1
|
Woodburn SC, Levitt CM, Koester AM, Kwan AC. Psilocybin Facilitates Fear Extinction: Importance of Dose, Context, and Serotonin Receptors. ACS Chem Neurosci 2024; 15:3034-3043. [PMID: 39087917 DOI: 10.1021/acschemneuro.4c00279] [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: 08/02/2024] Open
Abstract
A variety of classic psychedelics and MDMA have been shown to enhance fear extinction in rodent models. This has translational significance because a standard treatment for post-traumatic stress disorder (PTSD) is prolonged exposure therapy. However, few studies have investigated psilocybin's potential effect on fear learning paradigms. More specifically, the extents to which dose, timing of administration, and serotonin receptors may influence psilocybin's effect on fear extinction are not understood. In this study, we used a delay fear conditioning paradigm to determine the effects of psilocybin on fear extinction, extinction retention, and fear renewal in male and female mice. Psilocybin robustly enhances fear extinction when given acutely prior to testing for all doses tested. Psilocybin also exerts long-term effects to elevate extinction retention and suppress fear renewal in a novel context, although these changes were sensitive to dose. Analysis of sex differences showed that females may respond to a narrower range of doses than males. Administration of psilocybin prior to fear learning or immediately after extinction yielded no change in behavior, indicating that concurrent extinction experience is necessary for the drug's effects. Cotreatment with a 5-HT2A receptor antagonist blocked psilocybin's effects for extinction, extinction retention, and fear renewal, whereas 5-HT1A receptor antagonism attenuated only the effect on fear renewal. Collectively, these results highlight dose, context, and serotonin receptors as crucial factors in psilocybin's ability to facilitate fear extinction. The study provides preclinical evidence to support investigating psilocybin as a pharmacological adjunct for extinction-based therapy for PTSD.
Collapse
Affiliation(s)
- Samuel C Woodburn
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Caleb M Levitt
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Allison M Koester
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Alex C Kwan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
- Department of Psychiatry, Weill Cornell Medicine, New York, New York 10065, United States
| |
Collapse
|
2
|
Cao H, Sun J, Hua Q, Huang T, Wei Y, Zhan Y, Yao X, Zhang T, Yang Y, Xu W, Bai T, Tian Y, Zhang L, Wang K, Ji GJ. Decreased inter-hemispheric cooperation in major depressive disorder and its association with neurotransmitter profiles. J Affect Disord 2024; 359:109-116. [PMID: 38768823 DOI: 10.1016/j.jad.2024.05.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Inter-hemispheric cooperation is a prominent feature of the human brain, and previous neuroimaging studies have revealed aberrant inter-hemispheric cooperation patterns in patients with major depressive disorder (MDD). Typically, inter-hemispheric cooperation is examined by calculating the functional connectivity (FC) between each voxel in one hemisphere and its anatomical (structurally homotopic) counterpart in the opposite hemisphere. However, bilateral hemispheres are actually asymmetric in anatomy. METHODS In the present study, we utilized connectivity between functionally homotopic voxels (CFH) to investigate abnormal inter-hemispheric cooperation in 96 MDD patients compared to 173 age- and sex-matched healthy controls (HCs). In addition, we analyzed the spatial correlations between abnormal CFH and the density maps of 13 neurotransmitter receptors and transporters. RESULTS The CFH values in bilateral orbital frontal gyri and bilateral postcentral gyri were abnormally decreased in patients with MDD. Furthermore, these CFH abnormalities were correlated with clinical symptoms. In addition, the abnormal CFH pattern in MDD patients was spatially correlated with the distribution pattern of 5-HT1AR. LIMITATIONS drug effect; the cross-sectional research design precludes causal inferences; the neurotransmitter atlases selected were constructed from healthy individuals rather than MDD patients. CONCLUSION These findings characterized the abnormal inter-hemispheric cooperation in MDD using a novel method and the underlying neurotransmitter mechanism, which promotes our understanding of the pathophysiology of depression.
Collapse
Affiliation(s)
- Hai Cao
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China; Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui Province, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Jinmei Sun
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Qiang Hua
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Tongqing Huang
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Yuqing Wei
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Yuqian Zhan
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Xiaoqing Yao
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Ting Zhang
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China; Department of Psychiatry, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Department of Psychology and Sleep Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yinian Yang
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Wenqiang Xu
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Tongjian Bai
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Yanghua Tian
- Department of Psychology and Sleep Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lei Zhang
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China.
| | - Kai Wang
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China; Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China; Anhui Institute of Translational Medicine, Hefei, China.
| | - Gong-Jun Ji
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China; Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui Province, China; Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China; Anhui Institute of Translational Medicine, Hefei, China.
| |
Collapse
|
3
|
Bickle JG, Li Y, Millette A, Dixon R, Wu S, Arias EC, Luna VM, Anacker C. 5-HT 1A Receptors on Dentate Gyrus Granule Cells Confer Stress Resilience. Biol Psychiatry 2024; 95:800-809. [PMID: 37863245 PMCID: PMC10978305 DOI: 10.1016/j.biopsych.2023.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/19/2023] [Accepted: 10/09/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND Hyperactivity of granule cells in the ventral dentate gyrus (vDG) promotes vulnerability to chronic stress. However, which receptors in the vDG could be targeted to inhibit this hyperactivity and confer stress resilience is not known. The serotonin 1A receptor (5-HT1AR) is a Gi protein-coupled inhibitory receptor that has been implicated in stress adaptation, anxiety, depression, and antidepressant responses. 5-HT1ARs are highly expressed in the DG, but their potential to promote stress resilience by regulating granule cell activity has never been examined. METHODS We exposed male and female mice expressing 5-HT1ARs only in DG granule cells to 10 days of chronic social defeat stress (CSDS) and treated them with the 5-HT1AR agonist 8-OH-DPAT every day 30 minutes before each defeat throughout the CSDS paradigm. We then used whole-cell current clamp recordings, immunohistochemistry for the immediate early gene cFos, corticosterone immunoassays, and behavioral testing to determine how activating 5-HT1ARs on granule cells affects DG activity, neuroendocrine stress responses, and avoidance behavior. RESULTS We found that activating 5-HT1ARs hyperpolarized DG granule cells and reduced cFos+ granule cells in the vDG following CSDS, indicating that 5-HT1AR activation rescued stress-induced vDG hyperactivity. Moreover, 5-HT1AR activation dampened corticosterone responses to CSDS and prevented the development of stress-induced avoidance in the social interaction test and in the open field test. CONCLUSIONS Our findings show that activating 5-HT1ARs on DG granule cells can prevent stress-induced neuronal hyperactivity of the vDG and confer resilience to chronic stress.
Collapse
Affiliation(s)
- John Gregory Bickle
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University and Research Foundation for Mental Hygiene, Inc., New York State Psychiatric Institute, New York, New York
| | - Yifei Li
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University and Research Foundation for Mental Hygiene, Inc., New York State Psychiatric Institute, New York, New York
| | - Amira Millette
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University and Research Foundation for Mental Hygiene, Inc., New York State Psychiatric Institute, New York, New York
| | - Rushell Dixon
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University and Research Foundation for Mental Hygiene, Inc., New York State Psychiatric Institute, New York, New York
| | - Serena Wu
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University and Research Foundation for Mental Hygiene, Inc., New York State Psychiatric Institute, New York, New York
| | - Elena Carazo Arias
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University and Research Foundation for Mental Hygiene, Inc., New York State Psychiatric Institute, New York, New York
| | - Victor Mari Luna
- Alzheimer's Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Christoph Anacker
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University and Research Foundation for Mental Hygiene, Inc., New York State Psychiatric Institute, New York, New York; Columbia University Institute for Developmental Sciences, Department of Psychiatry, Columbia University Irving Medical Center, New York, New York; Columbia University Stem Cell Initiative, Columbia University Irving Medical Center, New York, New York.
| |
Collapse
|
4
|
Chda A, Bencheikh R. Flavonoids as G Protein-coupled Receptors Ligands: New Potential Therapeutic Natural Drugs. Curr Drug Targets 2023; 24:1346-1363. [PMID: 38037994 DOI: 10.2174/0113894501268871231127105219] [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] [Received: 06/24/2023] [Revised: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023]
Abstract
G protein coupled receptors (GPCRs) are among the largest family of cell surface receptors found in the human genome. They govern a wide range of physiological responses in both health and diseases, making them one of the potential targeted surface receptors for pharmaceuticals. Flavonoids can modulate GPCRs activity by acting as allosteric ligands. They can either enhance or reduce the GPCR's effect. Emerging research shows that individual flavonoids or mixtures of flavonoids from plant extracts can have relevant pharmacological effects against a number of diseases, particularly by influencing GPCRs. In the present review, we are considering to give a comprehensive overview of flavonoids and related compounds that exhibit GPCRs activity and to further explore which beneficial structural features. Molecular docking was used to strengthen experimental evidence and describe flavonoid-GPCRs interactions at molecular level.
Collapse
Affiliation(s)
- Alae Chda
- Laboratory of Microbial Biotechnology and Bioactive Molecules (LM2BM), Faculty of Science and Technology, Sidi Mohamed Ben Abdellah University, Road of Immouzer, PO Box 2202, Fez, Morocco
- Higher Institute of Nursing and Health Techniques - Fez. Ministry of Health and Social Protection, Fez, Morocco
| | - Rachid Bencheikh
- Laboratory of Microbial Biotechnology and Bioactive Molecules (LM2BM), Faculty of Science and Technology, Sidi Mohamed Ben Abdellah University, Road of Immouzer, PO Box 2202, Fez, Morocco
| |
Collapse
|
5
|
Tanaka M, Szabó Á, Spekker E, Polyák H, Tóth F, Vécsei L. Mitochondrial Impairment: A Common Motif in Neuropsychiatric Presentation? The Link to the Tryptophan-Kynurenine Metabolic System. Cells 2022; 11:2607. [PMID: 36010683 PMCID: PMC9406499 DOI: 10.3390/cells11162607] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/14/2022] [Accepted: 08/19/2022] [Indexed: 02/07/2023] Open
Abstract
Nearly half a century has passed since the discovery of cytoplasmic inheritance of human chloramphenicol resistance. The inheritance was then revealed to take place maternally by mitochondrial DNA (mtDNA). Later, a number of mutations in mtDNA were identified as a cause of severe inheritable metabolic diseases with neurological manifestation, and the impairment of mitochondrial functions has been probed in the pathogenesis of a wide range of illnesses including neurodegenerative diseases. Recently, a growing number of preclinical studies have revealed that animal behaviors are influenced by the impairment of mitochondrial functions and possibly by the loss of mitochondrial stress resilience. Indeed, as high as 54% of patients with one of the most common primary mitochondrial diseases, mitochondrial encephalomyopathy with lactic acidosis and stroke-like episodes (MELAS) syndrome, present psychiatric symptoms including cognitive impairment, mood disorder, anxiety, and psychosis. Mitochondria are multifunctional organelles which produce cellular energy and play a major role in other cellular functions including homeostasis, cellular signaling, and gene expression, among others. Mitochondrial functions are observed to be compromised and to become less resilient under continuous stress. Meanwhile, stress and inflammation have been linked to the activation of the tryptophan (Trp)-kynurenine (KYN) metabolic system, which observably contributes to the development of pathological conditions including neurological and psychiatric disorders. This review discusses the functions of mitochondria and the Trp-KYN system, the interaction of the Trp-KYN system with mitochondria, and the current understanding of the involvement of mitochondria and the Trp-KYN system in preclinical and clinical studies of major neurological and psychiatric diseases.
