1
|
Torres DB, Lopes A, Rodrigues AJ, Ventura-Silva AP, Sousa N, Gontijo JAR, Boer PA, Lopes MG. Early morphological and neurochemical changes of the bed nucleus of stria terminalis (BNST) in gestational protein-restricted male offspring. Nutr Neurosci 2024; 27:1250-1268. [PMID: 38576309 DOI: 10.1080/1028415x.2024.2320498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
BACKGROUND The bed nucleus of the stria terminalis (BNST) is a structure with a peculiar neurochemical composition involved in modulating anxietylike behavior and fear. AIM The present study investigated the effects on the BNST neurochemical composition and neuronal structure in critical moments of the postnatal period in gestational protein-restricted male rats' offspring. METHODS Dams were maintained during the pregnancy on isocaloric rodent laboratory chow with standard protein content [NP, 17%] or low protein content [LP, 6%]. BNST from male NP and age-matched LP offspring was studied using the isotropic fractionator method, Neuronal 3D reconstruction, dendritic-tree analysis, blotting analysis, and high-performance liquid chromatography. RESULTS Serum corticosterone levels were higher in male LP offspring than NP rats in 14-day-old offspring, without any difference in 7-day-old progeny. The BNST total cell number and anterodorsal BNST division volume in LP progeny were significantly reduced on the 14th postnatal day compared with NP offspring. The BNST HPLC analysis from 7 days-old LP revealed increased norepinephrine levels compared to NP progeny. The BNST blot analysis from 7-day-old LP revealed reduced levels of GR and BDNF associated with enhanced CRF1 expression compared to NP offspring. 14-day-old LP offspring showed reduced expression of MR and 5HT1A associated with decreased DOPAC and DOPA turnover levels relative to NP rats. In Conclusion, the BNST cellular and neurochemical changes may represent adaptation during development in response to elevated fetal exposure to maternal corticosteroid levels. In this way, gestational malnutrition alters the BNST content and structure and contributes to already-known behavioral changes.
Collapse
Affiliation(s)
- D B Torres
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | - A Lopes
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | - A J Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - A P Ventura-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - N Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - J A R Gontijo
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | - P A Boer
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | | |
Collapse
|
2
|
Ronan PJ, Korzan WJ, Johnson PL, Lowry CA, Renner KJ, Summers CH. Prior stress and vasopressin promote corticotropin-releasing factor inhibition of serotonin release in the central nucleus of the amygdala. Front Behav Neurosci 2023; 17:1148292. [PMID: 37064300 PMCID: PMC10098171 DOI: 10.3389/fnbeh.2023.1148292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/15/2023] [Indexed: 04/18/2023] Open
Abstract
Corticotropin-releasing factor (CRF) is essential for coordinating endocrine and neural responses to stress, frequently facilitated by vasopressin (AVP). Previous work has linked CRF hypersecretion, binding site changes, and dysfunctional serotonergic transmission with anxiety and affective disorders, including clinical depression. Crucially, CRF can alter serotonergic activity. In the dorsal raphé nucleus and serotonin (5-HT) terminal regions, CRF effects can be stimulatory or inhibitory, depending on the dose, site, and receptor type activated. Prior stress alters CRF neurotransmission and CRF-mediated behaviors. Lateral, medial, and ventral subdivisions of the central nucleus of the amygdala (CeA) produce CRF and coordinate stress responsiveness. The purpose of these experiments was to determine the effect of intracerebroventricular (icv) administration of CRF and AVP on extracellular 5-HT as an index of 5-HT release in the CeA, using in vivo microdialysis in freely moving rats and high performance liquid chromatography (HPLC) analysis. We also examined the effect of prior stress (1 h restraint, 24 h prior) on CRF- and AVP-mediated release of 5-HT within the CeA. Our results show that icv CRF infusion in unstressed animals had no effect on 5-HT release in the CeA. Conversely, in rats with prior stress, CRF caused a profound dose-dependent decrease in 5-HT release within the CeA. This effect was long-lasting (240 min) and was mimicked by CRF plus AVP infusion without stress. Thus, prior stress and AVP functionally alter CRF-mediated neurotransmission and sensitize CRF-induced inhibition of 5-HT release, suggesting that this is a potential mechanism underlying stress-induced affective reactivity in humans.
Collapse
Affiliation(s)
- Patrick J. Ronan
- Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD, United States
- Department of Psychiatry, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, United States
- Laboratory for Clinical and Translational Research in Psychiatry, Department of Veterans Affairs Medical Center, Denver, CO, United States
- Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
- Kenneth J. Renner,
| | - Wayne J. Korzan
- Department of Biological and Environmental Sciences, The University of West Alabama, Livingston, AL, United States
| | - Philip L. Johnson
- Department of Biology, University of South Dakota, Vermillion, SD, United States
| | - Christopher A. Lowry
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, CO, United States
| | - Kenneth J. Renner
- Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
- Department of Biology, University of South Dakota, Vermillion, SD, United States
- Patrick J. Ronan,
| | - Cliff H. Summers
- Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD, United States
- Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
- Department of Biology, University of South Dakota, Vermillion, SD, United States
- *Correspondence: Cliff H. Summers,
| |
Collapse
|
3
|
Young CE, Tong Q. Corticotropin Releasing Hormone Signaling in the Bed Nuclei of the Stria Terminalis as a Link to Maladaptive Behaviors. Front Neurosci 2021; 15:642379. [PMID: 33867924 PMCID: PMC8044981 DOI: 10.3389/fnins.2021.642379] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/01/2021] [Indexed: 11/30/2022] Open
Abstract
The bed nuclei of the stria terminalis (BST) is a limbic region in the extended amygdala that is heavily implicated in anxiety processing and hypothalamic-adrenal-pituitary (HPA) axis activation. The BST is complex, with many nuclei expressing different neurotransmitters and receptors involved in a variety of signaling pathways. One neurotransmitter that helps link its functions is corticotropin releasing hormone (CRH). BST CRH neuron activation may cause both anxiogenic and anxiolytic effects in rodents, and CRH neurons interact with other neuron types to influence anxiety-like responses as well as alcohol and drug–seeking behavior. This review covers the link between BST CRH neurons and thirteen other neurotransmitters and receptors and analyzes their effect on rodent behavior. Additionally, it covers the translational potential of targeting CRH signaling pathways for the treatment of human mental health disorders. Given the massive impact of anxiety, mood, and substance use disorders on our society, further research into BST CRH signaling is critical to alleviate the social and economic burdens of those disorders.
Collapse
Affiliation(s)
- Claire Emily Young
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Qingchun Tong
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Department of Neurobiology and Anatomy of McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,MD Anderson Cancer Center & UTHealth Graduate School of Biological Sciences, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
4
|
Kim JE, Chae S, Kim S, Jung YJ, Kang MG, Heo WD, Kim D. Cerebellar 5HT-2A receptor mediates stress-induced onset of dystonia. SCIENCE ADVANCES 2021; 7:7/10/eabb5735. [PMID: 33658190 PMCID: PMC7929497 DOI: 10.1126/sciadv.abb5735] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 01/15/2021] [Indexed: 06/12/2023]
Abstract
Stress is a key risk factor for dystonia, a debilitating motor disorder characterized by cocontractions of muscles leading to abnormal body posture. While the serotonin (5HT) system is known to control emotional responses to stress, its role in dystonia remains unclear. Here, we reveal that 5HT neurons in the dorsal raphe nuclei (DRN) send projections to the fastigial deep cerebellar nuclei (fDCN) and that photostimulation of 5HT-fDCN induces dystonia in wild-type mice. Moreover, we report that photoinhibition of 5HT-fDCN reduces dystonia in a1A tot/tot mice, a genetic model of stress-induced dystonia, and administration of a 5HT-2A receptor inverse agonist (MDL100907; 0.1 to 1 mg/kg) or shRNA-mediated knockdown of the ht2ar gene in fDCN can notably reduce the onset of dystonia in a1A tot/tot mice. These results support the serotonin theory of dystonia and suggest strategies for alleviating symptoms in human patients by blocking 5HT-2A receptors.
Collapse
Affiliation(s)
- Jung Eun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Sujin Chae
- KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Sungsoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Yeon-Joo Jung
- Bio Core Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Myoung-Goo Kang
- Department of Neuroscience, Cell Biology, and Anatomy, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
- KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Daesoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea.
- KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
- Bio Core Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| |
Collapse
|
5
|
CRF-5-HT interactions in the dorsal raphe nucleus and motivation for stress-induced opioid reinstatement. Psychopharmacology (Berl) 2021; 238:29-40. [PMID: 33231727 PMCID: PMC7796902 DOI: 10.1007/s00213-020-05652-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/20/2020] [Indexed: 01/17/2023]
Abstract
RATIONALE The serotonin (5-hydroxytryptamine, 5-HT) system plays an important role in stress-related psychiatric disorders and substance abuse. Our previous data show that stressors can inhibit 5-HT neuronal activity and release by stimulating the release of the stress neurohormone corticotropin-releasing factor (CRF) within the serotonergic dorsal raphe nucleus (DRN). The inhibitory effects of CRF on 5-HT DRN neurons are indirect, mediated by CRF-R1 receptors located on GABAergic afferents. OBJECTIVES We tested the hypothesis that DRN CRF-R1 receptors contribute to stress-induced reinstatement of morphine-conditioned place preference (CPP). We also examined the role of this circuitry in stress-induced negative affective state with 22-kHz distress ultrasonic vocalizations (USVs), which are naturally emitted by rats in response to environmental challenges such as pain, stress, and drug withdrawal. METHODS First, we tested if activation of CRF-R1 receptors in the DRN with the CRF-R1-preferring agonist ovine CRF (oCRF) would reinstate morphine CPP and then if blockade of CRF-R1 receptors in the DRN with the CRF-R1 antagonist NBI 35965 would attenuate swim stress-induced reinstatement of morphine CPP. Second, we tested if intra-DRN pretreatment with NBI 35965 would attenuate foot shock stress-induced 22-kHz USVs. RESULTS Intra-DRN injection of oCRF reinstated morphine CPP, while intra-DRN injection of NBI 35965 attenuated swim stress-induced reinstatement. Moreover, intra-DRN pretreatment with NBI 35965 significantly reduced 22-kHz distress calls induced by foot shock. CONCLUSIONS These data provide evidence that stress-induced negative affective state is mediated by DRN CRF-R1 receptors and may contribute to reinstatement of morphine CPP.
Collapse
|
6
|
Bryce CA, Floresco SB. Central CRF and acute stress differentially modulate probabilistic reversal learning in male and female rats. Behav Brain Res 2020; 397:112929. [PMID: 32998044 DOI: 10.1016/j.bbr.2020.112929] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/11/2020] [Accepted: 09/22/2020] [Indexed: 01/21/2023]
Abstract
Acute stress can have variable and sometimes sex-dependent effects on different executive functions, including cognitive flexibility, some of which may be mediated by increased corticotropin releasing factor (CRF). Previous studies on the effects of stress and CRF on cognitive flexibility have used procedures entailing deterministic rewards, yet how they may alter behavior when outcomes are probabilistic is unclear. The present study examined how acute stress and increased CRF activity alters probabilistic reversal learning (PRL) in male and female rats. Rats learned to discriminate between a 'correct' lever rewarded on 80 % of trials, and an "incorrect" lever delivering reward on 20 % of trials, with reward contingencies reversed after 8 consecutive correct choices. Separate groups received either intracerebroventricular infusions of CRF (3 μg) or restraint stress prior to a PRL session. Experiments examined how these manipulations affected learning when given prior to a one-day acquisition test or during performance in well-trained rats. Exogenous CRF, and to a lesser extent acute stress, impaired motivation across sexes, slowing deliberation times and increasing the number of trials omitted, particularly following a switch in reward contingencies. Neither manipulation significantly altered errors or reversal performance. However, increased CRF activity reduced negative feedback sensitivity. Across manipulations, females showed increased omissions and choice latencies, and were less sensitive to feedback than males. These results reveal the complexity with which stress, CRF, sex, and experience interact to alter aspects of motivation and probabilistic reinforcement learning and provide insight into how CRF activity may contribute to symptoms of stress-related disorders.
