1
|
Maita I, Roepke TA, Samuels BA. Chronic stress-induced synaptic changes to corticotropin-releasing factor-signaling in the bed nucleus of the stria terminalis. Front Behav Neurosci 2022; 16:903782. [PMID: 35983475 PMCID: PMC9378865 DOI: 10.3389/fnbeh.2022.903782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/05/2022] [Indexed: 11/22/2022] Open
Abstract
The sexually dimorphic bed nucleus of the stria terminalis (BNST) is comprised of several distinct regions, some of which act as a hub for stress-induced changes in neural circuitry and behavior. In rodents, the anterodorsal BNST is especially affected by chronic exposure to stress, which results in alterations to the corticotropin-releasing factor (CRF)-signaling pathway, including CRF receptors and upstream regulators. Stress increases cellular excitability in BNST CRF+ neurons by potentiating miniature excitatory postsynaptic current (mEPSC) amplitude, altering the resting membrane potential, and diminishing M-currents (a voltage-gated K+ current that stabilizes membrane potential). Rodent anterodorsal and anterolateral BNST neurons are also critical regulators of behavior, including avoidance of aversive contexts and fear learning (especially that of sustained threats). These rodent behaviors are historically associated with anxiety. Furthermore, BNST is implicated in stress-related mood disorders, including anxiety and Post-Traumatic Stress Disorders in humans, and may be linked to sex differences found in mood disorders.
Collapse
Affiliation(s)
- Isabella Maita
- Samuels Laboratory, Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Neuroscience Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Troy A. Roepke
- Roepke Laboratory, Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Benjamin A. Samuels
- Samuels Laboratory, Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| |
Collapse
|
2
|
Balogh B, Vecsernyés M, Veres-Székely A, Berta G, Stayer-Harci A, Tarjányi O, Sétáló G. Urocortin stimulates ERK1/2 phosphorylation and proliferation but reduces ATP production of MCF7 breast cancer cells. Mol Cell Endocrinol 2022; 547:111610. [PMID: 35219718 DOI: 10.1016/j.mce.2022.111610] [Citation(s) in RCA: 3] [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: 06/23/2021] [Revised: 12/15/2021] [Accepted: 02/22/2022] [Indexed: 11/29/2022]
Abstract
Urocortins are members of the stress-related corticotropin-releasing factor family. Small amounts of them are present in the circulation and they are produced locally in various tissues of higher vertebrates. Aside from regulating circulation, or food uptake they also influence, via auto- and paracrine mechanisms, cell proliferation. In the present study we investigated in MCF7 human breast cancer cells the effect of urocortin onto mitogenic signaling via ERK1/2. Our results revealed that already 10 nM urocortin could stimulate the phosphorylation of these kinases and cell proliferation of MCF7 cells while ATP production was reduced when kept in the presence of the peptide up to two days. We examined the expression and contribution of the specific receptors of urocortin to the activation of ERK1/2 and to cell proliferation, the intracellular distribution of phosphorylated ERK1/2, and the involvement of additional proteins like PKA, PKB/Akt, MEK, p53, Rb and E2F-1 behind the observed phenomena.
Collapse
Affiliation(s)
- Bálint Balogh
- Department of Medical Biology and Central Electron Microscope Laboratory, University of Pécs, Medical School, Pécs, H-7643, Pécs, Szigeti út 12, Hungary.
| | - Mónika Vecsernyés
- Department of Medical Biology and Central Electron Microscope Laboratory, University of Pécs, Medical School, Pécs, H-7643, Pécs, Szigeti út 12, Hungary; Signal Transduction Research Group, János Szentágothai Research Centre, Pécs, H-7624, Pécs, Ifjúság útja 20, Hungary.
| | - Apor Veres-Székely
- 1st Department of Pediatrics, Semmelweis University, Budapest, H-1083, Budapest, 53-54. Bókay Street, Hungary; ELKH-SE Pediatrics and Nephrology Research Group, Budapest, Hungary.
| | - Gergely Berta
- Department of Medical Biology and Central Electron Microscope Laboratory, University of Pécs, Medical School, Pécs, H-7643, Pécs, Szigeti út 12, Hungary; Signal Transduction Research Group, János Szentágothai Research Centre, Pécs, H-7624, Pécs, Ifjúság útja 20, Hungary.
| | - Alexandra Stayer-Harci
- Department of Medical Biology and Central Electron Microscope Laboratory, University of Pécs, Medical School, Pécs, H-7643, Pécs, Szigeti út 12, Hungary; Signal Transduction Research Group, János Szentágothai Research Centre, Pécs, H-7624, Pécs, Ifjúság útja 20, Hungary.
| | - Oktávia Tarjányi
- Department of Medical Biology and Central Electron Microscope Laboratory, University of Pécs, Medical School, Pécs, H-7643, Pécs, Szigeti út 12, Hungary; Signal Transduction Research Group, János Szentágothai Research Centre, Pécs, H-7624, Pécs, Ifjúság útja 20, Hungary.
| | - György Sétáló
- Department of Medical Biology and Central Electron Microscope Laboratory, University of Pécs, Medical School, Pécs, H-7643, Pécs, Szigeti út 12, Hungary; Signal Transduction Research Group, János Szentágothai Research Centre, Pécs, H-7624, Pécs, Ifjúság útja 20, Hungary.
| |
Collapse
|
3
|
Wu Q, Feng Y, Liu L, Liu Y, Liu X, Zhang L, Li Y, Wang L. Corticotropin-Releasing Factor Aggravates Ischemic Stroke Injury by the Inflammatory Activation of Microglia. Endocrinology 2022; 163:6523128. [PMID: 35137012 DOI: 10.1210/endocr/bqac013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Indexed: 11/19/2022]
Abstract
Ischemic stroke is the second leading cause of death worldwide. Therefore, exploring effective and emerging molecular targets for ischemic stroke is a primary task of basic and clinical research. The aim of the present study was to investigate the function of corticotropin-releasing factor (CRF) in ischemic stroke and its related mechanisms, to provide a reference for the treatment of ischemic stroke. CRF, antalarmin, or astressin-2B were used to activate or block the CRF1 (CRF receptor 1) or CRF2 (CRF receptor 2) in BV2 cells and adult male mice, thus constructing a distal middle cerebral artery occlusion (dMCAO) model. CRF not only accelerated microglial activity by promoting transcription and production of inflammatory factors, but also promoted the transformation of activated BV2 cells from a neuroprotective phenotype (M2) to cytotoxic phenotype (M1), and these effects were mediated by the TLR4/NF-κB signaling pathway. These effects can be blocked by antalarmin but not by astressin-2B. CRF significantly aggravated the neurological deficit, increased infarction volume, and exacerbated neuronal injuries. Additionally, CRF significantly improved the levels of TNF-α and phospho-NF-κB in the ischemia penumbra. Finally, CRF significantly increased the number of CD16/Iba-1-positive cells and decreased the number of CD206/Iba-1-positive cells in the ischemia penumbra. These results provide evidence of the proinflammatory role of CRF in an ischemic stroke model and a possible underlying mechanism, which may facilitate the elucidation of potential treatment approaches for ischemic stroke.
Collapse
Affiliation(s)
- Qiang Wu
- Department of Neurosurgery, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Yan Feng
- Department of Neurosurgery, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Ling Liu
- Department of Pathology, Children's Hospital of Hebei Province, Shijiazhuang, Hebei, 050031, China
| | - Yang Liu
- Department of Pathology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, China
| | - Xin Liu
- Department of Neurosurgery, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Liqiao Zhang
- Department of Neurosurgery, East Branch of Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Yanan Li
- Department of Neurosurgery, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Liqun Wang
- Department of Neurosurgery, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| |
Collapse
|
4
|
Bhargava A, Arnold AP, Bangasser DA, Denton KM, Gupta A, Hilliard Krause LM, Mayer EA, McCarthy M, Miller WL, Raznahan A, Verma R. Considering Sex as a Biological Variable in Basic and Clinical Studies: An Endocrine Society Scientific Statement. Endocr Rev 2021; 42:219-258. [PMID: 33704446 PMCID: PMC8348944 DOI: 10.1210/endrev/bnaa034] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Indexed: 02/08/2023]
Abstract
In May 2014, the National Institutes of Health (NIH) stated its intent to "require applicants to consider sex as a biological variable (SABV) in the design and analysis of NIH-funded research involving animals and cells." Since then, proposed research plans that include animals routinely state that both sexes/genders will be used; however, in many instances, researchers and reviewers are at a loss about the issue of sex differences. Moreover, the terms sex and gender are used interchangeably by many researchers, further complicating the issue. In addition, the sex or gender of the researcher might influence study outcomes, especially those concerning behavioral studies, in both animals and humans. The act of observation may change the outcome (the "observer effect") and any experimental manipulation, no matter how well-controlled, is subject to it. This is nowhere more applicable than in physiology and behavior. The sex of established cultured cell lines is another issue, in addition to aneuploidy; chromosomal numbers can change as cells are passaged. Additionally, culture medium contains steroids, growth hormone, and insulin that might influence expression of various genes. These issues often are not taken into account, determined, or even considered. Issues pertaining to the "sex" of cultured cells are beyond the scope of this Statement. However, we will discuss the factors that influence sex and gender in both basic research (that using animal models) and clinical research (that involving human subjects), as well as in some areas of science where sex differences are routinely studied. Sex differences in baseline physiology and associated mechanisms form the foundation for understanding sex differences in diseases pathology, treatments, and outcomes. The purpose of this Statement is to highlight lessons learned, caveats, and what to consider when evaluating data pertaining to sex differences, using 3 areas of research as examples; it is not intended to serve as a guideline for research design.
Collapse
Affiliation(s)
- Aditi Bhargava
- Center for Reproductive Sciences, San Francisco, CA, USA
- Department of Obstetrics and Gynecology, University of California, San Francisco, CA, USA
| | - Arthur P Arnold
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, USA
| | - Kate M Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Arpana Gupta
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lucinda M Hilliard Krause
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Emeran A Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, CA, USA
| | - Margaret McCarthy
- Department of Pharmacology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Walter L Miller
- Center for Reproductive Sciences, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institutes of Mental Health, Intramural Research Program, Bethesda, MD, USA
| | - Ragini Verma
- Diffusion and Connectomics In Precision Healthcare Research (DiCIPHR) lab, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
5
|
Sex differences in stress reactivity in arousal and attention systems. Neuropsychopharmacology 2019; 44:129-139. [PMID: 30022063 PMCID: PMC6235989 DOI: 10.1038/s41386-018-0137-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/21/2018] [Accepted: 06/15/2018] [Indexed: 01/04/2023]
Abstract
Women are more likely than men to suffer from psychiatric disorders with hyperarousal symptoms, including posttraumatic stress disorder (PTSD) and major depression. In contrast, women are less likely than men to be diagnosed with schizophrenia and attention deficit hyperactivity disorder (ADHD), which share attentional impairments as a feature. Stressful events exacerbate symptoms of the aforementioned disorders. Thus, researchers are examining whether sex differences in stress responses bias women and men towards different psychopathology. Here we review the preclinical literature suggesting that, compared to males, females are more vulnerable to stress-induced hyperarousal, while they are more resilient to stress-induced attention deficits. Specifically described are sex differences in receptors for the stress neuropeptide, corticotropin-releasing factor (CRF), that render the locus coeruleus arousal system of females more vulnerable to stress and less adaptable to CRF hypersecretion, a condition found in patients with PTSD and depression. Studies on the protective effects of ovarian hormones against CRF-induced deficits in sustained attention are also detailed. Importantly, we highlight how comparing males and females in preclinical studies can lead to the development of novel therapeutics to improve treatments for psychiatric disorders in both women and men.