Collapse
Affiliation(s)
- Masaru Tanaka
- ELKH-SZTE Neuroscience Research Group, Danube Neuroscience Research Laboratory, Eötvös Loránd Research Network, University of Szeged (ELKH-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - Ágnes Szabó
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, Korányi fasor 6, H-6720 Szeged, Hungary
| | - Eleonóra Spekker
- ELKH-SZTE Neuroscience Research Group, Danube Neuroscience Research Laboratory, Eötvös Loránd Research Network, University of Szeged (ELKH-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - Helga Polyák
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, Korányi fasor 6, H-6720 Szeged, Hungary
| | - Fanni Tóth
- ELKH-SZTE Neuroscience Research Group, Danube Neuroscience Research Laboratory, Eötvös Loránd Research Network, University of Szeged (ELKH-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - László Vécsei
- ELKH-SZTE Neuroscience Research Group, Danube Neuroscience Research Laboratory, Eötvös Loránd Research Network, University of Szeged (ELKH-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| |
Collapse
|
6
|
Hyperthermia and Serotonin: The Quest for a “Better Cyproheptadine”. Int J Mol Sci 2022; 23:ijms23063365. [PMID: 35328784 PMCID: PMC8952796 DOI: 10.3390/ijms23063365] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/18/2022] [Accepted: 03/18/2022] [Indexed: 02/04/2023] Open
Abstract
Fine temperature control is essential in homeothermic animals. Both hyper- and hypothermia can have deleterious effects. Multiple, efficient and partly redundant mechanisms of adjusting the body temperature to the value set by the internal thermostat exist. The neural circuitry of temperature control and the neurotransmitters involved are reviewed. The GABAergic inhibitory output from the brain thermostat in the preoptic area POA to subaltern neural circuitry of temperature control (Nucleus Raphe Dorsalis and Nucleus Raphe Pallidus) is a function of the balance between the (opposite) effects mediated by the transient receptor potential receptor TRPM2 and EP3 prostaglandin receptors. Activation of TRPM2-expressing neurons in POA favors hypothermia, while inhibition has the opposite effect. Conversely, EP3 receptors induce elevation in body temperature. Activation of EP3-expressing neurons in POA results in hyperthermia, while inhibition has the opposite effect. Agonists at TRPM2 and/or antagonists at EP3 could be beneficial in hyperthermia control. Activity of the neural circuitry of temperature control is modulated by a variety of 5-HT receptors. Based on the theoretical model presented the “ideal” antidote against serotonin syndrome hyperthermia appears to be an antagonist at the 5-HT receptor subtypes 2, 4 and 6 and an agonist at the receptor subtypes 1, 3 and 7. Very broadly speaking, such a profile translates in a sympatholytic effect. While a compound with such an ideal profile is presently not available, better matches than the conventional antidote cyproheptadine (used off-label in severe serotonin syndrome cases) appear to be possible and need to be identified.
Collapse
|
7
|
Lan MJ, Zanderigo F, Pantazatos SP, Sublette ME, Miller J, Ogden RT, Mann JJ. Serotonin 1A Receptor Binding of [11C]CUMI-101 in Bipolar Depression Quantified Using Positron Emission Tomography: Relationship to Psychopathology and Antidepressant Response. Int J Neuropsychopharmacol 2022; 25:534-544. [PMID: 34996114 PMCID: PMC9352178 DOI: 10.1093/ijnp/pyac001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/15/2021] [Accepted: 01/05/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The pathophysiology of bipolar disorder (BD) remains largely unknown despite it causing significant disability and suicide risk. Serotonin signaling may play a role in the pathophysiology, but direct evidence for this is lacking. Treatment of the depressed phase of the disorder is limited. Previous studies have indicated that positron emission tomography (PET) imaging of the serotonin 1A receptor (5HT1AR) may predict antidepressant response. METHODS A total of 20 participants with BD in a current major depressive episode and 16 healthy volunteers had PET imaging with [11C]CUMI-101, employing a metabolite-corrected input function for quantification of binding potential to the 5HT1AR. Bipolar participants then received an open-labeled, 6-week clinical trial with a selective serotonin reuptake inhibitor (SSRI) in addition to their mood stabilizer. Clinical ratings were obtained at baseline and during SSRI treatment. RESULTS Pretreatment binding potential (BPF) of [11C]CUMI-101 was associated with a number of pretreatment clinical variables within BD participants. Within the raphe nucleus, it was inversely associated with the baseline Montgomery Åsberg Rating Scale (P = .026), the Beck Depression Inventory score (P = .0023), and the Buss Durkee Hostility Index (P = .0058), a measure of lifetime aggression. A secondary analysis found [11C]CUMI-101 BPF was higher in bipolar participants compared with healthy volunteers (P = .00275). [11C]CUMI-101 BPF did not differ between SSRI responders and non-responders (P = .907) to treatment and did not predict antidepressant response (P = .580). Voxel-wise analyses confirmed the results obtained in regions of interest analyses. CONCLUSIONS A disturbance of serotonin system function is associated with both the diagnosis of BD and its severity of depression. Pretreatment 5HT1AR binding did not predict SSRI antidepressant outcome.The study was listed on clinicaltrials.gov with identifier NCT02473250.
Collapse
Affiliation(s)
- Martin J Lan
- Correspondence: Martin Lan, MD, PhD, 1051 Riverside Dr., Unit 42, New York, NY 10032, USA ()
| | - Francesca Zanderigo
- Department of Psychiatry, Vagelos College of Physicians and Surgeons at Columbia University, New York, NY, USA,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - Spiro P Pantazatos
- Department of Psychiatry, Vagelos College of Physicians and Surgeons at Columbia University, New York, NY, USA,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - M Elizabeth Sublette
- Department of Psychiatry, Vagelos College of Physicians and Surgeons at Columbia University, New York, NY, USA,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - Jeffrey Miller
- Department of Psychiatry, Vagelos College of Physicians and Surgeons at Columbia University, New York, NY, USA,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - R Todd Ogden
- Department of Psychiatry, Vagelos College of Physicians and Surgeons at Columbia University, New York, NY, USA,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - J John Mann
- Department of Psychiatry, Vagelos College of Physicians and Surgeons at Columbia University, New York, NY, USA,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA,Department of Radiology, Vagelos College of Physicians and Surgeons at Columbia University, New York, NY, USA
| |
Collapse
|
8
|
Xia B, Liu X, Li X, Wang Y, Wang D, Kou R, Zhang L, Shi R, Ye J, Bo X, Liu Q, Zhao B, Liu X. Sesamol Ameliorates Dextran Sulfate Sodium-induced Depression-like and Anxiety-like Behaviors in Colitis Mice:The potential involvement of Gut-Brain Axis. Food Funct 2022; 13:2865-2883. [DOI: 10.1039/d1fo03888e] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Inflammatory bowel disease (IBD) is accompanied by some psychiatric disorders, including anxiety and depression. Sesamol has been reported to alleviate colitis symptoms and depression-like behaviors caused by chronic unpredictable mild...
Collapse
|
9
|
Liu WG, Zhang LM, Yao JQ, Yin YY, Zhang XY, Li YF, Cao JB. Anti-PTSD Effects of Hypidone Hydrochloride (YL-0919): A Novel Combined Selective 5-HT Reuptake Inhibitor/5-HT 1A Receptor Partial Agonist/5-HT 6 Receptor Full Agonist. Front Pharmacol 2021; 12:625547. [PMID: 33643051 PMCID: PMC7902863 DOI: 10.3389/fphar.2021.625547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/06/2021] [Indexed: 01/17/2023] Open
Abstract
Posttraumatic stress disorder (PTSD) is a debilitating trauma and stressor-related disorder that has become a major neuropsychiatric problem, leading to substantial disruptions in individual health and societal costs. Our previous studies have demonstrated that hypidone hydrochloride (YL-0919), a novel combined selective 5-HT reuptake inhibitor/5-HT1A receptor partial agonist/5-HT6 receptor full agonist, exerts notable antidepressant- and anxiolytic-like as well as procognitive effects. However, whether YL-0919 exerts anti-PTSD effects and its underlying mechanisms are still unclear. In the present study, we showed that repeated treatment with YL-0919 caused significant suppression of contextual fear, enhanced anxiety and cognitive dysfunction induced by the time-dependent sensitization (TDS) procedure in rats and by inescapable electric foot-shock in a mouse model of PTSD. Furthermore, we found that repeated treatment with YL-0919 significantly reversed the accompanying decreased expression of the brain-derived neurotrophic factor (BDNF) and the synaptic proteins (synapsin1 and GluA1), and ameliorated the neuroplasticity disruption in the prefrontal cortex (PFC), including the dendritic complexity and spine density of pyramidal neurons. Taken together, the current study indicated that YL-0919 exerts clear anti-PTSD effects, which might be partially mediated by ameliorating the structural neuroplasticity by increasing the expression of BDNF and the formation of synaptic proteins in the PFC.
Collapse
Affiliation(s)
- Wen-Gang Liu
- Medical School of Chinese PLA, Beijing, China.,Department of Anesthesiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China.,Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Li-Ming Zhang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Jun-Qi Yao
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Yong-Yu Yin
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Xiao-Ying Zhang
- Department of Anesthesiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yun-Feng Li
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China.,Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jiang-Bei Cao
- Department of Anesthesiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
10
|
Bidirectional Regulation of Cognitive and Anxiety-like Behaviors by Dentate Gyrus Mossy Cells in Male and Female Mice. J Neurosci 2021; 41:2475-2495. [PMID: 33472828 DOI: 10.1523/jneurosci.1724-20.2021] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 12/04/2020] [Accepted: 01/04/2021] [Indexed: 02/08/2023] Open
Abstract
The dentate gyrus (DG) of the hippocampus is important for cognition and behavior. However, the circuits underlying these functions are unclear. DG mossy cells (MCs) are potentially important because of their excitatory synapses on the primary cell type, granule cells (GCs). However, MCs also activate GABAergic neurons, which inhibit GCs. We used viral delivery of designer receptors exclusively activated by designer drugs (DREADDs) in mice to implement a gain- and loss-of-function study of MCs in diverse behaviors. Using this approach, manipulations of MCs could bidirectionally regulate behavior. The results suggest that inhibiting MCs can reduce anxiety-like behavior and improve cognitive performance. However, not all cognitive or anxiety-related behaviors were influenced, suggesting specific roles of MCs in some, but not all, types of cognition and anxiety. Notably, several behaviors showed sex-specific effects, with females often showing more pronounced effects than the males. We also used the immediate early gene c-Fos to address whether DREADDs bidirectionally regulated MC or GC activity. We confirmed excitatory DREADDs increased MC c-Fos. However, there was no change in GC c-Fos, consistent with MC activation leading to GABAergic inhibition of GCs. In contrast, inhibitory DREADDs led to a large increase in GC c-Fos, consistent with a reduction in MC excitation of GABAergic neurons, and reduced inhibition of GCs. Together, these results suggest that MCs regulate anxiety and cognition in specific ways. We also raise the possibility that cognitive performance may be improved by reducing anxiety.SIGNIFICANCE STATEMENT The dentate gyrus (DG) has many important cognitive roles as well as being associated with affective behavior. This study addressed how a glutamatergic DG cell type called mossy cells (MCs) contributes to diverse behaviors, which is timely because it is known that MCs regulate the activity of the primary DG cell type, granule cells (GCs), but how MC activity influences behavior is unclear. We show, surprisingly, that activating MCs can lead to adverse behavioral outcomes, and inhibiting MCs have an opposite effect. Importantly, the results appeared to be task-dependent and showed that testing both sexes was important. Additional experiments indicated what MC and GC circuitry was involved. Together, the results suggest how MCs influence behaviors that involve the DG.
