1
|
Wang SC, Lin CC, Chen CC, Liu YP. Acute Restraint Stress Enhances Prosocial Behavior in Rats via Oxytocin and Fear-Related Circuits. J Integr Neurosci 2025; 24:33400. [PMID: 40302260 DOI: 10.31083/jin33400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 01/11/2025] [Accepted: 01/15/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Stress is a critical determinant of social behavior, with oxytocin playing a key role in buffering stress effects and facilitating social bonding. However, the relationship between stress-induced fear and oxytocin-associated sociability remains unclear, particularly in contexts reminiscent of prior stress. This study investigates whether acute restraint stress (ARS) alters anxiety-related behaviors and prosocial choices, and whether these effects can be modulated by pharmacological intervention targeting the oxytocin and corticotropin-releasing hormone (CRH) systems. METHODS Sprague-Dawley rats were subjected to ARS and assessed for anxiety-like behavior using the elevated T-maze (ETM) and for prosocial behavior using the social choice test (SCT). The effects of the oxytocin receptor antagonist L-368899 and CRH receptor antagonist antalarmin were evaluated in this paradigm. Plasma corticosterone was checked peripherally and the tissue concentrations of serotonin (5-HT), dopamine (DA), and norepinephrine (NE) were measured in the hippocampus, medial prefrontal cortex (mPFC), and amygdala to assess stress-related neurochemical changes in the fear circuit. RESULTS (i) ARS rats showed a significant increase in prosocial preference compared to control, an effect blocked by L-368899 or antalarmin. (ii) ARS rats exhibited reduced corticosterone levels, together with shorter avoidance latency, and longer escape latency in the ETM. (iii) Neurochemically, ARS rats had decreased DA and increased NE levels in the mPFC, both of which were normalized by L-368899 treatment. CONCLUSIONS Oxytocin modulates stress-induced alterations in monoaminergic activity within the mPFC, influencing social choice behavior. These findings provide new insights into the neurobiological mechanisms underlying stress-related sociability and the context-dependent role of oxytocin in fear memory and social behavior.
Collapse
Affiliation(s)
- Sheng-Chiang Wang
- Department of Psychiatry, School of Medicine, National Defense Medical Center, 114 Taipei
- Department of Psychiatry, Tri-Service General Hospital Songshan Branch, 105 Taipei
| | - Chen-Cheng Lin
- Laboratory of Cognitive Neuroscience, Department of Physiology, National Defense Medical Center, 114 Taipei
| | - Chun-Chuan Chen
- Laboratory of Cognitive Neuroscience, Department of Physiology, National Defense Medical Center, 114 Taipei
| | - Yia-Ping Liu
- Laboratory of Cognitive Neuroscience, Department of Physiology, National Defense Medical Center, 114 Taipei
- Department of Psychiatry, Cheng Hsin General Hospital, 112 Taipei
| |
Collapse
|
2
|
Iqbal J, Huang GD, Xue YX, Yang M, Jia XJ. The neural circuits and molecular mechanisms underlying fear dysregulation in posttraumatic stress disorder. Front Neurosci 2023; 17:1281401. [PMID: 38116070 PMCID: PMC10728304 DOI: 10.3389/fnins.2023.1281401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/13/2023] [Indexed: 12/21/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a stress-associated complex and debilitating psychiatric disorder due to an imbalance of neurotransmitters in response to traumatic events or fear. PTSD is characterized by re-experiencing, avoidance behavior, hyperarousal, negative emotions, insomnia, personality changes, and memory problems following exposure to severe trauma. However, the biological mechanisms and symptomatology underlying this disorder are still largely unknown or poorly understood. Considerable evidence shows that PTSD results from a dysfunction in highly conserved brain systems involved in regulating stress, anxiety, fear, and reward circuitry. This review provides a contemporary update about PTSD, including new data from the clinical and preclinical literature on stress, PTSD, and fear memory consolidation and extinction processes. First, we present an overview of well-established laboratory models of PTSD and discuss their clinical translational value for finding various treatments for PTSD. We then highlight the research progress on the neural circuits of fear and extinction-related behavior, including the prefrontal cortex, hippocampus, and amygdala. We further describe different molecular mechanisms, including GABAergic, glutamatergic, cholinergic, and neurotropic signaling, responsible for the structural and functional changes during fear acquisition and fear extinction processes in PTSD.
Collapse
Affiliation(s)
- Javed Iqbal
- Shenzhen Graduate School, Peking University Shenzhen, Guangdong, China
- Department of Addiction Medicine, Shenzhen Engineering Research Center for Precision Psychiatric Technology, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital and Shenzhen Mental Health Center; Clinical College of Mental Health, Shenzhen University Health Science Center; Affiliated Mental Health Center, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Geng-Di Huang
- Shenzhen Graduate School, Peking University Shenzhen, Guangdong, China
- Department of Addiction Medicine, Shenzhen Engineering Research Center for Precision Psychiatric Technology, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital and Shenzhen Mental Health Center; Clinical College of Mental Health, Shenzhen University Health Science Center; Affiliated Mental Health Center, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yan-Xue Xue
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Mei Yang
- Department of Addiction Medicine, Shenzhen Engineering Research Center for Precision Psychiatric Technology, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital and Shenzhen Mental Health Center; Clinical College of Mental Health, Shenzhen University Health Science Center; Affiliated Mental Health Center, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xiao-Jian Jia
- Department of Addiction Medicine, Shenzhen Engineering Research Center for Precision Psychiatric Technology, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital and Shenzhen Mental Health Center; Clinical College of Mental Health, Shenzhen University Health Science Center; Affiliated Mental Health Center, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
3
|
Ortiz-Nazario E, Denton-Ortiz CM, Soto-Escobar LDM, Mateo-Mayol Z, Colon-Romero M, Hernandez-Lopez A, Porter JT. Sex-dependent effects of angiotensin II type 1 receptor blocker on molecular and behavioral changes induced by single prolonged stress. Behav Brain Res 2023; 454:114639. [PMID: 37652238 PMCID: PMC10530531 DOI: 10.1016/j.bbr.2023.114639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a neuropsychiatric disorder that not only entails alterations in fear behavior and anxiety but also includes neuroendocrine dysfunctions involving the hypothalamic pituitary adrenal (HPA) axis and the renin-angiotensin system. Recent preclinical studies demonstrate that activation of the angiotensin type 1 receptor (AT1R) in the paraventricular region of the hypothalamus (PVR) promotes anxiety-like behaviors and enables microglia proliferation. An increase in microglia and anxiety-like behavior also occurs in the PTSD animal model single-prolonged stress (SPS). In the present study, we tested whether AT1Rs contribute to the effects of SPS on behavior and microglia in brain structures important for HPA axis regulation and fear behavior. To test this, male and female animals were exposed to SPS and then given the oral AT1R antagonist candesartan beginning one week later. Candesartan did not alter auditory fear conditioning or extinction in SPS-exposed male or female animals. However, we found that the male animals exposed to SPS showed increased anxiety-like behavior, which was reversed by candesartan. In contrast, neither SPS nor candesartan altered anxiety-like behavior in the female animals. At the molecular level, SPS increased the cellular expression of AT1Rs in the PVR of male animals and candesartan reversed this effect, whereas AT1Rs in the PVR of females were unaltered by either SPS or candesartan. Iba1-expressing microglia increased in the PVR after SPS exposure and was reversed by candesartan in both sexes suggesting that SPS stimulates AT1Rs to increase microglia in the PVR. Collectively, these results suggest that the contribution of AT1Rs to the molecular and behavioral effects of SPS is sex-dependent.
Collapse
Affiliation(s)
- Emily Ortiz-Nazario
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - Carla M Denton-Ortiz
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - Lawry D M Soto-Escobar
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - Zaira Mateo-Mayol
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - Maria Colon-Romero
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - Anixa Hernandez-Lopez
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - James T Porter
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico.
| |
Collapse
|
4
|
On making (and turning adaptive to) maladaptive aversive memories in laboratory rodents. Neurosci Biobehav Rev 2023; 147:105101. [PMID: 36804263 DOI: 10.1016/j.neubiorev.2023.105101] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/03/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
Fear conditioning and avoidance tasks usually elicit adaptive aversive memories. Traumatic memories are more intense, generalized, inflexible, and resistant to attenuation via extinction- and reconsolidation-based strategies. Inducing and assessing these dysfunctional, maladaptive features in the laboratory are crucial to interrogating posttraumatic stress disorder's neurobiology and exploring innovative treatments. Here we analyze over 350 studies addressing this question in adult rats and mice. There is a growing interest in modeling several qualitative and quantitative memory changes by exposing already stressed animals to freezing- and avoidance-related tests or using a relatively high aversive training magnitude. Other options combine aversive/fearful tasks with post-acquisition or post-retrieval administration of one or more drugs provoking neurochemical or epigenetic alterations reported in the trauma aftermath. It is potentially instructive to integrate these procedures and incorporate the measurement of autonomic and endocrine parameters. Factors to consider when defining the organismic and procedural variables, partially neglected aspects (sex-dependent differences and recent vs. remote data comparison) and suggestions for future research (identifying reliable individual risk and treatment-response predictors) are discussed.
Collapse
|
5
|
Sur B, Lee B. Ginsenoside Rg3 modulates spatial memory and fear memory extinction by the HPA axis and BDNF-TrkB pathway in a rat post-traumatic stress disorder. J Nat Med 2022; 76:821-831. [PMID: 35982366 DOI: 10.1007/s11418-022-01636-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 06/13/2022] [Indexed: 12/01/2022]
Abstract
Post-traumatic stress disorder (PTSD) is a serious mental disorder that can develop after exposure to extreme stress. Korean red ginseng, whose major active component is ginsenoside Rg3 (Rg3), is a widely used traditional antioxidant that has anti-inflammatory, anti-apoptotic and anxiolytics effects. This study investigated whether the administration of Rg3 ameliorated the memory deficit induced by a single prolonged stress (SPS) in rats. Male rats were dosed with Rg3 (25 or 50 mg/kg) once daily for 14 days after exposure to SPS. Rg3 administration improved fear memory and spatial memory might be involved in modulating the dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis and monoamine imbalance in the medial prefrontal cortex and hippocampus. It also increased the reduction in the brain-derived neurotrophic factor (BDNF) and tropomyosin-related kinase B (TrkB) mRNAs expression, and the ratio of p-Akt/Akt in the hippocampus. Thus, Rg3 exerted memory-improving actions might be involved in regulating HPA axis and activating BDNF-TrkB pathway. Our findings suggest that Rg3 could be useful for preventing traumatic stress, such as PTSD.
Collapse
Affiliation(s)
- Bongjun Sur
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Bombi Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
- Center for Converging Humanities, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
6
|
Pharmacological Implications of Adjusting Abnormal Fear Memory: Towards the Treatment of Post-Traumatic Stress Disorder. Pharmaceuticals (Basel) 2022; 15:ph15070788. [PMID: 35890087 PMCID: PMC9322538 DOI: 10.3390/ph15070788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a unique clinical mental abnormality presenting a cluster of symptoms in which patients primarily experience flashbacks, nightmares and uncontrollable thoughts about the event that triggered their PTSD. Patients with PTSD may also have comorbid depression and anxiety in an intractable and long-term course, which makes establishing a comprehensive treatment plan difficult and complicated. The present article reviews current pharmacological manipulations for adjusting abnormal fear memory. The roles of the central monoaminergic systems (including serotonin, norepinephrine and dopamine) within the fear circuit areas and the involvement of the hypothalamic-pituitary-adrenal (HPA) axis and glucocorticoid receptor (GR) are explored based on attempts to integrate current clinical and preclinical basic studies. In this review, we explain how these therapeutic paradigms function based on their connections to stages of the abnormal fear memory process from condition to extinction. This may provide useful translational interpretations for clinicians to manage PTSD.
