1
|
Liu L, Li R, Wu L, Guan Y, Miao M, Wang Y, Li C, Wu C, Lu G, Hu X, Sun L. (2R,6R)-hydroxynorketamine alleviates PTSD-like endophenotypes by regulating the PI3K/AKT signaling pathway in rats. Pharmacol Biochem Behav 2024; 245:173891. [PMID: 39369910 DOI: 10.1016/j.pbb.2024.173891] [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: 07/30/2024] [Revised: 09/08/2024] [Accepted: 10/01/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Patients diagnosed with post-traumatic stress disorder (PTSD) mainly exhibit enduring adverse emotions, heightening susceptibility to suicidal thoughts and behaviors. Notably, metabolites of ketamine, particularly (2R,6R)-hydroxyketamine (HNK), have demonstrated favorable antidepressant properties. However, the precise mechanism through which HNK exerts its therapeutic effects on negative emotional symptoms in PTSD patients should be fully elucidated. METHODS In this investigation, a model involving a single prolonged stress and plantar shock (SPS&S) was utilized, followed by the administration of (2R, 6R)-HNK into the lateral ventricle subsequent to the recovery phase. The evaluation of PTSD-related behaviors was conducted through the open field test (OFT), elevated plus maze test (EMPT), and forced swim test (FST). The expression of phosphatidylinositol 3-kinase (PI3K)/phosphokinase B (AKT) signaling pathway in rat brain regions was analyzed using molecular biology experiments. RESULTS SPS&S rats displayed adverse emotional behaviors characterized by depression and anxiety. Treatment with (2R, 6R)-HNK enhanced exploratory behavior and reversed negative emotional behaviors. This intervention mitigated disruptions in the expression levels of PI3K/AKT signaling pathway-associated proteins in the HIP and PFC, without influencing PI3K/AKT signaling in the AMY of SPS&S rats. CONCLUSION Traumatic stress can trigger negative emotional reactions in rats, potentially involving the PI3K/AKT signaling pathway in the HIP, PFC, and AMY. The (2R, 6R)-HNK compounds have demonstrated the potential to mitigate adverse emotions in rats subjected to the SPS&S paradigm. This effect may be attributed to the modulation of the PI3K/AKT signaling pathway in the HIP, and PFC, with a particularly notable impact observed in the HIP region.
Collapse
Affiliation(s)
- Lifen Liu
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Rui Li
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Lanxia Wu
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Yubo Guan
- School of Clinical Medicine, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Miao Miao
- School of Clinical Medicine, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Yuxuan Wang
- School of Clinical Medicine, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Changjiang Li
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Chunyan Wu
- Department of Neurology, Affiliated Hospital of Shandong Second Medical University, Weifang, PR China
| | - Guohua Lu
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Xinyu Hu
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China; CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, PR China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, PR China.
| | - Lin Sun
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China; Department of Neurosurgery, Shanting District People's Hospital, Beijing Road, New Town, Zaozhuang, Shandong 277200, PR China; Management Committee of Shanting Economic Development Zone, No.37, Fuqian Road, Zaozhuang, Shandong 277200, PR China.