Collapse
|
11
|
Saul’skaya NB, Marchuk OE, Puzanova MA, Trofimova NA. Activation of Serotonin System in the Medial Prefrontal Cortex by Sound Signals of Danger. NEUROCHEM J+ 2020. [DOI: 10.1134/s181971242004008x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
12
|
Sałaciak K, Głuch-Lutwin M, Siwek A, Szafarz M, Kazek G, Bednarski M, Nowiński L, Mitchell E, Jastrzębska-Więsek M, Partyka A, Wesołowska A, Kołaczkowski M, Szkaradek N, Marona H, Sapa J, Pytka K. The antidepressant-like activity of chiral xanthone derivatives may be mediated by 5-HT1A receptor and β-arrestin signalling. J Psychopharmacol 2020; 34:1431-1442. [PMID: 33103555 DOI: 10.1177/0269881120959605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Our previous studies showed that xanthone derivatives with N-(2-methoxyphenyl)piperazine fragment have an affinity to the 5-HT1A receptor and show antidepressant-like properties in rodents. In this study, we tested three xanthone derivatives, HBK-1 (R, S) and its enantiomers, in which we increased the distance between the piperazine and xanthone fragments by using a hydroxypropoxy linker. We hypothesized that this would increase the binding to the 5-HT1A receptor and consequently, pharmacological activity. AIMS We aimed to assess the in vitro and in vivo pharmacological activity of the xanthone derivatives. METHODS We evaluated the in vitro affinity for serotonin 5-HT1A and 5-HT2A receptors and serotonin transporter. We also determined the intrinsic activity at the 5-HT1A receptor. We investigated the antidepressant-like properties and safety after acute administration (dose range: 1.25-20 mg/kg) using the forced swim, tail suspension, locomotor activity, rotarod and chimney tests in mice. We also evaluated the basic pharmacokinetic parameters. RESULTS Our results indicated that the compounds showed a high affinity for the 5-HT1A receptor but very weak antagonistic properties in the Ca2+ mobilization assay; however, they showed significant agonistic properties in the β-arrestin recruitment assay. In both behavioural tests the studied xanthone derivatives showed antidepressant-like activity. Pre-treatment with p-chlorophenylalanine or WAY-100635 abolished their antidepressant-like activity. None of the compounds caused motor impairments at antidepressant-like doses. The racemate penetrated the blood-brain barrier and had a relatively high bioavailability after intraperitoneal administration. CONCLUSIONS Xanthone derivatives with N-(2-methoxyphenyl)piperazine fragment and hydroxypropoxy linker show increased binding to the 5-HT1A receptor and may represent an attractive putative treatment candidate for depression.
Collapse
Affiliation(s)
- Kinga Sałaciak
- Department of Pharmacodynamics, Jagiellonian University Medical College, Kraków, Poland
| | - Monika Głuch-Lutwin
- Department of Pharmacobiology, Jagiellonian University Medical College, Kraków, Poland
| | - Agata Siwek
- Department of Pharmacobiology, Jagiellonian University Medical College, Kraków, Poland
| | - Małgorzata Szafarz
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Grzegorz Kazek
- Department of Pharmacodynamics, Jagiellonian University Medical College, Kraków, Poland
| | - Marek Bednarski
- Department of Pharmacodynamics, Jagiellonian University Medical College, Kraków, Poland
| | - Leszek Nowiński
- Department of Pharmacodynamics, Jagiellonian University Medical College, Kraków, Poland
| | - Emma Mitchell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | | | - Anna Partyka
- Department of Clinical Pharmacy, Jagiellonian University Medical College Kraków, Kraków, Poland
| | - Anna Wesołowska
- Department of Clinical Pharmacy, Jagiellonian University Medical College Kraków, Kraków, Poland
| | - Marcin Kołaczkowski
- Department of Medicinal Chemistry, Jagiellonian University Medical College Kraków, Kraków, Poland
| | - Natalia Szkaradek
- Department of Bioorganic Chemistry, Jagiellonian University Medical College, Krakow, Poland
| | - Henryk Marona
- Department of Bioorganic Chemistry, Jagiellonian University Medical College, Krakow, Poland
| | - Jacek Sapa
- Department of Pharmacodynamics, Jagiellonian University Medical College, Kraków, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
13
|
Ito N, Sasaki K, Takemoto H, Kobayashi Y, Isoda H, Odaguchi H. Emotional Impairments and Neuroinflammation are Induced in Male Mice Invulnerable to Repeated Social Defeat Stress. Neuroscience 2020; 443:148-163. [PMID: 32707290 DOI: 10.1016/j.neuroscience.2020.07.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 11/26/2022]
Abstract
Prolonged stress triggers neuroinflammation, which plays a significant role in the development of depression; however, stressed people do not always suffer from depression because of individual differences in stress vulnerability. Negative cognitive bias (NCB) toward pessimistic judgment often underlies depressive episodes. However, a relationship between stress vulnerability, neuroinflammation, and NCB remains elusive. In addition, an animal model with all the traits would be a powerful tool for studying the etiology of depression and its therapeutic approaches. Accordingly, this study evaluated the effect of stress vulnerability on neuroinflammation and depression-related behaviors, including NCB in males, using a modified version of repeated social defeat stress (mRSDS) paradigm, a validated animal model of psychosocial stress. Exposure to mRSDS, consisting of 5 min of social defeat by unfamiliar CD-1 aggressor mice for five consecutive days, caused NCB, which co-occurred with depressive- and anxiety-like behaviors, and neuroinflammation in male BALB/c mice. Treatment with minocycline, an antibiotic with anti-inflammatory property, blocked mRSDS-induced depressive-like behaviors and neuroinflammation, but not NCB, indicating the limited effect of an anti-inflammatory intervention. In addition, marked differences were found in neuroinflammatory profiles and hippocampal gene expression patterns between resilient and unstressed mice, as well as between susceptible and resilient mice. Therefore, mice resilient to mRSDS are indeed not intact. Our findings provide insights into the unique features of the mRSDS model in male BALB/c mice, which could be used to investigate the etiological mechanisms underlying depression as well as bridge the gap in the relationship between stress vulnerability, neuroinflammation, and NCB in males.
Collapse
Affiliation(s)
- Naoki Ito
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Minato-ku, Tokyo 108-8642, Japan.
| | - Kazunori Sasaki
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba-shi, Ibaraki 305-8572, Japan; Interdisciplinary Research Center for Catalytic Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba-shi, Ibaraki 305-8565, Japan; Faculty of Pure and Applied Sciences, University of Tsukuba, Tsukuba-shi, Ibaraki 305-8571, Japan
| | - Hiroaki Takemoto
- School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8642, Japan
| | - Yoshinori Kobayashi
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Minato-ku, Tokyo 108-8642, Japan; School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8642, Japan
| | - Hiroko Isoda
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba-shi, Ibaraki 305-8572, Japan; Interdisciplinary Research Center for Catalytic Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba-shi, Ibaraki 305-8565, Japan; Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba-shi, Ibaraki 305-8572, Japan
| | - Hiroshi Odaguchi
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Minato-ku, Tokyo 108-8642, Japan
| |
Collapse
|
14
|
Petroianu GA, Lorke DE. The Role of Serotonin in Singultus: A Review. Front Neurosci 2020; 14:629. [PMID: 32765206 PMCID: PMC7378791 DOI: 10.3389/fnins.2020.00629] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/22/2020] [Indexed: 12/21/2022] Open
Affiliation(s)
- Georg A. Petroianu
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
- *Correspondence: Georg A. Petroianu,
| | - Dietrich E. Lorke
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| |
Collapse
|
15
|
Reduced serotonin impairs long-term depression in basolateral amygdala complex and causes anxiety-like behaviors in a mouse model of perimenopause. Exp Neurol 2019; 321:113030. [DOI: 10.1016/j.expneurol.2019.113030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 07/07/2019] [Accepted: 07/31/2019] [Indexed: 11/19/2022]
|
16
|
Attenuated palmitoylation of serotonin receptor 5-HT1A affects receptor function and contributes to depression-like behaviors. Nat Commun 2019; 10:3924. [PMID: 31477731 PMCID: PMC6718429 DOI: 10.1038/s41467-019-11876-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 07/24/2019] [Indexed: 12/20/2022] Open
Abstract
The serotonergic system and in particular serotonin 1A receptor (5-HT1AR) are implicated in major depressive disorder (MDD). Here we demonstrated that 5-HT1AR is palmitoylated in human and rodent brains, and identified ZDHHC21 as a major palmitoyl acyltransferase, whose depletion reduced palmitoylation and consequently signaling functions of 5-HT1AR. Two rodent models for depression-like behavior show reduced brain ZDHHC21 expression and attenuated 5-HT1AR palmitoylation. Moreover, selective knock-down of ZDHHC21 in the murine forebrain induced depression-like behavior. We also identified the microRNA miR-30e as a negative regulator of Zdhhc21 expression. Through analysis of the post-mortem brain samples in individuals with MDD that died by suicide we find that miR-30e expression is increased, while ZDHHC21 expression, as well as palmitoylation of 5-HT1AR, are reduced within the prefrontal cortex. Our study suggests that downregulation of 5-HT1AR palmitoylation is a mechanism involved in depression, making the restoration of 5-HT1AR palmitoylation a promising clinical strategy for the treatment of MDD. Palmitoylation is a post translational modification that regulates GPCR activity. Here the authors show that palmitoylation of 5-HT1AR by the palmitoyltransferase enzyme ZDHHC21 contributes to depression-like behaviour in rodents and might be implicated in major depressive disorder.
Collapse
|
17
|
Kurauchi Y, Devkota HP, Hori K, Nishihara Y, Hisatsune A, Seki T, Katsuki H. Anxiolytic activities of Matcha tea powder, extracts, and fractions in mice: Contribution of dopamine D1 receptor- and serotonin 5-HT1A receptor-mediated mechanisms. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.05.046] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
18
|
Yabuki Y, Fukunaga K. Clinical Therapeutic Strategy and Neuronal Mechanism Underlying Post-Traumatic Stress Disorder (PTSD). Int J Mol Sci 2019; 20:ijms20153614. [PMID: 31344835 PMCID: PMC6695947 DOI: 10.3390/ijms20153614] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/19/2019] [Accepted: 07/19/2019] [Indexed: 12/15/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is characterized by an exaggerated response to contextual memory and impaired fear extinction, with or without mild cognitive impairment, learning deficits, and nightmares. PTSD is often developed by traumatic events, such as war, terrorist attack, natural calamities, etc. Clinical and animal studies suggest that aberrant susceptibility of emotion- and fear-related neurocircuits, including the amygdala, prefrontal cortex (PFC), and hippocampus may contribute to the development and retention of PTSD symptoms. Psychological and pharmacological therapy, such as cognitive behavioral therapy (CBT), and treatment with anti-depressive agents and/or antipsychotics significantly attenuate PTSD symptoms. However, more effective therapeutics are required for improvement of quality of life in PTSD patients. Previous studies have reported that ω3 long-chain polyunsaturated fatty acid (LCPUFA) supplements can suppress the development of PTSD symptoms. Fatty acid binding proteins (FABPs) are essential for LCPUFA intracellular trafficking. In this review, we have introduced Fabp3 null mice as an animal model of PTSD with impaired fear extinction. Moreover, we have addressed the neuronal circuits and novel therapeutic strategies for PTSD symptoms.
Collapse
Affiliation(s)
- Yasushi Yabuki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan.
| |
Collapse
|
19
|
Fukunaga K, Yabuki Y, Takahata I, Matsuo K. [Neurological mechanism and therapeutic strategy for posttraumatic stress disorders]. Nihon Yakurigaku Zasshi 2019; 152:194-201. [PMID: 30298841 DOI: 10.1254/fpj.152.194] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Posttraumatic stress disorder (PTSD) is most often induced by traumatic events and serious public health problems. PTSD is characterized by excessive response to contextual memory and impaired fear extinction and also associated with mild cognitive impairment, attention and learning deficits. Clinical and animal studies suggest that increased susceptibility of emotion- and fear-related neuronal circuits, including those in the amygdala, prefrontal cortex and hippocampus, contributes to development and retention of PTSD symptoms. However, mechanisms underlying this susceptibility to fear are not known and the useful therapeutic approaches are limited. Recently, there have been reports that ω3 LCPUFA supplementation can prevent development of PTSD and significantly ameliorate symptoms in patients with PTSD after accidental injury such as motor vehicle accidents and natural calamities. Importantly, Fabp7 null mice exhibit enhancement of fear memory consolidation and anxiety-related behaviors that resemble PTSD-like behaviors in humans. In this review, we focused behavioral phenotype of PTSD in Fabp3 null mice. The Fabp3 null mice exhibit cognitive deficits, hyperlocomotion and impaired fear extinction, and thus show PTSD-like behaviors. Chronic administration of ramelteon, a melatonin receptor agonist, improved all PTSD-like behaviors tested in Fabp3-/- mice. Relevant to mechanisms underlying impaired fear extinction, we observed that Ca2+/calmodulin-dependent protein kinase II (CaMKII) autophosphorylation increases in the basolateral amygdala (BLA) but remained unchanges in the hippocampus of Fabp3-/- mice. Likewise, the number of c-Fos positive neurons in BLA significantly increased after exposure to contextual fear conditions. Finally, chronic ramelteon administration restored abnormal c-Fos expression and CaMKII autophosphorylation in the BLA of Fabp3-/- mice. Taken together, Fabp3-/- mice show PTSD-like behaviors, and ramelteon is an attractive candidate for PTSD therapeutics in human.
