PMCID: PMC3327853 DOI: 10.1038/npp.2011.334] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 12/20/2011] [Accepted: 12/20/2011] [Indexed: 12/14/2022]
Abstract
Increased anxiety is commonly observed in individuals who illicitly administer anabolic androgenic steroids (AAS). Behavioral effects of steroid abuse have become an increasing concern in adults and adolescents of both sexes. The dorsolateral bed nucleus of the stria terminalis (dlBnST) has a critical role in the expression of diffuse anxiety and is a key site of action for the anxiogenic neuromodulator, corticotropin releasing factor (CRF). Here we demonstrate that chronic, but not acute, exposure of female mice during adolescence to AAS augments anxiety-like behaviors; effects that were blocked by central infusion of the CRF receptor type 1 antagonist, antalarmin. AAS treatment selectively increased action potential (AP) firing in neurons of the central amygdala (CeA) that project to the dlBnST, increased the frequency of GABA(A) receptor-mediated spontaneous inhibitory postsynaptic currents (sIPSCs) in dlBnST target neurons, and decreased both c-FOS immunoreactivity (IR) and AP frequency in these postsynaptic cells. Acute application of antalarmin abrogated the enhancement of GABAergic inhibition induced by chronic AAS exposure whereas application of CRF to brain slices of naïve mice mimicked the actions of this treatment. These results, in concert with previous data demonstrating that chronic AAS treatment results in enhanced levels of CRF mRNA in the CeA and increased CRF-IR in the dlBnST neuropil, are consistent with a mechanism in which the enhanced anxiety elicited by chronic AAS exposure involves augmented inhibitory activity of CeA afferents to the dlBnST and CRF-dependent enhancement of GABAergic inhibition in this brain region.
Collapse
Affiliation(s)
- Joseph G Oberlander
- Department of Physiology & Neurobiology, Dartmouth Medical School, Hanover, NH, USA
| | - Leslie P Henderson
- Department of Physiology & Neurobiology, Dartmouth Medical School, Hanover, NH, USA
| |
Collapse
|
40
|
Stress-induced reinstatement of alcohol-seeking in rats is selectively suppressed by the neurokinin 1 (NK1) antagonist L822429. Psychopharmacology (Berl) 2011; 218:111-9. [PMID: 21340476 PMCID: PMC3192232 DOI: 10.1007/s00213-011-2201-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Accepted: 01/23/2011] [Indexed: 10/18/2022]
Abstract
RATIONALE Genetic inactivation or pharmacological antagonism of neurokinin 1 (NK1) receptors blocks morphine and alcohol reward in rodents, while NK1 antagonism decreases alcohol craving in humans. The role of the NK1 system for relapse-like behavior has not previously been examined. OBJECTIVE Divergence between human and rodent NK1 receptors has limited the utility of NK1 antagonists developed for the human receptor species for preclinical studies of addiction-related behaviors in rats. Here we used L822429, an NK1 antagonist specifically engineered to bind at high affinity to the rat receptor, to assess the effects of NK1 receptor antagonism on alcohol-seeking behaviors in rats. METHODS L822429 (15 and 30 mg/kg) was used to examine effects of NK1 receptor antagonism on operant self-administration of 10% alcohol in 30-min daily sessions, as well as intermittent footshock stress- and cue-induced reinstatement of alcohol-seeking after extinction of lever responding. RESULTS At the doses used, L822429 did not significantly affect alcohol self-administration or cue-induced reinstatement, but potently and dose dependently suppressed stress-induced reinstatement of alcohol seeking, with an essentially complete suppression at the highest dose. The effect of L822429 on stress-induced reinstatement was behaviorally specific. The drug had no effect on conditioned suppression of operant responding following fear conditioning, locomotor activity, or self-administration of a sucrose solution. CONCLUSIONS To the degree that the reinstatement model provides a model of drug relapse, the results provide support for NK1 antagonism as a promising mechanism for pharmacotherapy of alcoholism, acting through suppression of stress-induced craving and relapse.