Collapse
|
7
|
Sur B, Kwon S, Hahm DH, Lee B. The Anxiolytic-Like Effects of Protocatechuic Acid in an Animal Model of Post-Traumatic Stress Disorder. J Med Food 2022; 25:495-502. [PMID: 35561272 DOI: 10.1089/jmf.2021.k.0172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a serious psychiatric disorder characterized by impaired fear extinction, depression, and anxiety caused by dysregulation of the hypothalamic-pituitary-adrenal axis and an imbalance of monoamines. Protocatechuic acid (PCA; 3,4-dihydroxybenzoic acid), a major polyphenol metabolite, has various pharmacological effects, such as anti-inflammatory, antioxidant, anti-apoptotic, and neuroprotective activities. In this study, the efficacy of PCA for fear extinction, antidepressant, and anxiolytic effects in PTSD-mediated psychiatric disorders, were evaluated by exposing rats to single prolonged stress (SPS). Male rats were administered PCA (100 or 200 mg/kg) once daily for 14 days after exposure to SPS. PCA significantly decreased situational fear, depressive and anxiety-like behaviors, and corticosterone levels. In addition, PCA regulated the imbalance of serotonin and norepinephrine in the fear circuit region (i.e., the medial prefrontal cortex and hippocampus [Hipp]), and suppressed the decrease in brain-derived neurotrophic factor mRNA expression in the Hipp. The results showed that PCA administration improves freezing behavior and has antidepressant and anti-anxiety effects through modulation of the serotonergic nervous system and monoamines in rats. These results indicated that PCA may be useful as a food ingredient to prevent PTSD.
Collapse
Affiliation(s)
- Bongjun Sur
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Sunoh Kwon
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, Korea
| | - Dae-Hyun Hahm
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bombi Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.,Center for Converging Humanities, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
8
|
Lin CC, Cheng PY, Hsiao M, Liu YP. Effects of RU486 in Treatment of Traumatic Stress-Induced Glucocorticoid Dysregulation and Fear-Related Abnormalities: Early versus Late Intervention. Int J Mol Sci 2022; 23:ijms23105494. [PMID: 35628305 PMCID: PMC9141845 DOI: 10.3390/ijms23105494] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/08/2022] [Accepted: 05/08/2022] [Indexed: 02/06/2023] Open
Abstract
Central glucocorticoid receptor (GR) activity is enhanced following traumatic events, playing a key role in the stress-related cognitive abnormalities of posttraumatic stress disorder (PTSD). GR antagonists are expected to have potential as pharmacological agents to treat PTSD-related symptoms such as anxiety and fear memory disruption. However, an incubation period is usually required and stress-induced abnormalities do not develop immediately following the trauma; thus, the optimal intervention timing should be considered. Single prolonged stress (SPS) was employed as a rodent PTSD model to examine the effects of early or late (1–7 versus 8–14 days after the SPS) sub-chronic RU486 (a GR antagonist) administration. Behaviorally, fear conditioning and anxiety behavior were assessed using the fear-conditioning test and elevated T-maze (ETM), respectively. Neurochemically, the expressions of GR, FK506-binding proteins 4 and 5 (FKBP4 and FKBP5), and early growth response-1 (Egr-1) were assessed in the hippocampus, medial prefrontal cortex (mPFC), amygdala, and hypothalamus, together with the level of plasma corticosterone. Early RU486 administration could inhibit SPS-induced behavioral abnormalities and glucocorticoid system dysregulation by reversing the SPS-induced fear extinction deficit, and preventing SPS-reduced plasma corticosterone levels and SPS-induced Egr-1 overexpression in the hippocampus. Early RU486 administration following SPS also increased the FKBP5 level in the hippocampus and hypothalamus. Finally, both early and late RU486 administration inhibited the elevated hippocampal FKBP4 level and hypothalamus GR level in the SPS rats. Early intervention with a GR antagonist aids in the correction of traumatic stress-induced fear and anxiety dysregulation.
Collapse
Affiliation(s)
- Chen-Cheng Lin
- Laboratory of Cognitive Neuroscience, Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei 11490, Taiwan;
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan;
| | - Pao-Yun Cheng
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan;
| | - Yia-Ping Liu
- Laboratory of Cognitive Neuroscience, Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei 11490, Taiwan;
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei 11490, Taiwan;
- Department of Psychiatry, Cheng Hsin General Hospital, Taipei 11220, Taiwan
- Department of Psychiatry, Tri-Service General Hospital, Taipei 11490, Taiwan
- Correspondence:
| |
Collapse
|
9
|
Lee B. Neuroprotective Effect of Acupuncture against Single Prolonged Stress-Induced Memory Impairments and Inflammation in Rat Brain via Modulation of Brain-Derived Neurotrophic Factor Expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:4430484. [PMID: 35251208 PMCID: PMC8890831 DOI: 10.1155/2022/4430484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/25/2022] [Indexed: 01/04/2023]
Abstract
Posttraumatic stress disorder (PTSD) is a serious mental disorder that can appear after exposure to extreme stress. Acupuncture is an alternative therapy that is widely used to treat various neurodegenerative diseases, as well as cognitive and memory impairments. The aim of this study was to examine whether acupuncture stimulation at a specific acupoint (Shenmen or heart meridian, HT7) could improve memory defects caused by single prolonged stress (SPS) in rats. After exposure to SPS, acupuncture on the HT7 acupoint in male rats was performed, once daily for 21 days. We confirmed that this treatment improved fear memory, cognitive function, and spatial memory by modulating the neuroinflammation and expression of brain-derived neurotrophic factor (BDNF) mRNA in the brain. It also significantly inhibited the activation of proinflammatory cytokines, such as tumor necrosis factor-α and interleukin-1β and the enzyme cyclooxygenase-2 in the brain; it increased the expression of BDNF mRNA in the hippocampus. Our findings provide valuable information concerning the clinical usefulness of acupuncture in the treatment of PTSD.
Collapse
Affiliation(s)
- Bombi Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| |
Collapse
|
10
|
Ferland-Beckham C, Chaby LE, Daskalakis NP, Knox D, Liberzon I, Lim MM, McIntyre C, Perrine SA, Risbrough VB, Sabban EL, Jeromin A, Haas M. Systematic Review and Methodological Considerations for the Use of Single Prolonged Stress and Fear Extinction Retention in Rodents. Front Behav Neurosci 2021; 15:652636. [PMID: 34054443 PMCID: PMC8162789 DOI: 10.3389/fnbeh.2021.652636] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022] Open
Abstract
Posttraumatic stress disorder (PTSD) is a mental health condition triggered by experiencing or witnessing a terrifying event that can lead to lifelong burden that increases mortality and adverse health outcomes. Yet, no new treatments have reached the market in two decades. Thus, screening potential interventions for PTSD is of high priority. Animal models often serve as a critical translational tool to bring new therapeutics from bench to bedside. However, the lack of concordance of some human clinical trial outcomes with preclinical animal efficacy findings has led to a questioning of the methods of how animal studies are conducted and translational validity established. Thus, we conducted a systematic review to determine methodological variability in studies that applied a prominent animal model of trauma-like stress, single prolonged stress (SPS). The SPS model has been utilized to evaluate a myriad of PTSD-relevant outcomes including extinction retention. Rodents exposed to SPS express an extinction retention deficit, a phenotype identified in humans with PTSD, in which fear memory is aberrantly retained after fear memory extinction. The current systematic review examines methodological variation across all phases of the SPS paradigm, as well as strategies for behavioral coding, data processing, statistical approach, and the depiction of data. Solutions for key challenges and sources of variation within these domains are discussed. In response to methodological variation in SPS studies, an expert panel was convened to generate methodological considerations to guide researchers in the application of SPS and the evaluation of extinction retention as a test for a PTSD-like phenotype. Many of these guidelines are applicable to all rodent paradigms developed to model trauma effects or learned fear processes relevant to PTSD, and not limited to SPS. Efforts toward optimizing preclinical model application are essential for enhancing the reproducibility and translational validity of preclinical findings, and should be conducted for all preclinical psychiatric research models.
Collapse
Affiliation(s)
| | - Lauren E Chaby
- Cohen Veterans Bioscience, New York City, NY, United States
| | - Nikolaos P Daskalakis
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States.,McLean Hospital, Belmont, MA, United States
| | - Dayan Knox
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
| | - Israel Liberzon
- Department of Psychiatry, Texas A&M University, Bryan, TX, United States
| | - Miranda M Lim
- Departments of Neurology, Behavioral Neuroscience, Medicine, Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, United States.,Sleep Disorders Clinic, VA Portland Health Care System, Portland, OR, United States
| | - Christa McIntyre
- Department of Neuroscience, The University of Texas at Dallas, Richardson, TX, United States
| | - Shane A Perrine
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States.,Research Service, John. D. Dingell VA Medical Center, Detroit, MI, United States
| | - Victoria B Risbrough
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States.,Center for Excellence in Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, United States
| | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, United States
| | | | - Magali Haas
- Cohen Veterans Bioscience, New York City, NY, United States
| |
Collapse
|
11
|
Zhou P, Deng M, Wu J, Lan Q, Yang H, Zhang C. Ventral Tegmental Area Dysfunction and Disruption of Dopaminergic Homeostasis: Implications for Post-traumatic Stress Disorder. Mol Neurobiol 2021; 58:2423-2434. [PMID: 33428093 DOI: 10.1007/s12035-020-02278-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/30/2020] [Indexed: 12/27/2022]
Abstract
Post-traumatic stress disorder (PTSD) is a debilitating psychiatric condition characterized by intrusive recollections of the traumatic event, avoidance behaviors, hyper-arousal to event-related cues, cognitive disruption, and mood dysregulation. Accumulating preclinical and clinical evidence implicates dysfunction of the ventral tegmental area (VTA) dopaminergic system in PTSD pathogenesis. This article reviews recent advances in our knowledge of the relationship between dopaminergic dyshomeostasis and PTSD, including the contributions of specific dopaminergic gene variants to disease susceptibility, alterations in VTA dopamine neuron activity, dysregulation of dopaminergic transmission, and potential pharmacological and psychological interventions for PTSD targeting the dopaminergic system. An in-depth understanding of PTSD etiology is crucial for the development of innovative risk assessment, diagnostic, and treatment strategies following traumatic events.