Collapse
Affiliation(s)
- Kohji Fukunaga
- Department of Pharmacology, Tohoku University Graduate School of Pharmaceutical Sciences
| | - Yasushi Yabuki
- Department of Pharmacology, Tohoku University Graduate School of Pharmaceutical Sciences
| | - Ibuki Takahata
- Department of Pharmacology, Tohoku University Graduate School of Pharmaceutical Sciences
| | - Kazuya Matsuo
- Department of Pharmacology, Tohoku University Graduate School of Pharmaceutical Sciences
| |
Collapse
|
20
|
Zhang X, Huettel SA, Mullette-Gillman OA, Guo H, Wang L. Exploring common changes after acute mental stress and acute tryptophan depletion: Resting-state fMRI studies. J Psychiatr Res 2019; 113:172-180. [PMID: 30959228 DOI: 10.1016/j.jpsychires.2019.03.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/23/2019] [Accepted: 03/27/2019] [Indexed: 11/27/2022]
Abstract
Stress and low serotonin levels are important biological factors in depression and anxiety etiologies. Although studies indicate that low serotonin levels, stress, and other factors may interact in depression/anxiety psychopathology, few studies have investigated the potentially shared neural substrates. We conducted resting-state fMRI scans pre- and post-stress task, and under control and tryptophan depletion condition, to explore the common changes induced by acute mental stress (AMS) and acute tryptophan depletion (ATD). The present study targeted regions within core brain networks - default mode network, salience network, executive control network, and emotion network - reported altered in AMS and ATD, and used regional homogeneity (ReHo) and functional connectivity (FC) analyses to explore their overlapped effects. We additionally examined the relationships among core neural networks - operationalized as an index of resource allocation bias that quantifies the shift from internal to external modes of processing. We found both manipulations induced increased ReHo of the amygdala and decreased ReHo of the posterior cingulate cortex (PCC). The PCC-amygdala FC was negatively correlated with the change of negative affect, whereas the right dorsolateral prefrontal cortex and right anterior insula FC was positively associated with anxiety level. In addition, we found that a greater shift to an external mode was correlated with higher anxiety level under both conditions. Common changes induced by acute mental stress and acute tryptophan depletion confirmed our hypothesis that AMS and ATD induce changes in common neural pathways, which in turn might mark vulnerability to depression and anxiety.
Collapse
Affiliation(s)
- Xue Zhang
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, Tsinghua University School of Medicine, Beijing, China
| | - Scott A Huettel
- Center for Cognitive Neuroscience, Department of Psychology & Neuroscience, Duke University, Durham, USA
| | | | - Hua Guo
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, Tsinghua University School of Medicine, Beijing, China
| | - Lihong Wang
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, USA.
| |
Collapse
|
21
|
Berg L, Eckardt J, Masseck OA. Enhanced activity of pyramidal neurons in the infralimbic cortex drives anxiety behavior. PLoS One 2019; 14:e0210949. [PMID: 30677060 PMCID: PMC6345483 DOI: 10.1371/journal.pone.0210949] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 01/06/2019] [Indexed: 01/17/2023] Open
Abstract
We show that in an animal model of anxiety the overall excitation, particularly in the infralimbic region of the medial prefrontal cortex (IL), is increased and that the activity ratio between excitatory pyramidal neurons and inhibitory interneurons (AR PN/IN) is shifted towards excitation. The same change in AR PN/IN is evident for wildtype mice, which have been exposed to an anxiety stimulus. We hypothesize, that an elevated activity and the imbalance of excitation (PN) and inhibition (IN) within the neuronal microcircuitry of the prefrontal cortex is responsible for anxiety behaviour and employed optogenetic methods in freely moving mice to verify our findings. Consistent with our hypothesis elevation of pyramidal neuron activity in the infralimbic region of the prefrontal cortex significantly enhanced anxiety levels in several behavioural tasks by shifting the AR PN/IN to excitation, without affecting motor behaviour, thus revealing a novel mechanism by which anxiety is facilitated.
Collapse
Affiliation(s)
- Laura Berg
- Advanced Fluorescence Microscopy, Ruhr University Bochum, Bochum, Germany
| | - Josephine Eckardt
- Department of Systems Neuroscience Ruhr University Bochum, Bochum, Germany
| | - Olivia Andrea Masseck
- Advanced Fluorescence Microscopy, Ruhr University Bochum, Bochum, Germany
- University of Bremen, Synthetic Biology, Bremen, Germany
- * E-mail:
| |
Collapse
|
22
|
Asok A, Kandel ER, Rayman JB. The Neurobiology of Fear Generalization. Front Behav Neurosci 2019; 12:329. [PMID: 30697153 PMCID: PMC6340999 DOI: 10.3389/fnbeh.2018.00329] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 12/13/2018] [Indexed: 12/12/2022] Open
Abstract
The generalization of fear memories is an adaptive neurobiological process that promotes survival in complex and dynamic environments. When confronted with a potential threat, an animal must select an appropriate defensive response based on previous experiences that are not identical, weighing cues and contextual information that may predict safety or danger. Like other aspects of fear memory, generalization is mediated by the coordinated actions of prefrontal, hippocampal, amygdalar, and thalamic brain areas. In this review article, we describe the current understanding of the behavioral, neural, genetic, and biochemical mechanisms involved in the generalization of fear. Fear generalization is a hallmark of many anxiety and stress-related disorders, and its emergence, severity, and manifestation are sex-dependent. Therefore, to improve the dialog between human and animal studies as well as to accelerate the development of effective therapeutics, we emphasize the need to examine both sex differences and remote timescales in rodent models.
Collapse
Affiliation(s)
- Arun Asok
- Jerome L. Greene Science Center, Department of Neuroscience, Columbia University, New York, NY, United States
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
| | - Eric R. Kandel
- Jerome L. Greene Science Center, Department of Neuroscience, Columbia University, New York, NY, United States
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
- Howard Hughes Medical Institute (HHMI), Columbia University, New York, NY, United States
- Kavli Institute for Brain Science, Columbia University, New York, NY, United States
| | - Joseph B. Rayman
- Jerome L. Greene Science Center, Department of Neuroscience, Columbia University, New York, NY, United States
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
| |
Collapse
|
23
|
Park SC, Kim YK. A Novel Bio-Psychosocial-Behavioral Treatment Model of Panic Disorder. Psychiatry Investig 2019; 16:4-15. [PMID: 30301303 PMCID: PMC6354044 DOI: 10.30773/pi.2018.08.21.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/09/2018] [Accepted: 08/21/2018] [Indexed: 12/11/2022] Open
Abstract
To conceptualize a novel bio-psychosocial-behavioral treatment model of panic disorder (PD), it is necessary to completely integrate behavioral, psychophysiological, neurobiological, and genetic data. Molecular genetic research on PD is specifically focused on neurotransmitters, including serotonin, neuropeptides, glucocorticoids, and neurotrophins. Although pharmacological interventions for PD are currently available, the need for more effective, faster-acting, and more tolerable pharmacological interventions is unmet. Thus, glutamatergic receptor modulators, orexin receptor antagonists, corticotrophin-releasing factor 1 receptor antagonists, and other novel mechanism-based anti-panic therapeutics have been proposed. Research on the neural correlates of PD is focused on the dysfunctional "cross-talk" between emotional drive (limbic structure) and cognitive inhibition (prefrontal cortex) and the fear circuit, which includes the amygdala-hippocampus-prefrontal axis. The neural perspective regarding PD supports the idea that cognitive-behavioral therapy normalizes alterations in top-down cognitive processing, including increased threat expectancy and attention to threat. Consistent with the concept of "personalized medicine," it is speculated that Research Domain Criteria can enlighten further treatments targeting dysfunctions underlying PD more precisely and provide us with better definitions of moderators used to identify subgroups according to different responses to treatment. Structuring of the "negative valence systems" domain, which includes fear/anxiety, is required to define PD. Therefore, targeting glutamate- and orexin-related molecular mechanisms associated with the fear circuit, which includes the amygdala-hippocampus-prefrontal cortex axis, is required to define a novel bio-psychosocial-behavioral treatment model of PD.
Collapse
Affiliation(s)
- Seon-Cheol Park
- Department of Psychiatry, Inje University College of Medicine and Haeundae Paik Hospital, Busan, Republic of Korea
| | - Yong-Ku Kim
- Department of Psychiatry, College of Medicine, Korea University, Ansan Hospital, Ansan, Republic of Korea
| |
Collapse
|
24
|
Zhao Y, Bijlsma EY, ter Heegde F, Verdouw MP, Garssen J, Newman-Tancredi A, Groenink L. Activation of somatodendritic 5-HT 1A autoreceptors reduces the acquisition and expression of cued fear in the rat fear-potentiated startle test. Psychopharmacology (Berl) 2019; 236:1171-1185. [PMID: 30539269 PMCID: PMC6591185 DOI: 10.1007/s00213-018-5124-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 11/14/2018] [Indexed: 11/29/2022]
Abstract
RATIONALE Fear conditioning is an important factor in the etiology of anxiety disorders. Previous studies have demonstrated a role for serotonin (5-HT)1A receptors in fear conditioning. However, the relative contribution of somatodendritic 5-HT1A autoreceptors and post-synaptic 5-HT1A heteroreceptors in fear conditioning is still unclear. OBJECTIVE To determine the role of pre- and post-synaptic 5-HT1A receptors in the acquisition and expression of cued and contextual conditioned fear. METHODS We studied the acute effects of four 5-HT1A receptor ligands in the fear-potentiated startle test. Male Wistar rats were injected with the 5-HT1A receptors biased agonists F13714 (0-0.16 mg/kg, IP), which preferentially activates somatodendritic 5-HT1A autoreceptors, or F15599 (0-0.16 mg/kg, IP), which preferentially activates cortical post-synaptic 5-HT1A heteroreceptors, with the prototypical 5-HT1A receptor agonist R(+)8-OH-DPAT (0-0.3 mg/kg, SC) or the 5-HT1A receptor antagonist WAY100,635 (0-1.0 mg/kg, SC). RESULTS F13714 (0.16 mg/kg) and R(+)-8-OH-DPAT (0.03 mg/kg) injected before training reduced cued fear acquisition. Pre-treatment with F15599 or WAY100,635 had no effect on fear learning. In the fear-potentiated startle test, F13714 (0.04-0.16 mg/kg) and R(+)-8-OH-DPAT (0.1-0.3 mg/kg) reduced the expression of cued and contextual fear, whereas F15599 had no effect. WAY100,635 (0.03-1.0 mg/kg) reduced the overall startle response. CONCLUSIONS The current findings indicate that activation of somatodendritic 5-HT1A autoreceptors reduces cued fear learning, whereas 5-HT1A receptors seem not involved in contextual fear learning. Moreover, activation of somatodendritic 5-HT1A autoreceptors may reduce cued and contextual fear expression, whereas we found no evidence for the involvement of cortical 5-HT1A heteroreceptors in the expression of conditioned fear.