| |
Collapse
|
2
|
Denny RR, Connelly KL, Ghilotti MG, Meissler JJ, Yu D, Eisenstein TK, Unterwald EM. Artificial Intelligence Identified Resilient and Vulnerable Female Rats After Traumatic Stress and Ethanol Exposure: Investigation of Neuropeptide Y Pathway Regulation. Front Neurosci 2021; 15:772946. [PMID: 34975380 PMCID: PMC8716605 DOI: 10.3389/fnins.2021.772946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is initiated by traumatic-stress exposure and manifests into a collection of symptoms including increased anxiety, sleep disturbances, enhanced response to triggers, and increased sympathetic nervous system arousal. PTSD is highly co-occurring with alcohol use disorder. Only some individuals experiencing traumatic stress develop PTSD and a subset of individuals with PTSD develop co-occurring alcohol use disorder. To investigate the basis of these individual responses to traumatic stress, single prolonged stress (SPS) a rodent model of traumatic stress was applied to young adult female rats. Individual responses to SPS were characterized by measuring anxiety-like behaviors with open field and elevated plus maze tests. Rats were then allowed to drink ethanol under an intermittent two bottle choice procedure for 8 weeks, and ethanol consumption was measured. An artificial intelligence algorithm was built to predict resilient and vulnerable individuals based on data from anxiety testing and ethanol consumption. This model was implemented in a second cohort of rats that underwent SPS without ethanol drinking to identify resilient and vulnerable individuals for further study. Analysis of neuropeptide Y (NPY) levels and expression of its receptors Y1R and Y2R mRNA in the central nucleus of the amygdala (CeA), basolateral amygdala (BLA), and bed nucleus stria terminalis (BNST) were performed. Results demonstrate that resilient rats had higher expression of Y2R mRNA in the CeA compared with vulnerable and control rats and had higher levels of NPY protein in the BNST compared to controls. The results of the study show that an artificial intelligence algorithm can identify individual differences in response to traumatic stress which can be used to predict subsequent ethanol drinking, and the NPY pathway is differentially altered following traumatic stress exposure in resilient and vulnerable populations. Understanding neurochemical alterations following traumatic-stress exposure is critical in developing prevention strategies for the vulnerable phenotype and will help further development of novel therapeutic approaches for individuals suffering from PTSD and at risk for alcohol use disorder.
Collapse
Affiliation(s)
- Ray R. Denny
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Krista L. Connelly
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Marco G. Ghilotti
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Joseph J. Meissler
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Daohai Yu
- Center for Biostatistics and Epidemiology, Department of Biomedical Education and Data Science, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Toby K. Eisenstein
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States,Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Ellen M. Unterwald
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States,Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States,*Correspondence: Ellen M. Unterwald,
| |
Collapse
|
3
|
Pieper J, Chang DG, Mahasin SZ, Swan AR, Quinto AA, Nichols S, Diwakar M, Huang C, Swan J, Lee R, Baker DG, Huang M. Brain Amygdala Volume Increases in Veterans and Active-Duty Military Personnel With Combat-Related Posttraumatic Stress Disorder and Mild Traumatic Brain Injury. J Head Trauma Rehabil 2021; 35:E1-E9. [PMID: 31033749 PMCID: PMC6814512 DOI: 10.1097/htr.0000000000000492] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To identify amygdalar volumetric differences associated with posttraumatic stress disorder (PTSD) in individuals with comorbid mild traumatic brain injury (mTBI) compared with those with mTBI-only and to examine the effects of intracranial volume (ICV) on amygdala volumetric measures. SETTING Marine Corps Base and VA Healthcare System. PARTICIPANTS A cohort of veterans and active-duty military personnel with combat-related mTBI (N = 89). DESIGN Twenty-nine participants were identified with comorbid PTSD and mTBI. The remaining 60 formed the mTBI-only control group. Structural images of brains were obtained with a 1.5-T MRI scanner using a T1-weighted 3D-IR-FSPGR pulse sequence. Automatic segmentation was performed in Freesurfer. MAIN MEASURES Amygdala volumes with/without normalizations to ICV. RESULTS The comorbid mTBI/PTSD group had significantly larger amygdala volumes, when normalized to ICV, compared with the mTBI-only group. The right and left amygdala volumes after normalization to ICV were 0.122% ± 0.012% and 0.118% ± 0.011%, respectively, in the comorbid group compared with 0.115% ± 0.012% and 0.112% ± 0.009%, respectively, in the mTBI-only group (corrected P < .05). CONCLUSIONS The ICV normalization analysis performed here may resolve previous literature discrepancies. This is an intriguing structural finding, given the role of the amygdala in the challenging neuroemotive symptoms witnessed in casualties of combat-related mTBI and PTSD.