Collapse
Affiliation(s)
- Peiling Zhou
- School of Educational Sciences & Guangdong Provincial Key Laboratory of Development and Education for Special Needs Children, Lingnan Normal University, 29 Cunjing Road, Chikan District, Zhanjiang, 524048, China
| | - Meiping Deng
- School of Educational Sciences & Guangdong Provincial Key Laboratory of Development and Education for Special Needs Children, Lingnan Normal University, 29 Cunjing Road, Chikan District, Zhanjiang, 524048, China
| | - Jiashan Wu
- School of Educational Sciences & Guangdong Provincial Key Laboratory of Development and Education for Special Needs Children, Lingnan Normal University, 29 Cunjing Road, Chikan District, Zhanjiang, 524048, China
| | - Qinghui Lan
- School of Educational Sciences & Guangdong Provincial Key Laboratory of Development and Education for Special Needs Children, Lingnan Normal University, 29 Cunjing Road, Chikan District, Zhanjiang, 524048, China
| | - Huifang Yang
- School of Educational Sciences & Guangdong Provincial Key Laboratory of Development and Education for Special Needs Children, Lingnan Normal University, 29 Cunjing Road, Chikan District, Zhanjiang, 524048, China.
| | - Changzheng Zhang
- School of Educational Sciences & Guangdong Provincial Key Laboratory of Development and Education for Special Needs Children, Lingnan Normal University, 29 Cunjing Road, Chikan District, Zhanjiang, 524048, China. .,School of Psychology, Nanjing Normal University, 122 Ninghai Road, Gulou District, Nanjing, 210097, China.
| |
Collapse
|
12
|
Belda X, Fuentes S, Labad J, Nadal R, Armario A. Acute exposure of rats to a severe stressor alters the circadian pattern of corticosterone and sensitizes to a novel stressor: Relationship to pre-stress individual differences in resting corticosterone levels. Horm Behav 2020; 126:104865. [PMID: 32991887 DOI: 10.1016/j.yhbeh.2020.104865] [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: 05/23/2020] [Revised: 08/03/2020] [Accepted: 09/15/2020] [Indexed: 11/17/2022]
Abstract
Traumatic events have been proposed to be associated with hypo-activity of the hypothalamic-pituitary-adrenal (HPA) axis, but data in animal models exposed to severe stressors are controversial and have important methodological concerns. Individual differences in resting or stress levels of corticosterone might explain some of the inconsistencies. We then studied this issue in male rats exposed to 2 h immobilization on boards (IMO), a severe stressor. Thirty-six rats were blood sampled under resting conditions four times a day on three non-consecutive days. Then, they were assigned to control (n = 14) or IMO (n = 22) to study the HPA response to IMO, the stressor-induced alterations in the circadian pattern of corticosterone (CPCORT), and the behavioral and HPA responsiveness to an open-field. Individual differences in pre-IMO resting corticosterone were inconsistent, but averaging data markedly improved consistency. The CPCORT was markedly altered on day 1 post-IMO (higher trough and lower peak levels), less altered on day 3 and apparently normal on day 7. Importantly, when rats were classified in low and high resting corticosterone groups (LCORT and HCORT, respectively), on the basis of the area under the curve (AUC) of the averaged pre-IMO data, AUC differences between LCORT and HCORT groups were maintained in controls but disappeared in IMO rats during the post-IMO week. Open-field hypo-activity and corticosterone sensitization were similar in LCORT and HCORT groups nine days after IMO. A single IMO exposure causes long-lasting HPA alterations, some of them dependent on pre-stress resting corticosterone levels, with no evidence for post-IMO resting corticosterone hypo-activity.
Collapse
MESH Headings
- Adrenocorticotropic Hormone/blood
- Animals
- Circadian Rhythm/physiology
- Conditioning, Classical/physiology
- Corticosterone/blood
- Corticosterone/metabolism
- Hypothalamo-Hypophyseal System/metabolism
- Individuality
- Male
- Pituitary-Adrenal System/metabolism
- Rats
- Rats, Sprague-Dawley
- Rest/physiology
- Rest/psychology
- Restraint, Physical/physiology
- Restraint, Physical/psychology
- Stress Disorders, Post-Traumatic/blood
- Stress Disorders, Post-Traumatic/etiology
- Stress Disorders, Post-Traumatic/metabolism
- Stress Disorders, Post-Traumatic/psychology
- Stress, Psychological/blood
- Stress, Psychological/metabolism
Collapse
Affiliation(s)
- Xavier Belda
- Institut de Neurociències, Spain; Animal Physiology Unit (Department of Cellular Biology, Physiology and Immunology), Faculty of Biosciences, Universitat Autònoma de Barcelona, Spain
| | - Silvia Fuentes
- Institut de Neurociències, Spain; Psychobiology Unit, Faculty of Psychology, Universitat Autònoma de Barcelona, Spain
| | - Javier Labad
- Department of Mental Health, Parc Taulí Hospital Universitari, I3PT, Spain; Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Spain; CIBERSAM, Spain
| | - Roser Nadal
- Institut de Neurociències, Spain; Psychobiology Unit, Faculty of Psychology, Universitat Autònoma de Barcelona, Spain; CIBERSAM, Spain
| | - Antonio Armario
- Institut de Neurociències, Spain; Animal Physiology Unit (Department of Cellular Biology, Physiology and Immunology), Faculty of Biosciences, Universitat Autònoma de Barcelona, Spain; CIBERSAM, Spain.
| |
Collapse
|
13
|
Chaby LE, Sadik N, Burson NA, Lloyd S, O'Donnel K, Winters J, Conti AC, Liberzon I, Perrine SA. Repeated stress exposure in mid-adolescence attenuates behavioral, noradrenergic, and epigenetic effects of trauma-like stress in early adult male rats. Sci Rep 2020; 10:17935. [PMID: 33087769 PMCID: PMC7578655 DOI: 10.1038/s41598-020-74481-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022] Open
Abstract
Stress in adolescence can regulate vulnerability to traumatic stress in adulthood through region-specific epigenetic activity and catecholamine levels. We hypothesized that stress in adolescence would increase adult trauma vulnerability by impairing extinction-retention, a deficit in PTSD, by (1) altering class IIa histone deacetylases (HDACs), which integrate effects of stress on gene expression, and (2) enhancing norepinephrine in brain regions regulating cognitive effects of trauma. We investigated the effects of adolescent-stress on adult vulnerability to severe stress using the single-prolonged stress (SPS) model in male rats. Rats were exposed to either (1) adolescent-stress (33-35 postnatal days) then SPS (58-60 postnatal days; n = 14), or (2) no adolescent-stress and SPS (58-60 postnatal days; n = 14), or (3) unstressed conditions (n = 8). We then measured extinction-retention, norepinephrine, HDAC4, and HDAC5. As expected, SPS exposure induced an extinction-retention deficit. Adolescent-stress prior to SPS eliminated this deficit, suggesting adolescent-stress conferred resiliency to adult severe stress. Adolescent-stress also conferred region-specific resilience to norepinephrine changes. HDAC4 and HDAC5 were down-regulated following SPS, and these changes were also modulated by adolescent-stress. Regulation of HDAC levels was consistent with the pattern of cognitive effects of SPS; only animals exposed to SPS without adolescent-stress exhibited reduced HDAC4 and HDAC5 in the prelimbic cortex, hippocampus, and striatum. Thus, HDAC regulation caused by severe stress in adulthood interacts with stress history such that seemingly conflicting reports describing effects of adolescent stress on adult PTSD vulnerability may stem in part from dynamic HDAC changes following trauma that are shaped by adolescent stress history.
Collapse
MESH Headings
- Adolescent
- Adolescent Behavior/physiology
- Adolescent Behavior/psychology
- Animals
- Brain/metabolism
- Disease Models, Animal
- Epigenesis, Genetic
- Extinction, Psychological/physiology
- Histone Deacetylases/metabolism
- Humans
- Male
- Norepinephrine/metabolism
- Psychology, Adolescent
- Rats, Sprague-Dawley
- Retention, Psychology/physiology
- Stress Disorders, Post-Traumatic/etiology
- Stress Disorders, Post-Traumatic/genetics
- Stress Disorders, Post-Traumatic/metabolism
- Stress Disorders, Post-Traumatic/psychology
- Stress, Psychological
Collapse
Affiliation(s)
- Lauren E Chaby
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA.
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA.
| | - Nareen Sadik
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nicole A Burson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Scott Lloyd
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Research Service, John D. Dingell VA Medical Center, Detroit, MI, USA
| | - Kelly O'Donnel
- Department of Psychology, University of Colorado, Colorado Springs, CO, USA
| | - Jesse Winters
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Alana C Conti
- Research Service, John D. Dingell VA Medical Center, Detroit, MI, USA
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Israel Liberzon
- Department of Psychiatry, Texas A&M College of Medicine, Bryan, TX, USA
| | - Shane A Perrine
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Research Service, John D. Dingell VA Medical Center, Detroit, MI, USA
| |
Collapse
|
14
|
Lin CC, Cheng PY, Liu YP. Effects of early life social experience on fear extinction and related glucocorticoid profiles - behavioral and neurochemical approaches in a rat model of PTSD. Behav Brain Res 2020; 391:112686. [PMID: 32428628 DOI: 10.1016/j.bbr.2020.112686] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/30/2020] [Accepted: 04/30/2020] [Indexed: 01/06/2023]
Abstract
People may agonize over an intrusive fear-inducing memory even when the traumatic event has passed, which is the principle manifestation of posttraumatic stress disorder (PTSD). However, many traumatized people do not present symptoms of PTSD, implying that certain hidden factors help those individuals to cope with the traumatic stress. Increasing evidence suggests that early life experience may serve as a predisposing factor in the development of PTSD. For example, early life social deprivation disrupts the glucocorticoid system, one of the biological abnormalities of PTSD. By employing isolation rearing (IR) with a subsequent single prolonged stress (SPS) paradigm, we examined the hypothesis that early-life social experience may change the outcome of traumatic stress in both behavioral and neurochemical profiles. Behaviorally, the performance of rats on a Pavlovian fear conditioning test was measured to evaluate their retrieval ability of fear memory extinction. Neurochemically, plasma corticosterone levels and glucocorticoid receptor (GR), FK506-binding proteins 4 and 5 (FKBP4 and FKBP5) and early growth response-1 (Egr-1) expression were measured in GR-abundant brain areas, including the hypothalamus, medial prefrontal cortex, and hippocampus. Our results demonstrated an area-dependent IR effect on the SPS outcomes. IR prevented the SPS-impaired fear extinction retrieval ability and averted the SPS-elevated expression of GR, FKBP4, and Egr-1 in the hippocampus, whereas it did not change the SPS-reduced plasma corticosterone levels and SPS-enhanced GR activity in the mPFC and hypothalamus. The present study provides some new insights to support the hypothesis that early-life experience may play a role in the occurrence of PTSD.
Collapse
Affiliation(s)
- Chen-Cheng Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11490, Taiwan; Department of Psychiatry, Cheng Hsin General Hospital, Taipei 11220, Taiwan; Laboratory of Cognitive Neuroscience, Department of Physiology, National Defense Medical Center, Taipei 11490, Taiwan
| | - Pao-Yun Cheng
- Department of Physiology, National Defense Medical Center, Taipei 11490, Taiwan
| | - Yia-Ping Liu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11490, Taiwan; Department of Psychiatry, Cheng Hsin General Hospital, Taipei 11220, Taiwan; Laboratory of Cognitive Neuroscience, Department of Physiology, National Defense Medical Center, Taipei 11490, Taiwan.
| |
Collapse
|
15
|
Lin CC, Chen TY, Cheng PY, Liu YP. Early life social experience affects adulthood fear extinction deficit and associated dopamine profile abnormalities in a rat model of PTSD. Prog Neuropsychopharmacol Biol Psychiatry 2020; 101:109914. [PMID: 32165120 DOI: 10.1016/j.pnpbp.2020.109914] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/13/2020] [Accepted: 03/06/2020] [Indexed: 12/20/2022]
Abstract
Individuals may develop fear extinction deficits after life-threatening traumatic events; such deficits indicate posttraumatic stress disorder (PTSD). Because the occurrence of this disorder differs among people who have experienced trauma, hidden underlying factors should be determined. Increasing evidence suggests the involvement of neuronal dysregulation of information processes or cognitive function during development. This neuronal dysregulation is caused by disturbances in dopamine (DA) transmission within the fear circuit, which comprises the medial prefrontal cortex (mPFC), amygdala, and hippocampus. Single prolonged stress (SPS) combined with an isolation rearing (IR) paradigm was used to randomly assign rats to four groups [social rearing-no SPS (SR-NS), SR-SPS, IR-NS, and IR-SPS], and their performance in prepulse inhibition (PPI) and on Pavlovian fear conditioning tests was assessed. Tissue DA levels and the expression of DA receptors (D1R and D2R) in the fear circuit were measured at the end of the experiment. Our results indicated that PPI deficits and fear extinction problems were specific to rats subjected to IR and SPS, respectively. Furthermore, IR-induced PPI deficits were not influenced by SPS, but SPS-induced fear extinction retrieval impairment could be adjusted according to previous IR experiences. Neurochemically, tissue DA levels and D1R expression in the mPFC and amygdala were nonspecifically reduced by IR and SPS, whereas D2R expression in the mPFC and amygdala was higher in IR-SPS than in SR-SPS rats. These findings suggest that early life experiences may influence fear responses in adulthood through a change in DA profiles within the fear circuit.