Collapse
|
6
|
Bangasser DA, Wicks B. Sex-specific mechanisms for responding to stress. J Neurosci Res 2017; 95:75-82. [PMID: 27870416 DOI: 10.1002/jnr.23812] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/23/2016] [Accepted: 06/10/2016] [Indexed: 12/12/2022]
Abstract
Posttraumatic stress disorder and major depression share stress as an etiological contributor and are more common in women than in men. Traditionally, preclinical studies investigating the neurobiological underpinnings of stress vulnerability have used only male rodents; however, recent studies that include females are finding sex-specific mechanisms for responding to stress. This Mini-Review examines recent literature using a framework developed by McCarthy and colleagues (2012; J Neurosci 32:2241-2247) that highlights different types of sex differences. First, we detail how learned fear responses in rats are sexually dimorphic. Then, we contrast this finding with fear extinction, which is similar in males and females at the behavioral level but at the circuitry level is associated with sex-specific cellular changes and, thus, exemplifies a sex convergence. Next, sex differences in stress hormones are detailed. Finally, the effects of stress on learning, attention, and arousal are used to highlight the concept of a sex divergence in which the behavior of males and females is similar at baseline but diverges following stressor exposure. We argue that appreciating and investigating the diversity of sex differences in stress response systems will improve our understanding of vulnerability and resilience to stress-related psychiatric disorders and likely lead to the development of novel therapeutics for better treatment of these disorders in both men and women. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, Pennsylvania
| | - Brittany Wicks
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
7
|
Sex differences in the locus coeruleus-norepinephrine system and its regulation by stress. Brain Res 2015; 1641:177-88. [PMID: 26607253 DOI: 10.1016/j.brainres.2015.11.021] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 10/27/2015] [Accepted: 11/12/2015] [Indexed: 12/26/2022]
Abstract
Women are more likely than men to suffer from post-traumatic stress disorder (PTSD) and major depression. In addition to their sex bias, these disorders share stress as an etiological factor and hyperarousal as a symptom. Thus, sex differences in brain arousal systems and their regulation by stress could help explain increased vulnerability to these disorders in women. Here we review preclinical studies that have identified sex differences in the locus coeruleus (LC)-norepinephrine (NE) arousal system. First, we detail how structural sex differences in the LC can bias females towards increased arousal in response to emotional events. Second, we highlight studies demonstrating that estrogen can increase NE in LC target regions by enhancing the capacity for NE synthesis, while reducing NE degradation, potentially increasing arousal in females. Third, we review data revealing how sex differences in the stress receptor, corticotropin releasing factor 1 (CRF1), can increase LC neuronal sensitivity to CRF in females compared to males. This effect could translate into hyperarousal in women under conditions of CRF hypersecretion that occur in PTSD and depression. The implications of these sex differences for the treatment of stress-related psychiatric disorders are discussed. Moreover, the value of using information regarding biological sex differences to aid in the development of novel pharmacotherapies to better treat men and women with PTSD and depression is also highlighted. This article is part of a Special Issue entitled SI: Noradrenergic System.
Collapse
|
8
|
Bangasser DA, Valentino RJ. Sex differences in stress-related psychiatric disorders: neurobiological perspectives. Front Neuroendocrinol 2014; 35:303-19. [PMID: 24726661 PMCID: PMC4087049 DOI: 10.1016/j.yfrne.2014.03.008] [Citation(s) in RCA: 476] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 02/26/2014] [Accepted: 03/28/2014] [Indexed: 12/14/2022]
Abstract
Stress is associated with the onset and severity of several psychiatric disorders that occur more frequently in women than men, including posttraumatic stress disorder (PTSD) and depression. Patients with these disorders present with dysregulation of several stress response systems, including the neuroendocrine response to stress, corticolimbic responses to negatively valenced stimuli, and hyperarousal. Thus, sex differences within their underlying circuitry may explain sex biases in disease prevalence. This review describes clinical studies that identify sex differences within the activity of these circuits, as well as preclinical studies that demonstrate cellular and molecular sex differences in stress responses systems. These studies reveal sex differences from the molecular to the systems level that increase endocrine, emotional, and arousal responses to stress in females. Exploring these sex differences is critical because this research can reveal the neurobiological underpinnings of vulnerability to stress-related psychiatric disorders and guide the development of novel pharmacotherapies.
Collapse
Affiliation(s)
- Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States.
| | - Rita J Valentino
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
9
|
Ducarouge B, Pelissier-Rota M, Lainé M, Cristina N, Vachez Y, Scoazec JY, Bonaz B, Jacquier-Sarlin M. CRF2 signaling is a novel regulator of cellular adhesion and migration in colorectal cancer cells. PLoS One 2013; 8:e79335. [PMID: 24260200 PMCID: PMC3832608 DOI: 10.1371/journal.pone.0079335] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/30/2013] [Indexed: 12/14/2022] Open
Abstract
Stress has been proposed to be a tumor promoting factor through the secretion of specific neuromediators, such as Urocortin2 and 3 (Ucn2/3), however its role in colorectal cancer (CRC) remains elusive. We observed that Ucn2/3 and their receptor the Corticotropin Releasing Factor receptor 2 (CRF2) were up-regulated in high grade and poorly differentiated CRC. This suggests a role for CRF2 in the loss of cellular organization and tumor progression. Using HT-29 and SW620 cells, two CRC cell lines differing in their abilities to perform cell-cell contacts, we found that CRF2 signals through Src/ERK pathway to induce the alteration of cell-cell junctions and the shuttle of p120ctn and Kaiso in the nucleus. In HT-29 cells, this signaling pathway also leads to the remodeling of cell adhesion by i) the phosphorylation of Focal Adhesion Kinase and ii) a modification of actin cytoskeleton and focal adhesion complexes. These events stimulate cell migration and invasion. In conclusion, our findings indicate that CRF2 signaling controls cellular organization and may promote metastatic potential of human CRC cells through an epithelial-mesenchymal transition like process. This contributes to the comprehension of the tumor-promoting effects of stress molecules and designates Ucn2/3-CRF2 tandem as a target to prevent CRC progression and aggressiveness.
Collapse
Affiliation(s)
- Benjamin Ducarouge
- Equipe du Stress et des Interactions Neurodigestives, INSERM U836, Grenoble, France
- Université Joseph Fourier, Grenoble Institut des Neurosciences, Grenoble, France
| | - Marjolaine Pelissier-Rota
- Equipe du Stress et des Interactions Neurodigestives, INSERM U836, Grenoble, France
- Université Joseph Fourier, Grenoble Institut des Neurosciences, Grenoble, France
| | - Michèle Lainé
- Equipe du Stress et des Interactions Neurodigestives, INSERM U836, Grenoble, France
- Université Joseph Fourier, Grenoble Institut des Neurosciences, Grenoble, France
| | - Nadine Cristina
- Equipe du Stress et des Interactions Neurodigestives, INSERM U836, Grenoble, France
- Université Joseph Fourier, Grenoble Institut des Neurosciences, Grenoble, France
| | - Yvan Vachez
- Université Joseph Fourier, Grenoble Institut des Neurosciences, Grenoble, France
| | | | - Bruno Bonaz
- Equipe du Stress et des Interactions Neurodigestives, INSERM U836, Grenoble, France
- Université Joseph Fourier, Grenoble Institut des Neurosciences, Grenoble, France
- Centre Hospitalo Universitaire de Grenoble, Grenoble, France
| | - Muriel Jacquier-Sarlin
- Equipe du Stress et des Interactions Neurodigestives, INSERM U836, Grenoble, France
- Université Joseph Fourier, Grenoble Institut des Neurosciences, Grenoble, France
- * E-mail:
| |
Collapse
|
10
|
Rostkowski AB, Leitermann RJ, Urban JH. Differential activation of neuronal cell types in the basolateral amygdala by corticotropin releasing factor. Neuropeptides 2013; 47:273-80. [PMID: 23688647 PMCID: PMC3736811 DOI: 10.1016/j.npep.2012.12.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 12/05/2012] [Accepted: 12/14/2012] [Indexed: 11/30/2022]
Abstract
Enhanced corticotropin releasing factor (CRF) release in the basolateral amygdala (BLA) is strongly associated with the generation of behavioral stress responses through activation of the CRF-R1 receptor subtype. Stress and anxiety-like behavior are modulated in part by the balance of peptide actions such as excitatory CRF and inhibitory neuropeptide Y (NPY) receptor activation in the BLA. While the actions of CRF are clear, little is known about the cell type influenced by CRF receptor stimulation. These studies were designed to identify the cell types within the BLA activated by intra-BLA administration of CRF using multi-label immunohistochemistry for cFos and markers for pyramidal (CaMKII-immunopositive) and interneuronal [glutamic acid decarboxylase (GAD65)] cell populations. Administration of CRF into the BLA produced a dose-dependent increase in the expression of cFos-ir. Intra-BLA injection of CRF induced significant increases in cFos-ir in the CaMKII-ir population. Although increases in cFos-ir in GAD65-ir cells were observed, this did not reach statistical significance perhaps in part due to the decreased numbers of GAD65-ir cells within the BLA after CRF treatment. These findings demonstrate that CRF, when released into the BLA, activates projection neurons and that the activity of GABAergic interneurons is also altered by CRF treatment. Decreases in the number of GAD65-ir neurons could reflect either increased or decreased activity of these cells and future studies will more directly address these possibilities. The expression of cFos is associated with longer term regulation of gene expression which may be involved in the profound long term effects of neuropeptides, such as CRF, on the activity and plasticity of BLA pyramidal neurons.
Collapse
Affiliation(s)
- Amanda B. Rostkowski
- Interdepartmental Neuroscience Program, The Chicago Medical School/Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| | - Randy J. Leitermann
- Department of Physiology and Biophysics; The Chicago Medical School/Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| | - Janice H. Urban
- Interdepartmental Neuroscience Program, The Chicago Medical School/Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
- Department of Physiology and Biophysics; The Chicago Medical School/Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| |
Collapse
|
11
|
Kratzer S, Mattusch C, Metzger MW, Dedic N, Noll-Hussong M, Kafitz KW, Eder M, Deussing JM, Holsboer F, Kochs E, Rammes G. Activation of CRH receptor type 1 expressed on glutamatergic neurons increases excitability of CA1 pyramidal neurons by the modulation of voltage-gated ion channels. Front Cell Neurosci 2013; 7:91. [PMID: 23882180 PMCID: PMC3715697 DOI: 10.3389/fncel.2013.00091] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 05/25/2013] [Indexed: 01/16/2023] Open
Abstract
Corticotropin-releasing hormone (CRH) plays an important role in a substantial number of patients with stress-related mental disorders, such as anxiety disorders and depression. CRH has been shown to increase neuronal excitability in the hippocampus, but the underlying mechanisms are poorly understood. The effects of CRH on neuronal excitability were investigated in acute hippocampal brain slices. Population spikes (PS) and field excitatory postsynaptic potentials (fEPSP) were evoked by stimulating Schaffer-collaterals and recorded simultaneously from the somatic and dendritic region of CA1 pyramidal neurons. CRH was found to increase PS amplitudes (mean ± Standard error of the mean; 231.8 ± 31.2% of control; n = 10) while neither affecting fEPSPs (104.3 ± 4.2%; n = 10) nor long-term potentiation (LTP). However, when Schaffer-collaterals were excited via action potentials (APs) generated by stimulation of CA3 pyramidal neurons, CRH increased fEPSP amplitudes (119.8 ± 3.6%; n = 8) and the magnitude of LTP in the CA1 region. Experiments in slices from transgenic mice revealed that the effect on PS amplitude is mediated exclusively by CRH receptor 1 (CRHR1) expressed on glutamatergic neurons. The effects of CRH on PS were dependent on phosphatase-2B, L- and T-type calcium channels and voltage-gated potassium channels but independent on intracellular Ca2+-elevation. In patch-clamp experiments, CRH increased the frequency and decay times of APs and decreased currents through A-type and delayed-rectifier potassium channels. These results suggest that CRH does not affect synaptic transmission per se, but modulates voltage-gated ion currents important for the generation of APs and hence elevates by this route overall neuronal activity.