Collapse
|
41
|
Steiner MA, Lecourt H, Rakotoariniaina A, Jenck F. Favoured genetic background for testing anxiolytics in the fear-potentiated and light-enhanced startle paradigms in the rat. Behav Brain Res 2011; 221:34-42. [DOI: 10.1016/j.bbr.2011.02.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 02/04/2011] [Accepted: 02/12/2011] [Indexed: 12/31/2022]
|
42
|
Abstract
Previous findings suggest differences in the neuroanatomical substrates of short- (seconds) vs longer-duration (minutes) fear responses. We now report that phasic and sustained fear can also be differentiated pharmacologically, based on their response to several treatments that either are or are not clinically effective anxiolytics. For these experiments, short- or long-duration clicker stimuli were paired with footshock. Acoustic startle amplitude was later measured in the absence of the clicker, or within seconds (phasic fear) or minutes (sustained fear) of its onset. Before testing, rats received a single injection of vehicle, the benzodiazepine chlordiazepoxide, the 5HT(1A) agonist and dopamine D2 antagonist buspirone, the selective serotonin reuptake inhibitor fluoxetine, or a 3-week treatment with either vehicle or fluoxetine. Chlordiazepoxide blocked sustained, but not phasic startle increases. Acute buspirone, which is not anxiolytic in human beings, did not affect sustained startle increases, but did disrupt phasic increases. Chronic fluoxetine blocked sustained startle increases and unreliably reduced phasic increases; acute fluoxetine affected neither. The results indicate that phasic and sustained fear responses can be pharmacologically dissociated, further validating this distinction, and suggest that sustained startle increases may be especially useful as anxiety models and anxiolytic screens.
Collapse
|
43
|
Bijlsma E, van Leeuwen M, Westphal K, Olivier B, Groenink L. Local repeated corticotropin-releasing factor infusion exacerbates anxiety- and fear-related behavior: differential involvement of the basolateral amygdala and medial prefrontal cortex. Neuroscience 2011; 173:82-92. [DOI: 10.1016/j.neuroscience.2010.11.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 10/05/2010] [Accepted: 11/11/2010] [Indexed: 12/20/2022]
|
44
|
Restraint stress-induced reduction in prepulse inhibition in Brown Norway rats: role of the CRF2 receptor. Neuropharmacology 2010; 60:561-71. [PMID: 21185316 DOI: 10.1016/j.neuropharm.2010.12.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 12/15/2010] [Accepted: 12/15/2010] [Indexed: 11/22/2022]
Abstract
Stress plays a role in many psychiatric disorders that are characterized by deficits in prepulse inhibition (PPI), a form of sensorimotor gating. Corticotropin-releasing factor (CRF) is one of the most important neurotransmitters involved in behavioral components of the stress response. Central infusion of CRF reduces PPI in both rats and mice. In mice, it has been shown that CRF(1) receptor activation mediates the effect of exogenous CRF on PPI. However, the roles of the two CRF receptors in a stress-induced reduction in PPI are not known. We sought to determine whether CRF(1) and/or CRF(2) receptor blockade attenuates a stress-induced reduction of PPI in rats. In separate experiments, we assessed PPI in Brown Norway rats after exposure to 5 days of 2-h restraint, and after pretreatment with the CRF(1) receptor antagonist, CP-154,526 (20.0 mg/kg), or the CRF(2) receptor antagonist, antisauvagine-30 (10.0 μg). Repeated, but not acute, restraint decreased PPI and attenuated the increase in PPI caused by repeated PPI testing. Blockade of the CRF(1) receptor did not attenuate the effect of repeated restraint on PPI or grooming behavior. While CRF(2) receptor blockade did attenuate the effect of repeated restraint on PPI, repeated ICV infusion of the selective CRF(2) receptor agonist urocortin III, did not affect PPI. These findings demonstrate the effect of stress on sensorimotor gating and suggest that the CRF(2) receptor mediates this effect in rats.
Collapse
|
45
|
Chronic anabolic androgenic steroid exposure alters corticotropin releasing factor expression and anxiety-like behaviors in the female mouse. Psychoneuroendocrinology 2010; 35:1473-85. [PMID: 20537804 PMCID: PMC2937186 DOI: 10.1016/j.psyneuen.2010.04.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 04/05/2010] [Accepted: 04/27/2010] [Indexed: 01/18/2023]
Abstract
In the past several decades, the therapeutic use of anabolic androgenic steroids (AAS) has been overshadowed by illicit use of these drugs by elite athletes and a growing number of adolescents to enhance performance and body image. As with adults, AAS use by adolescents is associated with a range of behavioral effects, including increased anxiety and altered responses to stress. It has been suggested that adolescents, especially adolescent females, may be particularly susceptible to the effects of these steroids, but few experiments in animal models have been performed to test this assertion. Here we show that chronic exposure of adolescent female mice to a mixture of three commonly abused AAS (testosterone cypionate, nandrolone decanoate and methandrostenolone; 7.5 mg/kg/day for 5 days) significantly enhanced anxiety-like behavior as assessed by the acoustic startle response (ASR), but did not augment the fear-potentiated startle response (FPS) or alter sensorimotor gating as assessed by prepulse inhibition of the acoustic startle response (PPI). AAS treatment also significantly increased the levels of corticotropin releasing factor (CRF) mRNA and somal-associated CRF immunoreactivity in the central nucleus of the amygdala (CeA), as well as neuropil-associated immunoreactivity in the dorsal aspect of the anterolateral division of the bed nucleus of the stria terminalis (dBnST). AAS treatment did not alter CRF receptor 1 or 2 mRNA in either the CeA or the dBnST; CRF immunoreactivity in the ventral BnST, the paraventricular nucleus (PVN) or the median eminence (ME); or peripheral levels of corticosterone. These results suggest that chronic AAS treatment of adolescent female mice may enhance generalized anxiety, but not sensorimotor gating or learned fear, via a mechanism that involves increased CRF-mediated signaling from CeA neurons projecting to the dBnST.