Collapse
Affiliation(s)
- Chen-Cheng Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11490, Taiwan; Department of Psychiatry, Cheng Hsin General Hospital, Taipei 11220, Taiwan; Laboratory of Cognitive Neuroscience, Department of Physiology, National Defense Medical Center, Taipei 11490, Taiwan
| | - Tzung-Yan Chen
- Department of Psychiatry, Cheng Hsin General Hospital, Taipei 11220, Taiwan; Laboratory of Cognitive Neuroscience, Department of Physiology, National Defense Medical Center, Taipei 11490, Taiwan
| | - Pao-Yun Cheng
- Department of Physiology, National Defense Medical Center, Taipei 11490, Taiwan.
| | - Yia-Ping Liu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11490, Taiwan; Department of Psychiatry, Cheng Hsin General Hospital, Taipei 11220, Taiwan; Laboratory of Cognitive Neuroscience, Department of Physiology, National Defense Medical Center, Taipei 11490, Taiwan.
| |
Collapse
|
16
|
Lee B, Sur B, Lee H, Oh S. Korean Red Ginseng prevents posttraumatic stress disorder-triggered depression-like behaviors in rats via activation of the serotonergic system. J Ginseng Res 2020; 44:644-654. [PMID: 32617045 PMCID: PMC7322749 DOI: 10.1016/j.jgr.2019.09.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 09/04/2019] [Accepted: 09/25/2019] [Indexed: 01/28/2023] Open
Abstract
Background Posttraumatic stress disorder (PTSD), a mental disorder induced by traumatic stress and often accompanied by depression and/or anxiety, may involve an imbalance in the neurotransmitters associated with the fear response. Korean Red Ginseng (KRG) has long been used as a traditional medicine and is known to be involved in a variety of pharmacological activities. We used the open field test and forced swimming test to examine the effects of KRG on the depression-like response of rats after exposure to single prolonged stress (SPS), leading to activation of the serotonergic system. Methods Male rats received KRG (30, 50, and 100 mg/kg, intraperitoneal injection) once daily for 14 days after exposure to SPS. Results Daily KRG administration significantly improved depression-like behaviors in the forced swimming test, increased the number of lines crossed and time spent in the central zone in the open field test, and decreased freezing behavior in contextual and cued fear conditioning. KRG treatment attenuated SPS-induced decreases in serotonin (5-HT) tissue concentrations in the hippocampus and medial prefrontal cortex. The increased 5-HT concentration during KRG treatment may be partially attributable to the 5-hydroxyindoleacetic acid/5-HT ratio in the hippocampus of rats with PTSD. These effects may be caused by the activation of hippocampal genes encoding tryptophan hydroxylase-1 and 2 mRNA levels. Conclusion Our findings suggest that KRG has an antidepressant effect in rats subjected to SPS and may represent an effective use of traditional medicine for the treatment of PTSD.
Collapse
Affiliation(s)
- Bombi Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.,Center for Converging Humanities, Kyung Hee University, Seoul, Republic of Korea
| | - Bongjun Sur
- Department of Molecular medicine and TIDRC, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Hyejung Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Seikwan Oh
- Department of Molecular medicine and TIDRC, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
17
|
Mohammadi-Farani A, Pourmotabbed A, Ardeshirizadeh Y. Effects of HDAC inhibitors on spatial memory and memory extinction in SPS-induced PTSD rats. Res Pharm Sci 2020; 15:241-248. [PMID: 33088324 PMCID: PMC7540814 DOI: 10.4103/1735-5362.288426] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 02/06/2020] [Accepted: 02/25/2020] [Indexed: 11/04/2022] Open
Abstract
Background and purpose Neurobiological changes in memory processes seem to play a role in the pathophysiology of post-traumatic stress disorder (PTSD). Memory itself is influenced by PTSD, too. Histone deacetylase inhibitors (HDAIs) have shown promising results in the extinction of fear-related memories in animals and hence they seem to be important for the treatment of PTSD. Data are scarce about the effect of HDAIs in spatial memory formation/extinction in PTSD models. The main goal of the present work is to find the effect of sodium butyrate (NaBu), as an HDAI, on spatial memory and spatial memory extinction in rats exposed to single prolonged stress procedure (SPS). Experimental approach Different doses of NaBu were administered subcutaneously for 7 days in different groups of rats after SPS procedure. Learning, memory, and extinction of memory were evaluated in the Morris water maze test of spatial memory in 6 consecutive days. Findings / Results The results show that NaBu (0.5 mg/kg) alleviates impaired learning and memory in SPS rats. It also facilitates the extinction of newly formed memory in the animals. Conclusion and implications Our data suggest that the administration of HDAIs after a traumatic experience can prevent the aversive effects of SPS on spatial memory. It also reinforces the notion that extinction of spatial memory involves the same or similar brain circuitry that is involved in the extinction of fear memories in PTSD patients.
Collapse
Affiliation(s)
- Ahmad Mohammadi-Farani
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, I.R. Iran.,Department of Pharmacology, Toxicology and Medical Services, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, I.R. Iran
| | - Ali Pourmotabbed
- Department of Physiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, I.R. Iran
| | - Yazdan Ardeshirizadeh
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, I.R. Iran
| |
Collapse
|
18
|
Papalini S, Beckers T, Vervliet B. Dopamine: from prediction error to psychotherapy. Transl Psychiatry 2020; 10:164. [PMID: 32451377 PMCID: PMC7248121 DOI: 10.1038/s41398-020-0814-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 04/14/2020] [Accepted: 04/21/2020] [Indexed: 02/07/2023] Open
Abstract
Dopamine, one of the main neurotransmitters in the mammalian brain, has been implicated in the coding of prediction errors that govern reward learning as well as fear extinction learning. Psychotherapy too can be viewed as a form of error-based learning, because it challenges erroneous beliefs and behavioral patterns in order to induce long-term changes in emotions, cognitions, and behaviors. Exposure therapy, for example, relies in part on fear extinction principles to violate erroneous expectancies of danger and induce novel safety learning that inhibits and therefore reduces fear in the long term. As most forms of psychotherapy, however, exposure therapy suffers from non-response, dropout, and relapse. This narrative review focuses on the role of midbrain and prefrontal dopamine in novel safety learning and investigates possible pathways through which dopamine-based interventions could be used as an adjunct to improve both the response and the long-term effects of the therapy. Convincing evidence exists for an involvement of the midbrain dopamine system in the acquisition of new, safe memories. Additionally, prefrontal dopamine is emerging as a key ingredient for the consolidation of fear extinction. We propose that applying a dopamine prediction error perspective to psychotherapy can inspire both pharmacological and non-pharmacological studies aimed at discovering innovative ways to enhance the acquisition of safety memories. Additionally, we call for further empirical investigations on dopamine-oriented drugs that might be able to maximize consolidation of successful fear extinction and its long-term retention after therapy, and we propose to also include investigations on non-pharmacological interventions with putative prefrontal dopaminergic effects, like working memory training.
Collapse
Affiliation(s)
- Silvia Papalini
- Laboratory of Biological Psychology (LBP), Faculty of Psychology and Educational Sciences, KU Leuven, Leuven, Belgium. .,Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| | - Tom Beckers
- grid.5596.f0000 0001 0668 7884Leuven Brain Institute, KU Leuven, Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Centre for the Psychology of Learning and Experimental Psychopathology (CLEP), Faculty of Psychology and Educational Sciences, KU Leuven, Leuven, Belgium
| | - Bram Vervliet
- grid.5596.f0000 0001 0668 7884Laboratory of Biological Psychology (LBP), Faculty of Psychology and Educational Sciences, KU Leuven, Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Antidepressant-Like Effects of Hesperidin in Animal Model of Post-Traumatic Stress Disorder. Chin J Integr Med 2020; 27:39-46. [PMID: 32445019 DOI: 10.1007/s11655-020-2724-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Post-traumatic stress disorder (PTSD) is a psychiatric disorder characterized by depression and anxiety, that arises due to an imbalance of neurotransmitters in response to excessive stress. Hesperidin (HSD) is a naturally occurring flavonoid shown to exert a variety of biological activities, including antioxidant, anti-inflammatory, and neuroprotective effects. METHODS This study was used the open field test (OFT) and forced swimming test (FST) to examine the effects of HSD on the depression-like response of rats after exposure to a single prolonged stress (SPS) leading to the dysregulation of the serotonergic activation system. Male rats were given HSD (20, 50, and 100 mg/kg, intraperitoneal injection, n=6-7 per group) once daily for 14 days after exposure to SPS. The influence of administration of HSD on SPS-induced behavioral responses and concentrations of serotonin (5-HT), 5-hydroxyindoleacetic acid (5-HIAA), and monoamine oxidase-A (MAO-A) in the rat brain were also investigated using enzyme-linked immunoassays (ELISAs). RESULTS Daily HSD administration signifificantly improved depression-like behaviors in the FST (P0.05), increased the number of lines crossed in the central zone of the OFT (P0.01), and reduced freezing behavior both in contextual and cued fear conditioning. HSD treatment also attenuated the reduction in SPS-induced 5-HT concentrations in the hippocampus and amygdala. This increase in 5-HT concentrations during HSD treatment was partially attributed to a decrease in the 5-HIAA/5-HT ratio in the hippocampus of rats with PTSD. Furthermore, HSD treatment inhibited activity of MAO-A and decreases of tryptophan hydroxylase-1 expression in the hippocampus. CONCLUSION HSD was shown to exert antidepressant effects in rats exposed to SPS, suggesting that this natural flflavonoid may be an effective medicine for PTSD.
Collapse
|
20
|
Lee B, Choi GM, Shim I, Lee H. Genistein Prevents Single Prolonged Stress-Induced Cognitive Impairment in a Post-Traumatic Stress Disorder Rat Model via Activation of the Serotonergic System. J Med Food 2020; 23:476-484. [PMID: 32267780 DOI: 10.1089/jmf.2019.4519] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a stress-associated mental disorder characterized by an imbalance of neurotransmitters in response to traumatic events or fear. Genistein (GEN), a natural isoflavone, has been shown to exhibit neuroprotective effects. Here, we used the Morris water maze (MWM) and object recognition task (ORT) tests to examine the effects of GEN on cognitive impairment in rats after exposure to single prolonged stress (SPS), and its interaction with the serotonergic system. After exposure to SPS, male rats received GEN (2, 4, and 10 mg/kg, i.p.) for 14 days. Daily GEN administration significantly improved cognitive function in the ORT and MWM tests. GEN treatment also inhibited SPS-induced decreases in serotonin (5-HT) levels in the medial prefrontal cortex and hippocampus. These increased 5-HT concentrations in response to GEN treatment could be partially attributed to the ratio of 5-hydroxyindoleacetic acid/5-HT in the hippocampus. Our findings suggest that GEN significantly attenuates SPS-induced memory deficits in rats and may represent an effective therapeutic option for the treatment of PTSD.