Collapse
Affiliation(s)
- Courtney A Bryce
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2136 West Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Stan B Floresco
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2136 West Mall, Vancouver, BC, V6T 1Z4, Canada.
| |
Collapse
|
7
|
Steinberg LJ, Mann JJ. Abnormal stress responsiveness and suicidal behavior: A risk phenotype. Biomark Neuropsychiatry 2020. [DOI: 10.1016/j.bionps.2020.100011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
8
|
Lundy R. Comparison of GABA, Somatostatin, and Corticotrophin-Releasing Hormone Expression in Axon Terminals That Target the Parabrachial Nucleus. Chem Senses 2020; 45:275-282. [PMID: 32107535 DOI: 10.1093/chemse/bjaa010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Several forebrain areas have been shown to project to the parabrachial nucleus (PBN) and exert inhibitory and excitatory influences on taste processing. Some sources of descending input such as the central nucleus of the amygdala (CeA) might utilize somatostatin (Sst) and/or corticotrophin-releasing hormone (Crh) to influence taste processing in the PBN (Panguluri S, Saggu S, Lundy R. 2009. Comparison of somatostatin and corticotrophin-releasing hormone immunoreactivity in forebrain neurons projecting to taste-responsive and non-responsive regions of the parabrachial nucleus in rat. Brain Res 1298:57-69; Magableh A, Lundy R. 2014. Somatostatin and corticotrophin releasing hormone cell types are a major source of descending input from the forebrain to the parabrachial nucleus in mice. Chem Senses 39:673-682). Since the predominate effect of CeA stimulation on PBN taste-evoked responses is inhibition, this study used transgenic reporter lines (Sst/TdTomato and Crh/TdTomato) and electron microscopy to assess Sst/gamma aminobutyric acid (GABA) and Crh/GABA coexpression in axon terminals within the PBN. Robust expression of Sst and Crh axon terminals was observed in the PBN. The majority of Sst-positive axon terminals were positive for GABA expression, while the majority of Crh terminals were not. The results indicate that Sst-expressing neurons, but not Crh neurons, are a source of GABAergic input to the PBN. To assess whether the CeA is a source of GABAergic input to the PBN, the CeA of Sst-cre mice was injected with cre-dependent enhanced yellow fluorescent protein (EYFP) virus and PBN tissue processed for GABA and EYFP expression. Again, the majority of EYFP Sst-positive axon terminals in the PBN coexpressed GABA. Together, the present results suggest that CeA neurons marked by Sst expression represent a major extrinsic source of GABAergic input to the PBN and this could underlie the predominate inhibitory effect of CeA stimulation on taste-evoked responses in the PBN.
Collapse
Affiliation(s)
- Robert Lundy
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, 500 South Preston St., HSC A, rm 1003, Louisville, KY, USA
| |
Collapse
|
9
|
Effects of plus-maze experience and chlordiazepoxide on anxiety-like behavior and serotonin neural activity in the dorsal raphe nucleus in rats. Behav Pharmacol 2020; 30:208-219. [PMID: 30169377 DOI: 10.1097/fbp.0000000000000423] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The extent to which rats express anxiety-like behavior on the elevated plus-maze (EPM) depends on their previous maze experience. Open-arm avoidance develops in maze-experienced rats, and is often accompanied by a diminished anxiolytic response to benzodiazepines. Regions of the dorsal raphe nucleus (DRN) were examined in male Sprague-Dawley rats using c-Fos and serotonin immunohistochemistry following a single exposure, a second exposure or no exposure to the EPM. We then examined the effect of the benzodiazepine anxiolytic chlordiazepoxide (CDP, 5 mg/kg) on EPM behavior and DRN neural activity. Enhanced open-arm avoidance was evident on the second EPM trial in both experiments. The observed pattern of c-Fos expression suggests that the first exposure to the maze activates serotonin cells in the rostral and dorsal regions of the DRN and that only the dorsal subregion is activated by a second exposure. CDP increased open-arm exploration during the first trial, which corresponded to decreased 5-hydroxytryptamine (5-HT) activity in the rostral and ventral subregions of the DRN. However, 5-HT activity in the DRN was reduced in rats on the second maze trial compared with the first trial, when CDP had no effect on open-arm exploration. These results suggest that open-arm avoidance in maze-experienced rats can be characterized as a coping response that is mediated by specific populations of 5-HT neurons in the DRN.
Collapse
|
10
|
Hernández-Vázquez F, Garduño J, Hernández-López S. GABAergic modulation of serotonergic neurons in the dorsal raphe nucleus. Rev Neurosci 2019; 30:289-303. [PMID: 30173207 DOI: 10.1515/revneuro-2018-0014] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/18/2018] [Indexed: 11/15/2022]
Abstract
The dorsal raphe nucleus (DRN), located in the brainstem, is involved in several functions such as sleep, temperature regulation, stress responses, and anxiety behaviors. This nucleus contains the largest population of serotonin expressing neurons in the brain. Serotonergic DRN neurons receive tonic γ-aminobutyric acid (GABA)inhibitory inputs from several brain areas, as well as from interneurons within the same nucleus. Serotonergic and GABAergic neurons in the DRN can be distinguished by their size, location, pharmacological responses, and electrophysiological properties. GABAergic neurons regulate the excitability of DRN serotonergic neurons and the serotonin release in different brain areas. Also, it has been shown that GABAergic neurons can synchronize the activity of serotonergic neurons across functions such as sleep or alertness. Moreover, dysregulation of GABA signaling in the DRN has been linked to psychiatric disorders such as anxiety and depression. This review focuses on GABAergic transmission in the DRN. The interaction between GABAergic and serotonergic neurons is discussed considering some physiological implications. Also, the main electrophysiological and morphological characteristics of serotonergic and GABAergic neurons are described.
Collapse
Affiliation(s)
- Fabiola Hernández-Vázquez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, México
| | - Julieta Garduño
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, PO Box 70250, Ciudad de México 04510, México
| | - Salvador Hernández-López
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, PO Box 70250, Ciudad de México 04510, México, e-mail:
| |
Collapse
|
11
|
Huang KW, Ochandarena NE, Philson AC, Hyun M, Birnbaum JE, Cicconet M, Sabatini BL. Molecular and anatomical organization of the dorsal raphe nucleus. eLife 2019; 8:e46464. [PMID: 31411560 PMCID: PMC6726424 DOI: 10.7554/elife.46464] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 08/13/2019] [Indexed: 12/20/2022] Open
Abstract
The dorsal raphe nucleus (DRN) is an important source of neuromodulators and has been implicated in a wide variety of behavioral and neurological disorders. The DRN is subdivided into distinct anatomical subregions comprised of multiple cell types, and its complex cellular organization has impeded efforts to investigate the distinct circuit and behavioral functions of its subdomains. Here we used single-cell RNA sequencing, in situ hybridization, anatomical tracing, and spatial correlation analysis to map the transcriptional and spatial profiles of cells from the mouse DRN. Our analysis of 39,411 single-cell transcriptomes revealed at least 18 distinct neuron subtypes and 5 serotonergic neuron subtypes with distinct molecular and anatomical properties, including a serotonergic neuron subtype that preferentially innervates the basal ganglia. Our study lays out the molecular organization of distinct serotonergic and non-serotonergic subsystems, and will facilitate the design of strategies for further dissection of the DRN and its diverse functions.
Collapse
Affiliation(s)
- Kee Wui Huang
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Nicole E Ochandarena
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Adrienne C Philson
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Minsuk Hyun
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Jaclyn E Birnbaum
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Marcelo Cicconet
- Image and Data Analysis CoreHarvard Medical SchoolBostonUnited States
| | - Bernardo L Sabatini
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
12
|
Marcinkiewcz CA, Bierlein-De La Rosa G, Dorrier CE, McKnight M, DiBerto JF, Pati D, Gianessi CA, Hon OJ, Tipton G, McElligott ZA, Delpire E, Kash TL. Sex-Dependent Modulation of Anxiety and Fear by 5-HT 1A Receptors in the Bed Nucleus of the Stria Terminalis. ACS Chem Neurosci 2019; 10:3154-3166. [PMID: 31140276 DOI: 10.1021/acschemneuro.8b00594] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Serotonin (5-hydroxytryptamine; 5-HT) coordinates behavioral responses to stress through a variety of presynaptic and postsynaptic receptors distributed across functionally diverse neuronal networks in the central nervous system. Efferent 5-HT projections from the dorsal raphe nucleus (DRN) to the bed nucleus of the stria terminalis (BNST) are generally thought to enhance anxiety and aversive learning by activating 5-HT2C receptor (5-HT2CR) signaling in the BNST, although an opposing role for postsynaptic 5-HT1A receptors has recently been suggested. In the present study, we sought to delineate a role for postsynaptic 5-HT1A receptors in the BNST in aversive behaviors using a conditional knockdown of the 5-HT1A receptor. Both males and females were tested to dissect out sex-specific effects. We found that male mice have significantly reduced fear memory recall relative to female mice and inactivation of 5-HT1A receptor in the BNST increases contextual fear conditioning in male mice so that they resemble the females. This coincided with an increase in neuronal excitability in males, suggesting that 5-HT1A receptor deletion may enhance contextual fear recall by disinhibiting fear memory circuits in the BNST. Interestingly, 5-HT1A receptor knockdown did not significantly alter anxiety-like behavior in male or female mice, which is in agreement with previous findings that anxiety and fear are modulated by dissociable circuits in the BNST. Overall, these results suggest that BNST 5-HT1A receptors do not significantly alter behavior under basal conditions, but can act as a molecular brake that buffer against excessive activation of aversive circuits in more threatening contexts.
Collapse
Affiliation(s)
- Catherine A. Marcinkiewcz
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | | | - Cayce E. Dorrier
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Mackenzie McKnight
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States
| | - Jeffrey F. DiBerto
- Curriculum in Neurobiology, School of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Dipanwati Pati
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Carol A. Gianessi
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Olivia J. Hon
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Curriculum in Neurobiology, School of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Greg Tipton
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Zoe A. McElligott
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Curriculum in Neurobiology, School of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Eric Delpire
- Department of Anesthesiology, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Thomas L. Kash
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Curriculum in Neurobiology, School of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
13
|
Steinberg LJ, Rubin-Falcone H, Galfalvy HC, Kaufman J, Miller JM, Sublette ME, Cooper TB, Min E, Keilp JG, Stanley BH, Oquendo MA, Ogden RT, Mann JJ. Cortisol Stress Response and in Vivo PET Imaging of Human Brain Serotonin 1A Receptor Binding. Int J Neuropsychopharmacol 2019; 22:329-338. [PMID: 30927011 PMCID: PMC6499240 DOI: 10.1093/ijnp/pyz009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 12/07/2018] [Accepted: 02/15/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Abnormalities in the hypothalamic-pituitary-adrenal axis, serotonergic system, and stress response have been linked to the pathogenesis of major depressive disorder. State-dependent hyper-reactivity of the hypothalamic-pituitary-adrenal axis is seen in major depressive disorder, and higher binding to the serotonin 1A receptor is observed as a trait in both currently depressed and remitted untreated major depressive disorder. Here, we sought to examine whether a relationship exists between cortisol secretion in response to a stressor and serotonin 1A receptor binding throughout the brain, both in healthy controls and participants with major depressive disorder. METHODS Research participants included 42 medication-free, depressed subjects and 31 healthy volunteers. Participants were exposed to either an acute, physical stressor (radial artery catheter insertion) or a psychological stressor (Trier Social Stress Test). Levels of serotonin 1A receptor binding on positron emission tomography with [11C]WAY-100635 were also obtained from all participants. The relationship between [11C]WAY-100635 binding and cortisol was examined using mixed linear effects models with group (major depressive disorder vs control), cortisol, brain region, and their interactions as fixed effects and subject as a random effect. RESULTS We found a positive correlation between post-stress cortisol measures and serotonin 1A receptor ligand binding levels across multiple cortical and subcortical regions, independent of diagnosis and with both types of stress. The relationship between [11C]WAY-100635 binding and cortisol was homogenous across all a priori brain regions. In contrast, resting cortisol levels were negatively correlated with serotonin 1A receptor ligand binding levels independently of diagnosis, except in the RN. There was no significant difference in cortisol between major depressive disorder participants and healthy volunteers with either stressor. Similarly, there was no correlation between cortisol and depression severity in either stressor group. CONCLUSIONS This study suggests that there may be a common underlying mechanism that links abnormalities in the serotonin system and hypothalamic-pituitary-adrenal axis hyper-reactivity to stress. Future studies need to determine how hypothalamic-pituitary-adrenal axis dysfunction affects mood to increase the risk of suicide in major depression.