Collapse
|
46
|
Orexins in the paraventricular nucleus of the thalamus mediate anxiety-like responses in rats. Psychopharmacology (Berl) 2010; 212:251-65. [PMID: 20645079 DOI: 10.1007/s00213-010-1948-y] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 07/02/2010] [Indexed: 12/28/2022]
Abstract
RATIONALE Anatomical studies have shown that the paraventricular nucleus of the thalamus (PVT) innervates areas of the forebrain involved in the expression and regulation of emotional behaviors including fear and anxiety. In addition, the PVT is densely innervated by fibers containing orexin-A (OXA) and orexin-B (OXB), peptides that are well-known for their arousal effects on behavior. OBJECTIVES In this study, we investigate whether microinjections of orexin receptor agonists and antagonists in the PVT region alter expression of anxiety-like behaviors in the rat as measured in the elevated plus maze. RESULTS We report that microinjections of OXA and OXB in the PVT region elicited anxiety-like response as indicated by a reduction in open arm time and entries. In addition, OXA and OXB produced changes in ethological measures indicative of an anxiety state. Central administrations of antagonists for corticotropin releasing factor (CRF) or the opioid kappa receptors attenuated the anxiogenic effects produced by microinjections of OXA in the PVT region. We also provide evidence that endogenously released orexins act at the PVT to produce anxiety by showing that microinjections of TCSOX229, an orexin-2 receptor antagonist, in the PVT region attenuated the anxiogenic effects produced by a previous exposure to footshock stress. CONCLUSIONS This study indicates that endogenously released orexins act on the PVT to regulate anxiety levels through mechanisms involving the brain kappa and CRF receptors.
Collapse
|
47
|
Fendt M, Buchi M, Bürki H, Imobersteg S, Ricoux B, Suply T, Sailer AW. Neuropeptide S receptor deficiency modulates spontaneous locomotor activity and the acoustic startle response. Behav Brain Res 2010; 217:1-9. [PMID: 20888368 DOI: 10.1016/j.bbr.2010.09.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 09/17/2010] [Accepted: 09/21/2010] [Indexed: 10/19/2022]
Abstract
The present study investigated the phenotype of heterozygous and homozygous neuropeptide S receptor (Npsr) deficient C57BL/6 mice in NPS- and cocaine induced hyperactivity, spontaneous and reactive locomotor activity, elevated plus maze, conditioned fear, and prepulse inhibition of the acoustic startle response. In Npsr-deficient mice, a strong reduction of spontaneous locomotor activity and of the startle magnitude was observed; heterozygous mice had an intermediate phenotype. In the other experiments, Npsr deficiency leads to no or only a very modest phenotype. These results support an important role of neuropeptide S in regulating locomotor activity.
Collapse
Affiliation(s)
- Markus Fendt
- Novartis Institutes of BioMedical Research, Neuropsychiatry, GPCR Expertise Program, Basel, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
48
|
Kehne JH, Cain CK. Therapeutic utility of non-peptidic CRF1 receptor antagonists in anxiety, depression, and stress-related disorders: evidence from animal models. Pharmacol Ther 2010; 128:460-87. [PMID: 20826181 DOI: 10.1016/j.pharmthera.2010.08.011] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2010] [Accepted: 08/17/2010] [Indexed: 12/19/2022]
Abstract
Adaptive responding to threatening stressors is of fundamental importance for survival. Dysfunctional hyperactivation of corticotropin releasing factor type-1 (CRF(1)) receptors in stress response system pathways is linked to stress-related psychopathology and CRF(1) receptor antagonists (CRAs) have been proposed as novel therapeutic agents. CRA effects in diverse animal models of stress that detect anxiolytics and/or antidepressants are reviewed, with the goal of evaluating their potential therapeutic utility in depression, anxiety, and other stress-related disorders. CRAs have a distinct phenotype in animals that has similarities to, and differences from, those of classic antidepressants and anxiolytics. CRAs are generally behaviorally silent, indicating that CRF(1) receptors are normally in a state of low basal activation. CRAs reduce stressor-induced HPA axis activation by blocking pituitary and possibly brain CRF(1) receptors which may ameliorate chronic stress-induced pathology. In animal models sensitive to anxiolytics and/or antidepressants, CRAs are generally more active in those with high stress levels, conditions which may maximize CRF(1) receptor hyperactivation. Clinically, CRAs have demonstrated good tolerability and safety, but have thus far lacked compelling efficacy in major depressive disorder, generalized anxiety disorder, or irritable bowel syndrome. CRAs may be best suited for disorders in which stressors clearly contribute to the underlying pathology (e.g. posttraumatic stress disorder, early life trauma, withdrawal/abstinence from addictive substances), though much work is needed to explore these possibilities. An evolving literature exploring the genetic, developmental and environmental factors linking CRF(1) receptor dysfunction to stress-related psychopathology is discussed in the context of improving the translational value of current animal models.