Collapse
Affiliation(s)
- Bombi Lee
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul, Korea.,Center for Converging Humanities, Kyung Hee University, Seoul, Korea
| | - Gwang Muk Choi
- The Graduate School of Basic Science of Medicine, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Insop Shim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Hyejung Lee
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul, Korea
| |
Collapse
|
21
|
Skórzewska A, Lehner M, Wisłowska-Stanek A, Turzyńska D, Sobolewska A, Krząścik P, Szyndler J, Maciejak P, Chmielewska N, Kołosowska K, Płaźnik A. Individual susceptibility or resistance to posttraumatic stress disorder-like behaviours. Behav Brain Res 2020; 386:112591. [PMID: 32194190 DOI: 10.1016/j.bbr.2020.112591] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 12/20/2022]
Abstract
The aim of this study was to explore the neurobiological background of individual susceptibility and resistance to the development of posttraumatic stress disorder (PTSD)-like behaviours. Rats were divided into susceptible, PTSD(+), and resistant, PTSD(-), groups based on freezing duration during exposure to aversive context and the time spent in the central area in open field test one week after threefold stress experience (modified single prolonged stress). PTSD(-) rats showed increased concentrations of corticosterone in plasma and changes in GAD67 expression: decreased in the infralimbic cortex (IL) and increased in the lateral amygdala (LA), dentate gyrus (DG), and CA1 area of the hippocampus. Moreover, in this group, we found an increase in the number of CRF-positive nuclei in the parvocellular neurons of the paraventricular hypothalamic nucleus (pPVN). The PTSD(+) group, compared to PTSD(-) rats, had decreased concentrations of corticosterone in plasma and reduced CRF expression in the pPVN, higher CRF expression in the CA1, increased expression of CRF-positive nuclei and GR receptors in the CA3 area of the hippocampus, and increased expression of GR receptors in the DG and the central amygdala (CeA). Biochemical analysis showed higher concentrations of noradrenaline, glutamic acid in the dorsal hippocampus and amygdala and lower levels of dopamine and its metabolites in the amygdala of the PTSD(+) group than in the PTSD(-) group. The study revealed different behavioural and biochemical profiles of PTSD(+) and PTSD(-) rats and suggested that individual differences in hypothalamic-pituitary-adrenal (HPA) axis activity may determine hippocampal- and amygdala-dependent memory and fear processing.
Collapse
Affiliation(s)
- Anna Skórzewska
- Department of Neurochemistry, Institute of Psychiatry and Neurology, 9 Sobieskiego Street, 02-957, Warsaw, Poland.
| | - Małgorzata Lehner
- Department of Neurochemistry, Institute of Psychiatry and Neurology, 9 Sobieskiego Street, 02-957, Warsaw, Poland
| | - Aleksandra Wisłowska-Stanek
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, 1B Banacha Street, 02-097, Warsaw, Poland
| | - Danuta Turzyńska
- Department of Neurochemistry, Institute of Psychiatry and Neurology, 9 Sobieskiego Street, 02-957, Warsaw, Poland
| | - Alicja Sobolewska
- Department of Neurochemistry, Institute of Psychiatry and Neurology, 9 Sobieskiego Street, 02-957, Warsaw, Poland
| | - Paweł Krząścik
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, 1B Banacha Street, 02-097, Warsaw, Poland
| | - Janusz Szyndler
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, 1B Banacha Street, 02-097, Warsaw, Poland
| | - Piotr Maciejak
- Department of Neurochemistry, Institute of Psychiatry and Neurology, 9 Sobieskiego Street, 02-957, Warsaw, Poland; Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, 1B Banacha Street, 02-097, Warsaw, Poland
| | - Natalia Chmielewska
- Department of Neurochemistry, Institute of Psychiatry and Neurology, 9 Sobieskiego Street, 02-957, Warsaw, Poland
| | - Karolina Kołosowska
- Department of Neurochemistry, Institute of Psychiatry and Neurology, 9 Sobieskiego Street, 02-957, Warsaw, Poland
| | - Adam Płaźnik
- Department of Neurochemistry, Institute of Psychiatry and Neurology, 9 Sobieskiego Street, 02-957, Warsaw, Poland
| |
Collapse
|
22
|
Lee B, Choi GM, Sur B. Silibinin prevents depression-like behaviors in a single prolonged stress rat model: the possible role of serotonin. BMC Complement Med Ther 2020; 20:70. [PMID: 32143600 PMCID: PMC7076861 DOI: 10.1186/s12906-020-2868-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/26/2020] [Indexed: 01/22/2023] Open
Abstract
Background Post-traumatic stress disorder (PTSD) is an extreme mood disorder that occurs after experiencing extreme stress, and patients with this disorder are known to accompany with symptoms of depression, anxiety, and memory impairments. Silibinin (SIL) is a natural polyphenolic flavonoid and is the main active ingredient of silymarin, which is primarily extracted from the milk thistle. Although some studies have assessed the properties of this flavonoid, the potential of SIL as a treatment for PTSD patients and its mechanisms of action have yet to be fully elucidated. Methods After exposure to a model of single prolonged stress (SPS), the open field test (OFT) and forced swimming test (FST), were used to investigate the effects of SIL on anxiety- and depression-like symptoms in male rats. The rats received of SIL (25, 50, and 100 mg/kg) for 14 days following exposure to SPS. Results Administration of SIL significantly improved anxiety-like behaviors in the OFT, depression-like behaviors in the FST, and freezing behavior in fear conditioning test. SIL also increased levels of serotonin in the hippocampus (Hipp) and amygdala, and enhanced expression of tryptophan hydroxylase-1 mRNA in the Hipp. The administration of SIL also inhibited SPS-induced decreases dopamine levels and increases norepinephrine levels in the Hipp. Conclusions Taken together, the present findings suggest that SIL can be a useful therapeutic ingredient for the treatment of trauma stress-associated symptoms, including PTSD-induced anxiety and depression caused by PTSD.
Collapse
Affiliation(s)
- Bombi Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea. .,Center for Converging Humanities, Kyung Hee University, 02447, Seoul, Republic of Korea.
| | - Gwang Muk Choi
- The Graduate School of Basic Science of Medicine, College of Medicine, Kyung Hee University, 02447, Seoul, Republic of Korea
| | - Bongjun Sur
- Department of Molecular medicine and TIDRC, School of Medicine, Ewha Womans University, 07985, Seoul, Republic of Korea
| |
Collapse
|
23
|
Klinke CM, Fiedler D, Lange MD, Andreatta M. Evidence for impaired extinction learning in humans after distal stress exposure. Neurobiol Learn Mem 2020; 167:107127. [DOI: 10.1016/j.nlm.2019.107127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 10/25/2019] [Accepted: 11/20/2019] [Indexed: 12/19/2022]
|
24
|
Lee B, Yeom M, Shim I, Lee H, Hahm DH. Umbelliferone modulates depression-like symptoms by altering monoamines in a rat post-traumatic stress disorder model. J Nat Med 2019; 74:377-386. [DOI: 10.1007/s11418-019-01373-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/13/2019] [Indexed: 12/23/2022]
|
25
|
The ethanolic extract of Aralia continentalis ameliorates cognitive deficits via modifications of BDNF expression and anti-inflammatory effects in a rat model of post-traumatic stress disorder. Altern Ther Health Med 2019; 19:11. [PMID: 30621666 PMCID: PMC6323859 DOI: 10.1186/s12906-018-2417-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/19/2018] [Indexed: 01/16/2023]
Abstract
Abstract Background Post-traumatic stress disorder (PTSD) is a disease associated with that the experience of traumatic stress. The traumatic experience results in the development of a prolonged stress response that causes impaired memory function and increased inflammation in the hippocampus. Currently, antidepressants are the only approved therapy for PTSD. However, the efficacy of antidepressants in the treatment of PTSD is marginal. The ethanol extract of Aralia continentalis (AC) is traditionally used in oriental medicine, and has been showed to possess pharmacological properties, including anti-inflammatory, anti-cancer, anti-atherosclerotic, and anti-diabetic effects. Nevertheless, the effects of AC on cognitive memory and its mechanism of action in PTSD remain unclear. Given the necessity of further treatment options for PTSD, we investigated the effect of AC on the spatial cognitive impairment caused by single prolonged stress (SPS) in a rat model of PTSD. Methods Male rats were treated with various intraperitoneal (i.p.) doses of AC for 21 consecutive days after inducing chronic stress with the SPS procedure. Results Cognitive impairment caused by SPS were inhibited after treatment with 100 mg/kg AC, as measured by the Morris water maze test and an object recognition test. Additionally, AC treatment significantly alleviated memory-related decreases in brain-derived neurotrophic factor (BDNF) mRNA and protein levels in the hippocampus. Our results suggest that AC significantly inhibited the cognitive deficits caused by SPS via increased expression of pro-inflammatory cytokines, including tumor necrosis factor-α and interleukin-6, in the rat brain. Conclusions AC reversed the behavioral impairments and inflammation triggered by SPS-derived traumatic stress and should be further evaluated as a potential therapeutic drug for PTSD.
Collapse
|
26
|
Lin CC, Chang HA, Tai YM, Chen TY, Wan FJ, Chang CC, Tung CS, Liu YP. Subchronic administration of aripiprazole improves fear extinction retrieval of Pavlovian conditioning paradigm in rats experiencing psychological trauma. Behav Brain Res 2019; 362:181-187. [PMID: 30610908 DOI: 10.1016/j.bbr.2018.12.051] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/12/2018] [Accepted: 12/29/2018] [Indexed: 11/15/2022]
Abstract
People may suffer from an intruded fear memory when the attributable traumatic events no longer exist. This is of highly clinical relevance to trauma-induced mental disorders, such as posttraumatic stress disorder (PTSD). Mechanism underlying PTSD largely lies in the abnormal process of fear extinction and a functional imbalance within amygdala associated fear circuit areas. Previous evidence suggested central dopamine plays a key role in the regulation of the fear memory process, yet it remains unclear whether the intervention of dopamine modulators would be beneficial for the fear extinction abnormalities. The present study examined the performance of Pavlovian conditioned fear and the changes of dopamine profiles following a subchronic 14-day regimen of aripiprazole (a partial agonist of dopamine D2 receptors to normalize the condition caused by dopamine imbalance) in rats previously experienced a psychologically traumatic procedure of single prolonged stress (SPS). The results demonstrated that aripiprazole at 5.0 mg/kg reversed the SPS-impaired fear memory dysfunction and the SPS-reduced dopamine efflux in the amygdala. The present study suggests a therapeutic potential of subchronic treatment with aripiprazole in managing patients suffered from fear extinction problem.
