1
|
Smiley CE, Pate BS, Bouknight SJ, Francis MJ, Nowicki AV, Harrington EN, Wood SK. Estrogen receptor beta in the central amygdala regulates the deleterious behavioral and neuronal consequences of repeated social stress in female rats. Neurobiol Stress 2023; 23:100531. [PMID: 36879670 PMCID: PMC9984877 DOI: 10.1016/j.ynstr.2023.100531] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 02/02/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
While over 95% of the population has reported experiencing extreme stress or trauma, females of reproductive age develop stress-induced neuropsychiatric disorders at twice the rate of males. This suggests that ovarian hormones may facilitate neural processes that increase stress susceptibility and underlie the heightened rates of these disorders, like depression and anxiety, that result from stress exposure in females. However, there is contradicting evidence in the literature regarding estrogen's role in stress-related behavioral outcomes. Estrogen signaling through estrogen receptor beta (ERβ) has been traditionally thought of as anxiolytic, but recent studies suggest estrogen exhibits distinct effects in the context of stress. Furthermore, ERβ is found abundantly in many stress-sensitive brain loci, including the central amygdala (CeA), in which transcription of the vital stress hormone, corticotropin releasing factor (CRF), can be regulated by an estrogen response element. Therefore, these experiments sought to identify the role of CeA ERβ activity during stress on behavioral outcomes in naturally cycling, adult, female Sprague-Dawley rats. Rats were exposed to an ethological model of vicarious social stress, witness stress (WS), in which they experienced the sensory and psychological aspects of an aggressive social defeat encounter between two males. Following WS, rats exhibited stress-induced anxiety-like behaviors in the marble burying taskand brain analysis revealed increased ERβ and CRF specifically within the CeA following exposure to stress cues. Subsequent experiments were designed to target this receptor in the CeA using microinjections of the ERβ antagonist, PHTPP, prior to each stress session. During WS, estrogen signaling through ERβ was responsible for the behavioral sensitization to repeated social stress. Sucrose preference, acoustic startle, and marble burying tasks determined that blocking ERβ in the CeA during WS prevented the development of depressive-, anxiety-like, and hypervigilant behaviors. Additionally, brain analysis revealed a long-term decrease of intra-CeA CRF expression in PHTPP-treated rats. These experiments indicate that ERβ signaling in the CeA, likely through its effects on CRF, contributes to the development of negative valence behaviors that result from exposure to repeated social stress in female rats.
Collapse
Key Words
- ACTH, adrenocorticotropic hormone
- ASR, acoustic startle responding
- Anxiety
- BCA, bicinchoninic acid
- CON, control handing
- CORT, corticosterone
- CRF, corticotropin releasing factor
- CeA, central amygdala
- Central amygdala
- Corticotropin releasing factor
- EPM, elevated plus maze
- ERβ, estrogen receptor beta
- Estrogen receptor beta
- HPA, hypothalamic pituitary adrenal axis
- LC, locus coeruleus
- MB, marble burying
- PHTPP, 4-[2-Phenyl-5: 7-bis (trifluoromethyl) pyrazolo [1,5-a] pyrimidine-3- yl] phenol
- SPT, sucrose preference testing
- Social stress
- WS, witness stress
- dB, decibels
Collapse
Affiliation(s)
- Cora E. Smiley
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
- Dorn VA Medical Center, Columbia, SC, USA
| | - Brittany S. Pate
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
- University of South Carolina, Arnold School of Public Health, Department of Exercise Science, Columbia, SC, USA
| | - Samantha J. Bouknight
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
| | - Megan J. Francis
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
| | - Alexandria V. Nowicki
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
| | - Evelynn N. Harrington
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
| | - Susan K. Wood
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
- Dorn VA Medical Center, Columbia, SC, USA
| |
Collapse
|
2
|
Characterization of CRH-Binding Protein (CRHBP) in Chickens: Molecular Cloning, Tissue Distribution and Investigation of Its Role as a Negative Feedback Regulator within the Hypothalamus–Pituitary–Adrenal Axis. Genes (Basel) 2022; 13:genes13101680. [PMID: 36292565 PMCID: PMC9601729 DOI: 10.3390/genes13101680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 01/08/2023] Open
Abstract
Corticotropin (ACTH) is a pituitary hormone playing important roles in stress response within the hypothalamus–pituitary–adrenal (HPA) axis. The biosynthesis and secretion of ACTH are controlled by multiple factors, including corticotropin-releasing hormone (CRH). As a key hypothalamus-derived regulator, CRH binds to corticotropin-releasing hormone receptor 1 (CRHR1) in the anterior pituitary gland to regulate ACTH synthesis and release. Thus, CRH-binding protein (CRHBP), which binds CRH with high affinity to inhibit CRH-induced ACTH secretion from pituitary cells, draws wide attention. In contrast to the extensive investigation of CRHBP in mammals and other lower vertebrates, the gene structure, tissue expression and physiological functions of CRHBP in birds remain largely unknown. In the present study, using chicken (c-) as our animal model, we examined the gene structure, tissue expression and functionality of CRHBP. Our results showed that: (1) cCRHBP cDNA encodes a 345 amino acid precursor, which shares high sequence identity with that of mammals, reptiles, frogs and fish; (2) cCRHBP is abundantly expressed in the brain (cerebrum and hypothalamus), pituitary and ovary; (3) cCRHBP inhibits the signaling of cCRHRs induced by cCRH, thus reducing the cCRH-induced ACTH secretion from cultured chick pituitary cells; (4) stress mediators (e.g., glucocorticoids) and stress significantly upregulate CRHBP mRNA expression in chickens, supporting its role as a negative feedback regulator in the HPA axis. The present study enriches our understanding of the conserved roles of CRHBP across vertebrates. In addition, chicken is an important poultry animal with multiple economic traits which are tightly controlled by the HPA axis. The characterization of the chicken CRHBP gene helps to reveal the molecular basis of the chicken HPA axis and is thus beneficial to the poultry industry.
Collapse
|
3
|
Curley DE, Webb AE, Sheffler DJ, Haass-Koffler CL. Corticotropin Releasing Factor Binding Protein as a Novel Target to Restore Brain Homeostasis: Lessons Learned From Alcohol Use Disorder Research. Front Behav Neurosci 2021; 15:786855. [PMID: 34912198 PMCID: PMC8667027 DOI: 10.3389/fnbeh.2021.786855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Stress is well-known to contribute to the development of many psychiatric illnesses including alcohol and substance use disorder (AUD and SUD). The deleterious effects of stress have also been implicated in the acceleration of biological age, and age-related neurodegenerative disease. The physio-pathology of stress is regulated by the corticotropin-releasing factor (CRF) system, the upstream component of the hypothalamic-pituitary-adrenal (HPA) axis. Extensive literature has shown that dysregulation of the CRF neuroendocrine system contributes to escalation of alcohol consumption and, similarly, chronic alcohol consumption contributes to disruption of the stress system. The CRF system also represents the central switchboard for regulating homeostasis, and more recent studies have found that stress and aberrations in the CRF pathway are implicated in accelerated aging and age-related neurodegenerative disease. Corticotropin releasing factor binding protein (CRFBP) is a secreted glycoprotein distributed in peripheral tissues and in specific brain regions. It neutralizes the effects of CRF by sequestering free CRF, but may also possess excitatory function by interacting with CRF receptors. CRFBP’s dual role in influencing CRF bioavailability and CRF receptor signaling has been shown to have a major part in the HPA axis response. Therefore, CRFBP may represent a valuable target to treat stress-related illness, including: development of novel medications to treat AUD and restore homeostasis in the aging brain. This narrative review focuses on molecular mechanisms related to the role of CRFBP in the progression of addictive and psychiatric disorders, biological aging, and age-related neurodegenerative disease. We provide an overview of recent studies investigating modulation of this pathway as a potential therapeutic target for AUD and age-related neurodegenerative disease.
Collapse
Affiliation(s)
- Dallece E Curley
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI, United States.,Neuroscience Graduate Program, Department of Neuroscience, Brown University, Providence, RI, United States
| | - Ashley E Webb
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States.,Carney Institute for Brain Science, Brown University, Providence, RI, United States
| | - Douglas J Sheffler
- Cell and Molecular Biology of Cancer Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States.,Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Carolina L Haass-Koffler
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI, United States.,Carney Institute for Brain Science, Brown University, Providence, RI, United States.,Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, RI, United States.,Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI, United States
| |
Collapse
|
4
|
Cordner ZA, Marshall-Thomas I, Boersma GJ, Lee RS, Potash JB, Tamashiro KL. Fluoxetine and environmental enrichment similarly reverse chronic social stress-related depression- and anxiety-like behavior, but have differential effects on amygdala gene expression. Neurobiol Stress 2021; 15:100392. [PMID: 34568521 PMCID: PMC8449130 DOI: 10.1016/j.ynstr.2021.100392] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/20/2021] [Accepted: 09/08/2021] [Indexed: 01/26/2023] Open
Abstract
The adverse effects of stress on brain and behavior have long been known and well-studied, with abundant evidence linking stress to, among other things, mood and anxiety disorders. Likewise, many have investigated potential treatments for stress-related mood and anxiety phenotypes and demonstrated good response to standard antidepressant medications like selective serotonin reuptake inhibitors (SSRIs), as well as environmental manipulations like exercise or enrichment. However, the extent to which stress and various treatments act on overlapping pathways in the brain is less well understood. Here, we used a widely studied social defeat stress paradigm to induce a robust depression- and anxiety-like phenotype and chronic corticosterone elevation that persisted for at least 4 weeks in wild type male mice. When mice were treated with either the SSRI fluoxetine or an enriched environment, both led to similar behavioral recovery from social defeat. We then focused on the amygdala and assessed the effects of social defeat, fluoxetine, and enrichment on 168 genes broadly related to synaptic plasticity or oxidative stress. We found 24 differentially expressed genes in response to social defeat stress. Interestingly, fluoxetine led to broad normalization of the stress-induced expression pattern while enrichment led to expression changes in a separate set of genes. Together, this study provides additional insight into the chronic effects of social defeat stress on behavior and gene expression in the amygdala. The findings also suggest that, for a subset of genes assessed, fluoxetine and environmental enrichment have strikingly divergent effects on expression in the amygdala, despite leading to similar behavioral outcomes.