Collapse
Affiliation(s)
- Louisa J Steinberg
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY,Correspondence: Louisa J. Steinberg, MD, PhD, 1051 Riverside Drive, New York, NY 10032 ()
| | - Harry Rubin-Falcone
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY
| | - Hanga C Galfalvy
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY
| | - Joshua Kaufman
- Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY
| | - Jeffrey M Miller
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY
| | - M Elizabeth Sublette
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY
| | - Thomas B Cooper
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY,Nathan S. Kline Institute for Psychiatric Research, New York, NY
| | - Eli Min
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY
| | - John G Keilp
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY
| | - Barbara H Stanley
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY
| | - Maria A Oquendo
- Psychiatry Department, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - R Todd Ogden
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY,Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY
| | - J John Mann
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY,Department of Radiology, Columbia University, New York, NY
| |
Collapse
|
14
|
Loupy KM, Arnold MR, Hassell JE, Lieb MW, Milton LN, Cler KE, Fox JH, Siebler PH, Schmidt D, Noronha SISR, Day HEW, Lowry CA. Evidence that preimmunization with a heat-killed preparation of Mycobacterium vaccae reduces corticotropin-releasing hormone mRNA expression in the extended amygdala in a fear-potentiated startle paradigm. Brain Behav Immun 2019; 77:127-140. [PMID: 30597198 DOI: 10.1016/j.bbi.2018.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 12/21/2018] [Accepted: 12/22/2018] [Indexed: 01/16/2023] Open
Abstract
Posttraumatic stress disorder (PTSD) is a trauma and stressor-related disorder that is characterized by dysregulation of glucocorticoid signaling, chronic low-grade inflammation, and impairment in the ability to extinguish learned fear. Corticotropin-releasing hormone (Crh) is a stress- and immune-responsive neuropeptide secreted from the paraventricular nucleus of the hypothalamus (PVN) to stimulate the hypothalamic-pituitary-adrenal (HPA) axis; however, extra-hypothalamic sources of Crh from the central nucleus of the amygdala (CeA) and bed nucleus of the stria terminalis (BNST) govern specific fear- and anxiety-related defensive behavioral responses. We previously reported that preimmunization with a heat-killed preparation of the immunoregulatory environmental bacterium Mycobacterium vaccae NCTC 11659 enhances fear extinction in a fear-potentiated startle (FPS) paradigm. In this follow-up study, we utilized an in situ hybridization histochemistry technique to investigate Crh, Crhr1, and Crhr2 mRNA expression in the CeA, BNST, and PVN of the same rats from the original study [Fox et al., 2017, Brain, Behavior, and Immunity, 66: 70-84]. Here, we demonstrate that preimmunization with M. vaccae NCTC 11659 decreases Crh mRNA expression in the CeA and BNST of rats exposed to the FPS paradigm, and, further, that Crh mRNA expression in these regions is correlated with fear behavior during extinction training. These data are consistent with the hypothesis that M. vaccae promotes stress-resilience by attenuating Crh production in fear- and anxiety-related circuits. These data suggest that immunization with M. vaccae may be an effective strategy for prevention of fear- and anxiety-related disorders.
Collapse
Affiliation(s)
- Kelsey M Loupy
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Mathew R Arnold
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - James E Hassell
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Margaret W Lieb
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Lauren N Milton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Kristin E Cler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - James H Fox
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Philip H Siebler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Dominic Schmidt
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Sylvana I S R Noronha
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Heidi E W Day
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Department of Physical Medicine & Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center, Denver Veterans Affairs Medical Center (VAMC), Denver, CO 80045, USA; Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Denver, CO 80045, USA.
| |
Collapse
|
15
|
Hassell JE, Nguyen KT, Gates CA, Lowry CA. The Impact of Stressor Exposure and Glucocorticoids on Anxiety and Fear. Curr Top Behav Neurosci 2019; 43:271-321. [PMID: 30357573 DOI: 10.1007/7854_2018_63] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Anxiety disorders and trauma- and stressor-related disorders, such as posttraumatic stress disorder (PTSD), are common and are associated with significant economic and social burdens. Although trauma and stressor exposure are recognized as a risk factors for development of anxiety disorders and trauma or stressor exposure is recognized as essential for diagnosis of PTSD, the mechanisms through which trauma and stressor exposure lead to these disorders are not well characterized. An improved understanding of the mechanisms through which trauma or stressor exposure leads to development and persistence of anxiety disorders or PTSD may result in novel therapeutic approaches for the treatment of these disorders. Here, we review the current state-of-the-art theories, with respect to mechanisms through which stressor exposure leads to acute or chronic exaggeration of avoidance or anxiety-like defensive behavioral responses and fear, endophenotypes in both anxiety disorders and trauma- and stressor-related psychiatric disorders. In this chapter, we will explore physiological responses and neural circuits involved in the development of acute and chronic exaggeration of anxiety-like defensive behavioral responses and fear states, focusing on the role of the hypothalamic-pituitary-adrenal (HPA) axis and glucocorticoid hormones.
Collapse
Affiliation(s)
- J E Hassell
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - K T Nguyen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - C A Gates
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - C A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA.
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA.
- Department of Physical Medicine and Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center, Denver Veterans Affairs Medical Center (VAMC), Denver, CO, USA.
- Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Denver, CO, USA.
| |
Collapse
|
16
|
Culman J, Mühlenhoff S, Blume A, Hedderich J, Lützen U, Hunt SP, Rupniak NMJ, Zhao Y. The Hypothalamic-Pituitary-Adrenal Axis and Serotonin Metabolism in Individual Brain Nuclei of Mice with Genetic Disruption of the NK1 Receptor Exposed to Acute Stress. Cell Mol Neurobiol 2018; 38:1271-1281. [PMID: 29948553 DOI: 10.1007/s10571-018-0594-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/23/2018] [Indexed: 12/12/2022]
Abstract
Mice lacking the substance P (SP) neurokinin-1 (NK1) receptor (NK1R-/-mice) were used to investigate whether SP affects serotonin (5-HT) function in the brain and to assess the effects of acute immobilisation stress on the hypothalamic-pituitary-adrenocortical (HPA) axis and 5-HT turnover in individual brain nuclei. Basal HPA activity and the expression of hypothalamic corticotropin-releasing hormone (CRH) in wild-type (WT)- and NK1R-/- mice were identical. Stress-induced increases in plasma ACTH concentration were considerably higher in NK1R-/- mice than in WT mice while corticosterone concentrations were equally elevated in both mouse lines. Acute stress did not alter the expression of CRH. In the dorsal raphe nucleus (DRN), basal 5-HT turnover was increased in NK1R-/- mice and a 15 min stress further magnified 5-HT utilisation in this region. In the frontoparietal cortex, medial prefrontal cortex, central nucleus of amygdala, and the hippocampal CA1 region, stress increased 5-HT and/or 5-hydroxyindoleacetic acid (5-HIAA) concentrations to a similar extent in WT and NK1R-/- mice. 5-HT turnover in the hypothalamic paraventricular nucleus was not affected by stress, but stress induced similar increases in 5-HT and 5-HIAA in the ventromedial and dorsomedial hypothalamic nuclei in WT and NK1R-/- mice. Our findings indicate that NK1 receptor activation suppresses ACTH release during acute stress but does not exert sustained inhibition of the HPA axis. Genetic deletion of the NK1 receptor accelerates 5-HT turnover in DRN under basal and stress conditions. No differences between the responses of serotonergic system to acute stress in WT and NK1R-/- mice occur in forebrain nuclei linked to the regulation of anxiety and neuroendocrine stress responses.
Collapse
Affiliation(s)
- Juraj Culman
- Institute of Experimental and Clinical Pharmacology, University Hospital of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Strasse 3, 24105, Kiel, Germany.
- Department of Nuclear Medicine, Molecular Imaging, Diagnostics and Therapy, University Hospital of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Strasse 3, 24105, Kiel, Germany.
| | - Stephan Mühlenhoff
- Institute of Experimental and Clinical Pharmacology, University Hospital of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Strasse 3, 24105, Kiel, Germany
| | - Annegret Blume
- Institute of Experimental and Clinical Pharmacology, University Hospital of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Strasse 3, 24105, Kiel, Germany
| | - Jürgen Hedderich
- Institute of Medical Informatics and Statistics, University Hospital of Schleswig-Holstein, Campus Kiel, Brunswiker Strasse 10, 24105, Kiel, Germany
| | - Ulf Lützen
- Department of Nuclear Medicine, Molecular Imaging, Diagnostics and Therapy, University Hospital of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Strasse 3, 24105, Kiel, Germany
| | - Stephen P Hunt
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | | | - Yi Zhao
- Department of Nuclear Medicine, Molecular Imaging, Diagnostics and Therapy, University Hospital of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Strasse 3, 24105, Kiel, Germany
| |
Collapse
|
17
|
Nishii A, Amemiya S, Kubota N, Nishijima T, Kita I. Adaptive Changes in the Sensitivity of the Dorsal Raphe and Hypothalamic Paraventricular Nuclei to Acute Exercise, and Hippocampal Neurogenesis May Contribute to the Antidepressant Effect of Regular Treadmill Running in Rats. Front Behav Neurosci 2017; 11:235. [PMID: 29225572 PMCID: PMC5705550 DOI: 10.3389/fnbeh.2017.00235] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 11/13/2017] [Indexed: 01/20/2023] Open
Abstract
Increasing clinical evidence suggests that regular physical exercise can prevent or reduce the incidence of stress-related psychiatric disorders including depressive symptoms. Antidepressant effect of regular exercise may be implicated in monoaminergic transmission including serotonergic transmission, activation of the hypothalamic-pituitary-adrenal (HPA) axis, and hippocampal neurogenesis, but few general concepts regarding the optimal exercise regimen for stimulating neural mechanisms involved in antidepressant properties have been developed. Here, we examined how 4 weeks of treadmill running at different intensities (0, 15, 25 m/min, 60 min/day, 5 times/week) alters neuronal activity in the dorsal raphe nucleus (DRN), which is the major source of serotonin (5-HT) neurons in the central nervous system, and the hypothalamic paraventricular nucleus (PVN), in which corticotropin-releasing factor (CRF) neurons initiate the activation of the HPA axis, during one session of acute treadmill running at different speeds (0, 15, 25 m/min, 30 min) in male Wistar rats, using c-Fos immunohistochemistry. We also examined neurogenesis in the hippocampus using immunohistochemistry for doublecortin (DCX) and assessed depressive-like behavior using the forced swim test after regular exercise for 4 weeks. In the pre-training period, acute treadmill running at low speed, but not at high speed, increased c-Fos positive nuclei in the DRN compared with the sedentary control. The number of c-Fos positive nuclei in the PVN during acute treadmill running was increased in a running speed-dependent manner. Regular exercise for 4 weeks, regardless of the training intensity, induced an enhancement of c-Fos expression in the DRN during not only low-speed but also high-speed acute running, and generally reduced c-Fos expression in the PVN during acute running compared with pre-training. Furthermore, regular treadmill running for 4 weeks enhanced DCX immunoreactivity in the hippocampal dentate gyrus (DG), and resulted in decreased depressive-like behavior, regardless of the training intensity. These results suggest that long-term repeated exercise, regardless of the training intensity, improves depressive-like behavior through adaptive changes in the sensitivity of DRN and PVN neurons to acute exercise, and hippocampal neurogenesis.
Collapse
Affiliation(s)
- Ayu Nishii
- Laboratory of Behavioral Neuroscience, Department of Human Health Science, Tokyo Metropolitan University, Hachioji, Japan
| | - Seiichiro Amemiya
- Laboratory of Behavioral Neuroscience, Department of Human Health Science, Tokyo Metropolitan University, Hachioji, Japan
| | - Natsuko Kubota
- Laboratory of Behavioral Neuroscience, Department of Human Health Science, Tokyo Metropolitan University, Hachioji, Japan
| | - Takeshi Nishijima
- Laboratory of Behavioral Neuroscience, Department of Human Health Science, Tokyo Metropolitan University, Hachioji, Japan
| | - Ichiro Kita
- Laboratory of Behavioral Neuroscience, Department of Human Health Science, Tokyo Metropolitan University, Hachioji, Japan
| |
Collapse
|
18
|
Wong-Lin K, Wang DH, Moustafa AA, Cohen JY, Nakamura K. Toward a multiscale modeling framework for understanding serotonergic function. J Psychopharmacol 2017; 31:1121-1136. [PMID: 28417684 PMCID: PMC5606304 DOI: 10.1177/0269881117699612] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Despite its importance in regulating emotion and mental wellbeing, the complex structure and function of the serotonergic system present formidable challenges toward understanding its mechanisms. In this paper, we review studies investigating the interactions between serotonergic and related brain systems and their behavior at multiple scales, with a focus on biologically-based computational modeling. We first discuss serotonergic intracellular signaling and neuronal excitability, followed by neuronal circuit and systems levels. At each level of organization, we will discuss the experimental work accompanied by related computational modeling work. We then suggest that a multiscale modeling approach that integrates the various levels of neurobiological organization could potentially transform the way we understand the complex functions associated with serotonin.
Collapse
Affiliation(s)
- KongFatt Wong-Lin
- Intelligent Systems Research Centre, School of Computing and Intelligent Systems, University of Ulster, Magee Campus, Derry~Londonderry, UK
| | - Da-Hui Wang
- School of Systems Science, and National Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Ahmed A Moustafa
- School of Social Sciences and Psychology, and Marcs Institute for Brain and Behaviour, University of Western Sydney, Sydney, Australia
| | - Jeremiah Y Cohen
- Solomon H. Snyder Department of Neuroscience, Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Kae Nakamura
- Department of Physiology, Kansai Medical University, Hirakata, Osaka, Japan
| |
Collapse
|
19
|
Verdouw PM, van Esterik JC, Peeters BW, Millan MJ, Groenink L. CRF1 but not glucocorticoid receptor antagonists reduce separation-induced distress vocalizations in guinea pig pups and CRF overexpressing mouse pups. A combination study with paroxetine. Pharmacol Biochem Behav 2017; 154:11-19. [DOI: 10.1016/j.pbb.2017.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 01/05/2017] [Accepted: 01/09/2017] [Indexed: 01/04/2023]
|
20
|
Henckens MJAG, Deussing JM, Chen A. Region-specific roles of the corticotropin-releasing factor-urocortin system in stress. Nat Rev Neurosci 2016; 17:636-51. [PMID: 27586075 DOI: 10.1038/nrn.2016.94] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Dysregulation of the corticotropin-releasing factor (CRF)-urocortin (UCN) system has been implicated in stress-related psychopathologies such as depression and anxiety. It has been proposed that CRF-CRF receptor type 1 (CRFR1) signalling promotes the stress response and anxiety-like behaviour, whereas UCNs and CRFR2 activation mediate stress recovery and the restoration of homeostasis. Recent findings, however, provide clear evidence that this view is overly simplistic. Instead, a more complex picture has emerged that suggests that there are brain region- and cell type-specific effects of CRFR signalling that are influenced by the individual's prior experience and that shape molecular, cellular and ultimately behavioural responses to stressful challenges.