Collapse
Affiliation(s)
- Stephan Kratzer
- Department of Anesthesiology, Klinikum Rechts der Isar der Technischen Universität München Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Hough D, Swart P, Cloete S. Exploration of the Hypothalamic-Pituitary-Adrenal Axis to Improve Animal Welfare by Means of Genetic Selection: Lessons from the South African Merino. Animals (Basel) 2013; 3:442-74. [PMID: 26487412 PMCID: PMC4494397 DOI: 10.3390/ani3020442] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/06/2013] [Accepted: 05/13/2013] [Indexed: 11/25/2022] Open
Abstract
It is a difficult task to improve animal production by means of genetic selection, if the environment does not allow full expression of the animal's genetic potential. This concept may well be the future for animal welfare, because it highlights the need to incorporate traits related to production and robustness, simultaneously, to reach sustainable breeding goals. This review explores the identification of potential genetic markers for robustness within the hypothalamic-pituitary-adrenal axis (HPAA), since this axis plays a vital role in the stress response. If genetic selection for superior HPAA responses to stress is possible, then it ought to be possible to breed robust and easily managed genotypes that might be able to adapt to a wide range of environmental conditions whilst expressing a high production potential. This approach is explored in this review by means of lessons learnt from research on Merino sheep, which were divergently selected for their multiple rearing ability. These two selection lines have shown marked differences in reproduction, production and welfare, which makes this breeding programme ideal to investigate potential genetic markers of robustness. The HPAA function is explored in detail to elucidate where such genetic markers are likely to be found.
Collapse
Affiliation(s)
- Denise Hough
- Department of Biochemistry, Stellenbosch University, Stellenbosch 7602, South Africa.
| | - Pieter Swart
- Department of Biochemistry, Stellenbosch University, Stellenbosch 7602, South Africa.
| | - Schalk Cloete
- Department of Animal Sciences, Stellenbosch University, Stellenbosch 7602, South Africa.
- Institute for Animal Production, Elsenburg, Private Bag X1, Elsenburg 7607, South Africa.
| |
Collapse
|
13
|
Dunn HA, Walther C, Godin CM, Hall RA, Ferguson SSG. Role of SAP97 protein in the regulation of corticotropin-releasing factor receptor 1 endocytosis and extracellular signal-regulated kinase 1/2 signaling. J Biol Chem 2013; 288:15023-34. [PMID: 23576434 DOI: 10.1074/jbc.m113.473660] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The corticotropin-releasing factor (CRF) receptor 1 (CRFR1) is a target for the treatment of psychiatric diseases such as depression, schizophrenia, anxiety disorder, and bipolar disorder. The carboxyl-terminal tail of the CRFR1 terminates in a PDZ-binding motif that provides a potential site for the interaction of PSD-95/Discs Large/Zona Occludens 1 (PDZ) domain-containing proteins. In this study, we found that CRFR1 interacts with synapse-associated protein 97 (SAP97; also known as DLG1) by co-immunoprecipitation in human embryonic 293 (HEK 293) cells and cortical brain lysates and that this interaction is dependent upon an intact PDZ-binding motif at the end of the CRFR1 carboxyl-terminal tail. Similarly, we demonstrated that SAP97 is recruited to the plasma membrane in HEK 293 cells expressing CRFR1 and that mutation of the CRFR1 PDZ-binding motif results in the redistribution of SAP97 into the cytoplasm. Overexpression of SAP97 antagonized agonist-stimulated CRFR1 internalization, whereas single hairpin (shRNA) knockdown of endogenous SAP97 in HEK 293 cells resulted in increased agonist-stimulated CRFR1 endocytosis. CRFR1 was internalized as a complex with SAP97 resulting in the redistribution of SAP97 to endocytic vesicles. Overexpression or shRNA knockdown of SAP97 did not significantly affect CRFR1-mediated cAMP formation, but SAP97 knockdown did significantly attenuate CRFR1-stimulated ERK1/2 phosphorylation in a PDZ interaction-independent manner. Taken together, our studies show that SAP97 interactions with CRFR1 attenuate CRFR1 endocytosis and that SAP97 is involved in coupling G protein-coupled receptors to the activation of the ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Henry A Dunn
- J Allyn Taylor Centre for Cell Biology, Robarts Research Institute, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | | | | | | | | |
Collapse
|
14
|
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: 82] [Impact Index Per Article: 6.8] [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
|
15
|
Valadas JS, Batalha VL, Ferreira DG, Gomes R, Coelho JE, Sebastião AM, Diógenes MJ, Lopes LV. Neuroprotection afforded by adenosine A2A
receptor blockade is modulated by corticotrophin-releasing factor (CRF) in glutamate injured cortical neurons. J Neurochem 2012; 123:1030-40. [DOI: 10.1111/jnc.12050] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2012] [Revised: 07/21/2012] [Accepted: 10/07/2012] [Indexed: 11/29/2022]
Affiliation(s)
- Jorge S. Valadas
- Institute of Pharmacology and Neurosciences; Faculdade de Medicina da Universidade de Lisboa; Lisboa Portugal
- Instituto de Medicina Molecular; Faculdade de Medicina da Universidade de Lisboa; Lisboa Portugal
| | - Vânia L. Batalha
- Institute of Pharmacology and Neurosciences; Faculdade de Medicina da Universidade de Lisboa; Lisboa Portugal
- Instituto de Medicina Molecular; Faculdade de Medicina da Universidade de Lisboa; Lisboa Portugal
| | - Diana G. Ferreira
- Institute of Pharmacology and Neurosciences; Faculdade de Medicina da Universidade de Lisboa; Lisboa Portugal
- Instituto de Medicina Molecular; Faculdade de Medicina da Universidade de Lisboa; Lisboa Portugal
| | - Rui Gomes
- Instituto de Medicina Molecular; Faculdade de Medicina da Universidade de Lisboa; Lisboa Portugal
- Faculdade de Ciências da Universidade de Lisboa; Lisboa Portugal
| | - Joana E. Coelho
- Institute of Pharmacology and Neurosciences; Faculdade de Medicina da Universidade de Lisboa; Lisboa Portugal
- Instituto de Medicina Molecular; Faculdade de Medicina da Universidade de Lisboa; Lisboa Portugal
| | - Ana M. Sebastião
- Institute of Pharmacology and Neurosciences; Faculdade de Medicina da Universidade de Lisboa; Lisboa Portugal
- Instituto de Medicina Molecular; Faculdade de Medicina da Universidade de Lisboa; Lisboa Portugal
| | - Maria José Diógenes
- Institute of Pharmacology and Neurosciences; Faculdade de Medicina da Universidade de Lisboa; Lisboa Portugal
- Instituto de Medicina Molecular; Faculdade de Medicina da Universidade de Lisboa; Lisboa Portugal
| | - Luísa V. Lopes
- Institute of Pharmacology and Neurosciences; Faculdade de Medicina da Universidade de Lisboa; Lisboa Portugal
- Instituto de Medicina Molecular; Faculdade de Medicina da Universidade de Lisboa; Lisboa Portugal
| |
Collapse
|
16
|
Affiliation(s)
- Tracy L Bale
- Department of Animal Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| | | |
Collapse
|
17
|
Bangasser DA, Valentino RJ. Sex differences in molecular and cellular substrates of stress. Cell Mol Neurobiol 2012; 32:709-23. [PMID: 22488525 DOI: 10.1007/s10571-012-9824-4] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 02/22/2012] [Indexed: 12/20/2022]
Abstract
Women are twice as likely as men to suffer from stress-related psychiatric disorders, like unipolar depression and post-traumatic stress disorder. Although the underlying neural mechanisms are not well characterized, the pivotal role of stress in the onset and severity of these diseases has led to the idea that sex differences in stress responses account for this sex bias. Corticotropin-releasing factor (CRF) orchestrates stress responses by acting both as a neurohormone to initiate the hypothalamic-pituitary-adrenal (HPA) axis and as a neuromodulator in the brain. One target of CRF modulation is the locus coeruleus (LC)-norepinephrine system, which coordinates arousal components of the stress response. Hypersecretion of CRF and dysregulation of targets downstream from CRF, such as the HPA axis and LC-norepinephrine system, are characteristic features of many stress-related psychiatric diseases, suggesting a causal role for CRF and its targets in the development of these disorders. This review will describe sex differences in CRF and the LC-norepinephrine system that can increase stress sensitivity in females, making them vulnerable to stress-related disorders. Evidence for gonadal hormone regulation of hypothalamic CRF is discussed as an effect that can lead to increased HPA axis activity in females. Sex differences in the structure of LC neurons that create the potential for hyperarousal in response to emotional stimuli are described. Finally, sex differences at the molecular level of the CRF(1) receptor that make the LC-norepinephrine system more reactive in females are reviewed. The implications of these sex differences for the treatment of stress-related psychiatric disorders also will be discussed.
Collapse
Affiliation(s)
- Debra A Bangasser
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | |
Collapse
|
18
|
Williams TJ, Akama KT, Knudsen MG, McEwen BS, Milner TA. Ovarian hormones influence corticotropin releasing factor receptor colocalization with delta opioid receptors in CA1 pyramidal cell dendrites. Exp Neurol 2011; 230:186-96. [PMID: 21549703 DOI: 10.1016/j.expneurol.2011.04.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2010] [Revised: 03/14/2011] [Accepted: 04/14/2011] [Indexed: 11/19/2022]
Abstract
Stress interacts with addictive processes to increase drug use, drug seeking, and relapse. The hippocampal formation (HF) is an important site at which stress circuits and endogenous opioid systems intersect and likely plays a critical role in the interaction between stress and drug addiction. Our prior studies demonstrate that the stress-related neuropeptide corticotropin-releasing factor (CRF) and the delta-opioid receptor (DOR) colocalize in interneuron populations in the hilus of the dentate gyrus and stratum oriens of CA1 and CA3. While independent ultrastructural studies of DORs and CRF receptors suggest that each receptor is found in CA1 pyramidal cell dendrites and dendritic spines, whether DORs and CRF receptors colocalize in CA1 neuronal profiles has not been investigated. Here, hippocampal sections of adult male and proestrus female Sprague-Dawley rats were processed for dual label pre-embedding immunoelectron microscopy using well-characterized antisera directed against the DOR for immunoperoxidase and against the CRF receptor for immunogold. DOR-immunoreactivity (-ir) was found presynaptically in axons and axon terminals as well as postsynaptically in somata, dendrites and dendritic spines in stratum radiatum of CA1. In contrast, CRF receptor-ir was predominantly found postsynaptically in CA1 somata, dendrites, and dendritic spines. CRF receptor-ir frequently was observed in DOR-labeled dendritic profiles and primarily was found in the cytoplasm rather than at or near the plasma membrane. Quantitative analysis of CRF receptor-ir colocalization with DOR-ir in pyramidal cell dendrites revealed that proestrus females and males show comparable levels of CRF receptor-ir per dendrite and similar cytoplasmic density of CRF receptor-ir. In contrast, proestrus females display an increased number of dual-labeled dendritic profiles and an increased membrane density of CRF receptor-ir in comparison to males. We further examined the functional consequences of CRF receptor-ir colocalization with DOR-ir in the same neuron using the hormone responsive neuronal cell line NG108-15, which endogenously expresses DORs, and assayed intracellular cAMP production in response to CRF receptor and DOR agonists. Results demonstrated that short-term application of DOR agonist SNC80 inhibited CRF-induced cAMP accumulation in NG108-15 cells transfected with the CRF receptor. These studies provide new insights on opioid-stress system interaction in the hippocampus of both males and females and establish potential mechanisms through which DOR activation may influence CRF receptor activity.