Collapse
Affiliation(s)
- Zachary A. Cordner
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD, 21205, USA
| | - Isaiah Marshall-Thomas
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD, 21205, USA
| | - Gretha J. Boersma
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD, 21205, USA
| | - Richard S. Lee
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD, 21205, USA
| | - James B. Potash
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD, 21205, USA
| | - Kellie L.K. Tamashiro
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD, 21205, USA
- Cellular & Molecular Medicine Program, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD, 21205, USA
- Corresponding author. Department of Psychiatry & Behavioral Sciences Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD, 21205, USA.
| |
Collapse
|
5
|
Sbarski B, Akirav I. Cannabinoids as therapeutics for PTSD. Pharmacol Ther 2020; 211:107551. [PMID: 32311373 DOI: 10.1016/j.pharmthera.2020.107551] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 03/08/2020] [Indexed: 02/09/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a complex disorder that involves dysregulation of multiple neurobiological systems. The traumatic stressor plays a causal role in producing psychological dysfunction and the pattern of findings suggests that the hypothalamic-pituitary-adrenal (HPA) axis, which is instrumental for stress adaptation, is critically dysfunctional in PTSD. Given the lack of understanding of the basic mechanisms and underlying pathways that cause the disorder and its heterogeneity, PTSD poses challenges for treatment. Targeting the endocannabinoid (ECB) system to treat mental disorders, and PTSD in particular, has been the focus of research and interest in recent years. The ECB system modulates multiple functions, and drugs enhancing ECB signaling have shown promise as potential therapeutic agents in stress effects and other psychiatric and medical conditions. In this review, we focus on the interaction between the ECB-HPA systems in animal models for PTSD and in patients with PTSD. We summarize evidence supporting the use of cannabinoids in preventing and treating PTSD in preclinical and clinical studies. As the HPA system plays a key role in the mediation of the stress response and the pathophysiology of PTSD, we describe preclinical studies suggesting that enhancing ECB signaling is consistent with decreasing PTSD symptoms and dysfunction of the HPA axis. Overall, we suggest that a pharmacological treatment targeted at one system (e.g., HPA) may not be very effective because of the heterogeneity of the disorder. There are abnormalities across different neurotransmitter systems in the pathophysiology of PTSD and none of these systems function uniformly among all patients with PTSD. Hence, conceptually, enhancing ECB signaling may be a more effective avenue for pharmacological treatment.
Collapse
Affiliation(s)
- Brenda Sbarski
- School of Psychological Sciences, Integrated Brain and Behavior Research Center, University of Haifa, Haifa 3498838, Israel
| | - Irit Akirav
- School of Psychological Sciences, Integrated Brain and Behavior Research Center, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
6
|
Vandael D, Gounko NV. Corticotropin releasing factor-binding protein (CRF-BP) as a potential new therapeutic target in Alzheimer's disease and stress disorders. Transl Psychiatry 2019; 9:272. [PMID: 31641098 PMCID: PMC6805916 DOI: 10.1038/s41398-019-0581-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 07/30/2019] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease is the most common cause of dementia and one of the most complex human neurodegenerative diseases. Numerous studies have demonstrated a critical role of the environment in the pathogenesis and pathophysiology of the disease, where daily life stress plays an important role. A lot of epigenetic studies have led to the conclusion that chronic stress and stress-related disorders play an important part in the onset of neurodegenerative disorders, and an enormous amount of research yielded valuable discoveries but has so far not led to the development of effective treatment strategies for Alzheimer's disease. Corticotropin-releasing factor (CRF) is one of the major hormones and at the same time a neuropeptide acting in stress response. Deregulation of protein levels of CRF is involved in the pathogenesis of Alzheimer's disease, but little is known about the precise roles of CRF and its binding protein, CRF-BP, in neurodegenerative diseases. In this review, we summarize the key evidence for and against the involvement of stress-associated modulation of the CRF system in the pathogenesis of Alzheimer's disease and discuss how recent findings could lead to new potential treatment possibilities in Alzheimer's disease by using CRF-BP as a therapeutic target.
Collapse
Affiliation(s)
- Dorien Vandael
- VIB-KU Leuven Center for Brain and Disease Research, Electron Microscopy Platform, Herestraat 49, B-3000 Leuven, Belgium ,VIB Bioimaging Core Facility, Herestraat 49, B-3000 Leuven, Belgium ,KU Leuven Department of Neurosciences, Leuven Brain Institute, Herestraat 49, B-3000 Leuven, Belgium
| | - Natalia V. Gounko
- VIB-KU Leuven Center for Brain and Disease Research, Electron Microscopy Platform, Herestraat 49, B-3000 Leuven, Belgium ,VIB Bioimaging Core Facility, Herestraat 49, B-3000 Leuven, Belgium ,KU Leuven Department of Neurosciences, Leuven Brain Institute, Herestraat 49, B-3000 Leuven, Belgium
| |
Collapse
|
7
|
Pandey GN, Rizavi HS, Bhaumik R, Ren X. Increased protein and mRNA expression of corticotropin-releasing factor (CRF), decreased CRF receptors and CRF binding protein in specific postmortem brain areas of teenage suicide subjects. Psychoneuroendocrinology 2019; 106:233-243. [PMID: 31005044 PMCID: PMC7061258 DOI: 10.1016/j.psyneuen.2019.04.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/07/2019] [Accepted: 04/10/2019] [Indexed: 10/27/2022]
Abstract
Overactivity of hypothalamic-pituitary-adrenal (HPA) axis function has been implicated in depression and suicidal behavior. This is based on the observation of an abnormal dexamethasone (DEX) and DEX-adrenocorticotropic hormone (ACTH) test in patients with depression and suicidal behavior. Recently, some studies have also found abnormalities of glucocorticoid receptors (GR), mineralocorticoid receptors (MR), corticotropin releasing factor (CRF), CRF receptors (CRF-R) and CRF binding protein (CRF-BP) in depressed and suicidal patients. Some investigators have also observed increased levels of CRF in the cerebrospinal fluid (CSF) and altered levels of MR, GR and CRF in the postmortem brain of depressed and suicidal subjects. We have earlier reported decreased protein and mRNA expression of GR and GILZ, a chaperone protein, in the postmortem brain of teenage suicide subjects. We have further studied CRF and its receptors in different areas of the postmortem brain of suicide subjects, i.e., the prefrontal cortex (PFC), hippocampus (HIPPO), subiculum and amygdala (AMY) from teenage suicide subjects. The CRF and its receptors were determined in the PFC (Brodmann area 9), HIPPO, subiculum and different amygdaloid nuclei from 24 normal control subjects and 24 teenage suicide subjects. Protein expression of CRF, its receptors and CRF-BP was determined by immunolabeling using the Western blot technique and mRNA expression was determined by real-time PCR (qPCR) technique. We found that the mRNA levels of CRF were significantly increased in the PFC, in the central amygdaloid nucleus (CeAMY) and in the subiculum. mRNA levels of CRF-R1 and CRF-BP were significantly decreased in the PFC. We did not find any changes in the HIPPO of any of the CRF components we studied. When we compared the protein expression of CRF components we found that CRF was significantly increased and CRF-R1, CRF-R2 and CRF-BP significantly decreased in the PFC. On the other hand, there were no changes in the protein expression of CRF components in the HIPPO. Our results in the postmortem brain suggest that, as found by clinical studies in the CSF, there are significant alterations of CRF and its receptors in the postmortem brain of teenage suicide subjects. These alterations of CRF and its components were region-specific, as changes were not generally observed in the HIPPO.
Collapse
Affiliation(s)
- Ghanshyam N. Pandey
- Corresponding Author: Ghanshyam N. Pandey, Ph.D., University of Illinois at Chicago, 1601 West Taylor Street, Chicago, IL 60612, USA, Phone (312) 413-4540, Fax: (312) 413-4547,
| | | | | | | |
Collapse
|
8
|
Gołyszny M, Obuchowicz E. Are neuropeptides relevant for the mechanism of action of SSRIs? Neuropeptides 2019; 75:1-17. [PMID: 30824124 DOI: 10.1016/j.npep.2019.02.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/08/2019] [Accepted: 02/13/2019] [Indexed: 12/12/2022]
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are drugs of first choice in the therapy of moderate to severe depression and anxiety disorders. Their primary mechanism of action is via influence of the serotonergic (5-HT) system, but a growing amount of data provides evidence for other non-monoaminergic players in SSRI effects. It is assumed that neuropeptides, which play a role as neuromodulators in the CNS, are involved in their mechanism of action. In this review we focus on six neuropeptides: corticotropin-releasing factor - CRF, galanin - GAL, oxytocin - OT, vasopressin - AVP, neuropeptide Y - NPY, and orexins - OXs. First, information about their roles in depression and anxiety disorders are presented. Then, findings describing their interactions with the 5-HT system are summarized. These data provide background for analysis of the results of published preclinical and clinical studies related to SSRI effects on the neuropeptide systems. We also report findings showing how modulation of neuropeptide transmission influences behavioral and neurochemical effects of SSRIs. Finally, future research necessary for enriching our knowledge of SSRI mechanisms of action is proposed. Recognition of new molecular targets for antidepressants will have a significant effect on the development of novel therapeutic strategies for mood-related disorders.