Collapse
Affiliation(s)
- Marloes J A G Henckens
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel.,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany.,Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Jan M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Alon Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel.,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
21
|
Kim S, Kwok S, Mayes LC, Potenza MN, Rutherford HJV, Strathearn L. Early adverse experience and substance addiction: dopamine, oxytocin, and glucocorticoid pathways. Ann N Y Acad Sci 2016; 1394:74-91. [PMID: 27508337 DOI: 10.1111/nyas.13140] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/12/2016] [Accepted: 05/20/2016] [Indexed: 12/17/2022]
Abstract
Substance addiction may follow a chronic, relapsing course and critically undermine the physical and psychological well-being of the affected individual and the social units of which the individual is a member. Despite the public health burden associated with substance addiction, treatment options remain suboptimal, with relapses often seen. The present review synthesizes growing insights from animal and human research to shed light upon developmental and neurobiological pathways that may increase susceptibility to addiction. We examine the dopamine system, the oxytocin system, and the glucocorticoid system, as they are particularly relevant to substance addiction. Our aim is to delineate how early adverse experience may induce long-lasting alterations in each of these systems at molecular, neuroendocrine, and behavioral levels and ultimately lead to heightened vulnerability to substance addiction. We further discuss how substance addiction in adulthood may increase the risk of suboptimal caregiving for the next generation, perpetuating the intergenerational cycle of early adverse experiences and addiction.
Collapse
Affiliation(s)
- Sohye Kim
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas.,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas.,Attachment and Neurodevelopment Laboratory, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas
| | - Stephanie Kwok
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas
| | - Linda C Mayes
- Yale Child Study Center, Yale University School of Medicine, New Haven, Connecticut
| | - Marc N Potenza
- Yale Child Study Center, Yale University School of Medicine, New Haven, Connecticut.,Departments of Psychiatry and Neuroscience and the National Center on Addiction and Substance Abuse (CASAColumbia), Yale University School of Medicine, New Haven, Connecticut.,Connecticut Mental Health Center, New Haven, Connecticut
| | | | - Lane Strathearn
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas.,Attachment and Neurodevelopment Laboratory, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas.,Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
22
|
Mlinar B, Montalbano A, Piszczek L, Gross C, Corradetti R. Firing Properties of Genetically Identified Dorsal Raphe Serotonergic Neurons in Brain Slices. Front Cell Neurosci 2016; 10:195. [PMID: 27536220 PMCID: PMC4971071 DOI: 10.3389/fncel.2016.00195] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 07/22/2016] [Indexed: 11/13/2022] Open
Abstract
Tonic spiking of serotonergic neurons establishes serotonin levels in the brain. Since the first observations, slow regular spiking has been considered as a defining feature of serotonergic neurons. Recent studies, however, have revealed the heterogeneity of serotonergic neurons at multiple levels, comprising their electrophysiological properties, suggesting the existence of functionally distinct cellular subpopulations. In order to examine in an unbiased manner whether serotonergic neurons of the dorsal raphe nucleus (DRN) are heterogeneous, we used a non-invasive loose-seal cell-attached method to record α1 adrenergic receptor-stimulated spiking of a large sample of neurons in brain slices obtained from transgenic mice lines that express fluorescent marker proteins under the control of serotonergic system-specific Tph2 and Pet-1 promoters. We found wide homogeneous distribution of firing rates, well fitted by a single Gaussian function (r (2) = 0.93) and independent of anatomical location (P = 0.45), suggesting that in terms of intrinsic firing properties, serotonergic neurons in the DRN represent a single cellular population. Characterization of the population in terms of spiking regularity was hindered by its dependence on the firing rate. For instance, the coefficient of variation of the interspike intervals (ISI), a common measure of spiking irregularity, is of limited usefulness since it correlates negatively with the firing rate (r = -0.33, P < 0.0001). Nevertheless, the majority of neurons exhibited regular, pacemaker-like activity, with coefficient of variance of the ISI lower than 0.5 in ~97% of cases. Unexpectedly, a small percentage of neurons (~1%) exhibited a particular spiking pattern, characterized by low frequency (~0.02-0.1 Hz) oscillations in the firing rate. Transitions between regular and oscillatory firing were observed, suggesting that the oscillatory firing is an alternative firing pattern of serotonergic neurons.
Collapse
Affiliation(s)
- Boris Mlinar
- Department of Neuroscience, Psychology, Drug Research and Children's Health, University of Florence Florence, Italy
| | - Alberto Montalbano
- Department of Neuroscience, Psychology, Drug Research and Children's Health, University of Florence Florence, Italy
| | - Lukasz Piszczek
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| | - Cornelius Gross
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| | - Renato Corradetti
- Department of Neuroscience, Psychology, Drug Research and Children's Health, University of Florence Florence, Italy
| |
Collapse
|
23
|
Barra de la Tremblaye P, Plamondon H. Alterations in the corticotropin-releasing hormone (CRH) neurocircuitry: Insights into post stroke functional impairments. Front Neuroendocrinol 2016; 42:53-75. [PMID: 27455847 DOI: 10.1016/j.yfrne.2016.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/04/2016] [Accepted: 07/06/2016] [Indexed: 10/21/2022]
Abstract
Although it is well accepted that changes in the regulation of the hypothalamic-pituitary adrenal (HPA) axis may increase susceptibility to affective disorders in the general population, this link has been less examined in stroke patients. Yet, the bidirectional association between depression and cardiovascular disease is strong, and stress increases vulnerability to stroke. Corticotropin-releasing hormone (CRH) is the central stress hormone of the HPA axis pathway and acts by binding to CRH receptors (CRHR) 1 and 2, which are located in several stress-related brain regions. Evidence from clinical and animal studies suggests a role for CRH in the neurobiological basis of depression and ischemic brain injury. Given its importance in the regulation of the neuroendocrine, autonomic, and behavioral correlates of adaptation and maladaptation to stress, CRH is likely associated in the pathophysiology of post stroke emotional impairments. The goals of this review article are to examine the clinical and experimental data describing (1) that CRH regulates the molecular signaling brain circuit underlying anxiety- and depression-like behaviors, (2) the influence of CRH and other stress markers in the pathophysiology of post stroke emotional and cognitive impairments, and (3) context and site specific interactions of CRH and BDNF as a basis for the development of novel therapeutic targets. This review addresses how the production and release of the neuropeptide CRH within the various regions of the mesocorticolimbic system influences emotional and cognitive behaviors with a look into its role in psychiatric disorders post stroke.
Collapse
Affiliation(s)
- P Barra de la Tremblaye
- School of Psychology, Behavioral Neuroscience Program, University of Ottawa, 136 Jean-Jacques Lussier, Vanier Building, Ottawa, Ontario K1N 6N5, Canada
| | - H Plamondon
- School of Psychology, Behavioral Neuroscience Program, University of Ottawa, 136 Jean-Jacques Lussier, Vanier Building, Ottawa, Ontario K1N 6N5, Canada.
| |
Collapse
|
24
|
Belmer A, Patkar OL, Pitman KM, Bartlett SE. Serotonergic Neuroplasticity in Alcohol Addiction. Brain Plast 2016; 1:177-206. [PMID: 29765841 PMCID: PMC5928559 DOI: 10.3233/bpl-150022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Alcohol addiction is a debilitating disorder producing maladaptive changes in the brain, leading drinkers to become more sensitive to stress and anxiety. These changes are key factors contributing to alcohol craving and maintaining a persistent vulnerability to relapse. Serotonin (5-Hydroxytryptamine, 5-HT) is a monoamine neurotransmitter widely expressed in the central nervous system where it plays an important role in the regulation of mood. The serotonin system has been extensively implicated in the regulation of stress and anxiety, as well as the reinforcing properties of all of the major classes of drugs of abuse, including alcohol. Dysregulation within the 5-HT system has been postulated to underlie the negative mood states associated with alcohol use disorders. This review will describe the serotonergic (5-HTergic) neuroplastic changes observed in animal models throughout the alcohol addiction cycle, from prenatal to adulthood exposure. The first section will focus on alcohol-induced 5-HTergic neuroadaptations in offspring prenatally exposed to alcohol and the consequences on the regulation of stress/anxiety. The second section will compare alterations in 5-HT signalling induced by acute or chronic alcohol exposure during adulthood and following alcohol withdrawal, highlighting the impact on the regulation of stress/anxiety signalling pathways. The third section will outline 5-HTergic neuroadaptations observed in various genetically-selected ethanol preferring rat lines. Finally, we will discuss the pharmacological manipulation of the 5-HTergic system on ethanol- and anxiety/stress-related behaviours demonstrated by clinical trials, with an emphasis on current and potential treatments.
Collapse
Affiliation(s)
- Arnauld Belmer
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia.,Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, Australia
| | - Omkar L Patkar
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia.,Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, Australia
| | - Kim M Pitman
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia.,Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, Australia
| | - Selena E Bartlett
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia.,Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
25
|
Kuperman Y, Weiss M, Dine J, Staikin K, Golani O, Ramot A, Nahum T, Kühne C, Shemesh Y, Wurst W, Harmelin A, Deussing JM, Eder M, Chen A. CRFR1 in AgRP Neurons Modulates Sympathetic Nervous System Activity to Adapt to Cold Stress and Fasting. Cell Metab 2016; 23:1185-1199. [PMID: 27211900 PMCID: PMC4911344 DOI: 10.1016/j.cmet.2016.04.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 02/12/2016] [Accepted: 04/22/2016] [Indexed: 01/06/2023]
Abstract
Signaling by the corticotropin-releasing factor receptor type 1 (CRFR1) plays an important role in mediating the autonomic response to stressful challenges. Multiple hypothalamic nuclei regulate sympathetic outflow. Although CRFR1 is highly expressed in the arcuate nucleus (Arc) of the hypothalamus, the identity of these neurons and the role of CRFR1 here are presently unknown. Our studies show that nearly half of Arc-CRFR1 neurons coexpress agouti-related peptide (AgRP), half of which originate from POMC precursors. Arc-CRFR1 neurons are innervated by CRF neurons in the hypothalamic paraventricular nucleus, and CRF application decreases AgRP(+)CRFR1(+) neurons' excitability. Despite similar anatomy in both sexes, only female mice selectively lacking CRFR1 in AgRP neurons showed a maladaptive thermogenic response to cold and reduced hepatic glucose production during fasting. Thus, CRFR1, in a subset of AgRP neurons, plays a regulatory role that enables appropriate sympathetic nervous system activation and consequently protects the organism from hypothermia and hypoglycemia.
Collapse
Affiliation(s)
- Yael Kuperman
- Department of Veterinary Resources, Weizmann Institute of Science, 76100 Rehovot, Israel.
| | - Meira Weiss
- Department of Neurobiology, The Ruhman Family Laboratory for Research on the Neurobiology of Stress, Weizmann Institute of Science, 76100 Rehovot, Israel; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Julien Dine
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Katy Staikin
- Department of Neurobiology, The Ruhman Family Laboratory for Research on the Neurobiology of Stress, Weizmann Institute of Science, 76100 Rehovot, Israel; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Ofra Golani
- Biological Services Unit, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Assaf Ramot
- Department of Neurobiology, The Ruhman Family Laboratory for Research on the Neurobiology of Stress, Weizmann Institute of Science, 76100 Rehovot, Israel; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Tali Nahum
- Department of Neurobiology, The Ruhman Family Laboratory for Research on the Neurobiology of Stress, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Claudia Kühne
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Yair Shemesh
- Department of Neurobiology, The Ruhman Family Laboratory for Research on the Neurobiology of Stress, Weizmann Institute of Science, 76100 Rehovot, Israel; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Wolfgang Wurst
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Developmental Genetics, 85764 Neuherberg, Germany
| | - Alon Harmelin
- Department of Veterinary Resources, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Jan M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Matthias Eder
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Alon Chen
- Department of Neurobiology, The Ruhman Family Laboratory for Research on the Neurobiology of Stress, Weizmann Institute of Science, 76100 Rehovot, Israel; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany.
| |
Collapse
|
26
|
Early life adversity and serotonin transporter gene variation interact to affect DNA methylation of the corticotropin-releasing factor gene promoter region in the adult rat brain. Dev Psychopathol 2016; 27:123-35. [PMID: 25640835 DOI: 10.1017/s0954579414001345] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The interaction between childhood maltreatment and the serotonin transporter (5-HTT) gene linked polymorphic region has been associated with increased risk to develop major depression. This Gene × Environment interaction has furthermore been linked with increased levels of anxiety and glucocorticoid release upon exposure to stress. Both endophenotypes are regulated by the neuropeptide corticotropin-releasing factor (CRF) or hormone, which is expressed by the paraventricular nucleus of the hypothalamus, the bed nucleus of the stria terminalis, and the central amygdala (CeA). Therefore, we hypothesized that altered regulation of the expression of CRF in these areas represents a major neurobiological mechanism underlying the interaction of early life stress and 5-HTT gene variation. The programming of gene transcription by Gene × Environment interactions has been proposed to involve epigenetic mechanisms such as DNA methylation. In this study, we report that early life stress and 5-HTT genotype interact to affect DNA methylation of the Crf gene promoter in the CeA of adult male rats. Furthermore, we found that DNA methylation of a specific site in the Crf promoter significantly correlated with CRF mRNA levels in the CeA. Moreover, CeA CRF mRNA levels correlated with stress coping behavior in a learned helplessness paradigm. Together, our findings warrant further investigation of the link of Crf promoter methylation and CRF expression in the CeA with behavioral changes that are relevant for psychopathology.