Collapse
Affiliation(s)
- Chen-Cheng Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, 11490, Taiwan; Department of Psychiatry, Cheng Hsin General Hospital, Taipei, 11220, Taiwan
| | - Hsin-An Chang
- Department of Psychiatry, Tri-Service General Hospital, Taipei, 11490, Taiwan
| | - Yueh-Ming Tai
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Tsung-Yen Chen
- Department of Psychiatry, Cheng Hsin General Hospital, Taipei, 11220, Taiwan
| | - Fang-Jung Wan
- Department of Psychiatry, Tri-Service General Hospital, Taipei, 11490, Taiwan
| | - Chuan-Chia Chang
- Department of Psychiatry, Tri-Service General Hospital, Taipei, 11490, Taiwan
| | - Che-Se Tung
- Division of Medical Research and Education, Cheng Hsin General Hospital, Taipei, 11220, Taiwan
| | - Yia-Ping Liu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, 11490, Taiwan; Department of Psychiatry, Cheng Hsin General Hospital, Taipei, 11220, Taiwan; Department of Psychiatry, Tri-Service General Hospital, Taipei, 11490, Taiwan; Laboratory of Cognitive Neuroscience, Department of Physiology, Department of Physiology, National Defense Medical Center, Taipei, 11490, Taiwan.
| |
Collapse
|
27
|
Lucas EK, Wu WC, Roman-Ortiz C, Clem RL. Prazosin during fear conditioning facilitates subsequent extinction in male C57Bl/6N mice. Psychopharmacology (Berl) 2019; 236:273-279. [PMID: 30112577 PMCID: PMC6374171 DOI: 10.1007/s00213-018-5001-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 08/08/2018] [Indexed: 11/25/2022]
Abstract
RATIONALE Recovery from a traumatic experience requires extinction of cue-based fear responses, a process that is impaired in post-traumatic stress disorder. While studies suggest a link between fear behavioral flexibility and noradrenaline signaling, the role of specific receptors and brain regions in these effects is unclear. OBJECTIVES Here, we examine the role of prazosin, an α1-adrenergic receptor (α1-AR) antagonist, in auditory fear conditioning and extinction. METHODS C57Bl/6N mice were subjected to auditory fear conditioning and extinction in combination with systemic (0.1-2 mg/kg) or local microinjections (3 or 6 mM) of the α1-AR antagonist prazosin into the prelimbic division of medial prefrontal cortex or basolateral amygdala. Conditioned fear and anxiety-like behaviors were compared with vehicle-injected control animals. RESULTS Mice that received systemic prazosin prior to fear conditioning exhibited similar initial levels of cue-elicited freezing compared to vehicle controls on the following day. However, at all doses tested, fear that was acquired during prazosin treatment was more readily extinguished, whereas anxiety-like behavior on the day of extinction was unaffected. A similar pattern of results was observed when prazosin was microinjected into the basolateral amygdala but not the prelimbic cortex. In contrast to pre-conditioning injections, prazosin administration prior to extinction had no effect on freezing. CONCLUSIONS Our results indicate that α1-AR activity during aversive conditioning is dispensable for memory acquisition but renders conditioned fear more impervious to extinction. This suggests that behavioral flexibility is constrained by noradrenaline at the time of initial learning via activation of a specific AR isoform.
Collapse
Affiliation(s)
- Elizabeth K Lucas
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1065, New York, NY, 10029, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27606, USA
| | - Wan-Chen Wu
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1065, New York, NY, 10029, USA
| | - Ciorana Roman-Ortiz
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1065, New York, NY, 10029, USA
| | - Roger L Clem
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1065, New York, NY, 10029, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
28
|
Effects of Oxytocin on Fear Memory and Neuroinflammation in a Rodent Model of Posttraumatic Stress Disorder. Int J Mol Sci 2018; 19:ijms19123848. [PMID: 30513893 PMCID: PMC6321616 DOI: 10.3390/ijms19123848] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/26/2018] [Accepted: 11/29/2018] [Indexed: 12/27/2022] Open
Abstract
Posttraumatic stress disorder (PTSD) is a trauma-induced mental disorder characterized by fear extinction abnormalities, which involve biological dysfunctions among fear circuit areas in the brain. Oxytocin (OXT) is a neuropeptide that regulates sexual reproduction and social interaction and has recently earned specific attention due to its role in adjusting neurobiological and behavioral correlates of PTSD; however, the mechanism by which this is achieved remains unclear. The present study aimed to examine whether the effects of OXT on traumatic stress-induced abnormalities of fear extinction (specifically induced by single prolonged stress (SPS), an animal model of PTSD) are associated with pro-inflammatory cytokines. Seven days after SPS, rats received intranasal OXT 40 min before a cue-dependent Pavlovian fear conditioning-extinction test in which rats' freezing degree was used to reflect the outcome of fear extinction. We also measured mRNA expression of IL-1β, IFN-γ, and TNF-α in the medial prefrontal cortex (mPFC), hippocampus, and amygdala at the end of the study, together with plasma oxytocin, corticosterone, IL-1β, IFN-γ, and TNF-α, to reflect the central and peripheral changes of stress-related hormones and cytokines after SPS. Our results suggested that intranasal OXT effectively amends the SPS-impaired behavior of fear extinction retrieval. Moreover, it neurochemically reverses the SPS increase in pro-inflammatory cytokines; thus, IL-1β and IFN-γ can be further blocked by the OXT antagonist atosiban (ASB) in the hippocampus. Peripheral profiles revealed a similar response pattern to SPS of OXT and corticosterone (CORT), and the SPS-induced increase in plasma levels of IL-1β and TNF-α could be reduced by OXT. The present study suggests potential therapeutic effects of OXT in both behavioral and neuroinflammatory profiles of PTSD.
Collapse
|
29
|
Chikahisa S, Chida D, Shiuchi T, Harada S, Shimizu N, Otsuka A, Tanioka D, Séi H. Enhancement of fear learning in PPARα knockout mice. Behav Brain Res 2018; 359:664-670. [PMID: 30278189 DOI: 10.1016/j.bbr.2018.09.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/28/2018] [Accepted: 09/28/2018] [Indexed: 11/18/2022]
Abstract
Peroxisome proliferator-activated receptor alpha (PPARα) is a member of the nuclear receptor superfamily and regulates fatty acid oxidation. Although PPARα is expressed not only in the peripheral tissues but also in the brain, its role in higher brain function is unclear. In this study, we investigated the role of PPARα in the control of behavior, including memory/learning and mood change, using PPARα knockout (KO) mice. A significant difference between wild-type (WT) and KO mice was seen in the passive avoidance test, demonstrating that KO mice showed enhanced fear leaning. In the amygdala of KO mice, the levels of dopamine and its metabolites were increased, and the mRNA expression of dopamine degrading enzyme was decreased. When dopamine D1 receptor antagonist was administered, the enhanced fear learning observed in KO mice was attenuated. These results suggest that PPARα is involved in the regulation of emotional memory via the dopamine pathway in the amygdala.
Collapse
Affiliation(s)
- Sachiko Chikahisa
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8503, Japan.
| | - Daiki Chida
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8503, Japan; Student Lab, Tokushima University Faculty of Medicine, Tokushima, 770-8503, Japan
| | - Tetsuya Shiuchi
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8503, Japan
| | - Saki Harada
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8503, Japan; Student Lab, Tokushima University Faculty of Medicine, Tokushima, 770-8503, Japan
| | - Noriyuki Shimizu
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8503, Japan
| | - Airi Otsuka
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8503, Japan
| | - Daisuke Tanioka
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8503, Japan
| | - Hiroyoshi Séi
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8503, Japan
| |
Collapse
|
30
|
Lee B, Shim I, Lee H, Hahm DH. Effects of Epigallocatechin Gallate on Behavioral and Cognitive Impairments, Hypothalamic-Pituitary-Adrenal Axis Dysfunction, and Alternations in Hippocampal BDNF Expression Under Single Prolonged Stress. J Med Food 2018; 21:979-989. [PMID: 30273101 DOI: 10.1089/jmf.2017.4161] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a traumatic stress-related psychiatric disorder stimulated by experience. Green tea has potent antioxidative properties, due, in part, to the catechin (-) epigallocatechin-3-gallate (EGCG). EGCG is an important polyphenol with advantageous effects on anxiety and depression. Nevertheless, the mechanism about the inhibition of PTSD-like symptoms of EGCG is still unidentified. We examined whether EGCG improved learning and memory deficit stimulated in rats after single prolonged stress (SPS). Rats were administrated intraperitoneally (i.p.) with EGCG for 14 successive days after the SPS process. The SPS procedure stimulated cognitive deficit in the Morris water maze test and the object recognition task, and this impairment was improved by EGCG (25 mg/kg, i.p.). Daily EGCG administration significantly decreased the freezing response to contextual fear conditioning. The administration of EGCG also significantly moderated memory-related decreases in the alternation of cAMP-response element-binding protein and brain-derived neurotrophic factor in the hippocampus. Our results suggest that EGCG alleviated SPS-stimulated learning and memory deficit by inhibiting the increase of neuroinflammation in the rat brain. In addition, EGCG reversed the alternation of allopregnanolone and progesterone in the brain, and diminished simultaneously the hypothalamic-pituitary-adrenal axis dysfunction. Thus, EGCG reversed learning and memory-related behavioral dysfunction and molecular alternation accelerated by traumatic stress and may be a useful therapeutic material for PTSD.
Collapse
Affiliation(s)
- Bombi Lee
- 1 Center for Converging Humanities, Kyung Hee University , Seoul, Republic of Korea.,2 Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University , Seoul, Republic of Korea
| | - Insop Shim
- 3 Department of Physiology, College of Medicine, Kyung Hee University , Seoul, Republic of Korea
| | - Hyejung Lee
- 2 Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University , Seoul, Republic of Korea
| | - Dae-Hyun Hahm
- 3 Department of Physiology, College of Medicine, Kyung Hee University , Seoul, Republic of Korea
| |
Collapse
|
31
|
Qiu ZK, He JL, Liu X, Zeng J, Xiao W, Fan QH, Chai XM, Ye WH, Chen JS. Anxiolytic-like effects of paeoniflorin in an animal model of post traumatic stress disorder. Metab Brain Dis 2018; 33:1175-1185. [PMID: 29633071 DOI: 10.1007/s11011-018-0216-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 03/09/2018] [Indexed: 11/11/2022]
Abstract
Post-traumatic stress disorder (PTSD) is the serious psychiatric disorder. Paeoniflorin (PF) produces the antidepressant-like properties. However, few studies are concerned about its anti-PTSD-like effects and mechanisms. To investigate these, the single prolonged stress (SPS) model was utilized. PTSD-like behavioral deficits in rats after exposure to SPS were improved by PF (10 and 20 mg/kg, i.p.), evidenced by blocking increased freezing time in contextual fear paradigm (CFP) and increased time and entries in open arms in elevated plus maze (EPM) test without affecting the locomotor activity in open field (OF) test. We also found that increased levels of corticosterone (Cort), corticotropin releasing hormone (CRH) and adrenocorticotropic hormone (ACTH) after exposure to SPS were reversed by PF (10 and 20 mg/kg, i.p.) in serum, respectively. Moreover, the decreased levels of serotonin (5-HT) and 5-Hydroxyindoleacetic acid (5-HIAA) in prefrontal cortex and hippocampus were reversed by PF (10 and 20 mg/kg, i.p.), respectively. In summary, the anti-PTSD-like activities of PF were associated with the modulation of HPA axis and 5-HT system activation.