Collapse
Affiliation(s)
- Miłosz Gołyszny
- Department of Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Poniatowskiego 15, 40-055 Katowice, Poland
| | - Ewa Obuchowicz
- Department of Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Poniatowskiego 15, 40-055 Katowice, Poland.
| |
Collapse
|
9
|
Higuchi Y, Soga T, Parhar IS. Regulatory Pathways of Monoamine Oxidase A during Social Stress. Front Neurosci 2017; 11:604. [PMID: 29163009 PMCID: PMC5671571 DOI: 10.3389/fnins.2017.00604] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/16/2017] [Indexed: 12/18/2022] Open
Abstract
Social stress has a high impact on many biological systems in the brain, including serotonergic (5-HT) system-a major drug target in the current treatment for depression. Hyperactivity of hypothalamic-pituitary-adrenal (HPA) axis and monoamine oxidase A (MAO-A) are well-known stress responses, which are involved in the central 5-HT system. Although, many MAO-A inhibitors have been developed and used in the therapeutics of depression, effective management of depression by modulating the activity of MAO-A has not been achieved. Identifying the molecular pathways that regulate the activity of MAO-A in the brain is crucial for developing new drug targets for precise control of MAO-A activity. Over the last few decades, several regulatory pathways of MAO-A consisting of Kruppel like factor 11 (KLF11), Sirtuin1, Ring finger protein in neural stem cells (RINES), and Cell division cycle associated 7-like protein (R1) have been identified, and the influence of social stress on these regulatory factors evaluated. This review explores various aspects of these pathways to expand our understanding of the roles of the HPA axis and MAO-A regulatory pathways during social stress. The first part of this review introduces some components of the HPA axis, explains how stress affects them and how they interact with the 5-HT system in the brain. The second part summarizes the novel regulatory pathways of MAO-A, which have high potential as novel therapeutic targets for depression.
Collapse
Affiliation(s)
- Yuki Higuchi
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Tomoko Soga
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Ishwar S Parhar
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
10
|
Aisenberg N, Serova L, Sabban EL, Akirav I. The effects of enhancing endocannabinoid signaling and blocking corticotrophin releasing factor receptor in the amygdala and hippocampus on the consolidation of a stressful event. Eur Neuropsychopharmacol 2017; 27:913-927. [PMID: 28663121 DOI: 10.1016/j.euroneuro.2017.06.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/20/2017] [Accepted: 06/10/2017] [Indexed: 11/18/2022]
Abstract
Current clinical and pre-clinical data suggest that both cannabinoid agents and blockage of CRF through corticotrophin releasing factor receptor type 1 (CRFr1) may offer therapeutic benefits for post-traumatic stress disorder (PTSD). Here we aim to determine whether they are more effective when combined when microinjected into the basolateral amygdala (BLA) or CA1 area of the hippocampus after exposure to a stressful event in the shock/reminders rat model for PTSD. Injection of the fatty acid amide hydrolase (FAAH) inhibitor URB597 after the shock into either the BLA or CA1 facilitated extinction, and attenuated startle response and anxiety-like behavior. These preventive effects of URB597 were found to be mediated by the CB1 receptor. Intra-BLA and intra-CA1 microinjection of the CRFr1 antagonist, CP-154,526 attenuated startle response. When microinjected into the BLA, CP-154,526 also attenuated freezing behavior during exposure to the first reminder and decreased anxiety-like behavior. The combined treatment of URB597 and CP-154,526 was not more effective than the separate treatments. Finally, mRNA levels of CRF, CRFr1 and CB1r were significantly higher in the BLA of rats exposed to shock and reminders compared to non-shocked rats almost one month after the shock. Taken together, the results show that enhancing endocannabinoid signaling in the amygdala and hippocampus produced a more favorable spectrum of effects than those caused by the CRFr1 antagonist. The findings suggest that FAAH inhibitors may be used as a novel treatment for stress-related anxiety disorders.
Collapse
MESH Headings
- Amidohydrolases/antagonists & inhibitors
- Amidohydrolases/metabolism
- Animals
- Anxiety/drug therapy
- Anxiety/metabolism
- Basolateral Nuclear Complex/drug effects
- Basolateral Nuclear Complex/metabolism
- Benzamides/pharmacology
- CA1 Region, Hippocampal/drug effects
- CA1 Region, Hippocampal/metabolism
- Carbamates/pharmacology
- Disease Models, Animal
- Endocannabinoids/metabolism
- Male
- Memory Consolidation/drug effects
- Memory Consolidation/physiology
- Nootropic Agents/pharmacology
- Pyrimidines/pharmacology
- Pyrroles/pharmacology
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Receptor, Cannabinoid, CB1/metabolism
- Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors
- Receptors, Corticotropin-Releasing Hormone/metabolism
- Reflex, Startle/drug effects
- Reflex, Startle/physiology
- Stress Disorders, Post-Traumatic/drug therapy
- Stress Disorders, Post-Traumatic/metabolism
- Stress, Psychological/drug therapy
- Stress, Psychological/metabolism
Collapse
Affiliation(s)
- Nurit Aisenberg
- Department of Psychology, University of Haifa, Haifa 3498838, Israel
| | - Lidia Serova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA
| | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA
| | - Irit Akirav
- Department of Psychology, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
11
|
Ketchesin KD, Stinnett GS, Seasholtz AF. Corticotropin-releasing hormone-binding protein and stress: from invertebrates to humans. Stress 2017; 20:449-464. [PMID: 28436309 PMCID: PMC7885796 DOI: 10.1080/10253890.2017.1322575] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Corticotropin-releasing hormone (CRH) is a key regulator of the stress response. This peptide controls the hypothalamic-pituitary-adrenal (HPA) axis as well as a variety of behavioral and autonomic stress responses via the two CRH receptors, CRH-R1 and CRH-R2. The CRH system also includes an evolutionarily conserved CRH-binding protein (CRH-BP), a secreted glycoprotein that binds CRH with subnanomolar affinity to modulate CRH receptor activity. In this review, we discuss the current literature on CRH-BP and stress across multiple species, from insects to humans. We describe the regulation of CRH-BP in response to stress, as well as genetic mouse models that have been utilized to elucidate the in vivo role(s) of CRH-BP in modulating the stress response. Finally, the role of CRH-BP in the human stress response is examined, including single nucleotide polymorphisms in the human CRHBP gene that are associated with stress-related affective disorders and addiction. Lay summary The stress response is controlled by corticotropin-releasing hormone (CRH), acting via CRH receptors. However, the CRH system also includes a unique CRH-binding protein (CRH-BP) that binds CRH with an affinity greater than the CRH receptors. In this review, we discuss the role of this highly conserved CRH-BP in regulation of the CRH-mediated stress response from invertebrates to humans.
Collapse
Affiliation(s)
- Kyle D. Ketchesin
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109
| | - Gwen S. Stinnett
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109
| | - Audrey F. Seasholtz
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
12
|
Harris RBS. Repeated restraint stress lowers the threshold for response to third ventricle CRF administration. Horm Behav 2017; 89:64-68. [PMID: 28017597 PMCID: PMC5942218 DOI: 10.1016/j.yhbeh.2016.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/04/2016] [Accepted: 12/12/2016] [Indexed: 11/24/2022]
Abstract
Rats and mice exposed to repeated stress or a single severe stress exhibit a sustained increase in energetic, endocrine, and behavioral response to subsequent novel mild stress. This study tested whether the hyper-responsiveness was due to a lowered threshold of response to corticotropin releasing factor (CRF) or an exaggerated response to a standard dose of CRF. Male Sprague-Dawley rats were subjected to 3h of restraint on each of 3 consecutive days (RRS) or were non-restrained controls. RRS caused a temporary hypophagia but a sustained reduction in body weight. Eight days after the end of restraint, rats received increasing third ventricle doses of CRF (0-3.0μg). The lowest dose of CRF (0.25μg) increased corticosterone release in RRS, but not control rats. Higher doses caused the same stimulation of corticosterone in the two groups of rats. Fifteen days after the end of restraint, rats were food deprived during the light period and received increasing third ventricle doses of CRF at the start of the dark period. The lowest dose of CRF inhibited food intake during the first hour following infusion in RRS, but not control rats. All other doses of CRF inhibited food intake to the same degree in both RRS and control rats. The lowered threshold of response to central CRF is consistent with the chronic hyper-responsiveness to CRF and mild stress in RRS rats during the post-restraint period.
Collapse
Affiliation(s)
- Ruth B S Harris
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
13
|
Klampfl SM, Schramm MM, Stinnett GS, Bayerl DS, Seasholtz AF, Bosch OJ. Brain CRF-binding protein modulates aspects of maternal behavior under stressful conditions and supports a hypo-anxious state in lactating rats. Horm Behav 2016; 84:136-44. [PMID: 27368148 DOI: 10.1016/j.yhbeh.2016.06.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/06/2016] [Accepted: 06/26/2016] [Indexed: 12/28/2022]
Abstract
Reduced corticotropin-releasing factor (CRF) receptor activation in the postpartum period is essential for adequate maternal behavior. One of the factors contributing to this hypo-activity might be the CRF-binding protein (CRF-BP), which likely reduces the availability of free extracellular CRF/urocortin 1. Here, we investigated behavioral effects of acute CRF-BP inhibition using 5μg of CRF(6-33) administered either centrally or locally within different parts of the bed nucleus of the stria terminalis (BNST) in lactating rats. Additionally, we assessed CRF-BP expression in the BNST comparing virgin and lactating rats. Central CRF-BP inhibition increased maternal aggression during maternal defense but did not affect maternal care or anxiety-related behavior. CRF-BP inhibition in the medial-posterior BNST had no effect on maternal care under non-stress conditions but impaired the reinstatement of maternal care following stressor exposure. Furthermore, maternal aggression, particularly threat behavior, and anxiety-related behavior were elevated by CRF-BP inhibition in the medial-posterior BNST. In the anterior-dorsal BNST, CRF-BP inhibition increased only non-maternal behaviors following stress. Finally, CRF-BP expression was higher in the anterior compared to the posterior BNST but was not different between virgin and lactating rats in either region. Our study demonstrates a key role of the CRF-BP, particularly within the BNST, in modulating CRF's impact on maternal behavior. The CRF-BP is important for the reinstatement of maternal care after stress, for modulating threat behavior during an aggressive encounter and for maintaining a hypo-anxious state during lactation. Thus, the CRF-BP likely contributes to the postpartum-associated down-regulation of the CRF system in a brain region-dependent manner.