Collapse
|
27
|
Altered taste preference and loss of limbic-projecting serotonergic neurons in the dorsal raphe nucleus of chronically epileptic rats. Behav Brain Res 2016; 297:28-36. [DOI: 10.1016/j.bbr.2015.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/30/2015] [Accepted: 10/03/2015] [Indexed: 12/12/2022]
|
28
|
Hwa LS, Shimamoto A, Kayyali T, Norman KJ, Valentino RJ, DeBold JF, Miczek KA. Dissociation of μ-opioid receptor and CRF-R1 antagonist effects on escalated ethanol consumption and mPFC serotonin in C57BL/6J mice. Addict Biol 2016; 21:111-24. [PMID: 25262980 DOI: 10.1111/adb.12189] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Both the opioid antagonist naltrexone and corticotropin-releasing factor type-1 receptor (CRF-R1) antagonists have been investigated for the treatment of alcoholism. The current study examines the combination of naltrexone and CP154526 to reduce intermittent access ethanol drinking [intermittent access to alcohol (IAA)] in C57BL/6J male mice, and if these compounds reduce drinking via serotonergic mechanisms in the dorsal raphe nucleus (DRN). Systemic injections and chronic intracerebroventricular infusions of naltrexone, CP154526 or CP376395 transiently decreased IAA drinking. Immunohistochemistry revealed CRF-R1 or μ-opioid receptor immunoreactivity was co-localized in tryptophan hydroxylase (TPH)-immunoreactive neurons as well as non-TPH neurons in the DRN. Mice with a history of IAA or continuous access to alcohol were microinjected with artificial cerebral spinal fluid, naltrexone, CP154526 or the combination into the DRN or the median raphe nucleus (MRN). Either intra-DRN naltrexone or CP154526 reduced IAA in the initial 2 hours of fluid access, but the combination did not additively suppress IAA, suggesting a common mechanism via which these two compounds affect intermittent drinking. These alcohol-reducing effects were localized to the DRN of IAA drinkers, as intra-MRN injections only significantly suppressed water drinking, and continuous access drinkers were not affected by CRF-R1 antagonism. Extracellular serotonin was measured in the medial prefrontal cortex (mPFC) using in vivo microdialysis after intra-DRN microinjections in another group of mice. Intra-DRN CP154526 increased serotonin impulse flow to the mPFC while naltrexone did not. This suggests the mPFC may not be an essential location to intermittent drinking, as evidenced by different effects on serotonin signaling to the forebrain yet similar behavioral findings.
Collapse
Affiliation(s)
- Lara S. Hwa
- Department of Psychology; Tufts University; Medford MA USA
| | | | - Tala Kayyali
- Department of Psychology; Tufts University; Medford MA USA
| | | | - Rita J. Valentino
- Division of Stress Neurobiology; Children's Hospital of Philadelphia; Philadelphia PA USA
| | | | - Klaus A. Miczek
- Department of Psychology; Tufts University; Medford MA USA
- Department of Neuroscience; Tufts University; Medford MA USA
| |
Collapse
|
29
|
Otsuka T, Nishii A, Amemiya S, Kubota N, Nishijima T, Kita I. Effects of acute treadmill running at different intensities on activities of serotonin and corticotropin-releasing factor neurons, and anxiety- and depressive-like behaviors in rats. Behav Brain Res 2015; 298:44-51. [PMID: 26542811 DOI: 10.1016/j.bbr.2015.10.055] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/20/2015] [Accepted: 10/29/2015] [Indexed: 10/22/2022]
Abstract
Accumulating evidence suggests that physical exercise can reduce and prevent the incidence of stress-related psychiatric disorders, including depression and anxiety. Activation of serotonin (5-HT) neurons in the dorsal raphe nucleus (DRN) is implicated in antidepressant/anxiolytic properties. In addition, the incidence and symptoms of these disorders may involve dysregulation of the hypothalamic-pituitary-adrenal axis that is initiated by corticotropin-releasing factor (CRF) neurons in the hypothalamic paraventricular nucleus (PVN). Thus, it is possible that physical exercise produces its antidepressant/anxiolytic effects by affecting these neuronal activities. However, the effects of acute physical exercise at different intensities on these neuronal activation and behavioral changes are still unclear. Here, we examined the activities of 5-HT neurons in the DRN and CRF neurons in the PVN during 30 min of treadmill running at different speeds (high speed, 25 m/min; low speed, 15m/min; control, only sitting on the treadmill) in male Wistar rats, using c-Fos/5-HT or CRF immunohistochemistry. We also performed the elevated plus maze test and the forced swim test to assess anxiety- and depressive-like behaviors, respectively. Acute treadmill running at low speed, but not high speed, significantly increased c-Fos expression in 5-HT neurons in the DRN compared to the control, whereas high-speed running significantly enhanced c-Fos expression in CRF neurons in the PVN compared with the control and low-speed running. Furthermore, low-speed running resulted in decreased anxiety- and depressive-like behaviors compared with high-speed running. These results suggest that acute physical exercise with mild and low stress can efficiently induce optimal neuronal activation that is involved in the antidepressant/anxiolytic effects.
Collapse
Affiliation(s)
- Tomomi Otsuka
- Department of Human Health Science, Tokyo Metropolitan University, 1-1 Minami-ohsawa, Hachioji, Tokyo192-0397, Japan
| | - Ayu Nishii
- Department of Human Health Science, Tokyo Metropolitan University, 1-1 Minami-ohsawa, Hachioji, Tokyo192-0397, Japan
| | - Seiichiro Amemiya
- Department of Human Health Science, Tokyo Metropolitan University, 1-1 Minami-ohsawa, Hachioji, Tokyo192-0397, Japan
| | - Natsuko Kubota
- Department of Human Health Science, Tokyo Metropolitan University, 1-1 Minami-ohsawa, Hachioji, Tokyo192-0397, Japan
| | - Takeshi Nishijima
- Department of Human Health Science, Tokyo Metropolitan University, 1-1 Minami-ohsawa, Hachioji, Tokyo192-0397, Japan
| | - Ichiro Kita
- Department of Human Health Science, Tokyo Metropolitan University, 1-1 Minami-ohsawa, Hachioji, Tokyo192-0397, Japan.
| |
Collapse
|
30
|
Cohen JY, Amoroso MW, Uchida N. Serotonergic neurons signal reward and punishment on multiple timescales. eLife 2015; 4. [PMID: 25714923 PMCID: PMC4389268 DOI: 10.7554/elife.06346] [Citation(s) in RCA: 235] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 02/24/2015] [Indexed: 11/30/2022] Open
Abstract
Serotonin's function in the brain is unclear. One challenge in testing the numerous hypotheses about serotonin's function has been observing the activity of identified serotonergic neurons in animals engaged in behavioral tasks. We recorded the activity of dorsal raphe neurons while mice experienced a task in which rewards and punishments varied across blocks of trials. We ‘tagged’ serotonergic neurons with the light-sensitive protein channelrhodopsin-2 and identified them based on their responses to light. We found three main features of serotonergic neuron activity: (1) a large fraction of serotonergic neurons modulated their tonic firing rates over the course of minutes during reward vs punishment blocks; (2) most were phasically excited by punishments; and (3) a subset was phasically excited by reward-predicting cues. By contrast, dopaminergic neurons did not show firing rate changes across blocks of trials. These results suggest that serotonergic neurons signal information about reward and punishment on multiple timescales. DOI:http://dx.doi.org/10.7554/eLife.06346.001 Rewards and punishments can both encourage animals to change their immediate behavior and influence their mood over a longer term, particularly when given repeatedly. A region of the brain that increases its activity in response to rewards and punishments also contains many neurons that communicate with each other by releasing a chemical called serotonin. This chemical is commonly thought to produce feelings of happiness; however, it remains unclear exactly how these particular ‘serotonergic’ neurons help to process rewards and punishments. The ideal way to work out the role that a type of neuron plays in a behavior is to measure its electrical activity as the behavior is being performed. However, it is difficult to distinguish the activity of serotonergic neurons from the activity of the non-serotonergic neurons around them. To overcome this problem, Cohen et al. used viruses to force serotonergic neurons to make a type of ion channel that produces electrical currents in response to light. Shining light on these neurons via optical fibers and then measuring the neurons' responses helped to develop criteria that can identify which responses are generated by the serotonergic neurons. Cohen et al. then recorded the activity of serotonergic neurons in thirsty mice as they experienced a series of rewards (for example, a drop of water) or punishments (such as a puff of air to the eye). Each reward or punishment was preceded by a distinct odor, so that the mice learned to anticipate what was coming. These experiments revealed that serotonergic neurons respond to rewards and punishments by changing two aspects of their electrical activity: by producing short bursts of high activity, and by altering their baseline activity. Some of the serotonergic neurons fired rapidly in response to punishments, but not rewards; others fired rapidly when the mice detected a scent that meant that a reward was about to be given. The average level of reward or punishment the mice received also affected the baseline activity of many of the serotonergic neurons; this effect lasted for several minutes. Overall, Cohen et al. suggest that serotonergic neurons can affect how mice respond to rewards or punishments in both the short and long term. Future experiments should aim to understand the diversity of the signals that Cohen et al. observed, and to determine how these signals are used to drive behavior. Ultimately, understanding how neural circuits made up of different types of cells work may aid in understanding the neural basis of behavior. DOI:http://dx.doi.org/10.7554/eLife.06346.002
Collapse
Affiliation(s)
- Jeremiah Y Cohen
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, United States
| | - Mackenzie W Amoroso
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, United States
| | - Naoshige Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, United States
| |
Collapse
|
31
|
Prevention of alcohol-heightened aggression by CRF-R1 antagonists in mice: critical role for DRN-PFC serotonin pathway. Neuropsychopharmacology 2014; 39:2874-83. [PMID: 24917195 PMCID: PMC4200498 DOI: 10.1038/npp.2014.139] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 06/03/2014] [Accepted: 06/05/2014] [Indexed: 12/22/2022]
Abstract
Alcohol can escalate aggressive behavior in a significant subgroup of rodents, humans, and nonhuman primates. The present study investigated whether blockade of corticotropin-releasing factor receptor type 1 (CRF-R1) could prevent the emergence of alcohol-heightened aggression in mice. The serotonin (5-HT) pathway from the dorsal raphe nucleus (DRN) to the medial prefrontal cortex (mPFC) by CRF-R1 was investigated as a possible target for the prevention of alcohol-heightened aggressive behavior. Male CFW mice that reliably exhibited aggressive behaviors after consuming 1 g/kg of alcohol received systemic or intra-DRN administration of CRF-R1 antagonists, CP-154,526 or MTIP, before a confrontation with a male conspecific. Blockade of DRN CRF-R1 receptors with both antagonists significantly reduced only alcohol-heightened aggression, whereas systemic administration reduced both alcohol-heightened and species-typical aggression. Next, a 5-HT1A agonist, 8-OH-DPAT, was coadministered with CP-154,526 into the DRN to temporarily disrupt 5-HT activity. This manipulation abolished the antiaggressive effects of intra-DRN CP-154,526. In the mPFC, in vivo microdialysis revealed that extracellular 5-HT levels were increased in mice that consumed alcohol and were then injected with CP-154,526, both systemically or intra-DRN. Neither alcohol nor CP-154,526 alone affected 5-HT release in the mPFC. The present results suggest the DRN as a critical site for CRF-R1 to modulate alcohol-heightened aggression via action on the serotonergic DRN-PFC pathway.