Collapse
Affiliation(s)
- Yulong Zhao
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Elisabeth Y. Bijlsma
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Freija ter Heegde
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Monika P. Verdouw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - J. Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | | | - Lucianne Groenink
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, The Netherlands. .,Brain Center Rudolf Magnus (BCRM), UMC Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
25
|
Richter-Levin G, Stork O, Schmidt MV. Animal models of PTSD: a challenge to be met. Mol Psychiatry 2019; 24:1135-1156. [PMID: 30816289 PMCID: PMC6756084 DOI: 10.1038/s41380-018-0272-5] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 08/13/2018] [Accepted: 09/11/2018] [Indexed: 02/07/2023]
Abstract
Recent years have seen increased interest in psychopathologies related to trauma exposure. Specifically, there has been a growing awareness to posttraumatic stress disorder (PTSD) in part due to terrorism, climate change-associated natural disasters, the global refugee crisis, and increased violence in overpopulated urban areas. However, notwithstanding the increased awareness to the disorder, the increasing number of patients, and the devastating impact on the lives of patients and their families, the efficacy of available treatments remains limited and highly unsatisfactory. A major scientific effort is therefore devoted to unravel the neural mechanisms underlying PTSD with the aim of paving the way to developing novel or improved treatment approaches and drugs to treat PTSD. One of the major scientific tools used to gain insight into understanding physiological and neuronal mechanisms underlying diseases and for treatment development is the use of animal models of human diseases. While much progress has been made using these models in understanding mechanisms of conditioned fear and fear memory, the gained knowledge has not yet led to better treatment options for PTSD patients. This poor translational outcome has already led some scientists and pharmaceutical companies, who do not in general hold opinions against animal models, to propose that those models should be abandoned. Here, we critically examine aspects of animal models of PTSD that may have contributed to the relative lack of translatability, including the focus on the exposure to trauma, overlooking individual and sex differences, and the contribution of risk factors. Based on findings from recent years, we propose research-based modifications that we believe are required in order to overcome some of the shortcomings of previous practice. These modifications include the usage of animal models of PTSD which incorporate risk factors and of the behavioral profiling analysis of individuals in a sample. These modifications are aimed to address factors such as individual predisposition and resilience, thus taking into consideration the fact that only a fraction of individuals exposed to trauma develop PTSD. We suggest that with an appropriate shift of practice, animal models are not only a valuable tool to enhance our understanding of fear and memory processes, but could serve as effective platforms for understanding PTSD, for PTSD drug development and drug testing.
Collapse
Affiliation(s)
- Gal Richter-Levin
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel. .,The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, Haifa, Israel. .,Psychology Department, University of Haifa, Haifa, Israel.
| | - Oliver Stork
- 0000 0001 1018 4307grid.5807.aDepartment of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany ,grid.452320.2Center for Behavioral Brain Sciences, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Mathias V. Schmidt
- 0000 0000 9497 5095grid.419548.5Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
26
|
Abstract
Defensive motivation, broadly defined as the orchestrated optimization of defensive functions, encapsulates core components of threat-related psychopathology. The exact relationship between defensive functions and stress-induced symptoms, however, is not entirely clear. Here we review how some of the most important behavioral and neurological findings related to threat-related disorders -- lowering response threshold to threats, facilitated learning and generalization to new threatening cues, reduced appetitive sensitivity, and resistance to extinction of the defensive state -- map onto defensive motivational states, highlighting evidence that supports conjecturing threat-related disorders as persistent motivational states. We propose a mechanism for the perpetuation of the motivational state, progressively converting temporary defensive functions into persistent defensive states associated with distress and impairment.
Collapse
|
27
|
Katano T, Takao K, Abe M, Yamazaki M, Watanabe M, Miyakawa T, Sakimura K, Ito S. Distribution of Caskin1 protein and phenotypic characterization of its knockout mice using a comprehensive behavioral test battery. Mol Brain 2018; 11:63. [PMID: 30359304 PMCID: PMC6202847 DOI: 10.1186/s13041-018-0407-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/14/2018] [Indexed: 01/17/2023] Open
Abstract
Calcium/calmodulin-dependent serine protein kinase (CASK)-interacting protein 1 (Caskin1) is a direct binding partner of the synaptic adaptor protein CASK. Because Caskin1 forms homo-multimers and binds not only CASK but also other neuronal proteins in vitro, it is anticipated to have neural functions; but its exact role in mammals remains unclear. Previously, we showed that the concentration of Caskin1 in the spinal dorsal horn increases under chronic pain. To characterize this protein, we generated Caskin1-knockout (Caskin1-KO) mice and specific anti-Caskin1 antibodies. Biochemical and immunohistochemical analyses demonstrated that Caskin1 was broadly distributed in the whole brain and spinal cord, and that it primarily localized at synapses. To elucidate the neural function of Caskin1 in vivo, we subjected Caskin1-KO mice to comprehensive behavioral analysis. The mutant mice exhibited differences in gait, enhanced nociception, and anxiety-like behavior relative to their wild-type littermates. In addition, the knockouts exhibited strong freezing responses, with or without a cue tone, in contextual and cued-fear conditioning tests as well as low memory retention in the Barnes Maze test. Taken together, these results suggest that Caskin1 contributes to a wide spectrum of behavioral phenotypes, including gait, nociception, memory, and stress response, in broad regions of the central nervous system.
Collapse
Affiliation(s)
- Tayo Katano
- Department of Medical Chemistry, Kansai Medical University, Hirakata, 573-1010 Japan
| | - Keizo Takao
- Section of Behavior Patterns, National Institute of Physiological Sciences NINS, Okazaki, Aichi 444-8585 Japan
- Division of Experimental Animal Resource and Development, Life Science Research Center, University of Toyama, Toyama, 930-0194 Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585 Japan
| | - Maya Yamazaki
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585 Japan
- Department of Neurology, University of California, San Francisco, 94158 USA
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo, 060-8638 Japan
| | - Tsuyoshi Miyakawa
- Section of Behavior Patterns, National Institute of Physiological Sciences NINS, Okazaki, Aichi 444-8585 Japan
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi 470-1192 Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585 Japan
| | - Seiji Ito
- Department of Medical Chemistry, Kansai Medical University, Hirakata, 573-1010 Japan
| |
Collapse
|
28
|
Too Depressed to Swim or Too Afraid to Stop? A Reinterpretation of the Forced Swim Test as a Measure of Anxiety-Like Behavior. Neuropsychopharmacology 2018; 43:931-933. [PMID: 29210364 PMCID: PMC5854810 DOI: 10.1038/npp.2017.260] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/22/2017] [Accepted: 10/24/2017] [Indexed: 11/08/2022]
|
29
|
Kiryanova V, Smith VM, Antle MC, Dyck RH. Behavior of Adult 5-HT1A Receptor Knockout Mice Exposed to Stress During Prenatal Development. Neuroscience 2018; 371:16-28. [DOI: 10.1016/j.neuroscience.2017.11.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 10/27/2017] [Accepted: 11/21/2017] [Indexed: 12/29/2022]
|
30
|
Wu ZM, Yang LH, Cui R, Ni GL, Wu FT, Liang Y. Contribution of Hippocampal 5-HT 3 Receptors in Hippocampal Autophagy and Extinction of Conditioned Fear Responses after a Single Prolonged Stress Exposure in Rats. Cell Mol Neurobiol 2017; 37:595-606. [PMID: 27324798 DOI: 10.1007/s10571-016-0395-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/13/2016] [Indexed: 02/01/2023]
Abstract
One of the hypotheses about the pathogenesis of posttraumatic stress disorder (PTSD) is the dysfunction of serotonin (5-HT) neurotransmission. While certain 5-HT receptor subtypes are likely critical for the symptoms of PTSD, few studies have examined the role of 5-HT3 receptor in the development of PTSD, even though 5-HT3 receptor is critical for contextual fear extinction and anxiety-like behavior. Therefore, we hypothesized that stimulation of 5-HT3 receptor in the dorsal hippocampus (DH) could prevent hippocampal autophagy and the development of PTSD-like behavior in animals. To this end, we infused SR57227, selective 5-HT3 agonist, into the DH after a single prolonged stress (SPS) treatment in rats. Three weeks later, we evaluated the effects of this pharmacological treatment on anxiety-related behaviors and extinction of contextual fear memory. We also accessed hippocampal autophagy and the expression of 5-HT3A subunit, Beclin-1, LC3-I, and LC3-II in the DH. We found that SPS treatment did not alter anxiety-related behaviors but prolonged the extinction of contextual fear memory, and such a behavioral phenomenon was correlated with increased hippocampal autophagy, decreased 5-HT3A expression, and increased expression of Beclin-1 and LC3-II/LC3-I ratio in the DH. Furthermore, intraDH infusions of SR57227 dose-dependently promoted the extinction of contextual fear memory, prevented hippocampal autophagy, and decreased expression of Beclin-1 and LC3-II/LC3-I ratio in the DH. These results indicated that 5-HT3 receptor in the hippocampus may play a critical role in the pathogenesis of hippocampal autophagy, and is likely involved in the pathophysiology of PTSD.
Collapse
Affiliation(s)
- Zhong-Min Wu
- Department of Anatomy, Medical College of Taizhou University, 1139 Taizhou city government Avenue, Taizhou, 318000, China
- Department of Neurology, First People's Hospital of Linhai City, Linhai, 317000, China
| | - Li-Hua Yang
- Department of Neurology, Taizhou Hospital, Taizhou, 317000, China
| | - Rong Cui
- Department of Neurology, First People's Hospital of Linhai City, Linhai, 317000, China
| | - Gui-Lian Ni
- Department of Neurology, First People's Hospital of Linhai City, Linhai, 317000, China
| | - Feng-Tian Wu
- City College of Zhejiang University, Hangzhou, 310015, China
| | - Yong Liang
- Department of Anatomy, Medical College of Taizhou University, 1139 Taizhou city government Avenue, Taizhou, 318000, China.
| |
Collapse
|
31
|
Wirth A, Holst K, Ponimaskin E. How serotonin receptors regulate morphogenic signalling in neurons. Prog Neurobiol 2017; 151:35-56. [DOI: 10.1016/j.pneurobio.2016.03.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 03/09/2016] [Accepted: 03/19/2016] [Indexed: 11/25/2022]
|
32
|
Garcia AN, Bezner K, Depena C, Yin W, Gore AC. The effects of long-term estradiol treatment on social behavior and gene expression in adult female rats. Horm Behav 2017; 87:145-154. [PMID: 27871902 PMCID: PMC5203957 DOI: 10.1016/j.yhbeh.2016.11.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 11/04/2016] [Accepted: 11/14/2016] [Indexed: 12/24/2022]
Abstract
This study tested the effects of long-term estradiol (E2) replacement on social behavior and gene expression in brain nuclei involved in the regulation of these social behaviors in adult female rats. We developed an ultrasonic vocalization (USV) test and a sociability test to examine communications, social interactions, and social preference, using young adult female cagemates. All rats were ovariectomized (OVX) and implanted with a Silastic capsule containing E2 or vehicle, and housed in same-treatment pairs for a 3-month period. Then, rats were behaviorally tested, euthanized, and 5 nuclei in the brain's social decision-making circuit were selected for neuromolecular profiling by a multiplex qPCR method. Our novel USV test proved to be a robust tool to measure numbers and types of calls emitted by cagemates that had been reintroduced after a 1-week separation. Results also showed that E2-treated OVX rats had profoundly decreased numbers of USV calls compared to vehicle-treated OVX rats. In a test of sociability, in which a female was allowed to choose between her cagemate or a same-treatment novel rat, we found few effects of E2 compared to vehicle, although interestingly, rats chose the cagemate over an unfamiliar conspecific. Gene expression results revealed that the supraoptic nucleus had the greatest number of gene changes caused by E2: Oxt, Oxtr and Avp were increased, and Drd2, Htr1a, Grin2b, and Gabbr1 were decreased, by E2. No genes were affected in the prefrontal cortex, and 1-4 genes were changed in paraventricular nucleus (Pgr), bed nucleus of the stria terminalis (Oxtr, Esr2, Dnmt3a), and medial amygdala (Oxtr, Ar, Foxp1, Tac3). Thus, E2 changes communicative interactions between adult female rats, together with selected expression of genes in the brain, especially in the supraoptic nucleus.
Collapse
Affiliation(s)
- Alexandra N Garcia
- Department of Psychology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Kelsey Bezner
- Division of Pharmacology and Toxicology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Christina Depena
- Division of Pharmacology and Toxicology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Weiling Yin
- Division of Pharmacology and Toxicology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Andrea C Gore
- Department of Psychology, The University of Texas at Austin, Austin, TX 78712, USA; Division of Pharmacology and Toxicology, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
33
|
Davis MT, Holmes SE, Pietrzak RH, Esterlis I. Neurobiology of Chronic Stress-Related Psychiatric Disorders: Evidence from Molecular Imaging Studies. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2017; 1:2470547017710916. [PMID: 29862379 PMCID: PMC5976254 DOI: 10.1177/2470547017710916] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/30/2017] [Accepted: 05/01/2017] [Indexed: 01/12/2023]
Abstract
Chronic stress accounts for billions of dollars of economic loss annually in the United States alone, and is recognized as a major source of disability and mortality worldwide. Robust evidence suggests that chronic stress plays a significant role in the onset of severe and impairing psychiatric conditions, including major depressive disorder, bipolar disorder, and posttraumatic stress disorder. Application of molecular imaging techniques such as positron emission tomography and single photon emission computed tomography in recent years has begun to provide insight into the molecular mechanisms by which chronic stress confers risk for these disorders. The present paper provides a comprehensive review and synthesis of all positron emission tomography and single photon emission computed tomography imaging publications focused on the examination of molecular targets in individuals with major depressive disorder, posttraumatic stress disorder, or bipolar disorder to date. Critical discussion of discrepant findings and broad strengths and weaknesses of the current body of literature is provided. Recommended future directions for the field of molecular imaging to further elucidate the neurobiological substrates of chronic stress-related disorders are also discussed. This article is part of the inaugural issue for the journal focused on various aspects of chronic stress.