Collapse
Affiliation(s)
- Stefanie M Klampfl
- Department of Behavioural and Molecular Neurobiology, Institute of Zoology, University of Regensburg, Universitätsstr. 31, 93053 Regensburg, Germany.
| | - Milena M Schramm
- Department of Behavioural and Molecular Neurobiology, Institute of Zoology, University of Regensburg, Universitätsstr. 31, 93053 Regensburg, Germany.
| | - Gwen S Stinnett
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109-2200, USA.
| | - Doris S Bayerl
- Department of Behavioural and Molecular Neurobiology, Institute of Zoology, University of Regensburg, Universitätsstr. 31, 93053 Regensburg, Germany.
| | - Audrey F Seasholtz
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109-2200, USA; Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109-2200, USA.
| | - Oliver J Bosch
- Department of Behavioural and Molecular Neurobiology, Institute of Zoology, University of Regensburg, Universitätsstr. 31, 93053 Regensburg, Germany.
| |
Collapse
|
14
|
Reyes BAS, Kravets JL, Connelly KL, Unterwald EM, Van Bockstaele EJ. Localization of the delta opioid receptor and corticotropin-releasing factor in the amygdalar complex: role in anxiety. Brain Struct Funct 2016; 222:1007-1026. [PMID: 27376372 DOI: 10.1007/s00429-016-1261-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/22/2016] [Indexed: 12/24/2022]
Abstract
It is well established that central nervous system norepinephrine (NE) and corticotropin-releasing factor (CRF) systems are important mediators of behavioral responses to stressors. More recent studies have defined a role for delta opioid receptors (DOPR) in maintaining emotional valence including anxiety. The amygdala plays an important role in processing emotional stimuli, and has been implicated in the development of anxiety disorders. Activation of DOPR or inhibition of CRF in the amygdala reduces baseline and stress-induced anxiety-like responses. It is not known whether CRF- and DOPR-containing amygdalar neurons interact or whether they are regulated by NE afferents. Therefore, this study sought to better define interactions between the CRF, DOPR and NE systems in the basolateral (BLA) and central nucleus of the amygdala (CeA) of the male rat using anatomical and functional approaches. Irrespective of the amygdalar subregion, dual immunofluorescence microscopy showed that DOPR was present in CRF-containing neurons. Immunoelectron microscopy confirmed that DOPR was localized to both dendritic processes and axon terminals in the BLA and CeA. Semi-quantitative dual immunoelectron microscopy analysis of gold-silver labeling for DOPR and immunoperoxidase labeling for CRF revealed that 55 % of the CRF neurons analyzed contained DOPR in the BLA while 67 % of the CRF neurons analyzed contained DOPR in the CeA. Furthermore, approximately 41 % of DOPR-labeled axon terminals targeted BLA neurons that expressed CRF while 29 % of DOPR-labeled axon terminals targeted CeA neurons that expressed CRF. Triple label immunofluorescence microscopy revealed that DOPR and CRF were co-localized in common cellular profiles that were in close proximity to NE-containing fibers in both subregions. These anatomical results indicate significant interactions between DOPR and CRF in this critical limbic region and reveal that NE is poised to regulate these peptidergic systems in the amygdala. Functional studies were performed to determine if activation of DOPR could inhibit the anxiety produced by elevation of NE in the amygdala using the pharmacological stressor yohimbine. Administration of the DOPR agonist, SNC80, significantly attenuated elevated anxiogenic behaviors produced by yohimbine as measured in the rat on the elevated zero maze. Taken together, results from this study demonstrate the convergence of three important systems, NE, CRF, and DOPR, in the amygdala and provide insight into their functional role in modulating stress and anxiety responses.
Collapse
Affiliation(s)
- Beverly A S Reyes
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, 245 S. 15th Street, Philadelphia, PA, 19102, USA.
| | - J L Kravets
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, 245 S. 15th Street, Philadelphia, PA, 19102, USA
| | - K L Connelly
- Center for Substance Abuse Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - E M Unterwald
- Center for Substance Abuse Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - E J Van Bockstaele
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, 245 S. 15th Street, Philadelphia, PA, 19102, USA
| |
Collapse
|
15
|
Rahmani B, Ghasemi R, Dargahi L, Ahmadiani A, Haeri A. Neurosteroids; potential underpinning roles in maintaining homeostasis. Gen Comp Endocrinol 2016; 225:242-250. [PMID: 26432100 DOI: 10.1016/j.ygcen.2015.09.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 09/14/2015] [Accepted: 09/28/2015] [Indexed: 12/31/2022]
Abstract
The neuroactive steroids which are synthesized in the brain and nervous system are known as "Neurosteroids". These steroids have crucial functions such as contributing to the myelination and organization of the brain connectivity. Under the stressful circumstances, the concentrations of neurosteroid products such as allopregnanolone (ALLO) and allotetrahydrodeoxycorticosterone (THDOC) alter. It has been suggested that these stress-derived neurosteroids modulate the physiological response to stress. Moreover, it has been demonstrated that the hypothalamic-pituitary-adrenal (HPA) axis mediates the physiological adaptation following stress in order to maintain homeostasis. Although several regulatory pathways have been introduced, the exact role of neurosteroids in controlling HPA axis is not clear to date. In this review, we intend to discern specific pathways associated with regulation of HPA axis in which neuroactive steroids have the main role. In this respect, we propose pathways that may be initiated after neurosteroidogenesis in different brain subregions following acute stress which are potentially capable of activating or inhibiting the HPA axis.
Collapse
Affiliation(s)
- Behrouz Rahmani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Department of Physiology and Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ali Haeri
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
5-HT1A receptor blockade targeting the basolateral amygdala improved stress-induced impairment of memory consolidation and retrieval in rats. Neuroscience 2015; 300:609-18. [DOI: 10.1016/j.neuroscience.2015.05.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 05/08/2015] [Accepted: 05/12/2015] [Indexed: 01/20/2023]
|
17
|
Eraslan E, Akyazi I, Erg L-Ekiz E, Matur E. Noise stress changes mRNA expressions of corticotropin-releasing hormone, its receptors in amygdala, and anxiety-related behaviors. Noise Health 2015; 17:141-7. [PMID: 25913553 PMCID: PMC4918649 DOI: 10.4103/1463-1741.155838] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Noise is a psychological, environmental stressor that activates limbic sites in the brain. Limbic sites such as the amygdala and the amygdaloid corticotropin-releasing hormone (CRH) system play an important role in integrating stress response. We investigated the association between noise exposures, CRH-related molecules in the amygdala, and behavioral alterations. In total 54 Sprague-Dawley rats were divided into the following three groups: Control (CON), acute noise exposure (ANE), and chronic noise exposure (CNE). The ANE group was exposed to 100 dB white noise only once in 4 h and the CNE group was exposed to the same for 4 h per day for 30 days. Expression profiles of CRH and its receptors CRH-R1 and CRH-R2 were analyzed by quantitative real-time polymerase chain reaction (qPCR). The same stress procedure was applied to the ANE and CNE groups for behavior testing. The anxiety responses of the animals after acute and chronic stress exposure were measured in the defensive withdrawal test. CNE upregulated CRH and CRH-R1 mRNA levels but downregulated CRH-R2 mRNA levels. ANE led to a decrease in both CRH-R1 and CRH-R2 expression. In the defensive withdrawal test, while the ANE increased, CNE reduced anxiety-like behaviors. The present study shows that the exposure of rats to white noise (100 dB) leads to behavioral alterations and molecule-specific changes in the CRH system. Behavioral alterations can be related to these molecular changes in the amygdala.
Collapse
Affiliation(s)
- Evren Eraslan
- Department of Physiology, Faculty of Veterinary Medicine, Istanbul University, Istanbul, Turkey
| | | | | | | |
Collapse
|
18
|
Stinnett GS, Westphal NJ, Seasholtz AF. Pituitary CRH-binding protein and stress in female mice. Physiol Behav 2015; 150:16-23. [PMID: 25731977 DOI: 10.1016/j.physbeh.2015.02.050] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 02/05/2015] [Accepted: 02/23/2015] [Indexed: 11/30/2022]
Abstract
The CRH-binding protein (CRH-BP) binds CRH with very high affinity and inhibits CRH-mediated ACTH release from anterior pituitary cells in vitro, suggesting that the CRH-BP functions as a negative regulator of CRH activity. Our previous studies have demonstrated sexually dimorphic expression of CRH-BP in the murine pituitary. Basal CRH-BP expression is higher in the female pituitary, where CRH-BP mRNA is detected in multiple anterior pituitary cell types. In this study, we examined stress-induced changes in CRH-BP mRNA and protein expression in mouse pituitary and assessed the in vivo role of CRH-BP in modulating the stress response. Pituitary CRH-BP mRNA was greater than 200-fold more abundant in females than males, and restraint stress increased pituitary CRH-BP mRNA by 11.8-fold in females and 3.2-fold in males as assessed by qRT-PCR. In females, restraint stress increased CRH-BP mRNA levels not only in POMC-expressing cells, but also in PRL-expressing cells. The increase in female pituitary CRH-BP mRNA following stress resulted in significant increases in CRH-BP protein 4-6h after a 30-minute restraint stress as detected by [(125)I]-CRH:CRH-BP cross-linking analyses. Based on this temporal profile, the physiological role of CRH-BP was assessed using a stressor of longer duration. In lipopolysaccharide (LPS) stress studies, female CRH-BP-deficient mice showed elevated levels of stress-induced corticosterone release as compared to wild-type littermates. These studies demonstrate a role for the pituitary CRH-BP in attenuating the HPA response to stress in female mice.