Collapse
|
32
|
Paul ED, Johnson PL, Shekhar A, Lowry CA. The Deakin/Graeff hypothesis: focus on serotonergic inhibition of panic. Neurosci Biobehav Rev 2014; 46 Pt 3:379-96. [PMID: 24661986 PMCID: PMC4170046 DOI: 10.1016/j.neubiorev.2014.03.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 02/15/2014] [Accepted: 03/01/2014] [Indexed: 12/20/2022]
Abstract
The Deakin/Graeff hypothesis proposes that different subpopulations of serotonergic neurons through topographically organized projections to forebrain and brainstem structures modulate the response to acute and chronic stressors, and that dysfunction of these neurons increases vulnerability to affective and anxiety disorders, including panic disorder. We outline evidence supporting the existence of a serotonergic system originally discussed by Deakin/Graeff that is implicated in the inhibition of panic-like behavioral and physiological responses. Evidence supporting this panic inhibition system comes from the following observations: (1) serotonergic neurons located in the 'ventrolateral dorsal raphe nucleus' (DRVL) as well as the ventrolateral periaqueductal gray (VLPAG) inhibit dorsal periaqueductal gray-elicited panic-like responses; (2) chronic, but not acute, antidepressant treatment potentiates serotonin's panicolytic effect; (3) contextual fear activates a central nucleus of the amygdala-DRVL/VLPAG circuit implicated in mediating freezing and inhibiting panic-like escape behaviors; (4) DRVL/VLPAG serotonergic neurons are central chemoreceptors and modulate the behavioral and cardiorespiratory response to panicogenic agents such as sodium lactate and CO2. Implications of the panic inhibition system are discussed.
Collapse
Affiliation(s)
- Evan D Paul
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - Philip L Johnson
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - Anantha Shekhar
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - Christopher A Lowry
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| |
Collapse
|
33
|
Li H, Scholl JL, Tu W, Hassell JE, Watt MJ, Forster GL, Renner KJ. Serotonergic responses to stress are enhanced in the central amygdala and inhibited in the ventral hippocampus during amphetamine withdrawal. Eur J Neurosci 2014; 40:3684-92. [PMID: 25234335 DOI: 10.1111/ejn.12735] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 08/05/2014] [Accepted: 08/20/2014] [Indexed: 12/26/2022]
Abstract
Withdrawal from amphetamine increases anxiety and reduces the ability to cope with stress, which are factors that are believed to contribute to drug relapse. Stress-induced serotonergic transmission in the central nucleus of the amygdala is associated with anxiety states and fear. Conversely, stress-induced increases in ventral hippocampal serotonin (5-HT) levels have been linked to coping mechanisms. The goal of this study was to investigate the neurobiological changes induced by amphetamine that contribute to stress sensitivity during withdrawal. We tested the hypothesis that limbic serotonergic responses to restraint stress would be altered in male Sprague-Dawley rats chronically pretreated with amphetamine (2.5 mg/kg, intraperitoneal) and then subjected to 2 weeks of withdrawal. Amphetamine withdrawal resulted in increased stress-induced behavioral arousal relative to control treatment, suggesting that drug withdrawal induced greater sensitivity to the stressor. When microdialysis was used to determine the effects of restraint on extracellular 5-HT, stress-induced increases in 5-HT levels were abolished in the ventral hippocampus and augmented in the central amygdala during amphetamine withdrawal. Reverse dialysis of the glucocorticoid receptor antagonist mifepristone into the ventral hippocampus blocked the stress-induced increase in 5-HT levels in saline-pretreated rats, suggesting that glucocorticoid receptors mediate stress-induced increases in 5-HT levels in the ventral hippocampus. However, mifepristone had no effect on stress-induced increases in 5-HT levels in the central amygdala, indicating that stress increases 5-HT levels in this region independently of glucocorticoid receptors. During amphetamine withdrawal, the absence of stress-induced increases in ventral hippocampal 5-HT levels combined with enhanced stress-induced serotonergic responses in the central amygdala may contribute to drug relapse by decreasing stress-coping ability and heightening stress responsiveness.
Collapse
Affiliation(s)
- Hao Li
- Department of Biology & Center for Brain and Behavior Research, University of South Dakota, 414 E. Clark St, Vermillion, SD, 57069, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Magableh A, Lundy R. Somatostatin and corticotrophin releasing hormone cell types are a major source of descending input from the forebrain to the parabrachial nucleus in mice. Chem Senses 2014; 39:673-82. [PMID: 25086873 DOI: 10.1093/chemse/bju038] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The pontine parabrachial nucleus (PBN) receives substantial descending input from higher order forebrain regions that exerts inhibitory and excitatory influences on taste-evoked responses. Somatostatin (Sst) and corticotrophin releasing hormone (Crh) reporter mice were used in conjunction with injection of the retrograde tracer CTb-488 into the caudal PBN to determine the extent to which Sst and Crh cell types contribute to the descending pathways originating in the lateral hypothalamus (LH), central nucleus of the amygdala (CeA), bed nucleus of the stria terminalis (BNST), and insular cortex (IC). Five to 7 days following injections, the animals were euthanized and tissue sections prepared for confocal microscopy. Crh cell types in each forebrain site except IC project to the PBN with the greatest percentage originating in the BNST. For Sst cell types, the largest percentage of double-labeled cells was found in the CeA followed by the BNST. Few retrogradely labeled cells in the LH coexpressed Sst, whereas no double-labeled cells were observed in IC. The present results suggest that Sst and Crh cell types are a substantial component of the descending pathways from the amygdala and/or BNST to the PBN and are positioned to exert neuromodulatory effects on central taste processing.
Collapse
Affiliation(s)
- Ali Magableh
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Robert Lundy
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
35
|
Howerton AR, Roland AV, Fluharty JM, Marshall A, Chen A, Daniels D, Beck SG, Bale TL. Sex differences in corticotropin-releasing factor receptor-1 action within the dorsal raphe nucleus in stress responsivity. Biol Psychiatry 2014; 75:873-83. [PMID: 24289884 PMCID: PMC3997756 DOI: 10.1016/j.biopsych.2013.10.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/26/2013] [Accepted: 10/16/2013] [Indexed: 11/19/2022]
Abstract
BACKGROUND Women are twice as likely as men to suffer from stress-related affective disorders. Corticotropin-releasing factor (CRF) is an important link between stress and mood, in part through its signaling in the serotonergic dorsal raphe (DR). Development of CRF receptor-1 (CRFr1) antagonists has been a focus of numerous clinical trials but has not yet been proven efficacious. We hypothesized that sex differences in CRFr1 modulation of DR circuits might be key determinants in predicting therapeutic responses and affective disorder vulnerability. METHODS Male and female mice received DR infusions of the CRFr1 antagonist, NBI 35965, or CRF and were evaluated for stress responsivity. Sex differences in indices of neural activation (cFos) and colocalization of CRFr1 throughout the DR were examined. Whole-cell patch-clamp electrophysiology assessed sex differences in serotonin neuron membrane characteristics and responsivity to CRF. RESULTS Males showed robust behavioral and hypothalamic-pituitary-adrenal axis responses to DR infusion of NBI 35965 and CRF, whereas females were minimally responsive. Sex differences were also found for both CRF-induced DR cFos and CRFr1 co-localization throughout the DR. Electrophysiologically, female serotonergic neurons showed blunted membrane excitability and divergent inhibitory postsynaptic current responses to CRF application. CONCLUSIONS These studies demonstrate convincing sex differences in CRFr1 activity in the DR, where blunted female responses to NBI 35965 and CRF suggest unique stress modulation of the DR. These sex differences might underlie affective disorder vulnerability and differential sensitivity to pharmacologic treatments developed to target the CRF system, thereby contributing to a current lack of CRFr1 antagonist efficacy in clinical trials.
Collapse
Affiliation(s)
- Alexis R Howerton
- Department of Animal Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alison V Roland
- Department of Animal Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jessica M Fluharty
- Department of Animal Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anikò Marshall
- Department of Psychology, University at Buffalo, State University of New York, Buffalo, New York
| | - Alon Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Derek Daniels
- Department of Psychology, University at Buffalo, State University of New York, Buffalo, New York
| | - Sheryl G Beck
- Department of Anesthesia, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Tracy L Bale
- Department of Animal Biology, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
36
|
Carpenter RE, Maruska KP, Becker L, Fernald RD. Social opportunity rapidly regulates expression of CRF and CRF receptors in the brain during social ascent of a teleost fish, Astatotilapia burtoni. PLoS One 2014; 9:e96632. [PMID: 24824619 PMCID: PMC4019471 DOI: 10.1371/journal.pone.0096632] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 04/08/2014] [Indexed: 01/12/2023] Open
Abstract
In social animals, hierarchical rank governs food availability, territorial rights and breeding access. Rank order can change rapidly and typically depends on dynamic aggressive interactions. Since the neuromodulator corticotrophin releasing factor (CRF) integrates internal and external cues to regulate the hypothalamic-pituitary adrenal (HPA) axis, we analyzed the CRF system during social encounters related to status. We used a particularly suitable animal model, African cichlid fish, Astatotilapia burtoni, whose social status regulates reproduction. When presented with an opportunity to rise in rank, subordinate A. burtoni males rapidly change coloration, behavior, and their physiology to support a new role as dominant, reproductively active fish. Although changes in gonadotropin-releasing hormone (GnRH1), the key reproductive molecular actor, have been analyzed during social ascent, little is known about the roles of CRF and the HPA axis during transitions. Experimentally enabling males to ascend in social rank, we measured changes in plasma cortisol and the CRF system in specific brain regions 15 minutes after onset of social ascent. Plasma cortisol levels in ascending fish were lower than subordinate conspecifics, but similar to levels in dominant animals. In the preoptic area (POA), where GnRH1 cells are located, and in the pituitary gland, CRF and CRF1 receptor mRNA levels are rapidly down regulated in ascending males compared to subordinates. In the Vc/Vl, a forebrain region where CRF cell bodies are located, mRNA coding for both CRFR1 and CRFR2 receptors is lower in ascending fish compared to stable subordinate conspecifics. The rapid time course of these changes (within minutes) suggests that the CRF system is involved in the physiological changes associated with shifts in social status. Since CRF typically has inhibitory effects on the neuroendocrine reproductive axis in vertebrates, this attenuation of CRF activity may allow rapid activation of the reproductive axis and facilitate the transition to dominance.
Collapse
Affiliation(s)
- Russ E. Carpenter
- Biology Department, Stanford University, Stanford, California, United States of America
- * E-mail:
| | - Karen P. Maruska
- Biology Department, Stanford University, Stanford, California, United States of America
| | - Lisa Becker
- Biology Department, Stanford University, Stanford, California, United States of America
| | - Russell D. Fernald
- Biology Department, Stanford University, Stanford, California, United States of America
| |
Collapse
|
37
|
Tuckwell HC, Penington NJ. Computational modeling of spike generation in serotonergic neurons of the dorsal raphe nucleus. Prog Neurobiol 2014; 118:59-101. [PMID: 24784445 DOI: 10.1016/j.pneurobio.2014.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 04/14/2014] [Accepted: 04/21/2014] [Indexed: 01/14/2023]
Abstract
Serotonergic neurons of the dorsal raphe nucleus, with their extensive innervation of limbic and higher brain regions and interactions with the endocrine system have important modulatory or regulatory effects on many cognitive, emotional and physiological processes. They have been strongly implicated in responses to stress and in the occurrence of major depressive disorder and other psychiatric disorders. In order to quantify some of these effects, detailed mathematical models of the activity of such cells are required which describe their complex neurochemistry and neurophysiology. We consider here a single-compartment model of these neurons which is capable of describing many of the known features of spike generation, particularly the slow rhythmic pacemaking activity often observed in these cells in a variety of species. Included in the model are 11 kinds of ion channels: a fast sodium current INa, a delayed rectifier potassium current IKDR, a transient potassium current IA, a slow non-inactivating potassium current IM, a low-threshold calcium current IT, two high threshold calcium currents IL and IN, small and large conductance potassium currents ISK and IBK, a hyperpolarization-activated cation current IH and a leak current ILeak. In Sections 3-8, each current type is considered in detail and parameters estimated from voltage clamp data where possible. Three kinds of model are considered for the BK current and two for the leak current. Intracellular calcium ion concentration Cai is an additional component and calcium dynamics along with buffering and pumping is discussed in Section 9. The remainder of the article contains descriptions of computed solutions which reveal both spontaneous and driven spiking with several parameter sets. Attention is focused on the properties usually associated with these neurons, particularly long duration of action potential, steep upslope on the leading edge of spikes, pacemaker-like spiking, long-lasting afterhyperpolarization and the ramp-like return to threshold after a spike. In some cases the membrane potential trajectories display doublets or have humps or notches as have been reported in some experimental studies. The computed time courses of IA and IT during the interspike interval support the generally held view of a competition between them in influencing the frequency of spiking. Spontaneous activity was facilitated by the presence of IH which has been found in these neurons by some investigators. For reasonable sets of parameters spike frequencies between about 0.6Hz and 1.2Hz are obtained, but frequencies as high as 6Hz could be obtained with special parameter choices. Topics investigated and compared with experiment include shoulders, notches, anodal break phenomena, the effects of noradrenergic input, frequency versus current curves, depolarization block, effects of cell size and the effects of IM. The inhibitory effects of activating 5-HT1A autoreceptors are also investigated. There is a considerable discussion of in vitro versus in vivo firing behavior, with focus on the roles of noradrenergic input, corticotropin-releasing factor and orexinergic inputs. Location of cells within the nucleus is probably a major factor, along with the state of the animal.