Collapse
Affiliation(s)
- Joel Pieper
- Department of Internal Medicine, University of California, San Diego, CA, USA
| | - Douglas G. Chang
- Department of Orthopaedic Surgery, University of California, San Diego, CA, USA
| | | | - Ashley Robb Swan
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Radiology, University of California, San Diego, CA, USA
| | - Annemarie Angeles Quinto
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Radiology, University of California, San Diego, CA, USA
| | - Sharon Nichols
- Department of Neuroscience, University of California, San Diego, CA, USA
| | - Mithun Diwakar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Charles Huang
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - James Swan
- Department of Management Information Systems, San Diego State University, San Diego, CA, USA
| | - Roland Lee
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Radiology, University of California, San Diego, CA, USA
| | - Dewleen G. Baker
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, CA, USA
- VA Center of Excellence for Stress and Mental Health, San Diego, CA, USA
| | - Mingxiong Huang
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Radiology, University of California, San Diego, CA, USA
| |
Collapse
|
4
|
Yang M, Luo CH, Zhu YQ, Liu YC, An YJ, Iqbal J, Wang ZZ, Ma XM. 7, 8-Dihydroxy-4-methylcoumarin reverses depression model-induced depression-like behaviors and alteration of dendritic spines in the mood circuits. Psychoneuroendocrinology 2020; 119:104767. [PMID: 32563935 DOI: 10.1016/j.psyneuen.2020.104767] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 05/05/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022]
Abstract
Major depressive disorder (MDD) is a common mental disorder characterized by a persistent feeling of sadness, slow thought, impaired focus and loss of interest but the underlying mechanisms are largely unknown. Dendritic spines play an important role in the formation and maintenance of emotional circuits in the brain. Abnormalities in this process can lead to psychiatric diseases. 7,8-Dihydroxy-4-methylcoumarin (Dhmc), a precursor in the synthesis of derivatives of 4-methyl coumarin, plays an important role in protecting the nervous system from developing diseases and its most distinctive feature is safety. The aim of this study was to investigate whether Dhmc alleviates chronic unpredictable mild stress (CUMS)-induced depression-like behaviors and reverses CUMS-induced alterations in dendritic spines of principal neurons in brain areas of the emotional circuits including the hippocampus, medial prefrontal cortex (mPFC), nucleus accumbens (NAc) and basolateral amygdala (BLA) in male rats. Our results showed that CUMS-induced depression-like behaviors were accompanied by a decrease in spine density in pyramidal neurons of both the hippocampal CA3 area and the mPFC, and an increase in spine density in both the neurons of BLA and the medium spiny neurons (MSNs) of the NAc, as well as a decrease in the levels of the AMPA receptor subunit GluA1 and Kalirin-7 in the hippocampus compared with the control group. Intraperitoneal injection (i.p.) of Dhmc to the CUMS-exposed rats ameliorated CUMS-induced depression-like behaviors and reversed CUMS-mediated alterations in spine density and the levels of both GluA1 and Kalirin-7. Our results show an important role of Dhmc in reversing CUMS-induced depression-like behaviors and CUMS-mediated alterations in spine density.
Collapse
Affiliation(s)
- Mi Yang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710062, China; Yangling Vocational & Technical College, Yangling, 71210, China
| | - Chang-Hao Luo
- Yangling Vocational & Technical College, Yangling, 71210, China
| | - Ying-Qi Zhu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710062, China
| | - Yuan-Chu Liu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710062, China
| | - Ye-Juan An
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710062, China
| | - Javed Iqbal
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710062, China
| | - Zhe-Zhi Wang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710062, China.
| | - Xin-Ming Ma
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710062, China; Department of Neuroscience, University of Connecticut Health, Farmington, CT 06030, United States.