Collapse
Affiliation(s)
- Gwen S Stinnett
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Nicole J Westphal
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, United States
| | - Audrey F Seasholtz
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, United States; Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
19
|
Aubry JM. CRF system and mood disorders. J Chem Neuroanat 2013; 54:20-4. [DOI: 10.1016/j.jchemneu.2013.09.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 09/13/2013] [Indexed: 10/26/2022]
|
20
|
Bangasser DA. Sex differences in stress-related receptors: ″micro″ differences with ″macro″ implications for mood and anxiety disorders. Biol Sex Differ 2013; 4:2. [PMID: 23336736 PMCID: PMC3556142 DOI: 10.1186/2042-6410-4-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 12/27/2012] [Indexed: 11/10/2022] Open
Abstract
Stress-related psychiatric disorders, such as unipolar depression and post-traumatic stress disorder (PTSD), occur more frequently in women than in men. Emerging research suggests that sex differences in receptors for the stress hormones, corticotropin releasing factor (CRF) and glucocorticoids, contribute to this disparity. For example, sex differences in CRF receptor binding in the amygdala of rats may predispose females to greater anxiety following stressful events. Additionally, sex differences in CRF receptor signaling and trafficking in the locus coeruleus arousal center combine to make females more sensitive to low levels of CRF, and less adaptable to high levels. These receptor differences in females could lead to hyperarousal, a dysregulated state associated with symptoms of depression and PTSD. Similar to the sex differences observed in CRF receptors, sex differences in glucocorticoid receptor (GR) function also appear to make females more susceptible to dysregulation after a stressful event. Following hypothalamic pituitary adrenal axis activation, GRs are critical to the negative feedback process that inhibits additional glucocorticoid release. Compared to males, female rats have fewer GRs and impaired GR translocation following chronic adolescent stress, effects linked to slower glucocorticoid negative feedback. Thus, under conditions of chronic stress, attenuated negative feedback in females would result in hypercortisolemia, an endocrine state thought to cause depression. Together, these studies suggest that sex differences in stress-related receptors shift females more easily into a dysregulated state of stress reactivity, linked to the development of mood and anxiety disorders. The implications of these receptor sex differences for the development of novel pharmacotherapies are also discussed.
Collapse
Affiliation(s)
- Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, 1701 North 13th Street, 873 Weiss Hall, Philadelphia, 19122, PA.
| |
Collapse
|
21
|
Haass-Koffler CL, Bartlett SE. Stress and addiction: contribution of the corticotropin releasing factor (CRF) system in neuroplasticity. Front Mol Neurosci 2012; 5:91. [PMID: 22973190 PMCID: PMC3434418 DOI: 10.3389/fnmol.2012.00091] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 08/15/2012] [Indexed: 12/22/2022] Open
Abstract
Corticotropin releasing factor (CRF) has been shown to induce various behavioral changes related to adaptation to stress. Dysregulation of the CRF system at any point can lead to a variety of psychiatric disorders, including substance use disorders (SUDs). CRF has been associated with stress-induced drug reinforcement. Extensive literature has identified CRF to play an important role in the molecular mechanisms that lead to an increase in susceptibility that precipitates relapse to SUDs. The CRF system has a heterogeneous role in SUDs. It enhances the acute effects of drugs of abuse and is also responsible for the potentiation of drug-induced neuroplasticity evoked during the withdrawal period. We present in this review the brain regions and circuitries where CRF is expressed and may participate in stress-induced drug abuse. Finally, we attempt to evaluate the role of modulating the CRF system as a possible therapeutic strategy for treating the dysregulation of emotional behaviors that result from the acute positive reinforcement of substances of abuse as well as the negative reinforcement produced by withdrawal.
Collapse
Affiliation(s)
- Carolina L Haass-Koffler
- Ernest Gallo Clinic and Research Center at the University of California San Francisco Emeryville, CA, USA
| | | |
Collapse
|
22
|
Quantification of extracellular levels of corticosterone in the basolateral amygdaloid complex of freely-moving rats: A dialysis study of circadian variation and stress-induced modulation. Brain Res 2012; 1452:47-60. [DOI: 10.1016/j.brainres.2012.01.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 11/16/2011] [Accepted: 01/07/2012] [Indexed: 12/31/2022]
|
23
|
Differential gene expression between inbred Roman high- (RHA-I) and low- (RLA-I) avoidance rats. Neurosci Lett 2011; 504:265-70. [DOI: 10.1016/j.neulet.2011.09.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 09/10/2011] [Accepted: 09/19/2011] [Indexed: 11/18/2022]
|
24
|
Knoll AT, Muschamp JW, Sillivan SE, Ferguson D, Dietz DM, Meloni EG, Carroll FI, Nestler EJ, Konradi C, Carlezon WA. Kappa opioid receptor signaling in the basolateral amygdala regulates conditioned fear and anxiety in rats. Biol Psychiatry 2011; 70:425-33. [PMID: 21531393 PMCID: PMC3150294 DOI: 10.1016/j.biopsych.2011.03.017] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 03/07/2011] [Accepted: 03/10/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND The kappa opioid receptor (KOR) system contributes to the prodepressive and aversive consequences of stress and is implicated in the facilitation of conditioned fear and anxiety in rodents. Here, we sought to identify neural circuits that mediate KOR system effects on fear and anxiety in rats. METHODS We assessed whether fear conditioning induces plasticity in KOR or dynorphin (the endogenous KOR ligand) messenger RNA (mRNA) expression in the basolateral (BLA) and central (CeA) nuclei of the amygdala, hippocampus, or striatum. We then assessed whether microinfusions of the KOR antagonist JDTic (0-10 μg/side) into the BLA or CeA affect the expression of conditioned fear or anxiety. Finally, we examined whether fear extinction induces plasticity in KOR mRNA expression that relates to the quality of fear extinction. RESULTS Fear conditioning upregulated KOR mRNA in the BLA by 65% and downregulated it in the striatum by 22%, without affecting KOR levels in the CeA or hippocampus, or dynorphin levels in any region. KOR antagonism in either the BLA or CeA decreased conditioned fear in the fear-potentiated startle paradigm, whereas KOR antagonism in the BLA, but not the CeA, produced anxiolytic-like effects in the elevated plus maze. Effective fear extinction was associated with a 67% reduction in KOR mRNA in the BLA. CONCLUSIONS These findings suggest that fear conditioning and extinction dynamically regulate KOR expression in the BLA and provide evidence that the BLA and CeA are important neural substrates mediating the anxiolytic-like effects of KOR antagonists in models of fear and anxiety.
Collapse
MESH Headings
- Amygdala/drug effects
- Amygdala/metabolism
- Animals
- Anxiety/metabolism
- Conditioning, Psychological/drug effects
- Conditioning, Psychological/physiology
- Corpus Striatum/drug effects
- Corpus Striatum/metabolism
- Disease Models, Animal
- Dynorphins/physiology
- Extinction, Psychological/physiology
- Fear/drug effects
- Fear/physiology
- Gene Expression Regulation/physiology
- Hippocampus/drug effects
- Hippocampus/metabolism
- Male
- Maze Learning/drug effects
- Maze Learning/physiology
- Microinjections
- Piperidines/administration & dosage
- Piperidines/pharmacology
- Rats
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, kappa/biosynthesis
- Receptors, Opioid, kappa/physiology
- Reflex, Startle/drug effects
- Reflex, Startle/physiology
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Tetrahydroisoquinolines/administration & dosage
- Tetrahydroisoquinolines/pharmacology
Collapse
Affiliation(s)
- Allison T. Knoll
- Behavioral Genetics Laboratory, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478 (ATK, JWM, EGM, WAC)
| | - John W. Muschamp
- Behavioral Genetics Laboratory, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478 (ATK, JWM, EGM, WAC)
| | | | - Deveroux Ferguson
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, NY, 10029 (DF, DMD, EJN)
| | - David M. Dietz
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, NY, 10029 (DF, DMD, EJN)
| | - Edward G. Meloni
- Behavioral Genetics Laboratory, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478 (ATK, JWM, EGM, WAC)
| | - F. Ivy Carroll
- Research Triangle Institute, Organic and Medicinal Chemistry, Research Triangle Park, NC 27709 (FIC)
| | - Eric J. Nestler
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, NY, 10029 (DF, DMD, EJN)
| | - Christine Konradi
- Department of Pharmacology and Psychiatry, Vanderbilt University, Nashville, TN 37232 (CK)
| | - William A. Carlezon
- Behavioral Genetics Laboratory, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478 (ATK, JWM, EGM, WAC)
| |
Collapse
|
25
|
Skórzewska A, Lehner M, Hamed A, Wisłowska-Stanek A, Turzyńska D, Sobolewska A, Płaźnik A. The effect of CRF2 receptor antagonists on rat conditioned fear responses and c-Fos and CRF expression in the brain limbic structures. Behav Brain Res 2011; 221:155-65. [PMID: 21376756 DOI: 10.1016/j.bbr.2011.02.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 02/15/2011] [Accepted: 02/24/2011] [Indexed: 12/11/2022]
Abstract
The influence of intracerebroventricular-administered selective corticotropin-releasing factor receptor 2 (CRF(2)) antagonists (antisauvagine-30, astressin-2B), on rat anxiety-like behavior, expression levels of c-Fos and CRF, and plasma corticosterone levels were examined in the present study. In fear-conditioned animals, both CRF receptor antagonists enhanced a conditioned freezing fear response and increased the conditioned fear-elevated concentration of serum corticosterone. Exogenously administered antisauvagine-30 increased the aversive context-induced expression of c-Fos in the 1 and 2 areas of the cingulate cortex (Cg1, Cg2), the central amygdala (CeA) and parvocellular neurons of the paraventricular hypothalamic nucleus (pPVN), and it enhanced the effect of conditioned fear in the secondary motor cortex (M2) and medial amygdala (MeA). Immunocytochemistry demonstrated an increase in CRF expression in the Cg1, M2 areas of the cortex, and pPVN, and it revealed the effect of conditioned fear in the CeA 35 min after antisauvagine-30 administration and 10 min after the conditioned fear test. Furthermore, astressin-2B, another CRF(2) receptor antagonist, enhanced expression of c-Fos and CRF in the CeA and pPVN, and revealed the effect of conditioned fear in the Cg1. These data support a model in which an excess in CRF(1) receptor activation, combined with reduced CRF(2) receptor signaling, may contribute to stronger expression of anxiety-like responses.