Collapse
Affiliation(s)
- Margaret T. Davis
- Department of Psychiatry, Yale School of
Medicine, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical
Imaging, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Sophie E. Holmes
- Department of Psychiatry, Yale School of
Medicine, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical
Imaging, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Robert H. Pietrzak
- Department of Psychiatry, Yale School of
Medicine, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical
Imaging, Yale School of Medicine, Yale University, New Haven, CT, USA
- US Department of Veterans Affairs National
Center for Posttraumatic Stress Disorder, VA Connecticut Healthcare System, West Haven, CT,
USA
| | - Irina Esterlis
- Department of Psychiatry, Yale School of
Medicine, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical
Imaging, Yale School of Medicine, Yale University, New Haven, CT, USA
- US Department of Veterans Affairs National
Center for Posttraumatic Stress Disorder, VA Connecticut Healthcare System, West Haven, CT,
USA
| |
Collapse
|
34
|
Jaehne EJ, Ameti D, Paiva T, van den Buuse M. Investigating the Role of Serotonin in Methamphetamine Psychosis: Unaltered Behavioral Effects of Chronic Methamphetamine in 5-HT 1A Knockout Mice. Front Psychiatry 2017; 8:61. [PMID: 28473777 PMCID: PMC5397502 DOI: 10.3389/fpsyt.2017.00061] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/04/2017] [Indexed: 01/11/2023] Open
Abstract
Methamphetamine (Meth) is a widely abused stimulant drug, but this abuse is associated with an increased risk of developing psychosis. In addition to its well-known action on brain dopamine, Meth also affects serotonergic (5-HT) neurons. The aim of this study was to investigate this role in mice, which lack one of the main serotonin receptors, the 5-HT1A receptor, which has been implicated in both schizophrenia and Meth-induced psychosis. Male and female wild-type or 5-HT1A knockout (KO) mice received daily treatment with increasing doses of methamphetamine from 6 to 9 weeks of age (1-4 mg/kg/day twice a day). At least 2 weeks after the last injection, the mice underwent a battery of behavioral tests focusing on psychosis-related behaviors, including Meth-induced hyperactivity, prepulse inhibition (PPI), social interaction, elevated plus maze (EPM), and Y-maze. Meth pretreatment resulted in significantly increased hyperlocomotion in response to an acute Meth challenge, but this effect was independent of genotype. Chronic Meth treatment resulted in decreased levels of anxiety in the EPM in both sexes, as well as increased startle responses in female mice only, again independent of genotype. 5-HT1A KO mice showed an increased locomotor response to acute Meth in both sexes, as well as increased PPI and decreased startle responses in female mice only, independent of Meth pretreatment. In conclusion, the effects of chronic Meth appear unaffected by the absence of the 5-HT1A receptor. These results do not support a role of the 5-HT1A receptor in Meth-induced psychosis.
Collapse
Affiliation(s)
- Emily J Jaehne
- Department Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Dzeneta Ameti
- Department Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Tehani Paiva
- Department Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Maarten van den Buuse
- Department Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia.,Department of Pharmacology, University of Melbourne, Melbourne, VIC, Australia.,The College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| |
Collapse
|
35
|
Norris C, Loureiro M, Kramar C, Zunder J, Renard J, Rushlow W, Laviolette SR. Cannabidiol Modulates Fear Memory Formation Through Interactions with Serotonergic Transmission in the Mesolimbic System. Neuropsychopharmacology 2016; 41:2839-2850. [PMID: 27296152 PMCID: PMC5061893 DOI: 10.1038/npp.2016.93] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 06/06/2016] [Accepted: 06/08/2016] [Indexed: 12/20/2022]
Abstract
Emerging evidence suggests that the largest phytochemical component of cannabis, cannabidiol (CBD), may possess pharmacotherapeutic properties in the treatment of neuropsychiatric disorders. CBD has been reported to functionally interact with both the mesolimbic dopamine (DA) and serotonergic (5-HT) receptor systems. However, the underlying mechanisms by which CBD may modulate emotional processing are not currently understood. Using a combination of in vivo electrophysiological recording and fear conditioning in rats, the present study aimed to characterize the behavioral, neuroanatomical, and pharmacological effects of CBD within the mesolimbic pathway, and its possible functional interactions with 5-HT and DAergic transmission. Using targeted microinfusions of CBD into the shell region of the mesolimbic nucleus accumbens (NASh), we report that intra-NASh CBD potently blocks the formation of conditioned freezing behaviors. These effects were challenged with DAergic, cannabinoid CB1 receptor, and serotonergic (5-HT1A) transmission blockade, but only 5-HT1A blockade restored associative conditioned freezing behaviors. In vivo intra-ventral tegmental area (VTA) electrophysiological recordings revealed that behaviorally effective doses of intra-NASh CBD elicited a predominant decrease in spontaneous DAergic neuronal frequency and bursting activity. These neuronal effects were reversed by simultaneous blockade of 5-HT1A receptor transmission. Finally, using a functional contralateral disconnection procedure, we demonstrated that the ability of intra-NASh CBD to block the formation of conditioned freezing behaviors was dependent on intra-VTA GABAergic transmission substrates. Our findings demonstrate a novel NAcVTA circuit responsible for the behavioral and neuronal effects of CBD within the mesolimbic system via functional interactions with serotonergic 5-HT1A receptor signaling.
Collapse
Affiliation(s)
- Christopher Norris
- Addiction Research Group, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Michael Loureiro
- Addiction Research Group, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Cecilia Kramar
- Addiction Research Group, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Jordan Zunder
- Addiction Research Group, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Justine Renard
- Addiction Research Group, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Walter Rushlow
- Addiction Research Group, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada,Department of Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Steven R Laviolette
- Addiction Research Group, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada,Department of Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada,Department of Psychology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, 468 Medical Science Building, London, ON, Canada N6A 5C1, Tel: +1 519 661 2111, ext 80302, Fax: +1 519 661 3936, E-mail:
| |
Collapse
|
36
|
Poirier GL, Hitora-Imamura N, Sandi C. Emergence in extinction of enhanced and persistent responding to ambiguous aversive cues is associated with high MAOA activity in the prelimbic cortex. Neurobiol Stress 2016; 5:1-7. [PMID: 27981191 PMCID: PMC5145910 DOI: 10.1016/j.ynstr.2016.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/11/2016] [Accepted: 08/30/2016] [Indexed: 11/29/2022] Open
Abstract
There is a great deal of individual variability in the emotional outcomes of potentially traumatic events, and the underlying mechanisms are only beginning to be understood. In order to further our understanding of individual trajectories to trauma, its vulnerability and resilience, we adapted a model of fear expression to ambiguous vs perfect cues in adult male rats, and examined long-term fear extinction, 2, 3, and 50 days from acquisition. After the final conditioned fear test, mitochondrial enzyme monoamine oxidase A (MAOA) function was examined. In order to identify associations between this function and behavioral expression, an a posteri median segregation approach was adopted, and animals were classified as high or low responding according to level of freezing to the ambiguous cue at remote testing, long after the initial extinction. Those individuals characterized by their higher response showed a freezing pattern that persisted from their previous extinction sessions, in spite of their acquisition levels being equivalent to the low-freezing group. Furthermore, unlike more adaptive individuals, freezing levels of high-freezing animals even increased at initial extinction, to almost double their acquisition session levels. Controlling for perfect cue response at remote extinction, greater ambiguous threat cue response was associated with enhanced prelimbic cortex MAOA functional activity. These findings underscore MAOA as a potential target for the development of interventions to mitigate the impact of traumatic experiences. Potentially traumatic event outcomes vary and mechanisms are poorly understood. We examined fear extinction of perfect or ambiguous cues in adult male rats. Higher freezing to ambiguous cue in extinction yet followed equivalent acquisition. Ambiguous cue response was associated with higher prelimbic cortex MAOA function. These findings support targeting MAOA to mitigate impact of traumatic experiences.
Collapse
Affiliation(s)
- Guillaume L Poirier
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Natsuko Hitora-Imamura
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
37
|
Concordance and incongruence in preclinical anxiety models: Systematic review and meta-analyses. Neurosci Biobehav Rev 2016; 68:504-529. [PMID: 27328783 DOI: 10.1016/j.neubiorev.2016.04.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/19/2016] [Accepted: 04/18/2016] [Indexed: 12/14/2022]
Abstract
Rodent defense behavior assays have been widely used as preclinical models of anxiety to study possibly therapeutic anxiety-reducing interventions. However, some proposed anxiety-modulating factors - genes, drugs and stressors - have had discordant effects across different studies. To reconcile the effect sizes of purported anxiety factors, we conducted systematic review and meta-analyses of the literature on ten anxiety-linked interventions, as examined in the elevated plus maze, open field and light-dark box assays. Diazepam, 5-HT1A receptor gene knockout and overexpression, SERT gene knockout and overexpression, pain, restraint, social isolation, corticotropin-releasing hormone and Crhr1 were selected for review. Eight interventions had statistically significant effects on rodent anxiety, while Htr1a overexpression and Crh knockout did not. Evidence for publication bias was found in the diazepam, Htt knockout, and social isolation literatures. The Htr1a and Crhr1 results indicate a disconnect between preclinical science and clinical research. Furthermore, the meta-analytic data confirmed that genetic SERT anxiety effects were paradoxical in the context of the clinical use of SERT inhibitors to reduce anxiety.
Collapse
|
38
|
Abstract
The hippocampus plays an important role in emotional and cognitive processing, and both of these domains are affected in patients with major depressive disorder (MDD). Extensive preclinical research and the notion that modulation of serotonin (5-HT) neurotransmission plays a key role in the therapeutic efficacy of selective serotonin reuptake inhibitors (SSRIs) support the view that 5-HT is important for hippocampal function in normal and disease-like conditions. The hippocampus is densely innervated by serotonergic fibers, and the majority of 5-HT receptor subtypes are expressed there. Furthermore, hippocampal cells often co-express multiple 5-HT receptor subtypes that can have either complementary or opposing effects on cell function, adding to the complexity of 5-HT neurotransmission. Here we review the current knowledge of how 5-HT, through its various receptor subtypes, modulates hippocampal output and the activity of hippocampal pyramidal cells in rodents. In addition, we discuss the relevance of 5-HT modulation for cognitive processing in rodents and possible clinical implications of these results in patients with MDD. Finally, we review the data on how SSRIs and vortioxetine, an antidepressant with multimodal activity, affect hippocampal function, including cognitive processing, from both a preclinical and clinical perspective.