Collapse
Affiliation(s)
- Henry C Tuckwell
- Max Planck Institute for Mathematics in the Sciences, Inselstr. 22, 04103 Leipzig, Germany; School of Electrical and Electronic Engineering, University of Adelaide, Adelaide, South Australia 5005, Australia.
| | - Nicholas J Penington
- Department of Physiology and Pharmacology, State University of New York, Downstate Medical Center, Box 29, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA; Program in Neural and Behavioral Science and Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Medical Center, Box 29, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA
| |
Collapse
|
38
|
Issler O, Carter RN, Paul ED, Kelly PA, Olverman HJ, Neufeld-Cohen A, Kuperman Y, Lowry CA, Seckl JR, Chen A, Jamieson PM. Increased anxiety in corticotropin-releasing factor type 2 receptor-null mice requires recent acute stress exposure and is associated with dysregulated serotonergic activity in limbic brain areas. BIOLOGY OF MOOD & ANXIETY DISORDERS 2014; 4:1. [PMID: 24447313 PMCID: PMC4029322 DOI: 10.1186/2045-5380-4-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 12/11/2013] [Indexed: 11/28/2022]
Abstract
Background Corticotropin-releasing factor type 2 receptors (CRFR2) are suggested to facilitate successful recovery from stress to maintain mental health. They are abundant in the midbrain raphe nuclei, where they regulate serotonergic neuronal activity and have been demonstrated to mediate behavioural consequences of stress. Here, we describe behavioural and serotonergic responses consistent with maladaptive recovery from stressful challenge in CRFR2-null mice. Results CRFR2-null mice showed similar anxiety levels to control mice before and immediately after acute restraint stress, and also after cessation of chronic stress. However, they showed increased anxiety by 24 hours after restraint, whether or not they had been chronically stressed. Serotonin (5-HT) and 5-hydroxyindoleacetic acid (5-HIAA) contents were quantified and the level of 5-HIAA in the caudal dorsal raphe nucleus (DRN) was increased under basal conditions in CRFR2-null mice, indicating increased 5-HT turnover. Twenty-four hours following restraint, 5-HIAA was decreased only in CRFR2-null mice, suggesting that they had not fully recovered from the challenge. In efferent limbic structures, CRFR2-null mice showed lower levels of basal 5-HT in the lateral septum and subiculum, and again showed a differential response to restraint stress from controls. Local cerebral glucose utilization (LCMRglu) revealed decreased neuronal activity in the DRN of CRFR2-null mice under basal conditions. Following 5-HT receptor agonist challenge, LCMRglu responses indicated that 5-HT1A receptor responses in the DRN were attenuated in CRFR2-null mice. However, postsynaptic 5-HT receptor responses in forebrain regions were intact. Conclusions These results suggest that CRFR2 are required for proper functionality of 5-HT1A receptors in the raphe nuclei, and are key to successful recovery from stress. This disrupted serotonergic function in CRFR2-null mice likely contributes to their stress-sensitive phenotype. The 5-HT content in lateral septum and subiculum was notably altered. These areas are important for anxiety, and are also implicated in reward and the pathophysiology of addiction. The role of CRFR2 in stress-related psychopathologies deserves further consideration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Pauline M Jamieson
- Centre for Cardiovascular Science, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
39
|
Brambilla R. Using Viral-Mediated Gene Transfer to Study Depressive-Like Behavior. NEUROMETHODS 2014. [PMCID: PMC7122802 DOI: 10.1007/978-1-62703-610-8_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The use of viral-mediated gene transfer in animal behavioral experiments has become very popular over the last decade. Altering gene regulation and assessing subsequent behavior can be a useful tool in unraveling a specific gene’s contributions to complex behavioral processes such as depressive-like behavior. However, these types of experiments require detailed planning and designs to avoid pitfalls associated with issues such as surgical procedure or the peak of viral expression. This chapter is intended to be a primer on the design of such experiments and aims to discuss factors that must be considered in the early phases of experimental planning.
Collapse
Affiliation(s)
- Riccardo Brambilla
- Institute of Experimental Neurology, San Raffaele Scientific Institute Division of Neuroscience, Milan, Italy
| |
Collapse
|
40
|
Bethea CL, Reddy AP. The effect of long-term ovariectomy on midbrain stress systems in free ranging macaques. Brain Res 2012; 1488:24-37. [PMID: 23036275 PMCID: PMC3501558 DOI: 10.1016/j.brainres.2012.09.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 09/21/2012] [Accepted: 09/22/2012] [Indexed: 12/29/2022]
Abstract
Communication between the serotonin system and the CRF system plays a pivotal role in the mediation of stress and stress reactivity. CRF appears to be inhibitory of serotonin neurotransmission through the CRF receptor type 1 (CRF-R1). Serotonin neurons also detect the urocortins, which are thought to be anxiolytic. Components of the CRF system in the serotonergic dorsal raphe region were examined in macaques that were ovary-intact or ovariectomized for 3 years living in a relatively natural environment. Female Japanese macaques (Macaca fuscata) were ovariectomized or tubal-ligated (n=5/group) and returned to their natal troop for 3 years. Quantitation of (1) CRF innervation of the serotonergic dorsal raphe, (2) CRF-Receptor type 1 (CRF-R1) in the dorsal raphe, (3) Urocortin 1 (UCN1) cells near the Edinger-Westfal nucleus and (4) UCN1 axons, was obtained with immunocytochemical staining and image analysis. There was no statistical difference in CRF axonal staining in the dorsal raphe, or in UCN1 axonal staining near the dorsal raphe. However, the average number of detectable UCN1 postive cells was significantly lower in the Ovx group than in the Intact group (p=0.003). Average CRF-R1 positive pixel number and positive cell number were significantly higher in the Ovx group than in the Intact group (p=0.005 and 0.02, respectivly). The higher expression of CRF-R1 and lower expression of UCN1 in the Ovx group indicates they may be more vulnerable to stress. The greater expression of CRF-R1 could cause a greater inhibition of serotonin upon a stress-induced increase in CRF as well.
Collapse
Affiliation(s)
- Cynthia L Bethea
- Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, USA.
| | | |
Collapse
|
41
|
Hazra R, Guo JD, Dabrowska J, Rainnie DG. Differential distribution of serotonin receptor subtypes in BNST(ALG) neurons: modulation by unpredictable shock stress. Neuroscience 2012; 225:9-21. [PMID: 22922122 DOI: 10.1016/j.neuroscience.2012.08.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 07/30/2012] [Accepted: 08/09/2012] [Indexed: 11/17/2022]
Abstract
The bed nucleus of the stria terminalis (BNST) plays a critical role in regulating the behavioral response to stress. Stressors that activate the BNST also activate serotonergic (5-HT) systems. Hence, maladaptive changes of 5-HT receptor expression may contribute to stress-induced anxiety disorders. The BNST contains three neuronal types, Type I-III neurons. However, little is known about 5-HT receptor subtypes mRNA expression in these neurons, or whether it can be modulated by stress. Whole-cell patch clamp recording from Type I-III neurons was used in conjunction with single cell reverse transcriptase polymerase chain reaction (RT-PCR) to characterize 5-HT receptor mRNA expression, and examine the effects of stress on this expression. We report that Type I neurons expressed mRNA transcripts predominantly for 5-HT(1A) and 5-HT(7) receptors. Type II neurons expressed transcripts for every 5-HT receptor except the 5-HT(2C) receptor. Type II neurons were divided into three sub-populations: Type IIA in which transcripts for 5-HT(3) and 5-HT(7) receptors predominate, Type IIB that mainly express 5-HT(1B) and 5-HT(4) receptor transcripts, and Type IIC in which transcripts for 5-HT(1A) and 5-HT(2A) receptors predominate. Type III neurons were also subdivided into two sub-populations; one that predominantly expressed transcripts for 5-HT(1A), 5-HT(1B) and 5-HT(2A) receptors, and another that mainly expressed transcripts for 5-HT(2C) receptor. Unpredictable shock stress (USS) caused a long-lasting increase in anxiety-like behavior, and a concomitant decrease in 5-HT(1A) transcript expression in Type I-III neurons, as well as an up-regulation of a transcriptional repressor of 5-HT(1A) gene expression, deformed epidermal autoregulatory factor 1 (Deaf-1). Significantly USS decreased 5-HT(1A) protein level, and increased the level of Deaf-1. USS also increased 5-HT(1B) transcript expression in Type III neurons, as well as 5-HT(7) expression in Type I and II neurons. These data suggest that cell type-specific disruption of 5-HT receptor expression in BNST(ALG) neurons may contribute to stress-induced anxiety disorders.
Collapse
Affiliation(s)
- R Hazra
- Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, GA, USA
| | | | | | | |
Collapse
|
42
|
Bambico FR, Hattan PR, Garant JP, Gobbi G. Effect of delta-9-tetrahydrocannabinol on behavioral despair and on pre- and postsynaptic serotonergic transmission. Prog Neuropsychopharmacol Biol Psychiatry 2012; 38:88-96. [PMID: 22386778 DOI: 10.1016/j.pnpbp.2012.02.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 02/02/2012] [Accepted: 02/13/2012] [Indexed: 01/14/2023]
Abstract
Preclinical and clinical studies suggest that direct and indirect cannabinoid agonists, including enhancers of endocannabinoids, engender stress-relieving, anxiolytic and antidepressant effects, mediated by central CB(1) receptors (CB(1)Rs). The effect of the main pharmacologically active principle in cannabis, (-)-trans-Δ(9)-tetrahydrocannabinol (delta-9-THC), on depressive behavior and on the serotonin (5-HT) system, which is implicated in the mechanism of action of antidepressants, has not been extensively clarified. Here, we showed that repeated (5 days), but not single (acute) intraperitoneal (ip) treatment with delta-9-THC (1mg/kg) exerts antidepressant-like properties in the rat forced swim test (FST). This effect was CB(1)R-dependent because it was blocked by the CB(1)R antagonist rimonabant (1mg/kg, ip). Using in vivo electrophysiology, we demonstrated that delta-9-THC modulated dorsal raphe (DR) 5-HT neuronal activity through a CB(1)R-dependent mechanism. Acute intravenous delta-9-THC administration (0.1-1.5mg/kg) elicited a complex response profile, producing excitatory, inhibitory and inert responses of 5-HT neurons. Only excitatory responses were blocked by rimonabant. Finally, repeated but not single delta-9-THC administration (1mg/kg, ip) enhanced tonic 5-HT(1A) receptor activity in the hippocampus, a postsynaptic event commonly elicited by standard antidepressants. These results suggest that delta-9-THC, like other CB(1)R agonists and endocannabinoid enhancers, may possess antidepressant properties at low doses, and could modulate 5-HT transmission in the DR and hippocampus as standard antidepressants such as selective serotonin reuptake inhibitors.
Collapse
|
43
|
Jasinska AJ, Lowry CA, Burmeister M. Serotonin transporter gene, stress and raphe-raphe interactions: a molecular mechanism of depression. Trends Neurosci 2012; 35:395-402. [PMID: 22301434 DOI: 10.1016/j.tins.2012.01.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2011] [Revised: 12/02/2011] [Accepted: 01/01/2012] [Indexed: 12/16/2022]
Abstract
Reports of gene-environment interactions (GxE) between the serotonin transporter gene and stress on risk of depression have generated both excitement and controversy. The controversy persists in part because a mechanistic account of this GxE on serotonergic neurotransmission and risk of depression has been lacking. In this Opinion, we draw on recent discoveries in the functional neuroanatomy of the serotonergic dorsal raphe nucleus (DR) to propose such a mechanistic account. We argue that genetically produced variability in serotonin reuptake during stressor-induced raphe-raphe interactions alters the balance in the amygdala-ventromedial prefrontal cortex (VMPFC)-DR circuitry underlying stressor reactivity and emotion regulation. In particular, the recently characterized stressor-responsive serotonergic interneurons originating from the dorsolateral DR may hold a key to unlocking the GxE mechanism of depression.