Collapse
Affiliation(s)
- Tanya J Williams
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA.
| | | | | | | | | |
Collapse
|
19
|
Stern CM, Luoma JI, Meitzen J, Mermelstein PG. Corticotropin releasing factor-induced CREB activation in striatal neurons occurs via a novel Gβγ signaling pathway. PLoS One 2011; 6:e18114. [PMID: 21448293 PMCID: PMC3063246 DOI: 10.1371/journal.pone.0018114] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 02/25/2011] [Indexed: 02/06/2023] Open
Abstract
The peptide corticotropin-releasing factor (CRF) was initially identified as a critical component of the stress response. CRF exerts its cellular effects by binding to one of two cognate G-protein coupled receptors (GPCRs), CRF receptor 1 (CRFR1) or 2 (CRFR2). While these GPCRs were originally characterized as being coupled to Gα(s), leading to downstream activation of adenylyl cyclase (AC) and subsequent increases in cAMP, it has since become clear that CRFRs couple to and activate numerous other downstream signaling cascades. In addition, CRF signaling influences the activity of many diverse brain regions, affecting a variety of behaviors. One of these regions is the striatum, including the nucleus accumbens (NAc). CRF exerts profound effects on striatal-dependent behaviors such as drug addiction, pair-bonding, and natural reward. Recent data indicate that at least some of these behaviors regulated by CRF are mediated through CRF activation of the transcription factor CREB. Thus, we aimed to elucidate the signaling pathway by which CRF activates CREB in striatal neurons. Here we describe a novel neuronal signaling pathway whereby CRF leads to a rapid Gβγ- and MEK-dependent increase in CREB phosphorylation. These data are the first descriptions of CRF leading to activation of a Gβγ-dependent signaling pathway in neurons, as well as the first description of Gβγ activation leading to downstream CREB phosphorylation in any cellular system. Additionally, these data provide additional insight into the mechanisms by which CRF can regulate neuronal function.
Collapse
Affiliation(s)
- Christopher M. Stern
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jessie I. Luoma
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - John Meitzen
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Paul G. Mermelstein
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
20
|
Sex differences in corticotropin-releasing factor receptor signaling and trafficking: potential role in female vulnerability to stress-related psychopathology. Mol Psychiatry 2010; 15:877, 896-904. [PMID: 20548297 PMCID: PMC2935505 DOI: 10.1038/mp.2010.66] [Citation(s) in RCA: 268] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Although the higher incidence of stress-related psychiatric disorders in females is well documented, its basis is unknown. Here, we show that the receptor for corticotropin-releasing factor (CRF), the neuropeptide that orchestrates the stress response, signals and is trafficked differently in female rats in a manner that could result in a greater response and decreased adaptation to stressors. Most cellular responses to CRF in the brain are mediated by CRF receptor (CRFr) association with the GTP-binding protein, G(s). Receptor immunoprecipitation studies revealed enhanced CRFr-G(s) coupling in cortical tissue of unstressed female rats. Previous stressor exposure abolished this sex difference by increasing CRFr-G(s) coupling selectively in males. These molecular results mirrored the effects of sex and stress on sensitivity of locus ceruleus (LC)-norepinephrine neurons to CRF. Differences in CRFr trafficking were also identified that could compromise stress adaptation in females. Specifically, stress-induced CRFr association with beta-arrestin2, an integral step in receptor internalization, occurred only in male rats. Immunoelectron microscopy confirmed that stress elicited CRFr internalization in LC neurons of male rats exclusively, consistent with reported electrophysiological evidence for stress-induced desensitization to CRF in males. Together, these studies identified two aspects of CRFr function, increased cellular signaling and compromised internalization, which render CRF-receptive neurons of females more sensitive to low levels of CRF and less adaptable to high levels of CRF. CRFr dysfunction in females may underlie their increased vulnerability to develop stress-related pathology, particularly that related to increased activity of the LC-norepinephrine system, such as depression or post-traumatic stress disorder.
Collapse
|
21
|
Tanaka M, Watanabe Y, Yoshimoto K. Regulation of relaxin 3 gene expression via cAMP-PKA in a neuroblastoma cell line. J Neurosci Res 2009; 87:820-9. [PMID: 18831067 DOI: 10.1002/jnr.21895] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Relaxin 3 is expressed in neurons of the brain stem that inneravate wide areas of the forebrain. Relaxin 3 mRNA levels in these neurons are increased in response to restraint stress, and by central administration of corticotropin-releasing factor (CRF). In the present study, we observed that relaxin 3 was expressed in a mouse neuroblastoma cell line, Neuro2a, and investigated the intracellular signaling that activated relaxin 3 gene transcription in vitro. By means of a clone stably transfected with a relaxin 3 promoter-EGFP gene, we observed that dibutyryl cyclic AMP and forskolin increased the relaxin 3 promoter activity. These increases were inhibited by pretreatment with PKA inhibitors, H89 and KT5720. Moreover, the promoter activity was enhanced by CRF treatment after expression of CRF-R1 receptor on the cells. Taken together, these results indicate that relaxin 3 transcription is activated via the cAMP-PKA pathway in the downstream of CRF-R1.
Collapse
Affiliation(s)
- Masaki Tanaka
- Department of Cell Biology, Research Institute for Neurological Diseases and Geriatrics, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | | | | |
Collapse
|
22
|
Richardson HN, Zhao Y, Fekete ÉM, Funk CK, Wirsching P, Janda K, Zorrilla EP, Koob GF. MPZP: a novel small molecule corticotropin-releasing factor type 1 receptor (CRF1) antagonist. Pharmacol Biochem Behav 2008; 88:497-510. [PMID: 18031798 PMCID: PMC3319109 DOI: 10.1016/j.pbb.2007.10.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2007] [Revised: 10/13/2007] [Accepted: 10/17/2007] [Indexed: 10/22/2022]
Abstract
The extrahypothalamic stress peptide corticotropin-releasing factor (CRF) system is an important regulator of behavioral responses to stress. Dysregulation of CRF and the CRF type 1 receptor (CRF(1)) system is hypothesized to underlie many stress-related disorders. Modulation of the CRF(1) system by non-peptide antagonists currently is being explored as a therapeutic approach for anxiety disorders and alcohol dependence. Here, we describe a new, less hydrophilic (cLogP approximately 2.95), small molecule, non-peptide CRF(1) antagonist with high affinity (K(i)=4.9 nM) and specificity for CRF(1) receptors: N,N-bis(2-methoxyethyl)-3-(4-methoxy-2-methylphenyl)-2,5-dimethyl-pyrazolo[1,5-a] pyrimidin-7-amine (MPZP). The compound was systemically administered to adult male rats in two behavioral models dependent on the CRF(1) system: defensive burying (0, 5, 20 mg/kg, n=6-11 for each dose) and alcohol dependence (0, 5, 10, 20 mg/kg, n=8 for each self-administration group). Acute administration of MPZP reduced burying behavior in the defensive burying model of active anxiety-like behavior. MPZP also attenuated withdrawal-induced excessive drinking in the self-administration model of alcohol dependence without affecting nondependent alcohol drinking or water consumption. The present findings support the proposed significance of the CRF(1) system in anxiety and alcohol dependence and introduce a promising new compound for further development in the treatment of alcohol dependence and stress-related disorders.
Collapse
Affiliation(s)
- Heather N. Richardson
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, 92037 California, USA
| | - Yu Zhao
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, 92037 California, USA
| | - Éva M. Fekete
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, 92037 California, USA
- Institute of Physiology, Pécs University Medical School, 7602 Pécs, Hungary
| | - Cindy K. Funk
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, 92037 California, USA
- Institut National de la Sante et de la Recherche Medicale, France
| | - Peter Wirsching
- Department of Chemistry, The Scripps Research Institute, La Jolla, 92037 California, USA
| | - Kim Janda
- Department of Chemistry, The Scripps Research Institute, La Jolla, 92037 California, USA
| | - Eric P. Zorrilla
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, 92037 California, USA
- Harold L. Dorris Neurological Research Institute, The Scripps Research Institute, La Jolla, 92037 California, USA
| | - George F. Koob
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, 92037 California, USA
| |
Collapse
|
23
|
Grigoriadis DE, Heroux JA, De Souza EB. Characterization and regulation of corticotropin-releasing factor receptors in the central nervous, endocrine and immune systems. CIBA FOUNDATION SYMPOSIUM 2007; 172:85-101; discussion 101-7. [PMID: 8387906 DOI: 10.1002/9780470514368.ch5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Corticotropin-releasing factor (CRF) plays a major role in coordinating the endocrine, autonomic, behavioural and immune responses to stress through actions in the brain and in the periphery. CRF receptors identified in brain, pituitary and spleen have comparable kinetic and pharmacological characteristics, guanine nucleotide sensitivity and adenylate cyclase-stimulating activity. Differences were observed in the molecular mass of the CRF receptor complex between brain (58,000 Da) and pituitary and spleen (75,000 Da), which appeared to be due to differential glycosylation of the receptor proteins. In autoradiographic studies, CRF receptors were localized in highest densities in anterior and intermediate lobes of the pituitary, olfactory bulb, cerebral cortex, amygdala, cerebellum and the macrophage-rich marginal zones and red pulp regions of the spleen. CRF can modulate the number of CRF receptors in both the brain and pituitary in a reciprocal manner. The demonstration of functional CRF receptors in brain, pituitary and spleen suggests the importance of this neuropeptide in integrating the responses of the CNS, endocrine and immune systems to physiological, psychological and immunological stimuli.
Collapse
Affiliation(s)
- D E Grigoriadis
- Central Nervous System Diseases Research, Du Pont Merck Pharmaceutical Company, Experimental Station, Wilmington, DE 19880-0400
| | | | | |
Collapse
|
24
|
Kang JE, Cirrito JR, Dong H, Csernansky JG, Holtzman DM. Acute stress increases interstitial fluid amyloid-beta via corticotropin-releasing factor and neuronal activity. Proc Natl Acad Sci U S A 2007; 104:10673-8. [PMID: 17551018 PMCID: PMC1965571 DOI: 10.1073/pnas.0700148104] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Aggregation of the amyloid-beta (Abeta) peptide in the extracellular space of the brain is critical in the pathogenesis of Alzheimer's disease. Abeta is produced by neurons and released into the brain interstitial fluid (ISF), a process regulated by synaptic activity. To determine whether behavioral stressors can regulate ISF Abeta levels, we assessed the effects of chronic and acute stress paradigms in amyloid precursor protein transgenic mice. Isolation stress over 3 months increased Abeta levels by 84%. Similarly, acute restraint stress increased Abeta levels over hours. Exogenous corticotropin-releasing factor (CRF) but not corticosterone mimicked the effects of acute restraint stress. Inhibition of endogenous CRF receptors or neuronal activity blocked the effects of acute stress on Abeta. Thus, behavioral stressors can rapidly increase ISF Abeta through neuronal activity in a CRF-dependent manner, and the results suggest a mechanism by which behavioral stress may affect Alzheimer's disease pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | - David M. Holtzman
- Departments of *Neurology
- Molecular Biology and Pharmacology
- Hope Center for Neurological Disorders, and
- Alzheimer's Disease Research Center, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110
- **To whom correspondence should be addressed. E-mail:
| |
Collapse
|
25
|
Funk CK, Zorrilla EP, Lee MJ, Rice KC, Koob GF. Corticotropin-releasing factor 1 antagonists selectively reduce ethanol self-administration in ethanol-dependent rats. Biol Psychiatry 2007; 61:78-86. [PMID: 16876134 PMCID: PMC2741496 DOI: 10.1016/j.biopsych.2006.03.063] [Citation(s) in RCA: 233] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2005] [Revised: 03/09/2006] [Accepted: 03/09/2006] [Indexed: 12/27/2022]
Abstract
BACKGROUND Alcohol dependence is characterized by excessive alcohol consumption, loss of control over intake, and the presence of a withdrawal syndrome, which includes both motivational and physical symptoms. Similar to human alcoholics, ethanol-dependent animals display enhanced anxiety-like behaviors and enhanced ethanol self-administration during withdrawal, effects hypothesized to result from a dysregulation of corticotropin-releasing factor (CRF) stress systems. Here, we used an animal model of ethanol dependence to test the effects of CRF(1) receptor antagonists on excessive ethanol self-administration in dependent rats. METHODS Wistar rats, trained to orally self-administer ethanol, were exposed intermittently to ethanol vapors to induce ethanol dependence. Nondependent animals were exposed to control air. Following a 2-hour period of withdrawal, dependent and nondependent animals were systemically administered antalarmin, MJL-1-109-2, or R121919 (CRF(1) antagonists) and ethanol self-administration was measured. RESULTS The nonpeptide, small molecule CRF(1) antagonists selectively reduced excessive self-administration of ethanol in dependent animals during acute withdrawal. The antagonists had no effect on ethanol self-administration in nondependent rats. CONCLUSIONS These data demonstrate that CRF(1) receptors play an important role in mediating excessive ethanol self-administration in dependent rats, with no effect in nondependent rats. CRF(1) antagonists may be exciting new pharmacotherapeutic targets for the treatment of alcoholism in humans.