Collapse
|
39
|
Bar-Shira O, Maor R, Chechik G. Gene Expression Switching of Receptor Subunits in Human Brain Development. PLoS Comput Biol 2015; 11:e1004559. [PMID: 26636753 PMCID: PMC4670163 DOI: 10.1371/journal.pcbi.1004559] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 09/15/2015] [Indexed: 01/09/2023] Open
Abstract
Synaptic receptors in the human brain consist of multiple protein subunits, many of which have multiple variants, coded by different genes, and are differentially expressed across brain regions and developmental stages. The brain can tune the electrophysiological properties of synapses to regulate plasticity and information processing by switching from one protein variant to another. Such condition-dependent variant switch during development has been demonstrated in several neurotransmitter systems including NMDA and GABA. Here we systematically detect pairs of receptor-subunit variants that switch during the lifetime of the human brain by analyzing postmortem expression data collected in a population of donors at various ages and brain regions measured using microarray and RNA-seq. To further detect variant pairs that co-vary across subjects, we present a method to quantify age-corrected expression correlation in face of strong temporal trends. This is achieved by computing the correlations in the residual expression beyond a cubic-spline model of the population temporal trend, and can be seen as a nonlinear version of partial correlations. Using these methods, we detect multiple new pairs of context dependent variants. For instance, we find a switch from GLRA2 to GLRA3 that differs from the known switch in the rat. We also detect an early switch from HTR1A to HTR5A whose trends are negatively correlated and find that their age-corrected expression is strongly positively correlated. Finally, we observe that GRIN2B switch to GRIN2A occurs mostly during embryonic development, presumably earlier than observed in rodents. These results provide a systematic map of developmental switching in the neurotransmitter systems of the human brain. Synapses change their properties during development affecting information processing and learning. Most synaptic receptors consist of several proteins, each having several variants coded by closely related genes. These protein variants are similar in structure, yet often differ slightly in their biophysical attributes. Switching a synapse from using one variant to another provides the brain with a way to fine-tune electrophysiological properties of synapses and has been described in NMDA and GABA receptors. Here we describe a systematic approach to detect pairs of context-dependent variants at a genome-wide scale based on a set of post-mortem expression measurements taken from brains at multiple ages. We take into account both the profile of expression as it changes along life and also the detrended age-corrected correlation among genes. This method characterizes the landscape of developmental switches in brain transcriptome, putting forward new candidates pairs for deeper analysis. The abundance of switching between context-dependent variants through life suggests that it is a major mechanism by which the brain tunes its plasticity and information processing.
Collapse
Affiliation(s)
- Ossnat Bar-Shira
- Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Ronnie Maor
- Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Gal Chechik
- Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
- * E-mail:
| |
Collapse
|
40
|
Thomas SA. Neuromodulatory signaling in hippocampus-dependent memory retrieval. Hippocampus 2015; 25:415-31. [PMID: 25475876 DOI: 10.1002/hipo.22394] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2014] [Indexed: 12/15/2022]
Abstract
Considerable advances have been made toward understanding the molecular signaling events that underlie memory acquisition and consolidation. In contrast, less is known about memory retrieval, despite its necessity for utilizing learned information. This review focuses on neuromodulatory and intracellular signaling events that underlie memory retrieval mediated by the hippocampus, for which the most information is currently available. Among neuromodulators, adrenergic signaling is required for the retrieval of various types of hippocampus-dependent memory. Although they contribute to acquisition and/or consolidation, cholinergic and dopaminergic signaling are generally not required for retrieval. Interestingly, while not required for retrieval, serotonergic and opioid signaling may actually constrain memory retrieval. Roles for histamine and non-opioid neuropeptides are currently unclear but possible. A critical effector of adrenergic signaling in retrieval is reduction of the slow afterhyperpolarization mediated by β1 receptors, cyclic AMP, protein kinase A, Epac, and possibly ERK. In contrast, stress and glucocorticoids impair retrieval by decreasing cyclic AMP, mediated in part by the activation of β2 -adrenergic receptors. Clinically, alterations in neuromodulatory signaling and in memory retrieval occur in Alzheimer's disease, Down syndrome, depression, and post-traumatic stress disorder, and recent evidence has begun to link changes in neuromodulatory signaling with effects on memory retrieval.
Collapse
Affiliation(s)
- Steven A Thomas
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
41
|
Lazaroni TLDN, Bastos CP, Moraes MFD, Santos RS, Pereira GS. Angiotensin-(1-7)/Mas axis modulates fear memory and extinction in mice. Neurobiol Learn Mem 2015; 127:27-33. [PMID: 26642920 DOI: 10.1016/j.nlm.2015.11.012] [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] [Received: 08/28/2015] [Revised: 10/23/2015] [Accepted: 11/17/2015] [Indexed: 01/02/2023]
Abstract
Inappropriate defense-alerting reaction to fear is a common feature of neuropsychiatric diseases. Therefore, impairments in brain circuits, as well as in molecular pathways underlying the neurovegetative adjustments to fear may play an essential role on developing neuropsychiatric disorders. Here we tested the hypothesis that interfering with angiotensin-(1-7) [Ang-(1-7)]/Mas receptor axis homeostasis, which appears to be essential to arterial pressure control, would affect fear memory and extinction. Mas knockout (MasKO) mice, in FVB/N background, showed normal cued fear memory and extinction, but increased freezing in response to context. Next, as FVB/N has poor performance in contextual fear memory, we tested MasKO in mixed 129xC57BL/6 background. MasKO mice behaved similarly to wild-type (WT), but memory extinction was slower in contextual fear conditioning to a weak protocol (1CS/US). In addition, delayed extinction in MasKO mice was even more pronounced after a stronger protocol (3CS/US). We showed previously that Angiotensin II receptor AT1 antagonist, losantan, rescued object recognition memory deficit in MasKO mice. Here, losartan was also effective. Memory extinction was accelerated in MasKO mice after treatment with losartan. In conclusion, we showed for the first time that Ang-(1-7)/Mas axis may modulate fear memory extinction. Furthermore, we suggest MasKO mice as an animal model to study post-traumatic stress disorder (PTSD).
Collapse
Affiliation(s)
- Thiago Luiz do Nascimento Lazaroni
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil
| | - Cristiane Perácio Bastos
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil
| | - Márcio Flávio Dutra Moraes
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil
| | - Robson Souza Santos
- Laboratório de Hipertensão, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil
| | - Grace Schenatto Pereira
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil.
| |
Collapse
|
42
|
Ohno Y, Shimizu S, Tokudome K, Kunisawa N, Sasa M. New insight into the therapeutic role of the serotonergic system in Parkinson's disease. Prog Neurobiol 2015; 134:104-21. [DOI: 10.1016/j.pneurobio.2015.09.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 08/17/2015] [Accepted: 09/05/2015] [Indexed: 11/30/2022]
|
43
|
Chilmonczyk Z, Bojarski AJ, Pilc A, Sylte I. Functional Selectivity and Antidepressant Activity of Serotonin 1A Receptor Ligands. Int J Mol Sci 2015; 16:18474-506. [PMID: 26262615 PMCID: PMC4581256 DOI: 10.3390/ijms160818474] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 07/29/2015] [Accepted: 07/31/2015] [Indexed: 01/11/2023] Open
Abstract
Serotonin (5-HT) is a monoamine neurotransmitter that plays an important role in physiological functions. 5-HT has been implicated in sleep, feeding, sexual behavior, temperature regulation, pain, and cognition as well as in pathological states including disorders connected to mood, anxiety, psychosis and pain. 5-HT1A receptors have for a long time been considered as an interesting target for the action of antidepressant drugs. It was postulated that postsynaptic 5-HT1A agonists could form a new class of antidepressant drugs, and mixed 5-HT1A receptor ligands/serotonin transporter (SERT) inhibitors seem to possess an interesting pharmacological profile. It should, however, be noted that 5-HT1A receptors can activate several different biochemical pathways and signal through both G protein-dependent and G protein-independent pathways. The variables that affect the multiplicity of 5-HT1A receptor signaling pathways would thus result from the summation of effects specific to the host cell milieu. Moreover, receptor trafficking appears different at pre- and postsynaptic sites. It should also be noted that the 5-HT1A receptor cooperates with other signal transduction systems (like the 5-HT1B or 5-HT2A/2B/2C receptors, the GABAergic and the glutaminergic systems), which also contribute to its antidepressant and/or anxiolytic activity. Thus identifying brain specific molecular targets for 5-HT1A receptor ligands may result in a better targeting, raising a hope for more effective medicines for various pathologies.
Collapse
Affiliation(s)
- Zdzisław Chilmonczyk
- National Medicines Institute, Chełmska 30/34, 00-725 Warszawa, Poland.
- Institute of Nursing and Health Sciences, University of Rzeszów, W. Kopisto 2A, 35-310 Rzeszów, Poland.
| | - Andrzej Jacek Bojarski
- Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Kraków, Poland.
| | - Andrzej Pilc
- Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Kraków, Poland.
| | - Ingebrigt Sylte
- Faculty of Health Sciences, University of Tromsø-The Arctic University of Norway, No-9037 Tromsø, Norway.
| |
Collapse
|
44
|
Santos M, D'Amico D, Dierssen M. From neural to genetic substrates of panic disorder: Insights from human and mouse studies. Eur J Pharmacol 2015; 759:127-41. [PMID: 25818748 DOI: 10.1016/j.ejphar.2015.03.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 01/15/2015] [Accepted: 03/12/2015] [Indexed: 01/30/2023]
Abstract
Fear is an ancestral emotion, an intrinsic defensive response present in every organism. Although fear is an evolutionarily advantageous emotion, under certain pathologies such as panic disorder it might become exaggerated and non-adaptive. Clinical and preclinical work pinpoints that changes in cognitive processes, such as perception and interpretation of environmental stimuli that rely on brain regions responsible for high-level function, are essential for the development of fear-related disorders. This review focuses on the involvement of cognitive function to fear circuitry disorders. Moreover, we address how animal models are contributing to understand the involvement of human candidate genes to pathological fear and helping achieve progress in this field. Multidisciplinary approaches that integrate human genetic findings with state of the art genetic mouse models will allow to elucidate the mechanisms underlying pathology and to develop new strategies for therapeutic targeting.
Collapse
Affiliation(s)
- Mónica Santos
- Cellular & Systems Neurobiology, Systems Biology Program, Center for Genomic Regulation (CRG), E-08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), E-08003 Barcelona, Spain; CIBER de Enfermedades Raras (CIBERER), E-08003 Barcelona, Spain; Institute of Biology, Otto-von-Guericke University, 39120 Magdeburg, Germany.
| | - Davide D'Amico
- Cellular & Systems Neurobiology, Systems Biology Program, Center for Genomic Regulation (CRG), E-08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), E-08003 Barcelona, Spain; CIBER de Enfermedades Raras (CIBERER), E-08003 Barcelona, Spain; ZeClinics SL, E-08001 Barcelona, Spain.
| | - Mara Dierssen
- Cellular & Systems Neurobiology, Systems Biology Program, Center for Genomic Regulation (CRG), E-08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), E-08003 Barcelona, Spain; CIBER de Enfermedades Raras (CIBERER), E-08003 Barcelona, Spain.
| |
Collapse
|
45
|
Piszczek L, Piszczek A, Kuczmanska J, Audero E, Gross CT. Modulation of anxiety by cortical serotonin 1A receptors. Front Behav Neurosci 2015; 9:48. [PMID: 25759645 PMCID: PMC4338812 DOI: 10.3389/fnbeh.2015.00048] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 02/09/2015] [Indexed: 01/29/2023] Open
Abstract
Serotonin (5-HT) plays an important role in the modulation of behavior across animal species. The serotonin 1A receptor (Htr1a) is an inhibitory G-protein coupled receptor that is expressed both on serotonin and non-serotonin neurons in mammals. Mice lacking Htr1a show increased anxiety behavior suggesting that its activation by serotonin has an anxiolytic effect. This outcome can be mediated by either Htr1a population present on serotonin (auto-receptor) or non-serotonin neurons (hetero-receptor), or both. In addition, both transgenic and pharmacological studies have shown that serotonin acts on Htr1a during development to modulate anxiety in adulthood, demonstrating a function for this receptor in the maturation of anxiety circuits in the brain. However, previous studies have been equivocal about which Htr1a population modulates anxiety behavior, with some studies showing a role of Htr1a hetero-receptor and others implicating the auto-receptor. In particular, cell-type specific rescue and suppression of Htr1a expression in either forebrain principal neurons or brainstem serotonin neurons reached opposite conclusions about the role of the two populations in the anxiety phenotype of the knockout. One interpretation of these apparently contradictory findings is that the modulating role of these two populations depends on each other. Here we use a novel Cre-dependent inducible allele of Htr1a in mice to show that expression of Htr1a in cortical principal neurons is sufficient to modulate anxiety. Together with previous findings, these results support a hetero/auto-receptor interaction model for Htr1a function in anxiety.