Collapse
Affiliation(s)
- John H Kehne
- Translational Neuropharmacology Consulting, LLC, 9710 Traville Gateway Drive #307, Rockville, MD 20850-7408, USA.
| | | |
Collapse
|
49
|
Pickens CL, Navarre BM, Nair SG. Incubation of conditioned fear in the conditioned suppression model in rats: role of food-restriction conditions, length of conditioned stimulus, and generality to conditioned freezing. Neuroscience 2010; 169:1501-10. [PMID: 20600654 DOI: 10.1016/j.neuroscience.2010.06.036] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2010] [Revised: 06/14/2010] [Accepted: 06/16/2010] [Indexed: 11/19/2022]
Abstract
We recently adapted the conditioned suppression of operant responding method to study fear incubation. We found that food-restricted rats show low fear 2 days after extended (10 d; 100 30-s tone-shock pairings) fear training and high fear after 1-2 months. Here, we studied a potential mechanism of fear incubation: extended food-restriction stress. We also studied whether fear incubation is observed after fear training with a prolonged-duration (6-min) tone conditioned stimulus (CS), and whether conditioned freezing incubates after extended training in rats with or without a concurrent operant task. Conditioned fear was assessed 2 days and 1 month after training. In the conditioned suppression method, fear incubation was reliably observed in rats under moderate food-restriction conditions (18-20 g food/day) that allowed for weight gain, and after extended (10 d), but not limited (1 d), fear training with the 6-min CS. Incubation of conditioned freezing was observed after extended fear training in rats lever-pressing for food and, to a lesser degree, in rats not performing an operant task. Results indicate that prolonged hunger-related stress does not account for fear incubation in the conditioned suppression method, and that fear incubation occurs to a longer-duration (6-min) fear CS. Extended training also leads to robust fear incubation of conditioned freezing in rats performing an operant task and weaker fear incubation in rats not performing an operant task.
Collapse
Affiliation(s)
- C L Pickens
- Behavioral Neuroscience Branch, Intramural Research Program-National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD 21218, USA.
| | | | | |
Collapse
|
50
|
Sanford LD, Yang L, Wellman LL, Liu X, Tang X. Differential effects of controllable and uncontrollable footshock stress on sleep in mice. Sleep 2010; 33:621-30. [PMID: 20469804 DOI: 10.1093/sleep/33.5.621] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
STUDY OBJECTIVES Inescapable shock (IS), an uncontrollable stressor, and presentation of fearful contexts associated with IS produce prominent reductions in REM sleep. We compared sleep in animals trained with IS to that in animals trained with escapable shock (ES), a controllable stressor, in a paradigm in which animals always received shock but could terminate it by their actions. DESIGN Male BALB/cJ mice were implanted with telemetry transmitters for recording EEG and activity. After recovery from surgery, baseline sleep recordings were obtained for 2 days. The mice were then randomly assigned to receive ES (n=9) or IS (n=9). ES mice could escape a footshock (20 trials; 0.5 mA; 5.0 sec maximum duration; 1.0 min intervals) by moving to the unoccupied chamber in a shuttlebox. Yoked-control IS mice in a separate shuttlebox received identical footshock. The mice received 2 days of shock training (ST1; ST2) and were re-exposed to the shuttlebox without footshock (context alone). SETTING NA. PATIENTS OR PARTICIPANTS NA. INTERVENTIONS NA. MEASUREMENTS AND RESULTS On each training and test day, the mice were returned to their home cages, and EEG and activity were recorded for 20 h. Freezing was scored in the context alone. Compared to baseline, ES mice showed significantly increased REM, and IS mice showed significantly decreased REM after ST1, ST2, and context alone. Total NREM was decreased after shock training only in IS mice. Contextual freezing was enhanced in both ES and IS mice. CONCLUSIONS The directionally opposite changes in REM suggest that stressor controllability is an important factor in the effects of stress and stressful memories on sleep.
Collapse
Affiliation(s)
- Larry D Sanford
- Sleep Research Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA 23501, USA.
| | | | | | | | | |
Collapse
|