Collapse
Affiliation(s)
- Cindy K Funk
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, California 92037, USA.
| | | | | | | | | |
Collapse
|
26
|
Funk CK, O'Dell LE, Crawford EF, Koob GF. Corticotropin-releasing factor within the central nucleus of the amygdala mediates enhanced ethanol self-administration in withdrawn, ethanol-dependent rats. J Neurosci 2006; 26:11324-32. [PMID: 17079660 PMCID: PMC6674550 DOI: 10.1523/jneurosci.3096-06.2006] [Citation(s) in RCA: 317] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Revised: 09/01/2006] [Accepted: 09/21/2006] [Indexed: 11/21/2022] Open
Abstract
Alcohol dependence is characterized by excessive consumption, loss of control over intake, and the presence of a withdrawal syndrome, including both motivational and physical symptoms. The motivational symptoms, including anxiety, have been hypothesized to be important factors eliciting excessive drinking during abstinence. Previous work has shown that ethanol-dependent rats also display enhanced anxiety-like behaviors and enhanced ethanol self-administration during withdrawal, likely resulting from dysregulation of brain corticotropin-releasing factor (CRF) stress systems. The present study was designed to explore the brain sites within the extended amygdala [central nucleus of the amygdala (CeA), lateral bed nucleus of the stria terminalis (BNST), and nucleus accumbens shell (NAcSh)] that mediate the increased ethanol self-administration observed during withdrawal. Ethanol-dependent animals showed an increase in ethanol self-administration after acute withdrawal relative to nondependent controls. The CRF antagonist D-Phe-CRF(12-41) ([D-Phe(12),Nle(21,38),C alpha MeLeu(37)]-rCRF(12-41)) was administered into the CeA, lateral BNST, or NAcSh of acute-withdrawn dependent and nondependent rats. Administered into the CeA, the antagonist reduced ethanol self-administration in dependent animals, with no effect in nondependent animals. Administration of D-Phe-CRF(12-41) into the lateral BNST and NAcSh was without effect on ethanol self-administration in dependent and nondependent animals. At the same time point of withdrawal, there was a decrease in CRF immunoreactivity within the CeA, suggesting an increased extracellular release of CRF during withdrawal. There was no change in CRF immunoreactivity in the BNST or NAcSh. These results indicate that CRF, specifically within the CeA, plays a role in mediating excessive ethanol consumption in ethanol-dependent animals.
Collapse
Affiliation(s)
- Cindy K Funk
- The Scripps Research Institute, La Jolla, California 92037, USA.
| | | | | | | |
Collapse
|
27
|
Gallopin T, Geoffroy H, Rossier J, Lambolez B. Cortical sources of CRF, NKB, and CCK and their effects on pyramidal cells in the neocortex. ACTA ACUST UNITED AC 2005; 16:1440-52. [PMID: 16339088 DOI: 10.1093/cercor/bhj081] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In order to investigate how neuropeptide transmission can modulate the neocortical network, we mapped the expression of neurokinin (NK) B, cholecystokinin (CCK), and corticotropin-releasing factor (CRF) and their receptors to neuronal types using patch-clamp and single-cell reverse transcription-polymerase chain reaction in acute slices of rat neocortex. Classification of neurons by unsupervised clustering based on the analysis of multiple electrophysiological and molecular properties disclosed 3 GABAergic interneuron clusters and 1 pyramidal cell cluster. The 3 neuropeptides were expressed in a cluster of interneurons characteristically expressing vasoactive intestinal peptide. CRF was additionally found in a cluster containing almost exclusively somatostatin-expressing interneurons, whereas CCK was present in all clusters. The respective receptors of these peptides, NK-3, CCK-B, and CRF-1, were essentially expressed in pyramidal cells. At -60 mV, pyramidal cells were weakly depolarized by each of these peptides. When pyramidal neurons were maintained to about 5 mV below spike threshold, depolarization induced by each peptide resulted in a long-lasting action potential discharge. Neuropeptide effects were prevented by selective antagonists of NK-3, CCK-B, and CRF-1 receptors. These results suggest that pyramidal neurons are the primary target of NKB, CCK, and CRF in the neocortex. They further indicate that specific interneuron types coordinate the release of these peptides and can induce a long-lasting increase of the excitability of the neocortical network.
Collapse
Affiliation(s)
- Thierry Gallopin
- Laboratoire de Neurobiologie et Diversité Cellulaire, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7637, Ecole Supérieure de Physique et de Chimie Industrielles, 10 rue Vauquelin, 75005 Paris, France
| | | | | | | |
Collapse
|
28
|
Duman CH, Duman RS. Neurobiology and treatment of anxiety: signal transduction and neural plasticity. Handb Exp Pharmacol 2005:305-34. [PMID: 16594263 DOI: 10.1007/3-540-28082-0_11] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The stress-dependence and chronic nature of anxiety disorders along with the anxiolytic effectiveness of antidepressant drugs suggests that neuronal plasticity may play a role in the pathophysiology of anxiety. Intracellular signaling pathways are known in many systems to be critical links in the cascades from surface signals to the molecular alterations that result in functional plasticity. Chronic antidepressant treatments can regulate intracellular signaling pathways and can induce molecular, cellular, and structural changes over time. These changes may be important to the anxiolytic effectiveness of these drugs. In addition, the signaling proteins implicated in the actions of chronic antidepressant action, such as cAMP response element binding protein (CREB), have also been implicated in conditioned fear and in anxiety. The cellular mechanisms underlying conditioned fear indicate roles for additional signaling pathways; however, less is known about such mechanisms in anxiety. The challenge to identify intracellular signaling pathways and related molecular and structural changes that are critical to the etiology and treatment of anxiety will further establish the importance of mechanisms of neuronal plasticity in functional outcome and improve treatment strategies.
Collapse
Affiliation(s)
- C H Duman
- Laboratory of Molecular Psychiatry, Departments of Psychiatry and Pharmacology, Yale University School of Medicine, 34 Park Street, New Haven CT, 06508, USA
| | | |
Collapse
|
29
|
Maya-Núñez G, Castro-Fernández C, Méndez JP. CRH-stimulation of cyclic adenosine 5'-monophosphate pathway is partially inhibited by the coexpression of CRH-R1 and CRH-R2alpha. Endocrine 2005; 27:67-73. [PMID: 16077174 DOI: 10.1385/endo:27:1:067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2005] [Revised: 05/18/2005] [Accepted: 06/02/2005] [Indexed: 01/21/2023]
Abstract
Corticotropin-releasing hormone (CRH) is one of the major proteins responsible for brain stress regulation. Two well-known receptors have been described: type 1 and type 2alpha, both members of the receptor superfamily of G protein-coupled receptors (GPCR). We investigated receptor regulation when both CRH receptor subtypes are coexpressed in the same mammalian cell line. When both types of receptors are coexpressed, cAMP second messenger production is partially inhibited compared to when receptors are expressed separately. However, neither binding kinetics nor internalization rates are modified by coexpression of these receptors. To our knowledge this is the first demonstration of receptor interaction that results in the modification of CRH-mediated signal transduction pathway. Because CRH-R1 and CRH-R2alpha have overlapping mRNA expression patterns in the brain, these receptors may be coexpressed in neurons, suggesting that receptor interaction may play an important role in the effect evoked by CRH, contributing to the complexity of differential coupling of the CRH receptors in different endocrine and stress behavior responses.
Collapse
Affiliation(s)
- G Maya-Núñez
- Research Unit in Developmental Biology, Hospital de Pediatría, Centro Médico Nacional Siglo SXXI, Instituto Mexicano del Seguro Social, Mexico
| | | | | |
Collapse
|
30
|
Lyubimov YE, Emel'yanov NA, Izvarina NL. The effects of corticotropin-releasing factor on anoxia-induced changes in evoked potentials in living slices of rat olfactory cortex. ACTA ACUST UNITED AC 2004; 34:557-61. [PMID: 15368900 DOI: 10.1023/b:neab.0000028284.55405.8f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Experiments were performed to address the dynamics of evoked focal potentials in slices of rat olfactory cortex during 10-min anoxia and subsequent reoxygenation. These experiments showed that perfusion with corticotropin-releasing factor (CRF)--corticoliberin--at concentrations of 1, 10, and 100 nM had no effect on changes in EPSP parameters before or during anoxia. However, CRF (10 and 100 nM) significantly aided recovery of the amplitude and slope of EPSP during reoxygenation. Application of the competitive NMDA receptor blocker APV (50 microM) during reoxygenation did not eliminate the protective effects of CRF on neuronal activity.
Collapse
Affiliation(s)
- Ya E Lyubimov
- Laboratory for the Regulation of Brain Neuron Function, I. P. Pavlov Institute of Physiology, Russian Academy of Sciences, 6 Makarov Bank, 199034 St. Petersburg, Russia
| | | | | |
Collapse
|
31
|
Sheehan TP, Chambers RA, Russell DS. Regulation of affect by the lateral septum: implications for neuropsychiatry. ACTA ACUST UNITED AC 2004; 46:71-117. [PMID: 15297155 DOI: 10.1016/j.brainresrev.2004.04.009] [Citation(s) in RCA: 402] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2004] [Indexed: 11/17/2022]
Abstract
Substantial evidence indicates that the lateral septum (LS) plays a critical role in regulating processes related to mood and motivation. This review presents findings from the basic neuroscience literature and from some clinically oriented research, drawing from behavioral, neuroanatomical, electrophysiological, and molecular studies in support of such a role, and articulates models and hypotheses intended to advance our understanding of these functions. Neuroanatomically, the LS is connected with numerous regions known to regulate affect, such as the hippocampus, amygdala, and hypothalamus. Through its connections with the mesocorticolimbic dopamine system, the LS regulates motivation, both by stimulating the activity of midbrain dopamine neurons and regulating the consequences of this activity on the ventral striatum. Evidence that LS function could impact processes related to schizophrenia and other psychotic spectrum disorders, such as alterations in LS function following administration of antipsychotics and psychotomimetics in animals, will also be presented. The LS can also diminish or enable fear responding when its neural activity is stimulated or inhibited, respectively, perhaps through its projections to the hypothalamus. It also regulates behavioral manifestations of depression, with antidepressants stimulating the activity of LS neurons, and depression-like phenotypes corresponding to blunted activity of LS neurons; serotonin likely plays a key role in modulating these functions by influencing the responsiveness of the LS to hippocampal input. In conclusion, a better understanding of the LS may provide important and useful information in the pursuit of better treatments for a wide range of psychiatric conditions typified by disregulation of affective functions.