| |
Collapse
|
5
|
Sher LD, Geddie H, Olivier L, Cairns M, Truter N, Beselaar L, Essop MF. Chronic stress and endothelial dysfunction: mechanisms, experimental challenges, and the way ahead. Am J Physiol Heart Circ Physiol 2020; 319:H488-H506. [PMID: 32618516 DOI: 10.1152/ajpheart.00244.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although chronic stress is an important risk factor for cardiovascular diseases (CVD) onset, the underlying mechanisms driving such pathophysiological complications remain relatively unknown. Here, dysregulation of innate stress response systems and the effects of downstream mediators are strongly implicated, with the vascular endothelium emerging as a primary target of excessive glucocorticoid and catecholamine action. Therefore, this review article explores the development of stress-related endothelial dysfunction by focusing on the following: 1) assessing the phenomenon of stress and complexities surrounding this notion, 2) discussing mechanistic links between chronic stress and endothelial dysfunction, and 3) evaluating the utility of various preclinical models currently employed to study mechanisms underlying the onset of stress-mediated complications such as endothelial dysfunction. The data reveal that preclinical models play an important role in our efforts to gain an increased understanding of mechanisms underlying stress-mediated endothelial dysfunction. It is our understanding that this provides a good foundation going forward, and we propose that further efforts should be made to 1) more clearly define the concept of stress and 2) standardize protocols of animal models with specific guidelines to better indicate the mental complications that are simulated.
Collapse
Affiliation(s)
- Lucien Derek Sher
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Hannah Geddie
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Lukas Olivier
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Megan Cairns
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Nina Truter
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Leandrie Beselaar
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - M Faadiel Essop
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
6
|
Nikbakhtzadeh M, Borzadaran FM, Zamani E, Shabani M. Protagonist Role of Opioidergic System on Post-Traumatic Stress Disorder and Associated Pain. Psychiatry Investig 2020; 17:506-516. [PMID: 32492768 PMCID: PMC7324730 DOI: 10.30773/pi.2020.0002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/19/2020] [Accepted: 03/24/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Post-traumatic stress disorder (PTSD) and chronic pain often co-occur. Studies have shown an interaction between pain and PTSD. In this narrative review, we aim to support conducting comprehensive studies by describing PTSD, pain and determining whether opioidergic system, its agonist and antagonist manipulation could positively or negatively affect PTSD symptoms and concurrent pain. METHODS Term searches was done in Google Scholar, Scopus, ScienceDirect, Web of Science and PubMed databases as well as hand searching in key resource journals from 1979-2019. RESULTS There are a lot of contradictions and disputes when endogenous opioidergic system and opioidergic antagonist system are studied in PTSD patients. Exogenous morphine administration in PTSD patients can decrease the symptoms of PTSD but it doesn't have a pain reduction effect to an acceptable level. Beta-endorphin as an endogenous opioid is effective in pain reduction in the moment of events but after minutes to hours, the endorphins withdrawal syndrome leads to exaggerated intrusive thoughts and flashbacks of PTSD, which exacerbate the pain. It has also been shown that naloxone, as an opioidergic antagonist, can reduce or increase the PTSD symptoms and its associated pain. CONCLUSION Data suggest different roles of opioidergic system and their antagonist in pain control and mood in PTSD. However, further investigations need to be done in order to reveal the role of endogenous opioidergic system and opioidergic antagonist system as a mediator in PTSD patients suffering from acute or chronic pain.
Collapse
Affiliation(s)
- Marjan Nikbakhtzadeh
- Department of Physiology, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Fatemeh Mohtashami Borzadaran
- Kerman Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Zamani
- Department of Physiology, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Mohammad Shabani
- Kerman Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
7
|
Zhang HH, Meng SQ, Guo XY, Zhang JL, Zhang W, Chen YY, Lu L, Yang JL, Xue YX. Traumatic Stress Produces Delayed Alterations of Synaptic Plasticity in Basolateral Amygdala. Front Psychol 2019; 10:2394. [PMID: 31708835 PMCID: PMC6824323 DOI: 10.3389/fpsyg.2019.02394] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 10/07/2019] [Indexed: 12/19/2022] Open
Abstract
Acute traumatic event exposure is a direct cause of post-traumatic stress disorder (PTSD). Amygdala is suggested to be associated with the development of PTSD. In our previous findings, different activation patterns of GABAergic neurons and glutamatergic neurons in early or late stages after stress were found. However, the neural plastic mechanism underlying the role of basolateral amygdala (BLA) in post-traumatic stress disorder remains unclear. Therefore, this study mainly aimed at investigating time-dependent morphologic and electrophysiological changes in BLA during the development of PTSD. We used single prolonged stress (SPS) procedure to establish PTSD model of rats. The rats showed no alterations in anxiety behavior as well as in dendritic spine density or synaptic transmission in BLA 1 day after SPS. However, 10 days after SPS, rats showed enhancement of anxiety behavior, and spine density and frequency of miniature excitatory and inhibitory postsynaptic currents in BLA. Our results suggested that after traumatic stress, BLA displayed delayed increase in both spinogenesis and synaptic transmission, which seemed to facilitate the development of PTSD.