Collapse
Affiliation(s)
- Lukasz Piszczek
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| | - Agnieszka Piszczek
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| | - Joanna Kuczmanska
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| | - Enrica Audero
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| | - Cornelius T Gross
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| |
Collapse
|
46
|
Samuels BA, Mendez-David I, Faye C, David SA, Pierz KA, Gardier AM, Hen R, David DJ. Serotonin 1A and Serotonin 4 Receptors: Essential Mediators of the Neurogenic and Behavioral Actions of Antidepressants. Neuroscientist 2014; 22:26-45. [PMID: 25488850 DOI: 10.1177/1073858414561303] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Selective serotonin reuptake inhibitors are the mostly widely used treatment for major depressive disorders and also are prescribed for several anxiety disorders. However, similar to most antidepressants, selective serotonin reuptake inhibitors suffer from two major problems: They only show beneficial effects after 2 to 4 weeks and only about 33% of patients show remission to first-line treatment. Thus, there is a considerable need for development of more effective antidepressants. There is a growing body of evidence supporting critical roles of 5-HT1A and 5-HT4 receptor subtypes in mediating successful depression treatments. In addition, appropriate activation of these receptors may be associated with a faster onset of the therapeutic response. This review will examine the known roles of 5-HT1A and 5-HT4 receptors in mediating both the pathophysiology of depression and anxiety and the treatment of these mood disorders. At the end of the review, the role of these receptors in the regulation of adult hippocampal neurogenesis will also be discussed. Ultimately, we propose that novel antidepressant drugs that selectively target these serotonin receptors could be developed to yield improvements over current treatments for major depressive disorders.
Collapse
Affiliation(s)
- Benjamin Adam Samuels
- Research Foundation for Mental Hygiene, New York State Psychiatric Institute and Department of Psychiatry, Columbia University, New York, NY, USA
| | - Indira Mendez-David
- EA3544 "Pharmacologie des troubles anxio-depressifs et Neurogenese", Faculté de Pharmacie, Université Paris-Sud, 5 Rue J-B Clement, Tour D1, 2e etage, F-92296 Chatenay-Malabry, France
| | - Charlène Faye
- EA3544 "Pharmacologie des troubles anxio-depressifs et Neurogenese", Faculté de Pharmacie, Université Paris-Sud, 5 Rue J-B Clement, Tour D1, 2e etage, F-92296 Chatenay-Malabry, France
| | | | | | - Alain M Gardier
- EA3544 "Pharmacologie des troubles anxio-depressifs et Neurogenese", Faculté de Pharmacie, Université Paris-Sud, 5 Rue J-B Clement, Tour D1, 2e etage, F-92296 Chatenay-Malabry, France
| | - René Hen
- Research Foundation for Mental Hygiene, New York State Psychiatric Institute and Department of Psychiatry, Columbia University, New York, NY, USA
| | - Denis J David
- EA3544 "Pharmacologie des troubles anxio-depressifs et Neurogenese", Faculté de Pharmacie, Université Paris-Sud, 5 Rue J-B Clement, Tour D1, 2e etage, F-92296 Chatenay-Malabry, France
| |
Collapse
|
47
|
Straube B, Reif A, Richter J, Lueken U, Weber H, Arolt V, Jansen A, Zwanzger P, Domschke K, Pauli P, Konrad C, Gerlach AL, Lang T, Fydrich T, Alpers GW, Ströhle A, Wittmann A, Pfleiderer B, Wittchen HU, Hamm A, Deckert J, Kircher T. The functional -1019C/G HTR1A polymorphism and mechanisms of fear. Transl Psychiatry 2014; 4:e490. [PMID: 25514753 PMCID: PMC4270311 DOI: 10.1038/tp.2014.130] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 10/23/2014] [Accepted: 10/26/2014] [Indexed: 01/01/2023] Open
Abstract
Serotonin receptor 1A gene (HTR1A) knockout mice show pronounced defensive behaviour and increased fear conditioning to ambiguous conditioned stimuli. Such behaviour is a hallmark of pathological human anxiety, as observed in panic disorder with agoraphobia (PD/AG). Thus, variations in HTR1A might contribute to neurophysiological differences within subgroups of PD/AG patients. Here, we tested this hypothesis by combining genetic with behavioural techniques and neuroimaging. In a clinical multicentre trial, patients with PD/AG received 12 sessions of manualized cognitive-behavioural therapy (CBT) and were genotyped for HTR1A rs6295. In four subsamples of this multicentre trial, exposure behaviour (n=185), defensive reactivity measured using a behavioural avoidance test (BAT; before CBT: n=245; after CBT: n=171) and functional magnetic resonance imaging (fMRI) data during fear conditioning were acquired before and after CBT (n=39). HTR1A risk genotype (GG) carriers more often escaped during the BAT before treatment. Exploratory fMRI results suggest increased activation of the amygdala in response to threat as well as safety cues before and after treatment in GG carriers. Furthermore, GG carriers demonstrated reduced effects of CBT on differential conditioning in regions including the bilateral insulae and the anterior cingulate cortex. Finally, risk genotype carriers demonstrated reduced self-initiated exposure behaviour to aversive situations. This study demonstrates the effect of HTR1A variation on defensive behaviour, amygdala activity, CBT-induced neural plasticity and normalization of defence behaviour in PD/AG. Our results, therefore, translate evidence from animal studies to humans and suggest a central role for HTR1A in differentiating subgroups of patients with anxiety disorders.
Collapse
Affiliation(s)
- B Straube
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany,Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Rudolf-Bultmann-Strasse 8, 35039 Marburg, Germany. E-mail:
| | - A Reif
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - J Richter
- Department of Biological and Clinical Psychology, University of Greifswald, Greifswald, Germany
| | - U Lueken
- Institute of Clinical Psychology and Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - H Weber
- Department of Biological and Clinical Psychology, University of Greifswald, Greifswald, Germany
| | - V Arolt
- Department of Psychiatry, University of Münster, Münster, Germany
| | - A Jansen
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - P Zwanzger
- Department of Psychiatry, University of Münster, Münster, Germany
| | - K Domschke
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - P Pauli
- Department of Psychology, University of Würzburg, Würzburg, Germany
| | - C Konrad
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany,Department of Psychiatry, University of Münster, Münster, Germany
| | - A L Gerlach
- Department of Psychology, University of Cologne, Cologne, Germany
| | - T Lang
- Institute of Clinical Psychology and Psychotherapy, Technische Universität Dresden, Dresden, Germany,University of Bremen and Christoph-Dornier Foundation for Clinical Psychology, Bremen, Germany
| | - T Fydrich
- Department of Psychology, Humboldt University, Berlin, Germany
| | - G W Alpers
- Department of Psychology, Clinical and Biological Psychology, School of Social Sciences, Mannheim, Germany
| | - A Ströhle
- Department of Psychiatry and Psychotherapy, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - A Wittmann
- Department of Psychiatry and Psychotherapy, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - B Pfleiderer
- Department of Clinical Radiology, University of Münster, Münster, Germany
| | - H-U Wittchen
- Institute of Clinical Psychology and Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - A Hamm
- Department of Biological and Clinical Psychology, University of Greifswald, Greifswald, Germany
| | - J Deckert
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - T Kircher
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
48
|
Baas JMP, Heitland I. The impact of cue learning, trait anxiety and genetic variation in the serotonin 1A receptor on contextual fear. Int J Psychophysiol 2014; 98:506-14. [PMID: 25448266 DOI: 10.1016/j.ijpsycho.2014.10.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 10/07/2014] [Accepted: 10/28/2014] [Indexed: 01/09/2023]
Abstract
In everyday life, aversive events are usually associated with certain predictive cues. Normally, the acquisition of these contingencies enables organisms to appropriately respond to threat. Presence of a threat cue clearly signals 'danger', whereas absence of such cues signals a period of 'safety'. Failure to identify threat cues may lead to chronic states of anxious apprehension in the context in which the threat has been imminent, which may be instrumental in the pathogenesis of anxiety disorders. In this study, existing data from 150 healthy volunteers in a cue and context virtual reality fear conditioning paradigm were reanalyzed. The aim was to further characterize the impact of cue acquisition and trait anxiety, and of a single nucleotide polymorphism in the serotonin 1A receptor gene (5-HTR1A, rs6295), on cued fear and contextual anxiety before and after fear contingencies were explicitly introduced. Fear conditioned responding was quantified with fear potentiation of the eyeblink startle reflex and subjective fear ratings. First, we replicated previous findings that the inability to identify danger cues during acquisition leads to heightened anxious apprehension in the threat context. Second, in subjects who did not identify the danger cue initially, contextual fear was associated with trait anxiety after the contingencies were explicitly instructed. Third, genetic variability within 5-HTR1A (rs6295) was associated with contextual fear independent of awareness or trait anxiety. These findings confirm that failure to acquire cue contingencies impacts contextual fear responding, in association with trait anxiety. The observed 5-HTR1A effect is in line with models of anxiety, but needs further replication.
Collapse
Affiliation(s)
- Johanna M P Baas
- Experimental Psychology and Helmholtz Institute, Utrecht University, The Netherlands; Helmholtz Research Institute, Utrecht, The Netherlands.
| | - Ivo Heitland
- Experimental Psychology and Helmholtz Institute, Utrecht University, The Netherlands; Helmholtz Research Institute, Utrecht, The Netherlands.
| |
Collapse
|
49
|
Islam M, Moriguchi S, Tagashira H, Fukunaga K. Rivastigmine improves hippocampal neurogenesis and depression-like behaviors via 5-HT1A receptor stimulation in olfactory bulbectomized mice. Neuroscience 2014; 272:116-30. [DOI: 10.1016/j.neuroscience.2014.04.046] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 04/23/2014] [Accepted: 04/23/2014] [Indexed: 11/29/2022]
|
50
|
Garcia-Garcia A, Tancredi AN, Leonardo ED. 5-HT(1A) [corrected] receptors in mood and anxiety: recent insights into autoreceptor versus heteroreceptor function. Psychopharmacology (Berl) 2014; 231:623-36. [PMID: 24337875 PMCID: PMC3927969 DOI: 10.1007/s00213-013-3389-x] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 11/26/2013] [Indexed: 11/26/2022]
Abstract
RATIONALE Serotonin (5-HT) neurotransmission is intimately linked to anxiety and depression and a diverse body of evidence supports the involvement of the main inhibitory serotonergic receptor, the serotonin-1A (5-HT(1A)) subtype, in both disorders. OBJECTIVES In this review, we examine the function of 5-HT(1A) receptor subpopulations and re-interpret our understanding of their role in mental illness in light of new data, separating both spatial (autoreceptor versus heteroreceptor) and the temporal (developmental versus adult) roles of the endogenous 5-HT(1A) receptors, emphasizing their distinct actions in mediating anxiety and depression-like behaviors. RESULTS It is difficult to unambiguously distinguish the effects of different populations of the 5-HT(1A) receptors with traditional genetic animal models and pharmacological approaches. However, with the advent of novel genetic systems and subpopulation-selective pharmacological agents, direct evidence for the distinct roles of these populations in governing emotion-related behavior is emerging. CONCLUSIONS There is strong and growing evidence for a functional dissociation between auto- and heteroreceptor populations in mediating anxiety and depressive-like behaviors, respectively. Furthermore, while it is well established that 5-HT(1A) receptors act developmentally to establish normal anxiety-like behaviors, the developmental role of 5-HT(1A) heteroreceptors is less clear, and the specific mechanisms underlying the developmental role of each subpopulation are likely to be key elements determining mood control in adult subjects.
Collapse
Affiliation(s)
- Alvaro Garcia-Garcia
- Department of Psychiatry, Division of Integrative Neuroscience, Columbia University and the New York State Psychiatric Institute, 1051 Riverside Dr. Box 87, New York, NY 10032
- Correspondence should be addressed to either AGG at or EDL at , Telephone: (001) 212-543-5266, Fax: (001) 212-543-5129
| | | | - E. David Leonardo
- Department of Psychiatry, Division of Integrative Neuroscience, Columbia University and the New York State Psychiatric Institute, 1051 Riverside Dr. Box 87, New York, NY 10032
- Correspondence should be addressed to either AGG at or EDL at , Telephone: (001) 212-543-5266, Fax: (001) 212-543-5129
| |
Collapse
|