Collapse
Affiliation(s)
- A Skórzewska
- Department of Neurochemistry, Institute of Psychiatry and Neurology, 9 Sobieskiego Street, 02-957 Warsaw, Poland.
| | | | | | | | | | | | | |
Collapse
|
26
|
Qi C, Roseboom PH, Nanda SA, Lane JC, Speers JM, Kalin NH. Anxiety-related behavioral inhibition in rats: a model to examine mechanisms underlying the risk to develop stress-related psychopathology. GENES BRAIN AND BEHAVIOR 2011; 9:974-84. [PMID: 20738409 DOI: 10.1111/j.1601-183x.2010.00636.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Behavioral inhibition (BI) is an adaptive defensive response to threat; however, children who display extreme BI as a stable trait are at risk for development of anxiety disorders and depression. The present study validates a rodent model of BI based on an ethologically relevant predator exposure paradigm. We show that individual differences in rat BI are stable and trait-like from adolescence into adulthood. Using in situ hybridization to quantify expression of the immediate early genes homer1a and fos as measures of neuronal activation, we show that individual differences in BI are correlated with the activation of various stress-responsive brain regions that include the paraventricular nucleus of the hypothalamus and CA3 region of the hippocampus. Further supporting the concept that threat-induced BI in rodents reflects levels of anxiety, we also show that BI is decreased by administration of the anxiolytic, diazepam. Finally, we developed criteria for identifying extreme BI animals that are stable in their expression of high levels of BI and also show that high BI (HBI) individuals exhibit maladaptive appetitive responses following stress exposure. These findings support the use of predator threat as a stimulus and HBI rats as a model to study mechanisms underlying extreme and stable BI in humans.
Collapse
Affiliation(s)
- C Qi
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53719-1176, USA
| | | | | | | | | | | |
Collapse
|
27
|
Ronan PJ, Summers CH. Molecular Signaling and Translational Significance of the Corticotropin Releasing Factor System. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 98:235-92. [DOI: 10.1016/b978-0-12-385506-0.00006-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
28
|
Giardino WJ, Pastor R, Anacker AMJ, Spangler E, Cote DM, Li J, Stenzel-Poore MP, Phillips TJ, Ryabinin AE. Dissection of corticotropin-releasing factor system involvement in locomotor sensitivity to methamphetamine. GENES BRAIN AND BEHAVIOR 2010; 10:78-89. [PMID: 20731720 DOI: 10.1111/j.1601-183x.2010.00641.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Sensitivity to the euphoric and locomotor-activating effects of drugs of abuse may contribute to risk for excessive use and addiction. Repeated administration of psychostimulants such as methamphetamine (MA) can result in neuroadaptive consequences that manifest behaviorally as a progressive escalation of locomotor activation, termed psychomotor sensitization. The present studies addressed the involvement of specific components of the corticotropin-releasing factor (CRF) system in locomotor activation and psychomotor sensitization induced by MA (1, 2 mg/kg) by utilizing pharmacological approaches, as well as a series of genetic knockout (KO) mice, each deficient for a single component of the CRF system: CRF-R1, CRF-R2, CRF, or the CRF-related peptide Urocortin 1 (Ucn1). CRF-R1 KO mice did not differ from wild-type mice in sensitization to MA, and pharmacological blockade of CRF-R1 with CP-154,526 (15, 30 mg/kg) in DBA/2J mice did not selectively attenuate either the acquisition or expression of MA-induced sensitization. Deletion of either of the endogenous ligands of CRF-R1 (CRF, Ucn1) either enhanced or had no effect on MA-induced sensitization, providing further evidence against a role for CRF-R1 signaling. Interestingly, deletion of CRF-R2 attenuated MA-induced locomotor activation, elucidating a novel contribution of the CRF system to MA sensitivity, and suggesting the participation of the endogenous urocortin peptides Ucn2 and Ucn3. Immunohistochemistry for Fos was used to visualize neural activation underlying CRF-R2-dependent sensitivity to MA, identifying the basolateral and central nuclei of the amygdala as neural substrates involved in this response. Our results support further examination of CRF-R2 involvement in neural processes associated with MA addiction.
Collapse
Affiliation(s)
- W J Giardino
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
The effect of CRF and α-helical CRF(9–41) on rat fear responses and amino acids release in the central nucleus of the amygdala. Neuropharmacology 2009; 57:148-56. [DOI: 10.1016/j.neuropharm.2009.04.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 04/01/2009] [Accepted: 04/28/2009] [Indexed: 11/15/2022]
|
30
|
Pitts MW, Todorovic C, Blank T, Takahashi LK. The central nucleus of the amygdala and corticotropin-releasing factor: insights into contextual fear memory. J Neurosci 2009; 29:7379-88. [PMID: 19494159 PMCID: PMC2771694 DOI: 10.1523/jneurosci.0740-09.2009] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Revised: 04/30/2009] [Accepted: 05/02/2009] [Indexed: 11/21/2022] Open
Abstract
The central nucleus of the amygdala (CeA) has been traditionally viewed in fear conditioning to serve as an output neural center that transfers conditioned information formed in the basolateral amygdala to brain structures that generate emotional responses. Recent studies suggest that the CeA may also be involved in fear memory consolidation. In addition, corticotropin-releasing factor systems were shown to facilitate memory consolidation in the amygdala, which contains a high density of CRF immunoreactive cell bodies and fibers in the lateral part of the CeA (CeAl). However, the involvement of CeA CRF in contextual fear conditioning remains poorly understood. Therefore, we first conducted a series of studies using fiber-sparing lesion and reversible inactivation methods to assess the general role of the CeA in contextual fear. We then used identical training and testing procedures to compare and evaluate the specific function of CeA CRF using CRF antisense oligonucleotides (CRF ASO). Rats microinjected with ibotenic acid, muscimol, or a CRF ASO into the CeA before contextual fear conditioning showed typical levels of freezing during acquisition training but exhibited significant reductions in contextual freezing in a retention test 48 h later. Furthermore, CeA inactivation induced by either muscimol or CRF ASO administration immediately before retention testing did not impair freezing, suggesting that the previously observed retention deficits were caused by inhibition of consolidation rather than fear expression. Collectively, our results suggest CeA involvement in the consolidation of contextual fear memory and specifically implicate CeA CRF as an important mediator.
Collapse
Affiliation(s)
| | - Cedomir Todorovic
- Specialized Neuroscience Research Project 2, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96813, and
| | - Thomas Blank
- Specialized Neuroscience Research Project 2, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96813, and
| | - Lorey K. Takahashi
- Department of Cell & Molecular Biology and
- Department of Psychology, University of Hawaii, Honolulu, Hawaii 96822
| |
Collapse
|
31
|
Gender differences in corticotropin and corticosterone secretion and corticotropin-releasing factor mRNA expression in the paraventricular nucleus of the hypothalamus and the central nucleus of the amygdala in response to footshock stress or psychological stress in rats. Psychoneuroendocrinology 2009; 34:226-237. [PMID: 18849120 DOI: 10.1016/j.psyneuen.2008.09.003] [Citation(s) in RCA: 168] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Revised: 07/14/2008] [Accepted: 09/03/2008] [Indexed: 11/20/2022]
Abstract
Anorexia nervosa is mostly seen in adolescent females, although the gender-differentiation mechanism is unclear. Corticotropin-releasing factor (CRF), a key peptide for stress responses such as inhibition of food intake, increases in arousal and locomotor activity, and gonadal dysfunction, is thought to be involved in the pathophysiology of anorexia nervosa. CRF in the paraventricular nucleus of the hypothalamus (PVN) and CRF in the central nucleus of the amygdala (CeA) are involved in the regulation of stress responses, and gender differences in CRF mRNA expression in these regions in response to various stressors are controversial. We therefore examined CRF gene expression in the PVN and CeA as well as corticotropin (ACTH) and corticosterone secretion in response to a 60-min period of electric footshock (FS) or psychological stress (PS) induced by a communication box in both male and female rats in proestrus or diestrus in an effort to elucidate the mechanism underlying the gender difference in the activity of the hypothalamic-pituitary-adrenal (HPA) axis and the mechanism underlying the remarkable prevalence of anorexia nervosa in females. Female rats in proestrus showed higher basal plasma ACTH and CRF mRNA expression levels in the PVN and CeA than males. Females more rapidly showed higher plasma ACTH and corticosterone levels and a higher CRF mRNA expression level in the PVN in response to FS than males. Although females in both proestrus and diestrus showed significant increases in plasma ACTH and corticosterone and CRF mRNA expression in the PVN in response to PS, no significant responses of the HPA axis to PS were found in males. FS significantly increased CRF mRNA expression in the CeA in both females and males, with significantly higher peaks in females in proestrus than in males, while PS significantly increased CRF mRNA expression in the CeA only in males. These results suggest that gender affects differentially the function of the stress-related regions such as the PVN and CeA. The finding that CRF gene expression in the PVN responds to PS only in females may be a clue to elucidation of the neurobiological mechanism underlying the gender-differential prevalence of anorexia nervosa.
Collapse
|
32
|
Gammie SC, Seasholtz AF, Stevenson SA. Deletion of corticotropin-releasing factor binding protein selectively impairs maternal, but not intermale aggression. Neuroscience 2008; 157:502-12. [PMID: 18929624 DOI: 10.1016/j.neuroscience.2008.09.026] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Revised: 09/17/2008] [Accepted: 09/18/2008] [Indexed: 11/25/2022]
Abstract
Corticotropin-releasing factor (CRF) binding protein (CRF-BP) is a secreted protein that acts to bind and limit the activity of the neuropeptides, CRF and urocortin (Ucn) 1. We previously selected for high maternal defense (protection of offspring) in mice and found CRF-BP to be elevated in the CNS of selected mice. We also previously determined that both CRF and Ucn 1 are potent inhibitors of offspring protection when administered centrally. Thus, elevated CRF-BP could promote defense by limiting endogenous actions of CRF or Ucn 1. To test this hypothesis, we crossed the deletion for CRF-BP into the mice selected for high maternal defense and evaluated offspring protection and other maternal behaviors. CRF-BP knockout (KO) mice exhibited significant deficits in maternal aggression relative to wild-type (WT) mice in three different measures. Other maternal features were almost identical between groups, including dam and pup weight, litter size, nursing time, and pup retrieval. Both groups performed similarly in a forced swim stress test and aggression in both groups was reduced following the swim test. Virgin KO female mice exhibited higher levels of anxiety-like behavior in terms of decreased time in the light portion of the light/dark box test. For males, no differences in light/dark box or swim test were found. However, increased anxiety-like behavior in male KO mice was identified in terms of contact and approach to a novel object both with and without previous exposure to the swim test. No differences in isolation induced resident intruder male aggression were found between groups. Together, these results indicate that loss of CRF-BP selectively impairs maternal, but not intermale aggression and that loss of the gene induces anxiety-like behavior in males and females, but there are sex differences in terms of how that anxiety is revealed.