Collapse
Affiliation(s)
- Zhi-Kun Qiu
- Pharmaceutical Department of the First Affiliated Hospital of Guangdong Pharmaceutical University, Clinical Pharmacy Department of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China
- Guangdong Provincial Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, People's Republic of China
| | - Jia-Li He
- Department of Endocrinology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, People's Republic of China
| | - Xu Liu
- Pharmacy Department, General Hospital of Chinese People's Armed Police Forces, Beijing, 100039, People's Republic of China
| | - Jia Zeng
- Pharmaceutical Department of the First Affiliated Hospital of Guangdong Pharmaceutical University, Clinical Pharmacy Department of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China
| | - Wei Xiao
- Guangdong Provincial Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, People's Republic of China
| | - Qing-Hong Fan
- Guangdong Provincial Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, People's Republic of China
| | - Xiao-Meng Chai
- Guangdong Provincial Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, People's Republic of China
| | - Wei-Hai Ye
- The Affiliated Chencun Hospital of Shunde Hospital, Southern Medical University, Foshan, 528313, People's Republic of China.
| | - Ji-Sheng Chen
- Pharmaceutical Department of the First Affiliated Hospital of Guangdong Pharmaceutical University, Clinical Pharmacy Department of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
32
|
Lin CC, Huang KL, Tung CS, Liu YP. Hyperbaric oxygen therapy restored traumatic stress-induced dysregulation of fear memory and related neurochemical abnormalities. Behav Brain Res 2018; 359:861-870. [PMID: 30056129 DOI: 10.1016/j.bbr.2018.07.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 07/13/2018] [Accepted: 07/19/2018] [Indexed: 01/12/2023]
Abstract
Individuals with posttraumatic stress disorder (PTSD) are characterized by fear memory problems and hypocortisolemia of which traumatic stress-induced monoaminergic disruption over infralimbic (IL) cortex is considered the key mechanism. Hyperbaric oxygen therapy (HBOT) has recently proven its utility in treating several mental disorders but remains unexplored for PTSD. The present study aimed to examine the effects of 5-day HBO paradigm on traumatic stress (single prolonged stress, SPS, an animal model of PTSD)-induced dysregulation of fear memory/anxiety profiles and related abnormalities in IL monoamines and plasma corticosterone. Rats were randomly assigned to four groups (CON-sham, CON-HBOT, SPS-sham, and SPS-HBOT) and received Pavlovian fear conditioning test or elevated-T maze (ETM). The extracellular and tissue levels of monoamines over the IL cortex and the activity of the hypothalamus-pituitary-adrenal axis (i.e., the plasma corticosterone level and expression of the glucocorticoid receptor (GR) in the IL, hippocampus, amygdala, and hypothalamus) were measured. The results demonstrated that HBOT restored behaviorally the SPS-impaired fear extinction retrieval ability and SPS-induced conditioned anxiety, and neurochemically the SPS-reduced IL monoamines efflux level, and the corticosterone profiles. The present study shows some positive effects of HBOT in both behavioral and neurochemical profiles of PTSD outcomes.
Collapse
Affiliation(s)
- Chen-Cheng Lin
- Department of Psychiatry, Cheng Hsin General Hospital, Taipei, Taiwan; Laboratory of Cognitive Neuroscience, Department of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Hyperbaric Oxygen Therapy Center, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Che-Se Tung
- Division of Medical Research and Education, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Yia-Ping Liu
- Department of Psychiatry, Cheng Hsin General Hospital, Taipei, Taiwan; Laboratory of Cognitive Neuroscience, Department of Physiology, National Defense Medical Center, Taipei, Taiwan; Department of Psychiatry, Tri-Service General Hospital, Taipei, Taiwan.
| |
Collapse
|
33
|
Lee B, Shim I, Lee H, Hahm DH. Melatonin ameliorates cognitive memory by regulation of cAMP-response element-binding protein expression and the anti-inflammatory response in a rat model of post-traumatic stress disorder. BMC Neurosci 2018; 19:38. [PMID: 29973144 PMCID: PMC6032787 DOI: 10.1186/s12868-018-0439-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 06/28/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Post-traumatic stress disorder (PTSD) is an important psychological disease that can develop following the physical experience or witnessing of traumatic events. The psychopathological response to traumatic stressors increases inflammation in the hippocampus and induces memory deficits. Melatonin (MTG) plays critical roles in circadian rhythm disorders, Alzheimer's disease, and other neurological disorders. However, the cognitive efficiency of MTG and its mechanisms of action in the treatment of PTSD remain unclear. Thus, the present study investigated the effects of MTG on spatial cognitive impairments stimulated by single prolonged stress (SPS) in rats, an animal model of PTSD. Male rats received intraperitoneal (i.p.) administration of various doses of MTG for 21 consecutive days after the SPS procedure. RESULTS SPS-stimulated cognitive impairments in the object recognition task and Morris water maze were reversed by MTG treatment (25 mg/kg, i.p). Additionally, MTG significantly increased cognitive memory-related decreases in cAMP-response element-binding (CREB) protein and mRNA levels in the hippocampus. Our results also demonstrate that MTG significantly inhibited SPS-stimulated cognitive memory impairments by inhibiting the expression of proinflammatory cytokines, including tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) in the rat brain. CONCLUSION The present results indicate that MTG can be beneficial for SPS-stimulated memory impairments via changes in CREB expression and proinflammatory mediators. Thus, MTG may be a prophylactic strategy for the prevention or mitigation of the progression of some features of the PTSD pathology.
Collapse
Affiliation(s)
- Bombi Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447 Republic of Korea
- Center for Converging Humanities, Kyung Hee University, Seoul, 02447 Republic of Korea
| | - Insop Shim
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447 Republic of Korea
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, 02447 Republic of Korea
| | - Hyejung Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447 Republic of Korea
| | - Dae-Hyun Hahm
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447 Republic of Korea
- Center for Converging Humanities, Kyung Hee University, Seoul, 02447 Republic of Korea
| |
Collapse
|
34
|
Giustino TF, Maren S. Noradrenergic Modulation of Fear Conditioning and Extinction. Front Behav Neurosci 2018; 12:43. [PMID: 29593511 PMCID: PMC5859179 DOI: 10.3389/fnbeh.2018.00043] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/26/2018] [Indexed: 12/12/2022] Open
Abstract
The locus coeruleus norepinephrine (LC-NE) system plays a broad role in learning and memory. Here we begin with an overview of the LC-NE system. We then consider how both direct and indirect manipulations of the LC-NE system affect cued and contextual aversive learning and memory. We propose that NE dynamically modulates Pavlovian conditioning and extinction, either promoting or impairing learning aversive processes under different levels of behavioral arousal. We suggest that under high levels of stress (e.g., during/soon after fear conditioning) the locus coeruleus (LC) promotes cued fear learning by enhancing amygdala function while simultaneously blunting prefrontal function. Under low levels of arousal, the LC promotes PFC function to promote downstream inhibition of the amygdala and foster the extinction of cued fear. Thus, LC-NE action on the medial prefrontal cortex (mPFC) might be described by an inverted-U function such that it can either enhance or hinder learning depending on arousal states. In addition, LC-NE seems to be particularly important for the acquisition, consolidation and extinction of contextual fear memories. This may be due to dense adrenoceptor expression in the hippocampus (HPC) which encodes contextual information, and the ability of NE to regulate long-term potentiation (LTP). Moreover, recent work reveals that the diversity of LC-NE functions in aversive learning and memory are mediated by functionally heterogeneous populations of LC neurons that are defined by their projection targets. Hence, LC-NE function in learning and memory is determined by projection-specific neuromodulation that accompanies various states of behavioral arousal.
Collapse
Affiliation(s)
- Thomas F Giustino
- Department of Psychological and Brain Sciences, Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| | - Stephen Maren
- Department of Psychological and Brain Sciences, Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| |
Collapse
|
35
|
Lee B, Shim I, Lee H, Hahm DH. Berberine alleviates symptoms of anxiety by enhancing dopamine expression in rats with post-traumatic stress disorder. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018. [PMID: 29520171 PMCID: PMC5840077 DOI: 10.4196/kjpp.2018.22.2.183] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Post-traumatic stress disorder (PTSD) is a trauma-induced psychiatric disorder characterized by impaired fear extermination, hyperarousal, anxiety, depression, and amnesic symptoms that may involve the release of monoamines in the fear circuit. The present study measured several anxiety-related behavioral responses to examine the effects of berberine (BER) on symptoms of anxiety in rats after single prolonged stress (SPS) exposure, and to determine if BER reversed the dopamine (DA) dysfunction. Rats received BER (10, 20, or 30 mg/kg, intraperitoneally, once daily) for 14 days after SPS exposure. BER administration significantly increased the time spent in the open arms and reduced grooming behavior during the elevated plus maze test, and increased the time spent in the central zone and the number of central zone crossings in the open field test. BER restored neurochemical abnormalities and the SPS-induced decrease in DA tissue levels in the hippocampus and striatum. The increased DA concentration during BER treatment may partly be attributed to mRNA expression of tyrosine hydroxylase and the DA transporter in the hippocampus, while BER exerted no significant effects on vesicular monoamine transporter mRNA expression in the hippocampus of rats with PTSD. These results suggest that BER had anxiolytic-like effects on behavioral and biochemical measures associated with anxiety. These findings support a role for reduced anxiety altered DAergic transmission and reduced anxiety in rats with PTSD. Thus, BER may be a useful agent to treat or alleviate psychiatric disorders like those observed in patients with PTSD.
Collapse
Affiliation(s)
- Bombi Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea.,Center for Converging Humanities, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Insop Shim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Hyejung Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Dae-Hyun Hahm
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
36
|
Camp R, Stier CT, Serova LI, McCloskey J, Edwards JG, Reyes-Zaragoza M, Sabban EL. Cardiovascular responses to intranasal neuropeptide Y in single prolonged stress rodent model of post-traumatic stress disorder. Neuropeptides 2018; 67:87-94. [PMID: 29169656 DOI: 10.1016/j.npep.2017.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/26/2017] [Accepted: 11/08/2017] [Indexed: 01/25/2023]
Abstract
Delivery of neuropeptide Y (NPY) to the brain by intranasal administration shows promise as non-invasive means for preventing or treating PTSD symptoms. Here, radiotelemetry and echocardiography were used to determine effects of intranasal NPY on cardiovascular functions in absence and presence of stress. Male adult Sprague Dawley rats were implanted with radiotelemetric probes, and subjected to single prolonged stress (SPS), followed by intranasal vehicle (V) or NPY (150μg) under conditions shown to prevent development of many of the behavioral neuroendocrine and biochemical impairments. In both groups, mean arterial pressure (MAP) rose rapidly peaking at about 125mmHg, remaining near maximal levels for 1h. SPS also elicited robust rise in heart rate (HR) which was mitigated by intranasal NPY, and significantly lower than V-treated rats 12-50min after exposure to SPS stressors. In the first hr. after SPS, locomotor activity was elevated but only in the V-treated group. By 3h, MAP returned to pre-stress levels in both groups with no further change when monitored for 6days. HR remained elevated during the 6h remaining light phase after SPS. Subsequently HR was at pre-SPS levels during the remaining days. However dark phase HR was low following SPS, gradually recovered over 6days and was associated with reduced activity. When administered in the absence of further stress, intranasal NPY or V elicited similar much smaller, short-lived rises in MAP and HR. Echocardiography revealed no change in HR, stroke volume (SV) or cardiac output (Q) with intranasal NPY in the absence of stress. SPS led to reduced SV and Q but was not affected by NPY. Overall the results demonstrate no major cardiovascular effects of intranasal NPY and indicate possible benefit from transient amelioration of HR response in line with its translational potential to combat PTSD and comorbid impairments.