Collapse
Affiliation(s)
- Teige P Sheehan
- Department of Psychology, Brown University, P.O. Box 1853, Providence, RI 02912, USA.
| | | | | |
Collapse
|
32
|
Bayatti N, Zschocke J, Behl C. Brain region-specific neuroprotective action and signaling of corticotropin-releasing hormone in primary neurons. Endocrinology 2003; 144:4051-60. [PMID: 12933679 DOI: 10.1210/en.2003-0168] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CRH regulates the body's response to stressful stimuli by modulating the activity of the hypothalamic pituitary axis. In primary cultures and cell lines, CRH also acts as a potent neuroprotective factor in response to a number of toxins. Using primary neuronal cultures from the cerebellum, cerebral cortex, and hippocampus, we demonstrate that CRH exerts a brain region-specific neuroprotective effect on amyloid beta 25-35 toxicity. At low CRH concentrations (10(-8) M), neuroprotective effects can be observed only in cerebellar and hippocampal cultures, but a higher CRH concentration (10(-7) M) additionally led to the protection of cortical neurons. These neuroprotective effects were inhibited by H89, a specific protein kinase A inhibitor. Western blot analysis, carried out using phospho-specific antibodies directed against MAPK, cAMP response element-binding protein (CREB), and glycogen synthase kinase (GSK)3 beta also resulted in brain legion-specific differences regarding intracellular signaling. Correlating with cell survival, low CRH concentrations resulted in activation of the CREB pathway and inactivation of GSK3 beta in cerebellar and hippocampal cultures, but higher concentrations additionally resulted in activated CREB and inactivated GSK3 beta in cortical cultures. In contrast, MAPK activation occurred only in cortical neurons. Differences in signaling were found to be independent of receptor expression levels because RT-PCR analysis indicated no region-specific differences in CRHR1 mRNA expression.
Collapse
Affiliation(s)
- Nadhim Bayatti
- Independent Research Group Neurodegeneration, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Chemistry and Pathobiochemistry, Johannes Gutenberg University Mainz, 55099 Mainz, Germany
| | | | | |
Collapse
|
33
|
Kasckow J, Mulchahey JJ, Aguilera G, Pisarska M, Nikodemova M, Chen HC, Herman JP, Murphy EK, Liu Y, Rizvi TA, Dautzenberg FM, Sheriff S. Corticotropin-releasing hormone (CRH) expression and protein kinase A mediated CRH receptor signalling in an immortalized hypothalamic cell line. J Neuroendocrinol 2003; 15:521-9. [PMID: 12694378 DOI: 10.1046/j.1365-2826.2003.01026.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Corticotropin-releasing hormone (CRH) is a 41 amino acid neuropeptide which plays an important role in the stress response in the hypothalamus. We describe the development of an immortalized hypothalamic cell line which expresses CRH. We hypothesized that this cell line would possess the relevant characteristics of parvocellular CRH-expressing neurones such as glucocorticoid receptor (GR) expression and vasopressin (VP) coexpression. For production of hypothalamic cells, embryonic day 19 rat pup hypothalami were dissected and dissociated into tissue culture dishes. They were immortalized by retrovirus-mediated transfer of the SV40 large T antigen gene at 3 days of culture and then screened for expression of CRH following dilution cloning. One cell line was chosen (IVB) which exhibited CRH-like immunoreactivity (CRH-LI) and expressed CRH, VP and CRH1 receptor RNA via the reverse transcriptase-polymerase chain reaction. In addition, the cell line expressed the neuronal marker, microtubule-associated protein-2. We verified that the CRH-LI from IVB cell lysates coeluted with CRH standard via reversed-phase high-performance liquid chromatography (HPLC). Furthermore, oxidation of the lysate converted its HPLC profile to that identical with oxidized CRH standard. In addition, IVB cells exhibited high affinity binding to CRH. Incubation of IVB cells with CRH lead to increases in cAMP levels and protein kinase A activity in a concentration-dependent manner. Incubation of IVB cells with CRH also resulted in increases in phospho-cyclic-AMP response element binding protein (CREB) immunostaining as detected by immunocytochemical analysis. Finally, CRH treatment of IVB cell lines has been linked to CREB-mediated gene expression as determined via the PathDetect CREB trans-reporting system. The characteristics of IVB cells, such as CRH and VP coexpression, GR expression and a biologically active CRH-R1-mediated signalling pathway, suggest that this neuronal cell line may serve as model of parvocellular CRH neurones.
Collapse
Affiliation(s)
- J Kasckow
- Cincinnati VAMC, Psychiatry Service (116A), and School of Medicine, Department of Psychiatry, and Neurosciences Program, University of Cincinnati, OH 45267-0559, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ladds G, Davis K, Hillhouse EW, Davey J. Modified yeast cells to investigate the coupling of G protein-coupled receptors to specific G proteins. Mol Microbiol 2003; 47:781-92. [PMID: 12535076 DOI: 10.1046/j.1365-2958.2003.03336.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
G protein-coupled receptors (GPCRs) help to regulate the physiology of all the major organ systems. They respond to a multitude of ligands and activate a range of effector proteins to bring about the appropriate cellular response. The choice of effector is largely determined by the interaction of individual GPCRs with different G proteins. Several factors influence this interaction, and a better understanding of the process may enable a more rational approach to identifying compounds that affect particular signalling pathways. A number of systems have been developed for the analysis of GPCRs. All provide useful information, but the genetic amenability and relative simplicity of yeast makes them a particularly attractive option for ligand identification and pharmaceutical screening. Many, but not all, GPCRs are functional in the budding yeast Saccharomyces cerevisiae, and we have developed reporter strains of the fission yeast Schizosaccharomyces pombe as an alternative host. To provide a more generic system for investigating GPCRs, we created a series of yeast-human Galpha-transplants, in which the last five residues at the C-terminus of the yeast Galpha-subunit are replaced with the corresponding residues from different human G proteins. These enable GPCRs to be coupled to the Sz. pombe signalling machinery so that stimulation with an appropriate ligand induces the expression of a signal-dependent lacZ reporter gene. We demonstrate the specificity of the system using corticotropin releasing factor (CRF) and CRF-related peptides on two CRF receptors. We find that different combinations of ligand and receptor activate different Galpha-transplants, and the specificity of the coupling is similar to that in mammalian systems. Thus, CRF signalled through the Gs- and Gi-transplants, consistent with its regulation of adenylate cyclase, and was more active against the CRF-R1A receptor than against the CRF-R2B receptor. In contrast, urocortin II and urocortin III were selective for the CRF-R2B receptors. Furthermore, urocortin, but not CRF, induced signalling through the CRF-R1A receptor and the Gq-transplant. This is the first time that human GPCRs have been coupled to the signalling pathway in Sz. pombe, and the strains described in this study will complement the other systems available for studying this important family of receptors.
Collapse
Affiliation(s)
- Graham Ladds
- Department of Biological Sciences, University of Warwick, Coventry, UK
| | | | | | | |
Collapse
|
35
|
Corticotropin-releasing factor receptors couple to multiple G-proteins to activate diverse intracellular signaling pathways in mouse hippocampus: role in neuronal excitability and associative learning. J Neurosci 2003. [PMID: 12533630 DOI: 10.1523/jneurosci.23-02-00700.2003] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Corticotropin-releasing factor (CRF) exerts a key neuroregulatory control on stress responses in various regions of the mammalian brain, including the hippocampus. Using hippocampal slices, extracts, and whole animals, we investigated the effects of human/rat CRF (h/rCRF) on hippocampal neuronal excitability and hippocampus-dependent learning in two mouse inbred strains, BALB/c and C57BL/6N. Intracellular recordings from slices revealed that application of h/rCRF increased the neuronal activity in both mouse inbred strains. Inhibition of protein kinase C (PKC) by bisindolylmaleimide I (BIS-I) prevented the h/rCRF effect only in hippocampal slices from BALB/c mice but not in slices from C57BL/6N mice. Inhibition of cAMP-dependent protein kinase (PKA) by H-89 abolished the h/rCRF effect in slices from C57BL/6N mice, with no effect in slices from BALB/c mice. Accordingly, h/rCRF elevated PKA activity in hippocampal slices from C57BL/6N mice but increased only PKC activity in the hippocampus of BALB/c mice. These differences in h/rCRF signal transduction were also observed in hippocampal membrane suspensions from both mouse strains. In BALB/c mice, hippocampal CRF receptors coupled to G(q/11) during stimulation by h/rCRF, whereas they coupled to G(s), G(q/11), and G(i) in C57BL/6N mice. As expected on the basis of the slice experiments, h/rCRF improved context-dependent fear conditioning of BALB/c mice in behavioral experiments, and BIS-I prevented this effect. However, although h/rCRF increased neuronal spiking in slices from C57BL/6N mice, it did not enhance conditioned fear. These results indicate that the CRF system activates different intracellular signaling pathways in mouse hippocampus and may have distinct effects on associative learning depending on the mouse strain investigated.
Collapse
|
36
|
Rebaudo R, Melani R, Balestrino M, Izvarina N. Electrophysiological effects of sustained delivery of CRF and its receptor agonists in hippocampal slices. Brain Res 2001; 922:112-7. [PMID: 11730708 DOI: 10.1016/s0006-8993(01)03160-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The corticotropin-releasing factor (CRF) is a hypothalamic peptide that regulates the release of adrenocorticotropic hormone (ATCH) and of beta-endorphin. It has been suggested that it modulates learning and memory processes in rat. However, the electrophysiological effects that CRF produces on hippocampal neurons have been so far little investigated. In particular, the effects of CRF on long-term potentiation (LTP), a phenomenon which is thought to be the substrate of memory processes, are unknown. We studied the effects of sustained administration of CRF and of two of its receptor agonists on basal neuronal activity and on in vitro hippocampal LTP. The two receptor agonists were D-Glu-20-CRF and D-Pro-5-CRF, selective for the CRF-R1 and the CRF-R2 receptors, respectively. We found that CRF, D-Pro-5-CRF and D-Glu-20-CRF at the concentration of 1 nM diminish the amplitude of hippocampal population spike and prevent the onset of LTP. Higher concentrations of CFR have less depressing effects on neuronal activity, yet they still prevent the occurrence of LTP.