Collapse
Affiliation(s)
- S C Gammie
- Department of Zoology, University of Wisconsin, 1117 West Johnson Street, Madison, WI 53706, USA.
| | | | | |
Collapse
|
33
|
Roozendaal B, Schelling G, McGaugh JL. Corticotropin-releasing factor in the basolateral amygdala enhances memory consolidation via an interaction with the beta-adrenoceptor-cAMP pathway: dependence on glucocorticoid receptor activation. J Neurosci 2008; 28:6642-51. [PMID: 18579737 PMCID: PMC2586417 DOI: 10.1523/jneurosci.1336-08.2008] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2007] [Revised: 04/28/2008] [Accepted: 05/15/2008] [Indexed: 11/21/2022] Open
Abstract
Extensive evidence indicates that stress hormone effects on the consolidation of emotionally influenced memory involve noradrenergic activation of the basolateral complex of the amygdala (BLA). The present experiments examined whether corticotropin-releasing factor (CRF) modulates memory consolidation via an interaction with the beta-adrenoceptor-cAMP system in the BLA. In a first experiment, male Sprague Dawley rats received bilateral infusions of the CRF-binding protein ligand inhibitor CRF(6-33) into the BLA either alone or together with the CRF receptor antagonist alpha-helical CRF(9-41) immediately after inhibitory avoidance training. CRF(6-33) induced dose-dependent enhancement of 48 h retention latencies, which was blocked by coadministration of alpha-helical CRF(9-41), suggesting that CRF(6-33) enhances memory consolidation by displacing CRF from its binding protein, thereby increasing "free" endogenous CRF concentrations. In a second experiment, intra-BLA infusions of atenolol (beta-adrenoceptor antagonist) and Rp-cAMPS (cAMP inhibitor), but not prazosin (alpha(1)-adrenoceptor antagonist), blocked CRF(6-33)-induced retention enhancement. In a third experiment, the CRF receptor antagonist alpha-helical CRF(9-41) administered into the BLA immediately after training attenuated the dose-response effects of concurrent intra-BLA infusions of clenbuterol (beta-adrenoceptor agonist). In contrast, alpha-helical CRF(9-41) did not alter retention enhancement induced by posttraining intra-BLA infusions of either cirazoline (alpha(1)-adrenoceptor agonist) or 8-br-cAMP (cAMP analog). These findings suggest that CRF facilitates the memory-modulatory effects of noradrenergic stimulation in the BLA via an interaction with the beta-adrenoceptor-cAMP cascade, at a locus between the membrane-bound beta-adrenoceptor and the intracellular cAMP formation site. Moreover, consistent with evidence that glucocorticoids enhance memory consolidation via a similar interaction with the beta-adrenoceptor-cAMP cascade, a last experiment found that the CRF and glucocorticoid systems within the BLA interact in influencing beta-adrenoceptor-cAMP effects on memory consolidation.
Collapse
Affiliation(s)
- Benno Roozendaal
- Center for the Neurobiology of Learning and Memory, Department of Neurobiology and Behavior, University of California, Irvine, California 92697-3800, USA.
| | | | | |
Collapse
|
34
|
Nanda SA, Qi C, Roseboom PH, Kalin NH. Predator stress induces behavioral inhibition and amygdala somatostatin receptor 2 gene expression. GENES BRAIN AND BEHAVIOR 2008; 7:639-48. [PMID: 18363859 DOI: 10.1111/j.1601-183x.2008.00401.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Psychological stressors precipitate and maintain stress-induced psychopathology, and it is likely that altered amygdala function underlies some of the deleterious effects of psychological stress. To understand the mechanisms underlying the linkage between the response to psychological stressors and maladaptive or psychopathological responses, we have focused on amygdala responsivity in animal models employing species-specific psychological stressors. In the present study, we characterized the effects of a 15-min exposure to a natural predator, the ferret, on rat behavior and the expression of the somatostatin family of genes in the amygdala. We examined the somatostatin family of genes because substantial evidence shows that central somatostatin systems are altered in various neuropsychiatric illnesses. We report that rats respond to acute ferret exposure with a significant increase in fearful and anxious behaviors that is accompanied by robust amygdala activation and an increase in somatostatin receptor 2 (sst2) messenger RNA expression within the amygdala and anterior cingulate cortex. These studies are the first to show stress-induced changes in amygdala sst2 expression and may represent one mechanism by which psychological stress is linked to adaptive and maladaptive behavioral responses.
Collapse
Affiliation(s)
- S A Nanda
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI 53719, USA.
| | | | | | | |
Collapse
|
35
|
Alderman SL, Raine JC, Bernier NJ. Distribution and regional stressor-induced regulation of corticotrophin-releasing factor binding protein in rainbow trout (Oncorhynchus mykiss). J Neuroendocrinol 2008; 20:347-58. [PMID: 18208552 DOI: 10.1111/j.1365-2826.2008.01655.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The corticotrophin-releasing factor (CRF) system plays a key role in the co-ordination of the physiological response to stress in vertebrates. Although the binding protein (BP) for CRF-related peptides, CRF-BP, is an important player in the many functions of the CRF system, the distribution of CRF-BP and the impact of stressors on its expression in fish are poorly understood. In the present study, we describe the distribution of CRF-BP in the brain and peripheral tissues of rainbow trout (Oncorhynchus mykiss) using a combination of real-time reverse transcriptase-polymerase chain reaction, in situ hybridisation and immunohistochemistry. Our results indicate a widespread and highly localised distribution of CRF-BP in the central nervous system, but do not support a significant peripheral production of the protein. Major expression sites in the brain include the area ventralis telencephali, nucleus preopticus, anterior and lateral tuberal nuclei, and the posterior region of the pituitary pars distalis. We further characterise changes in CRF-BP gene expression in three discrete brain regions after exposure to 8 h and 24 h of social stress or hypoxia. The plasma cortisol concentration in subordinate fish was much higher than in dominant fish and controls, and was indicative of a relatively severe stressor. By contrast, the increase in plasma cortisol concentration in fish exposed to hypoxia was characteristic of the response to a mild stressor. Changes in CRF-BP gene expression were only observed after 24 h of either stressor, and were region-specific. CRF-BP mRNA in the telencephalon increased in both subordinate fish and fish exposed to hypoxia, but CRF-BP in the preoptic area only increased after 24 h of hypoxia exposure. In the hypothalamus, CRF-BP mRNA levels decreased in dominant fish relative to controls after 24 h. Taken together, our results support a diverse role for CRF-BP in the central actions of the fish CRF system, but a negligible role in the peripheral functions of circulating CRF-related peptides. Furthermore, the differential changes in forebrain CRF-BP mRNA appear to occur independently of the hypothalamic-pituitary-inter-renal axis.
Collapse
Affiliation(s)
- S L Alderman
- Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada
| | | | | |
Collapse
|
36
|
Wang B, You ZB, Rice KC, Wise RA. Stress-induced relapse to cocaine seeking: roles for the CRF(2) receptor and CRF-binding protein in the ventral tegmental area of the rat. Psychopharmacology (Berl) 2007; 193:283-94. [PMID: 17437087 DOI: 10.1007/s00213-007-0782-3] [Citation(s) in RCA: 161] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Accepted: 03/21/2007] [Indexed: 11/30/2022]
Abstract
RATIONALE Footshock reinstates cocaine seeking in cocaine-experienced rats by inducing corticotropin-releasing factor (CRF) and glutamate release in the ventral tegmental area (VTA) and thus activating VTA dopaminergic neurons. Footshock-induced VTA glutamate release, dopamine activation and reinstatements are blocked by VTA administration of a alpha-helical CRF, a nonselective CRF receptor antagonist. The effects of selective CRF antagonists have not yet been reported. OBJECTIVE The present studies were designed to explore the roles of VTA CRF receptor subtypes and CRF-BP in these effects induced by footshock. METHODS Rats were first trained to lever-press for intravenous cocaine (1 mg/infusion/0.13 ml, FR-1 schedule), and then tested under extinction conditions until response rates returned to the pretraining baseline. Reinstatements, VTA glutamate and dopamine levels [microdialysis with high performance liquid chromatography (HPLC)] were then assessed, under various pharmacological conditions, after mild inescapable footshock. RESULTS Footshock-induced reinstatement of cocaine seeking and release of VTA glutamate and dopamine were blocked by selective blockade of VTA CRF(2) receptors (CRF(2)Rs) but not CRF(1)Rs. VTA perfusion of CRF or CRF(2)R agonists that have strong affinity for CRF-BP mimicked the effects induced by footshock while CRFR agonists that do not bind CRF-BP were ineffective. CRF(6-33), which competes for the CRF binding site on CRF-BP, attenuated the effects of CRF or urocortin I on VTA glutamate and dopamine release and on reinstatement of cocaine seeking. CONCLUSIONS The present studies revealed a role of VTA CRF-BP and suggest an involvement of CRF(2)R in the effectiveness of stress in triggering glutamate and dopamine release and cocaine seeking in drug-experienced animals.