Collapse
Affiliation(s)
- Robert Camp
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York 10595, USA
| | - Charles T Stier
- Department of Pharmacology, New York Medical College, Valhalla, New York 10595, USA
| | - Lidia I Serova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York 10595, USA
| | - Jaclyn McCloskey
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York 10595, USA
| | - John G Edwards
- Department of Physiology, New York Medical College, Valhalla, New York 10595, USA
| | - Miguel Reyes-Zaragoza
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York 10595, USA
| | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York 10595, USA.
| |
Collapse
|
37
|
Lee B, Shim I, Lee H, Hahm DH. The polymethoxylated flavone, Tangeretin improves cognitive memory in rats experiencing a single episode of prolonged post-traumatic stress. Anim Cells Syst (Seoul) 2018. [DOI: 10.1080/19768354.2018.1426627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Bombi Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Center for Converging Humanities, Kyung Hee University, Seoul, Republic of Korea
| | - Insop Shim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyejung Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Dae-Hyun Hahm
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
38
|
β-Adrenoceptor Blockade in the Basolateral Amygdala, But Not the Medial Prefrontal Cortex, Rescues the Immediate Extinction Deficit. Neuropsychopharmacology 2017; 42:2537-2544. [PMID: 28462941 PMCID: PMC5686500 DOI: 10.1038/npp.2017.89] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/21/2017] [Accepted: 04/24/2017] [Indexed: 01/09/2023]
Abstract
Early psychological interventions, such as exposure therapy, rely on extinction learning to reduce the development of stress- and trauma-related disorders. However, recent research suggests that extinction often fails to reduce fear when administered soon after trauma. This immediate extinction deficit (IED) may be due to stress-induced dysregulation of neural circuits involved in extinction learning. We have shown that systemic β-adrenoceptor blockade with propranolol rescues the IED, but impairs delayed extinction. Here we sought to determine the neural locus of these effects. Rats underwent auditory fear conditioning and then received either immediate (30 min) or delayed (24 h) extinction training. We used bilateral intracranial infusions of propranolol into either the infralimbic division of the medial prefrontal cortex (mPFC) or the basolateral amygdala (BLA) to examine the effects of β-adrenoceptor blockade on immediate and delayed extinction learning. Interestingly, intra-BLA, but not intra-mPFC, propranolol rescued the IED; animals receiving intra-BLA propranolol prior to immediate extinction showed less spontaneous recovery of fear during extinction retrieval. Importantly, this was not due to impaired consolidation of the conditioning memory. In contrast, neither intra-BLA nor intra-mPFC propranolol affected delayed extinction learning. Overall, these data contribute to a growing literature suggesting dissociable roles for key nodes in the fear extinction circuit depending on the timing of extinction relative to conditioning. These data also suggest that heightened noradrenergic activity in the BLA underlies stress-induced extinction deficits. Propranolol may be a useful adjunct to behavioral therapeutic interventions in recently traumatized individuals who are at risk for developing trauma-related disorders.
Collapse
|
39
|
Souza RR, Noble LJ, McIntyre CK. Using the Single Prolonged Stress Model to Examine the Pathophysiology of PTSD. Front Pharmacol 2017; 8:615. [PMID: 28955225 PMCID: PMC5600994 DOI: 10.3389/fphar.2017.00615] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 08/23/2017] [Indexed: 01/08/2023] Open
Abstract
The endurance of memories of emotionally arousing events serves the adaptive role of minimizing future exposure to danger and reinforcing rewarding behaviors. However, following a traumatic event, a subset of individuals suffers from persistent pathological symptoms such as those seen in posttraumatic stress disorder (PTSD). Despite the availability of pharmacological treatments and evidence-based cognitive behavioral therapy, a considerable number of PTSD patients do not respond to the treatment, or show partial remission and relapse of the symptoms. In controlled laboratory studies, PTSD patients show deficient ability to extinguish conditioned fear. Failure to extinguish learned fear could be responsible for the persistence of PTSD symptoms such as elevated anxiety, arousal, and avoidance. It may also explain the high non-response and dropout rates seen during treatment. Animal models are useful for understanding the pathophysiology of the disorder and the development of new treatments. This review examines studies in a rodent model of PTSD with the goal of identifying behavioral and physiological factors that predispose individuals to PTSD symptoms. Single prolonged stress (SPS) is a frequently used rat model of PTSD that involves exposure to several successive stressors. SPS rats show PTSD-like symptoms, including impaired extinction of conditioned fear. Since its development by the Liberzon lab in 1997, the SPS model has been referred to by more than 200 published papers. Here we consider the findings of these studies and unresolved questions that may be investigated using the model.
Collapse
Affiliation(s)
- Rimenez R Souza
- Texas Biomedical Device Center, School of Behavioral and Brain Sciences, University of Texas at Dallas, RichardsonTX, United States
| | - Lindsey J Noble
- Texas Biomedical Device Center, School of Behavioral and Brain Sciences, University of Texas at Dallas, RichardsonTX, United States.,Cognition and Neuroscience Program, School of Behavioral and Brain Sciences, University of Texas at Dallas, RichardsonTX, United States
| | - Christa K McIntyre
- Cognition and Neuroscience Program, School of Behavioral and Brain Sciences, University of Texas at Dallas, RichardsonTX, United States
| |
Collapse
|
40
|
Lee B, Shim I, Lee H, Hahm DH. Effect of oleuropein on cognitive deficits and changes in hippocampal brain-derived neurotrophic factor and cytokine expression in a rat model of post-traumatic stress disorder. J Nat Med 2017; 72:44-56. [PMID: 28884427 DOI: 10.1007/s11418-017-1103-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/05/2017] [Indexed: 11/24/2022]
Abstract
Post-traumatic stress disorder (PTSD) is a condition that develops after an individual has experienced a major trauma. This psychopathological response to traumatic stressors induces learning and memory deficits in rats. Oleuropein (OLE), a major compound in olive leaves, has been reported to possess several pharmacological properties, including anti-cancer, anti-diabetic, anti-atherosclerotic and neuroprotective activities. However, the cognitive effects of OLE and its mechanism of action have remained unclear in PTSD. In this study, we examined whether OLE improved spatial cognitive impairment induced in rats following single prolonged stress (SPS), an animal model of PTSD. Male rats were treated intraperitoneally (i.p.) with vehicle or various doses of OLE for 14 consecutive days after the SPS procedure. The SPS procedure resulted in cognitive impairment in the object recognition task and the Morris water maze test, which was reversed by OLE (100 mg/kg, i.p). Additionally, as assessed by immunohistochemistry and reverse transcription-polymerase chain reaction analysis, the administration of OLE significantly alleviated memory-associated decreases in the levels of brain-derived neurotrophic factor and cAMP response element-binding protein and mRNA in the hippocampus. Together, these findings suggest that OLE attenuated SPS-induced cognitive impairment significantly by inhibiting the expression of pro-inflammatory mediators in the rat brain. Thus, OLE reversed several behavioral impairments triggered by the traumatic stress of SPS and might be a potential useful therapeutic intervention for PTSD.
Collapse
Affiliation(s)
- Bombi Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | - Insop Shim
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyejung Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Dae-Hyun Hahm
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
41
|
Malikowska N, Fijałkowski Ł, Nowaczyk A, Popik P, Sałat K. Antidepressant-like activity of venlafaxine and clonidine in mice exposed to single prolonged stress - A model of post-traumatic stress disorder. Pharmacodynamic and molecular docking studies. Brain Res 2017; 1673:1-10. [PMID: 28797691 DOI: 10.1016/j.brainres.2017.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 07/14/2017] [Accepted: 08/02/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Post-traumatic stress disorder (PTSD) is a growing issue worldwide characterized by stress and anxiety in response to re-experiencing traumatic events which strongly impair patient's quality of life and social functions. Available antidepressant and anxiolytic drugs are not efficacious in the majority of treated individuals. This necessitates a significant medical demand to develop novel therapeutic strategies for PTSD. EXPERIMENTAL APPROACH Animal model of PTSD was induced using a mouse single prolonged stress protocol (mSPS). To assess the activity of venlafaxine and clonidine, the forced swim test (FST) was used repeatedly 24h, 3days, 8days, 15days and 25days after mSPS. To get insight into a possible mechanism of anti-PTSD action, molecular docking procedure was utilized for the most active drug. This in silico part comprised molecular docking of enantiomers of venlafaxine to human transporters for serotonin (hSERT), norepinephrine (hNET) and dopamine (hDAT). KEY RESULTS In mSPS-subjected mice FST revealed the effectiveness of venlafaxine, however in non SPS-subjected mice both venlafaxine and clonidine were active. Molecular docking studies indicated that the affinity of venlafaxine to monoamine transporters is growing in the following rank order: hDAT<hNET<hSERT. Both venlafaxine enantiomers present different selectivity and binding mode. CONCLUSION AND IMPLICATIONS Venlafaxine but not clonidine was effective in an animal model of PTSD. Its mechanism of action, i.e., SERT, NET and DAT inhibition indicates potential drug targets for PTSD treatment. We expect that these results will contribute to a broader application of VLX in PTSD patients.
Collapse
Affiliation(s)
- Natalia Malikowska
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland
| | - Łukasz Fijałkowski
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 2 dr. A. Jurasza St., 85-094 Bydgoszcz, Poland
| | - Alicja Nowaczyk
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 2 dr. A. Jurasza St., 85-094 Bydgoszcz, Poland
| | - Piotr Popik
- Department of Behavioral Neuroscience and Drug Development, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St., 31-343 Krakow, Poland
| | - Kinga Sałat
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland.
| |
Collapse
|
42
|
Park SH, Kim YJ, Park JC, Han JS, Choi SY. Intranasal Oxytocin following Uncontrollable Stress Blocks Impairments in Hippocampal Plasticity and Recognition Memory in Stressed Rats. Int J Neuropsychopharmacol 2017; 20:861-866. [PMID: 28977526 PMCID: PMC5632307 DOI: 10.1093/ijnp/pyx061] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Nasal pretreatment with the neuropeptide oxytocin has been reported to prevent stress-induced impairments in hippocampal synaptic plasticity and spatial memory in rats. However, no study has asked if oxytocin application following a stress experience is effective in rescuing stress-induced impairments. METHODS Synaptic plasticity was measured in hippocampal Schaffer collateral-CA1 synapses of rats subjected to uncontrollable stress; their cognitive function was examined using an object recognition task. RESULTS Impaired induction of long-lasting, long-term potentiation by uncontrollable stress was rescued, as demonstrated both in rats and hippocampal slices. Intranasal oxytocin after experiencing uncontrollable stress blocked cognitive impairments in stressed rats and in stressed hippocampal slices treated with a perfused bath solution containing oxytocin. CONCLUSIONS These results indicated that posttreatment with oxytocin after experiencing a stressful event can keep synaptic plasticity and cognition function intact, indicating the therapeutic potential of oxytocin for stress-related disorders, including posttraumatic stress disorder.
Collapse
Affiliation(s)
- Seong-Hae Park
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (Dr Park, Ms Kim, and Dr Choi); Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea (Mr Park and Dr Han)
| | - Yoon-Jung Kim
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (Dr Park, Ms Kim, and Dr Choi); Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea (Mr Park and Dr Han)
| | - Jung-Cheol Park
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (Dr Park, Ms Kim, and Dr Choi); Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea (Mr Park and Dr Han)
| | - Jung-Soo Han
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (Dr Park, Ms Kim, and Dr Choi); Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea (Mr Park and Dr Han)
| | - Se-Young Choi
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (Dr Park, Ms Kim, and Dr Choi); Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea (Mr Park and Dr Han).,Correspondence: Se-Young Choi, PhD, Department of Physiology, Seoul National University School of Dentistry, Seoul 110–749, Republic of Korea ()
| |
Collapse
|