Collapse
Affiliation(s)
- R Rebaudo
- Department of Neurological and Vision Sciences, University of Genoa, Via De Toni 5, 16132 Genoa, Italy
| | | | | | | |
Collapse
|
37
|
Pohl S, Darlison MG, Clarke WC, Lederis K, Richter D. Cloning and functional pharmacology of two corticotropin-releasing factor receptors from a teleost fish. Eur J Pharmacol 2001; 430:193-202. [PMID: 11711031 DOI: 10.1016/s0014-2999(01)01391-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Although it is well established that fish possess corticotropin-releasing factor (CRF) and a CRF-like peptide, urotensin I, comparatively little is known about the pharmacology of their cognate receptors. Here we report the isolation and functional expression of two complementary DNAs (cDNAs), from the chum salmon Oncorhynchus keta, which encode orthologues of the mammalian and amphibian CRF type 1 (CRF(1)) and type 2 (CRF(2)) receptors. Radioligand competition binding experiments have revealed that the salmon CRF(1) and CRF(2) receptors bind urotensin I with approximately 8-fold higher affinity than rat/human CRF. These two peptides together with two related CRF-like peptides, namely, sauvagine and urocortin, were also tested in cAMP assays; for cells expressing the salmon CRF(1) receptor, EC(50) values for the stimulation of cAMP production were between 4.5+/-1.8 and 15.3+/-3.1 nM. For the salmon CRF(2) receptor, the corresponding values were: rat/human CRF, 9.4+/-0.4 nM; urotensin I, 21.2+/-2.1 nM; sauvagine, 0.7+/-0.1 nM; and urocortin, 2.2+/-0.7 nM. We have also functionally coupled the O. keta CRF(1) receptor, in Xenopus laevis oocytes, to the endogenous Ca(2+)-activated chloride conductance by co-expression with the G-protein alpha subunit, G(alpha16). The EC(50) value for channel activation by rat/human CRF (11.2+/-2.6 nM) agrees well with that obtained in cAMP assays (15.3+/-3.1 nM). We conclude that although sauvagine is 13- and 30-fold more potent than rat/human CRF and urotensin I, respectively, in activating the salmon CRF(2) receptor, neither receptor appears able to discriminate between the native ligands CRF and urotensin I.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Binding, Competitive/drug effects
- Cell Line
- Cloning, Molecular
- Corticotropin-Releasing Hormone/metabolism
- Corticotropin-Releasing Hormone/pharmacology
- Cyclic AMP/metabolism
- DNA, Complementary/chemistry
- DNA, Complementary/genetics
- Dose-Response Relationship, Drug
- Female
- Gene Expression
- Humans
- Male
- Membrane Potentials/drug effects
- Molecular Sequence Data
- Oncorhynchus keta/genetics
- Oocytes
- Phylogeny
- RNA/genetics
- RNA/metabolism
- Rats
- Receptors, Corticotropin-Releasing Hormone/drug effects
- Receptors, Corticotropin-Releasing Hormone/genetics
- Receptors, Corticotropin-Releasing Hormone/metabolism
- Sequence Alignment
- Sequence Analysis, DNA
- Sequence Homology, Amino Acid
- Tissue Distribution
- Urotensins/metabolism
- Urotensins/pharmacology
- Xenopus laevis
Collapse
Affiliation(s)
- S Pohl
- Institut für Zellbiochemie und Klinische Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Universität Hamburg, Martinistrasse 52, 20246, Hamburg, Germany
| | | | | | | | | |
Collapse
|
38
|
Onali P, Olianas MC. Beta gamma-mediated enhancement of corticotropin-releasing hormone-stimulated adenylyl cyclase activity by activation of gamma-aminobutyric acid(B) receptors in membranes of rat frontal cortex. Biochem Pharmacol 2001; 62:183-90. [PMID: 11389876 DOI: 10.1016/s0006-2952(01)00659-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A number of studies have shown that activation of gamma-aminobutyric acid(B) (GABA(B)) receptors potentiates neurotransmitter-induced accumulation of cyclic AMP in brain slices, but the mechanisms involved in the facilitatory effect have not been fully elucidated. In the present study, we showed that in membranes of rat frontal cortex the GABA(B) receptor agonist (-)baclofen increased basal adenylyl cyclase activity and potentiated the maximal enzyme stimulation elicited by corticotropin-releasing hormone (CRH). The less active enantiomer (+)baclofen had no effect on cyclic AMP formation, whereas the natural agonist GABA mimicked the stimulatory action of (-)baclofen. In radioligand-binding experiments, the affinity and maximal binding capacity of (125)I-Tyr-CRH was not affected by (-)baclofen. The GABA(B) receptor antagonist CGP 55845A competitively counteracted the (-)baclofen potentiation of CRH-stimulated adenylyl cyclase activity with a pA(2) value of 6.70. Moreover, both (-)baclofen and GABA, but not (+)baclofen, caused a concentration-dependent stimulation of [(35)S]GTP gamma S binding to membrane G-proteins. The intracerebral injection of pertussis toxin significantly reduced the facilitatory effects of (-)baclofen on both basal and CRH-stimulated adenylyl cyclase activities. Moreover, membrane incubation with the GDP-bound form of the alpha subunit of transducin, a scavenger of G protein beta gamma subunits, blocked the stimulatory effects of (-)baclofen. The data indicate that in rat frontal cortex activation of GABA(B) receptors potentiates the CRH stimulation of adenylyl cyclase activity through a mechanism involving the beta gamma subunits of the pertussis toxin-sensitive G protein G(i)/G(o).
Collapse
Affiliation(s)
- P Onali
- Section of Biochemical Pharmacology, Department of Neuroscience, University of Cagliari, 09124 Cagliari, Italy.
| | | |
Collapse
|
39
|
Cibelli G, Corsi P, Diana G, Vitiello F, Thiel G. Corticotropin-releasing factor triggers neurite outgrowth of a catecholaminergic immortalized neuron via cAMP and MAP kinase signalling pathways. Eur J Neurosci 2001; 13:1339-48. [PMID: 11298794 DOI: 10.1046/j.0953-816x.2001.01510.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Corticotropin-releasing factor (CRF), a neuropeptide of 41 amino acids, acts as the major physiological regulator of the basal and stress-induced release of corticotropin (ACTH), beta-endorphin and other proopiomelanocortin-derived peptides from the anterior pituitary gland. In addition to its endocrine activity, CRF displays extrahypophysiotropic effects, mainly as a regulator of stress responses. We show here that CRF may additionally function as a differentiating factor in immortalized noradrenergic neuronal CATH.a cells that express CRF receptor type I and resemble locus coeruleus-derived neurons. CRF triggers morphological changes in CATH.a cells including the appearance of extended long, slender neurites with prominent growth cones. CRF-treated CATH.a cells exhibit a morphology similar to locus coeruleus neurons in primary culture. CRF-induced neurite outgrowth of CATH.a cells was blocked by addition of inhibitors for cAMP-dependent protein kinase or extracellular signal-regulated protein kinase (ERK), a subtype of the mitogen-activated protein kinases. The participation of ERK within the CRF signalling cascade was further confirmed by Western blot experiments, with antibodies directed against the phosphorylated form of ERK, and also with transcription-based assays. We conclude that CRF functions as a differentiating factor of CATH.a cells via the cAMP and the MAP kinase signalling pathways.
Collapse
Affiliation(s)
- G Cibelli
- Department of Pharmacology and Human Physiology, Medical Faculty, University of Bari, I-70124 Bari, Italy
| | | | | | | | | |
Collapse
|
40
|
King JS, Madtes P, Bishop GA, Overbeck TL. The distribution of corticotropin-releasing factor (CRF), CRF binding sites and CRF1 receptor mRNA in the mouse cerebellum. PROGRESS IN BRAIN RESEARCH 2001; 114:55-66. [PMID: 9193138 DOI: 10.1016/s0079-6123(08)63358-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The purpose of the present study is to determine the distribution of CRF containing afferents, and correlate these findings with the distribution of CRF binding sites and the neuronal localization of mRNA for the CRF1 receptor in the cerebellum of a single species, the mouse. Corticotropin releasing factor (CRF) has been localized within climbing fibers and mossy fibers throughout the cerebellar cortex of the mouse using immunohistochemistry. CRF immunoreactive, axonal varicosities also are present within all four of the cerebellar nuclei. 125I-labeled CRF binding sites are evident throughout all three layers of the cerebellar cortex (molecular, Purkinje and granule cell layers), but are not seen within the cerebellar nuclei. In situ hybridization histochemistry was employed using an antisense riboprobe corresponding to the full length sequence of the rat mRNA for the CRF1 receptor. Positive signal is present throughout the cerebellum in Purkinje cells and the granule cell layer. CRF1 receptor mRNA also is expressed within all four of the cerebellar nuclei. Further experiments are required to reconcile the lack of CRF binding sites in the cerebellar nuclei with the positive mRNA receptor expression and the presence of immunoreactive axonal varicosities. In previous physiological experiments, iontophoretic application of CRF enhances spontaneous as well as quisqualate-induced activity of Purkinje cells in slice preparations of the mouse cerebellum. When the results of the anatomical techniques are compared to the physiological data, there is convergent evidence to suggest that CRF influences the firing rate or responsiveness of Purkinje cells directly via release of the peptide from the climbing fiber system and indirectly via the mossy fiber-granule cell-parallel fiber circuit. Taken together, these anatomical and physiological data provide strong evidence to suggest that, in the adult cerebellum, CRF functions as a neuromodulator.
Collapse
Affiliation(s)
- J S King
- Department of Cell Biology, Neurobiology and Anatomy, Ohio State University, Columbus 43210, USA
| | | | | | | |
Collapse
|
41
|
Grammatopoulos DK, Randeva HS, Levine MA, Kanellopoulou KA, Hillhouse EW. Rat cerebral cortex corticotropin-releasing hormone receptors: evidence for receptor coupling to multiple G-proteins. J Neurochem 2001; 76:509-19. [PMID: 11208914 DOI: 10.1046/j.1471-4159.2001.00067.x] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The wide distribution of corticotrophin-releasing hormone (CRH) receptors in brain and periphery appear to be important in integrating the responses of the brain, endocrine and immune systems to physiological, psychological and immunological stimuli. The type 1 receptors are highly expressed throughout the cerebral cortex, a region involved in cognitive function and modulation of stress responses, where they are coupled to the adenylyl cyclase system. Using techniques that analyse receptor-mediated guanine-nucleotide binding protein (G-proteins) activation, we recently demonstrated that expressed type 1alpha CRH receptors are capable of activating multiple G-proteins, which suggests that CRH can regulate multiple signalling pathways. In an effort to characterize the intracellular signals generated by CRH in the rat cerebral cortex we sought to identify G-proteins activated by CRH in a physiological membrane environment. Rat cerebral cortical membrane suspensions were analysed for the ability of CRH to stimulate incorporation of [alpha-32P]-GTP-gamma-azidoanilide to various G-protein alpha-chains. Our results show that CRH receptors are coupled to and activate at least five different G-proteins (Gs, Gi, Gq/11, Go and Gz) with subsequent stimulation of at least two intracellular signalling cascades. In addition, the photoaffinity experiments indicated that the CRH receptors preferentially activate the 45 kDa form of the Gs alpha-protein. This data may help elucidate the intracellular signalling pathways mediating the multiple actions of CRH especially under different physiological conditions.
Collapse
Affiliation(s)
- D K Grammatopoulos
- Sir Quinton Hazell Molecular Medicine Research Centre, Department of Biological Sciences, The University of Warwick, Coventry, UK.
| | | | | | | | | |
Collapse
|
42
|
Engler D, Redei E, Kola I. The corticotropin-release inhibitory factor hypothesis: a review of the evidence for the existence of inhibitory as well as stimulatory hypophysiotropic regulation of adrenocorticotropin secretion and biosynthesis. Endocr Rev 1999; 20:460-500. [PMID: 10453355 DOI: 10.1210/edrv.20.4.0376] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- D Engler
- Laboratory of Molecular Genetics and Development, Institute of Reproduction and Development, Monash Medical Centre, Clayton, Victoria, Australia
| | | | | |
Collapse
|
43
|
Thiel G, Cibelli G. Corticotropin-releasing factor and vasoactive intestinal polypeptide activate gene transcription through the cAMP signaling pathway in a catecholaminergic immortalized neuron. Neurochem Int 1999; 34:183-91. [PMID: 10355485 DOI: 10.1016/s0197-0186(98)00086-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Corticotropin-releasing factor (CRF) and vasoactive intestinal polypeptide (VIP) are neuropeptides displaying a variety of short-term effects in the nervous system. It is shown here in transfection experiments of an immortalized noradrenergic locus coeruleus-like cell line that both CRF and VIP also trigger a signaling cascade capable of activating gene transcription. To elucidate the signaling pathway leading to transcriptional induction, cells were transfected with an inhibitor for cAMP-dependent protein kinase, targeted to the nucleus via a nuclear-localization signal. Transcriptional induction of a reporter gene by CRF and VIP was blocked in these cells, indicating that the cAMP-dependent protein kinase is required for transducing CRF and VIP generated signals into the nucleus. Additionally, transfection experiments with a reporter gene containing cAMP response elements in its regulatory region demonstrate that CRF and VIP receptor activation induce transcription through this genetic regulatory element. We conclude that long-term effects of CRF and VIP in neurons are likely to be mediated by the transcriptional regulation of CRF and VIP-responsive genes via the cAMP signaling pathway.