Collapse
Affiliation(s)
- Agnes J Jasinska
- Department of Psychology, University of Michigan, Ann Arbor, Michigan, USA.
| | | | | |
Collapse
|
44
|
Staub D, Lunden J, Cathel A, Dolben E, Kirby L. Morphine history sensitizes postsynaptic GABA receptors on dorsal raphe serotonin neurons in a stress-induced relapse model in rats. Psychoneuroendocrinology 2012; 37:859-70. [PMID: 22047957 PMCID: PMC3319501 DOI: 10.1016/j.psyneuen.2011.10.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2011] [Revised: 09/02/2011] [Accepted: 10/05/2011] [Indexed: 01/05/2023]
Abstract
The serotonin (5-hydroxytryptamine, 5-HT) system plays an important role in stress-related psychiatric disorders and substance abuse. Previous work has shown that the dorsal raphe nucleus (DR)-5-HT system is inhibited by swim stress via stimulation of GABA synaptic activity by the stress neurohormone corticotropin-releasing factor (CRF). Additionally, the DR 5-HT system is regulated by opioids. The present study tests the hypothesis that the DR 5-HT system regulates stress-induced opioid relapse. In the first experiment, electrophysiological recordings of GABA synaptic activity in 5-HT DR neurons were conducted in brain slices from Sprague-Dawley rats that were exposed to swim stress-induced reinstatement of previously extinguished morphine conditioned place preference (CPP). Behavioral data indicate that swim stress triggers reinstatement of morphine CPP. Electrophysiology data indicate that 5-HT neurons in the morphine-conditioned group exposed to stress had increased amplitude of inhibitory postsynaptic currents (IPSCs), which would indicate greater postsynaptic GABA receptor density and/or sensitivity, compared to saline controls exposed to stress. In the second experiment, rats were exposed to either morphine or saline CPP and extinction, and then 5-HT DR neurons from both groups were examined for sensitivity to CRF in vitro. CRF induced a greater inward current in 5-HT neurons from morphine-conditioned subjects compared to saline-conditioned subjects. These data indicate that morphine history sensitizes 5-HT DR neurons to the GABAergic inhibitory effects of stress as well as to some of the effects of CRF. These mechanisms may sensitize subjects with a morphine history to the dysphoric effects of stressors and ultimately confer an enhanced vulnerability to stress-induced opioid relapse.
Collapse
Affiliation(s)
| | | | | | | | - L.G. Kirby
- Corresponding Author: Lynn G. Kirby, Ph.D., Center for Substance Abuse Research, Temple University School of Medicine, 3400 N. Broad St., Philadelphia, PA 19140, (215) 707-8566 (phone), (215) 707-9468 (fax)
| |
Collapse
|
45
|
Zorrilla EP, Wee S, Zhao Y, Specio S, Boutrel B, Koob GF, Weiss F. Extended access cocaine self-administration differentially activates dorsal raphe and amygdala corticotropin-releasing factor systems in rats. Addict Biol 2012; 17:300-8. [PMID: 21762287 DOI: 10.1111/j.1369-1600.2011.00329.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cocaine-induced neuroadaptation of stress-related circuitry and increased access to cocaine each putatively contribute to the transition from cocaine use to cocaine dependence. The present study tested the hypothesis that rats receiving extended versus brief daily access to cocaine would exhibit regional differences in levels of the stress-regulatory neuropeptide corticotropin-releasing factor (CRF). A secondary goal was to explore how CRF levels change in relation to the time since cocaine self-administration. Male Wistar rats acquired operant self-administration of cocaine and were assigned to receive daily long access (6 hours/day, LgA, n=20) or short access (1 hour/day, ShA, n=18) to intravenous cocaine self-administration (fixed ratio 1, ~0.50 mg/kg/infusion). After at least 3 weeks, tissue CRF immunoreactivity was measured at one of three timepoints: pre-session, post-session or 3 hours post-session. LgA, but not ShA, rats showed increased total session and first-hour cocaine intake. CRF immunoreactivity increased within the dorsal raphe (DR) and basolateral, but not central, nucleus of the amygdala (BLA, CeA) of ShA rats from pre-session to 3 hours post-session. In LgA rats, CRF immunoreactivity increased from pre-session to 3 hours post-session within the CeA and DR but tended to decrease in the BLA. LgA rats showed higher CRF levels than ShA rats in the DR and, pre-session, in the BLA. Thus, voluntary cocaine intake engages stress-regulatory CRF systems of the DR and amygdala. Increased availability of cocaine promotes greater tissue CRF levels in these extrahypothalamic brain regions, changes associated here with a model of cocaine dependence.
Collapse
Affiliation(s)
- Eric P Zorrilla
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Kubota N, Amemiya S, Motoki C, Otsuka T, Nishijima T, Kita I. Corticotropin-releasing factor antagonist reduces activation of noradrenalin and serotonin neurons in the locus coeruleus and dorsal raphe in the arousal response accompanied by yawning behavior in rats. Neurosci Res 2012; 72:316-23. [PMID: 22285921 DOI: 10.1016/j.neures.2012.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 12/20/2011] [Accepted: 01/04/2012] [Indexed: 12/11/2022]
Abstract
We previously reported that intracerebroventricular (icv) administration of corticotropin-releasing factor (CRF) antagonist attenuates the arousal response during yawning behavior in rats. However, the CRF-related pathway involved in the arousal response during yawning is still unclear. In the present study, we assessed the involvement of the CRF-containing pathway from the hypothalamic paraventricular nucleus (PVN) to the locus coeruleus (LC) and the dorsal raphe nucleus (DRN) in the arousal response during frequent spontaneous yawning, which was induced by several microinjections of l-glutamate into the PVN in anesthetized rats, using c-Fos immunohistochemistry. The PVN stimulation showed significant increases in activation of PVN CRF neurons, LC noradrenalin (NA) neurons and DRN serotonin (5-HT) neurons as well as arousal response during yawning. But icv administration of a CRF receptor antagonist, α-helical CRF (9-41), significantly inhibited the activation of both LC NA neurons and DRN 5-HT neurons except the activation of CRF neurons in the PVN, and significantly suppressed the arousal response during yawning. These results suggest that the CRF-containing pathway from PVN CRF neurons to LC NA neurons and DRN 5-HT neurons can be involved in the arousal response during yawning behavior.
Collapse
Affiliation(s)
- Natsuko Kubota
- Department of Human Health Science, Tokyo Metropolitan University, Japan
| | | | | | | | | | | |
Collapse
|
47
|
Hubbard CS, Rose JD. Brainstem neuronal and behavioral activation by corticotropin-releasing hormone depend on the behavioral state of the animal. Horm Behav 2012; 61:121-33. [PMID: 22137972 PMCID: PMC4465356 DOI: 10.1016/j.yhbeh.2011.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 11/03/2011] [Accepted: 11/09/2011] [Indexed: 10/15/2022]
Abstract
Central administration of corticotropin-releasing hormone (CRH) is known to enhance locomotion across a wide range of vertebrates, including the roughskin newt, Taricha granulosa. The present study aimed to identify the CRH effects on locomotor-controlling medullary neurons that underlie the peptide's behavioral stimulating actions. Single neurons were recorded from the rostral medullary reticular formation before and after intraventricular infusion of CRH in freely behaving newts and newts paralyzed with a myoneural blocking agent. In behaving newts, most medullary neurons showed increased firing 3-23 min after CRH infusion. Decreases in firing were less common. Of particular importance was the finding that in behaving newts, medullary neurons showed a cyclic firing pattern that was strongly associated with an increase in the incidence of walking bouts, an effect blocked by pretreatment with the CRH antagonist, alpha-helical CRH and not seen following vehicle administration. In contrast, the majority of medullary neurons sampled in immobilized newts lacked temporal cyclicity in their firing patterns following intraventricular infusion of CRH. That is, there was no evidence for a fictive locomotor activity pattern. Our results indicate that the actual expression of locomotion is a critical factor in regulating the behavior-activating effects of CRH and underscore the importance of using an awake, unrestrained animal for analysis of a hormone's neurobehavioral actions.
Collapse
|
48
|
Waselus M, Valentino RJ, Van Bockstaele EJ. Collateralized dorsal raphe nucleus projections: a mechanism for the integration of diverse functions during stress. J Chem Neuroanat 2011; 41:266-80. [PMID: 21658442 PMCID: PMC3156417 DOI: 10.1016/j.jchemneu.2011.05.011] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 05/18/2011] [Accepted: 05/20/2011] [Indexed: 01/01/2023]
Abstract
The midbrain dorsal raphe nucleus (DR) is the origin of the central serotonin (5-HT) system, a key neurotransmitter system that has been implicated in the expression of normal behaviors and in diverse psychiatric disorders, particularly affective disorders such as depression and anxiety. One link between the DR-5-HT system and affective disorders is exposure to stressors. Stress is a major risk factor for affective disorders, and stressors alter activity of DR neurons in an anatomically specific manner. Stress-induced changes in DR neuronal activity are transmitted to targets of the DR via ascending serotonergic projections, many of which collateralize to innervate multiple brain regions. Indeed, the collateralization of DR efferents allows for the coordination of diverse components of the stress response. This review will summarize our current understanding of the organization of the ascending DR system and its collateral projections. Using the neuropeptide corticotropin-releasing factor (CRF) system as an example of a stress-related initiator of DR activity, we will discuss how topographic specificity of afferent regulation of ascending DR circuits serves to coordinate activity in functionally diverse target regions under appropriate conditions.
Collapse
Affiliation(s)
- Maria Waselus
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States.
| | | | | |
Collapse
|
49
|
Bethea CL, Lima FB, Centeno ML, Weissheimer KV, Senashova O, Reddy AP, Cameron JL. Effects of citalopram on serotonin and CRF systems in the midbrain of primates with differences in stress sensitivity. J Chem Neuroanat 2011; 41:200-18. [PMID: 21683135 DOI: 10.1016/j.jchemneu.2011.05.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 05/02/2011] [Accepted: 05/16/2011] [Indexed: 10/24/2022]
Abstract
This chapter reviews the neurobiological effects of stress sensitivity and s-citalpram (CIT) treatment observed in our nonhuman primate model of functional hypothalamic amenorrhea (FHA). This type of infertility, also known as stress-induced amenorrhea, is exhibited by cynomolgus macaques. In small populations, some individuals are stress-sensitive (SS) and others are highly stress-resilient (HSR). The SS macaques have suboptimal secretion of estrogen and progesterone during normal menstrual cycles. SS monkeys also have decreased serotonin gene expression and increased CRF expression compared to HSR monkeys. Recently, we found that CIT treatment improved ovarian steroid secretion in SS monkeys, but had no effect in HSR monkeys. Examination of the serotonin system revealed that SS monkeys had significantly lower Fev (fifth Ewing variant, rodent Pet1), TPH2 (tryptophan hydroxylase 2), 5HT1A autoreceptor and SERT (serotonin reuptake transporter) expression in the dorsal raphe than SR monkeys. However, CIT did not alter the expression of either Fev, TPH2, SERT or 5HT1A mRNAs. In contrast, SS monkeys tended to have a higher density of CRF fiber innervation of the dorsal raphe than HSR monkeys, and CIT significantly decreased the CRF fiber density in SS animals. In addition, CIT increased CRF-R2 gene expression in the dorsal raphe. We speculate that in a 15-week time frame, the therapeutic effect of S-citalopram may be achieved through a mechanism involving extracellular serotonin inhibition of CRF and stimulation of CRF-R2, rather than alteration of serotonin-related gene expression.
Collapse
Affiliation(s)
- Cynthia L Bethea
- Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, United States.
| | | | | | | | | | | | | |
Collapse
|
50
|
Backström T, Pettersson A, Johansson V, Winberg S. CRF and urotensin I effects on aggression and anxiety-like behavior in rainbow trout. ACTA ACUST UNITED AC 2011; 214:907-14. [PMID: 21346117 DOI: 10.1242/jeb.045070] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Corticotropin-releasing factor (CRF) is central in the stress response but also modulates several behaviors including anxiety-related behaviors and aggression. In this study, juvenile rainbow trout (Oncorhynchus mykiss) were tested for competitive ability, determined during dyadic fights for dominance, after intracerebroventricular (i.c.v.) administration of CRF, urotensin I (UI), the non-specific CRF antagonist α-helical RF(9-41) (ahCRF) or the CRF receptor subtype 1-specific antagonist antalarmin, when paired with a mass-matched con-specific injected with saline. In addition, isolated fish received the same substances. Plasma cortisol and brain monoamines were monitored in all fish. Most fish receiving CRF showed a conspicuous behavior consisting of flaring the opercula, opening the mouth and violent shaking of the head from side to side. When this occurred, the fish immediately forfeited the fight. Similar behavior was observed in most fish receiving UI but no effect on outcome of dyadic fights was noted. This behavior seems similar to non-ambulatory motor activity seen in rats and could be anxiety related. Furthermore, fish receiving CRF at a dose of 1000 ng became subordinate, whereas all other treatments had no effects on the outcome of dyadic fights. In addition, isolated fish receiving ahCRF had lower brain stem concentrations of 5-hydroxyindoleacetic acid, serotonin, 3,4-dihydroxyphenylacetic acid and dopamine. In conclusion, CRF seems to attenuate competitive ability, and both CRF and UI seem to induce anxiety-like behavior.
Collapse
Affiliation(s)
- Tobias Backström
- Evolutionary Biology Centre, Comparative Physiology, Uppsala University, Norbyvägen 18A, Uppsala, Sweden
| | | | | | | |
Collapse
|