Collapse
Affiliation(s)
- Bin Wang
- Behavioral Neuroscience Branch, Intramural Research Program (IRP), National Institute on Drug Abuse (NIDA), National Institutes of Health, 5500 Nathan Shock Drive, Baltimore, MD 21224, USA
| | | | | | | |
Collapse
|
37
|
Roseboom PH, Nanda SA, Bakshi VP, Trentani A, Newman SM, Kalin NH. Predator threat induces behavioral inhibition, pituitary-adrenal activation and changes in amygdala CRF-binding protein gene expression. Psychoneuroendocrinology 2007; 32:44-55. [PMID: 17116372 PMCID: PMC1847640 DOI: 10.1016/j.psyneuen.2006.10.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2006] [Revised: 09/29/2006] [Accepted: 10/02/2006] [Indexed: 10/23/2022]
Abstract
Behavioral inhibition (BI) is an adaptive defensive response to threat; however, extreme BI is associated with anxiety-related psychopathology. When rats are exposed to a natural predator they display stress- and anxiety-related behavioral alterations and physiological activation. To develop a preclinical rodent model to study mechanisms underlying human BI and anxiety, we examined the extent to which ferret exposure elicits anxiety-related BI and HPA and amygdala activation of the CRF system. In the first experiment, BI and other behaviors were assessed in the presence or absence of a ferret. In the second experiment, ferret-induced corticosterone release and changes in brain c-fos expression were assessed. In the final experiment, gene chip and quantitative real time-PCR analyses were performed on amygdala tissue from control and ferret-exposed rats. Ferret exposure increased BI and submissive posturing, as well as plasma corticosterone and the number of Fos-positive cells in several brain regions including the amygdala. Gene expression analysis revealed increased amygdalar mRNA for CRF-binding protein, but not the CRF1 receptor, CRF2 receptor or CRF. In rodents, ferret exposure can be used to elicit anxiety-related BI, which is associated with HPA and amygdala activation. Since the amygdala and the CRF system have been implicated in adaptive and maladaptive anxiety responses in humans, these data support use of our rodent model to further investigate mechanisms underlying anxiety-related psychopathology in humans.
Collapse
Affiliation(s)
- Patrick H Roseboom
- Neuroscience Training Program, University of Wisconsin-Madison, 6001 Research Park Blvd., Madison, WI 53719, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Herringa RJ, Roseboom PH, Kalin NH. Decreased amygdala CRF-binding protein mRNA in post-mortem tissue from male but not female bipolar and schizophrenic subjects. Neuropsychopharmacology 2006; 31:1822-31. [PMID: 16482088 DOI: 10.1038/sj.npp.1301038] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Stressful life events are commonly associated with the onset and maintenance of psychopathology and much research has focused on the role of the corticotropin-releasing factor (CRF) system in mediating psychopathology. Since CRF serves to integrate the stress response, it is possible that the CRF system plays a role as a neurochemical linkage between stress and psychopathology. CRF-binding protein (CRF-BP) is thought to modulate CRF activity by decreasing its actions. Therefore, in some psychopathological states, alterations in CRF-BP function may contribute to dysregulation of the CRF system. Since the amygdala CRF system mediates stress- and anxiety-related behaviors and alterations in amygdala function are associated with psychopathology, we examined amygdala CRF-BP gene expression in post-mortem brains from subjects with major depression, bipolar disorder, and schizophrenia as well as in controls. In addition to characterizing the anatomic distribution of CRF-BP mRNA in the human amygdala and medial temporal lobe region, we found a significant decrease in CRF-BP mRNA levels in the basolateral amygdala of male bipolar and male schizophrenic subjects and the lateral amygdala of male bipolar subjects. These results raise the possibility that men with decreased amygdala CRF-BP may be more vulnerable to the effects of stress exposure on the etiology or maintenance of bipolar disorder or schizophrenia.
Collapse
Affiliation(s)
- Ryan J Herringa
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | | | | |
Collapse
|
39
|
Herringa RJ, Mackenrodt DB, Barlow JD, Roseboom PH, Nanda SA, Kalin NH. Corticotropin-releasing factor (CRF), but not corticosterone, increases basolateral amygdala CRF-binding protein. Brain Res 2006; 1083:21-8. [PMID: 16545343 DOI: 10.1016/j.brainres.2006.01.122] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2005] [Revised: 01/26/2006] [Accepted: 01/30/2006] [Indexed: 11/28/2022]
Abstract
Corticotropin-releasing factor (CRF) is a key mediator of the behavioral, autonomic, and endocrine responses to stress. CRF binds two receptors and a CRF-binding protein (CRF-BP), which may inactivate or modulate the actions of CRF at its receptors. The amygdala is an important anatomical substrate for CRF and contains CRF, its receptors, and CRF-BP. Our previous studies demonstrated that acute stress increases basolateral amygdala (BLA) CRF-BP mRNA. However, factors that may be responsible for this increase remain unclear. Both CRF and corticosterone are released during stress and are known to increase CRF-BP in vitro. However, the effects of these agents in vivo on brain CRF-BP have not been studied. Therefore, we examined the effects of CRF and corticosterone administration on BLA CRF-BP mRNA in rats. The findings demonstrate that intracerebroventricular CRF (5 microg) significantly increases BLA CRF-BP mRNA 9 h post-infusion, a time point consistent with that observed for the effects of acute stress-induced increases in CRF-BP. In contrast, injection of corticosterone at a dose mimicking acute stress (6.5 mg/kg sc) failed to increase BLA CRF-BP mRNA 9 h post-injection. Surprisingly, two different CRF antagonists failed to block CRF-induced increases in CRF-BP mRNA. These results suggest that CRF, but not corticosterone, may be responsible for stress-induced increases in BLA CRF-BP gene expression. Furthermore, this effect appears to be mediated by mechanisms other than the identified CRF receptors.
Collapse
MESH Headings
- Amygdala/drug effects
- Amygdala/metabolism
- Animals
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Corticosterone/blood
- Corticosterone/pharmacology
- Corticotropin-Releasing Hormone/metabolism
- Corticotropin-Releasing Hormone/pharmacology
- Dose-Response Relationship, Drug
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/genetics
- Male
- Protein Binding/drug effects
- Protein Binding/genetics
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors
- Receptors, Corticotropin-Releasing Hormone/genetics
- Receptors, Corticotropin-Releasing Hormone/metabolism
- Stress, Physiological/blood
- Stress, Physiological/genetics
- Stress, Physiological/physiopathology
- Up-Regulation/drug effects
- Up-Regulation/physiology
Collapse
Affiliation(s)
- Ryan J Herringa
- Wisconsin Psychiatric Institute and Clinics, 6001 Research Park Boulevard, Madison, WI 53719-1176, USA
| | | | | | | | | | | |
Collapse
|
40
|
Shekhar A, Truitt W, Rainnie D, Sajdyk T. Role of stress, corticotrophin releasing factor (CRF) and amygdala plasticity in chronic anxiety. Stress 2005; 8:209-19. [PMID: 16423710 DOI: 10.1080/10253890500504557] [Citation(s) in RCA: 162] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Stress initiates a series of neuronal responses that prepare an organism to adapt to new environmental challenges. However, chronic stress may lead to maladaptive responses that can result in psychiatric syndromes such as anxiety and depressive disorders. Corticotropin-releasing factor (CRF) has been identified as a key neuropeptide responsible for initiating many of the endocrine, autonomic and behavioral responses to stress. The amygdala expresses high concentrations of CRF receptors and is itself a major extrahypothalamic source of CRF containing neurons. Within the amygdala, the basolateral nucleus (BLA) has an important role in regulating anxiety and affective responses. During periods of stress, CRF is released into the amygdala and local CRF receptor activation has been postulated as a substrate for stress-induced alterations in affective behavior. Previous studies have suggested that synaptic plasticity in the BLA contributes to mechanisms underlying long-term changes in the regulation of affective behaviors. Several studies have shown that acute glutamate receptor-mediated activation, by either GABA-mediated disinhibition or CRF-mediated excitation, induces long-term synaptic plasticity and increases the excitability of BLA neurons. This review summarizes some of the data supporting the hypotheses that stress induced plasticity within the amygdala may be a critical step in the pathophysiology of the development of chronic anxiety states. It is further proposed that such a change in the limbic neural circuitry is involved in the transition from normal vigilance responses to pathological anxiety, leading to syndromes such as panic and post-traumatic stress disorders.
Collapse
Affiliation(s)
- Anantha Shekhar
- Indiana University School of Medicine, Department of Psychiatry, 1111 West 10th Street, Indianapolis, IN 46202, USA.
| | | | | | | |
Collapse
|
41
|
Van Den Eede F, Van Broeckhoven C, Claes SJ. Corticotropin-releasing factor-binding protein, stress and major depression. Ageing Res Rev 2005; 4:213-39. [PMID: 15996902 DOI: 10.1016/j.arr.2005.02.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2005] [Accepted: 02/22/2005] [Indexed: 11/26/2022]
Abstract
Major depressive disorder (MDD) is characterized by a dysregulation of the stress response system. A corticotropin-releasing factor (CRF) hyperdrive is a consistent and well-documented finding. CRF-binding protein (CRF-BP) may play a role in the pathogenesis of MDD. CRF-BP reduces the availability of CRF by binding free CRF and inhibits CRF function at the pituitary level. Moreover, CRF-BP expression increases in the pituitary and amygdala in response to acute stress, providing an additional feedback mechanism to maintain the homeostasis of the stress response. There are different regulatory elements of the expression of CRF-BP gene that are implicated in the pathophysiology of MDD, including CRF, glucocorticoids, cytokines and estrogens. A specific haplotype within the CRF-BP gene has been associated with MDD, but confirmation of this finding is necessary. Currently, the possible role of CRF-BP in the pathophysiology of conditions that have been associated with a hypofunction of the CRF system and immune dysfunctions is unclear. Implications of the function of CRF-BP for therapeutic strategies in MDD are being discussed. An important advantage of ligands that target CRF-BP is that concentrations of free CRF can be altered without acting directly on the transmission of CRF through its receptor.
Collapse
Affiliation(s)
- Filip Van Den Eede
- Department of Molecular Genetics VIB8, Flanders Interuniversity Institute for Biotechnology, University of Antwerp (UA), Universiteitsplein 1/Building T, B-2610 Antwerpen, Belgium
| | | | | |
Collapse
|