Collapse
Affiliation(s)
- G Thiel
- Medical Biochemistry and Molecular Biology, University of the Saarland, Medical School, Homburg, Germany.
| | | |
Collapse
|
44
|
Schilling L, Kanzler C, Schmiedek P, Ehrenreich H. Characterization of the relaxant action of urocortin, a new peptide related to corticotropin-releasing factor in the rat isolated basilar artery. Br J Pharmacol 1998; 125:1164-71. [PMID: 9863643 PMCID: PMC1565693 DOI: 10.1038/sj.bjp.0702182] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
In addition to its well established neuroendocrine and neurotransmitter effects, corticotropin releasing factor (CRF) exerts a potent vasorelaxant action. Recently, a CRF-related peptide, urocortin, has been identified in the mammalian brain. In the present study, the cerebral vasomotor action of this peptide and the mechanism underlying its relaxant effect are characterized. Ring segments obtained from the rat basilar artery were used for measurement of isometric force. The relaxant action of urocortin, CRF and sauvagine was studied in segments with a functionally intact endothelium. In segments precontracted with prostaglandin F2alpha, urocortin, CRF and sauvagine induced concentration-related relaxation. The order of potency was as follows (pD2+/-s.e.m. given in brackets): urocortin (9.32+/-0.07) > sauvagine (9.08+/-0.08) > CRF (7.50+/-0.07). Complete relaxation was achieved with each agonist. Relaxation was not affected by removal of the endothelium but was markedly attenuated in segments precontracted with 50 mM K+ Krebs solution. The relaxant effect of urocortin was inhibited by astressin in an apparently competitive manner. A pA2 value of 7.52 was estimated for astressin. Inhibition of urocortin-induced relaxation was also observed in the presence of the adenylate cyclase inhibitor SQ22536 (pD2 in the presence of 300 microM SQ22536, 9.36+/-0.05) and the K+ channel blockers tetraethylammonium (10 mM; pD2, 8.65+/-0.07), iberiotoxin (100 nM; pD2, 8.88+/-0.08) and apamin (10 nM; pD2, 8.94+/-0.07). However, the inhibitory actions of SQ22536 and apamin or iberiotoxin were not additive. The results suggest that urocortin induces relaxation of cerebral arteries by activating CRF-R2 receptors present in the vascular wall. Relaxation appears to be mediated by adenylate cyclase stimulation and activation of Ca2+-dependent K+ channels.
Collapse
Affiliation(s)
- L Schilling
- Department of Neurosurgery, Faculty of Clinical Medicine Mannheim, University Heidelberg, Germany
| | | | | | | |
Collapse
|
45
|
Asaba K, Makino S, Hashimoto K. Effect of urocortin on ACTH secretion from rat anterior pituitary in vitro and in vivo: comparison with corticotropin-releasing hormone. Brain Res 1998; 806:95-103. [PMID: 9739115 DOI: 10.1016/s0006-8993(98)00747-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Both urocortin (UCN) and corticotropin-releasing hormone (CRH) are known to stimulate secretion of adrenocorticotropic hormone (ACTH) by corticotroph cells via type-1 corticotropin-releasing hormone receptor (CRHR-1). We extensively examined UCN effects on the anterior pituitary (AP), particularly on proopiomelanocortin (POMC) mRNA and CRHR-1 mRNA as well as ACTH secretion in vivo. Moreover, signal transduction with UCN exposure was assessed in AP cell cultures in comparison with transduction following CRH exposure. Intravenously administered of UCN (5 microg/kg) increased ACTH and corticosterone secretion. Similarly, intravenous administration of UCN increased POMC mRNA and decreased CRHR-1 mRNA in the AP. These UCN effects were more potent and long-lasting than those of CRH. The prominent effect of UCN on ACTH secretion in vivo was confirmed in AP cell cultures, where application of UCN stimulated ACTH release approximately 7 times more strongly than CRH. The effect of UCN on ACTH release was enhanced by phorbol esters which activate protein kinase C, but was reduced by the selective cAMP-dependent protein kinase inhibitor, H-89. These results suggest that, as with CRH, UCN stimulates ACTH production and/or release through cAMP-dependent mechanisms, and that protein kinase C-dependent mechanism has a synergistic effect upon UCN-induced ACTH release. The more potent effects of UCN relative to CRH may be attributable to UCN's higher affinity for CRHR-1.
Collapse
Affiliation(s)
- K Asaba
- Second Department of Internal Medicine, Kochi Medical School, Okoh-cho, Nankoku-city, Kochi 783-8505, Japan
| | | | | |
Collapse
|
46
|
Lehnert H, Schulz C, Dieterich K. Physiological and neurochemical aspects of corticotropin-releasing factor actions in the brain: the role of the locus coeruleus. Neurochem Res 1998; 23:1039-52. [PMID: 9704593 DOI: 10.1023/a:1020751817723] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Corticotropin-releasing factor (CRF) is both a major regulator of the hypothalamo-pituitary-adrenal (HPA) axis and the activity of the autonomic nervous system. Besides, it exerts numerous effects on other physiological functions such as appetite control, motor and cognitive behavior and immune function. The basis for these effects is constituted by its distribution in hypothalamic and extra-hypothalamic brain areas, the latter being represented by limbic structures such as the central nucleus of the amygdala or by brain stem neurons such as the locus coeruleus (LC) or nucleus of the solitary tract (NTS). The effects of CRF are mediated through recently described CRF-receptor subtypes, whose molecular biology, biochemistry and pharmacological regulation are discussed in detail. In the second part of this review, we will focus on the physiology of CRF-systems in the brain, with a particular emphasis on cardiovascular regulation, respiration, appetite control and stress-related behavior. Finally, the role of the locus coeruleus in the control of CRF-mediated behavioral activities is discussed. The interaction of noradrenergic and CRF-neurons clearly implies that CRF appears to directly activate LC neurons in a stressful situation, thus ultimately coordinating the bodily response to a stressful stimulus.
Collapse
Affiliation(s)
- H Lehnert
- Dept. of Endocrinology and Metabolism University Hospital of Magdeburg, Germany.
| | | | | |
Collapse
|
47
|
Grammatopoulos D, Hillhouse EW. Solubilization and biochemical characterization of the human myometrial corticotrophin-releasing hormone receptor. Mol Cell Endocrinol 1998; 138:185-98. [PMID: 9685227 DOI: 10.1016/s0303-7207(97)00238-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
We have solubilized an active form of the myometrial corticotrophin-releasing hormone (CRH) receptor using 1% w/v digitonin. The solubilized receptor retains its capacity for high-affinity binding as demonstrated by Scatchard analysis, although there was a shift in dissociation constant (Kd) from 83.6 +/- 15-195 +/- 35 pM for the membrane-bound and soluble receptor respectively. There was no difference in the maximum binding site concentrations (Bmax) of 13 +/- 5 and 21.5 +/- 6 fmol/mg protein for the membrane-bound and soluble receptor respectively. Sauvagine unlike CRH had no effect on radiolabeled CRH binding which suggests that the CRH-R2 receptor is not present in the myometrium. The solubilized receptor did not retain guanine-nucleotide sensitivity. The isoelectric focusing (IEF) profile of the human myometrial CRH receptors was significantly different from that of the rat cerebral cortex. Furthermore, solubilization of human myometrial membrane proteins followed by gel filtration and SDS-PAGE revealed a specifically labeled protein with an apparent molecular weight of 42000-47000 kDa. Our results suggest that during solubilization the human myometrial CRH receptor is dissociated from the guanine nucleotide-binding protein (Gs) and that high affinity binding for soluble CRH receptors is not dependent on the coupling of a guanine nucleotide-binding protein.
Collapse
Affiliation(s)
- D Grammatopoulos
- Department of Biological Sciences, The University of Warwick, Coventry, UK
| | | |
Collapse
|
48
|
Birnbaum SG, Davis M. Modulation of the acoustic startle reflex by infusion of corticotropin-releasing hormone into the nucleus reticularis pontis caudalis. Brain Res 1998; 782:318-23. [PMID: 9519280 DOI: 10.1016/s0006-8993(97)01309-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The amplitude of the acoustic startle reflex can be modulated by exposure to aversive stimuli or other conditions which evoke a state of fear. The neurotransmitters involved in this modulation are currently being investigated. Unilateral local infusion of corticotropin-releasing hormone (CRH; 0, 10, 20, 40 and 80 ng) into the nucleus reticularis pontis caudalis (PnC), an obligatory synapse in the acoustic startle reflex, significantly elevated startle amplitude in a dose-dependent manner. The facilitation of startle began immediately following infusion, reached asymptote approximately 20-25 min later, and persisted throughout the remaining 60 min test session. This CRH-enhanced startle effect was blocked by infusion of the CRH antagonist, alpha-helical CRH9-41, immediately prior to CRH infusion. These results support an involvement of CRH at the level of the PnC in modulating the acoustic startle reflex.
Collapse
Affiliation(s)
- S G Birnbaum
- Interdept. Neuroscience Program, Yale University School of Medicine, Connecticut Mental Health Center, New Haven 06508, USA.
| | | |
Collapse
|
49
|
Walker DL, Cassella JV, Lee Y, De Lima TC, Davis M. Opposing roles of the amygdala and dorsolateral periaqueductal gray in fear-potentiated startle. Neurosci Biobehav Rev 1997; 21:743-53. [PMID: 9415899 DOI: 10.1016/s0149-7634(96)00061-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The whole-body acoustic startle response is a short-latency reflex mediated by a relatively simple neural circuit in the lower brainstem and spinal cord. The amplitude of this reflex is markedly enhanced by moderate fear levels, and less effectively increased by higher fear levels. Extensive evidence indicates that the amygdala plays a key role in the potentiation of startle by moderate fear. More recent evidence suggests that the periaqueductal gray is involved in the loss of potentiated startle at higher levels of fear. The influence of both structures may be mediated by anatomical connections with the acoustic startle circuit, perhaps at the level of the nucleus reticularis pontis caudalis. The present chapter reviews these data.
Collapse
Affiliation(s)
- D L Walker
- Ribicoff Research Facilities of the Connecticut Mental Health Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06508, USA
| | | | | | | | | |
Collapse
|
50
|
Iwakiri Y, Chijiiwa Y, Motomura Y, Osame M, Nawata H. Presence of functional receptors for corticotropin releasing hormone in caecal circular smooth muscle cells of guinea pig. Life Sci 1997; 60:857-64. [PMID: 9076325 DOI: 10.1016/s0024-3205(97)00014-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The presence of specific binding sites for corticotropin releasing hormone (CRH) in caecal circular smooth muscle cells of guinea pig was investigated by binding and pharmacological studies. The specific binding of 125I-CRH to these muscle cells reached an equilibrium after 90 minutes. Several peptides structurally unrelated to CRH did not affect the specific binding of 125I-CRH to these muscle cells. Unlabeled CRH completely inhibited the specific binding of 125I-CRH in a concentration-dependent manner, with an IC50 value of 13.5 nM. A CRH receptor antagonist, alpha-helical CRH (9-41), inhibited the specific binding of 125I-CRH in a concentration-dependent manner with a lower affinity than CRH. In pharmacological study, CRH inhibited the contractile response of these muscle cells to 1 nM cholecystokinin-octapeptide in a concentration-dependent manner, with an IC50 value of 14.1 nM. A CRH receptor antagonist, alpha-helical CRH (9-41), significantly antagonized this inhibitory effect produced by CRH. These results strongly suggest the presence of functional receptors for CRH that mediate relaxation of caecal circular smooth muscle cells of guinea pig.
Collapse
Affiliation(s)
- Y Iwakiri
- Third Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan
| | | | | | | | | |
Collapse
|