Collapse
Affiliation(s)
- Huan-Huan Zhang
- Department of Psychiatry, Tianjin Medical University, Tianjin, China
- Department of Clinical Psychology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shi-Qiu Meng
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Xin-Yi Guo
- Department of Psychiatry, Tianjin Medical University, Tianjin, China
- Department of Clinical Psychology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing-Liang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, Peking University School of Pharmaceutical Sciences, Beijing, China
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy and Purdue Institute for Integrative Neuroscience, West Lafayette, IN, United States
| | - Wen Zhang
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Ya-Yun Chen
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Lin Lu
- National Institute on Drug Dependence, Peking University, Beijing, China
- Peking University Sixth Hospital/Peking University Institute of Mental Health, Peking University, Beijing, China
| | - Jian-Li Yang
- Department of Psychiatry, Tianjin Medical University, Tianjin, China
- Department of Clinical Psychology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan-Xue Xue
- National Institute on Drug Dependence, Peking University, Beijing, China
| |
Collapse
|
8
|
Chen X, Jiang Y, Wang J, Liu Y, Xiao M, Song C, Bai Y, Yinuo Han N, Han F. Synapse impairment associated with enhanced apoptosis in post-traumatic stress disorder. Synapse 2019; 74:e22134. [PMID: 31562782 DOI: 10.1002/syn.22134] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 12/31/2022]
Abstract
Synapse impairment is associated with post-traumatic stress disorder (PTSD), which is characterized by enhanced apoptosis in the hippocampus, amygdala, and other brain regions. However, there are no detailed studies on the relationship between apoptosis and synaptic connectivity in PTSD. In this review, we discuss results from various studies describing the synaptic changes observed in the PTSD brain. A decreased number of dendrites/spines or increased number of immature spines in the hippocampus, medial prefrontal cortex, and other brain regions has been reported. Studies on axon guidance, myelination, and the cytoskeleton suggest that PTSD may involve axon overgrowth and overbranching. Apoptosis affects synapse formation; low levels of caspase maintain the balance between growth cone attraction and repulsion and inhibit axon elongation. PTSD enhances neuronal apoptosis through caspase activation, which disrupts the balance between growth cone attraction and repulsion and alters growth cone trajectory, leading to axon mistargeting. Meanwhile, caspase activation induces dendritic pruning and dendrite degeneration. These events contribute to the formation of fewer and aberrant synapses, which is associated with enhanced apoptosis in PTSD.
Collapse
Affiliation(s)
- Xinzhao Chen
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Yifan Jiang
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Jiayu Wang
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Yishu Liu
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Menglei Xiao
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Congshan Song
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Yu Bai
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Nancy Yinuo Han
- Faculty of Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fang Han
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| |
Collapse
|
9
|
Klaming R, Spadoni AD, Veltman DJ, Simmons AN. Expansion of hippocampal and amygdala shape in posttraumatic stress and early life stress. NEUROIMAGE-CLINICAL 2019; 24:101982. [PMID: 31437724 PMCID: PMC6706650 DOI: 10.1016/j.nicl.2019.101982] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/09/2019] [Accepted: 08/12/2019] [Indexed: 01/03/2023]
Abstract
OBJECTIVE The aim of this study was to examine the effect of Posttraumatic Stress Disorder (PTSD) and childhood adversity on brain structure. We assessed hippocampal and amygdala shape in veterans with varying levels of PTSD symptom severity and exposure to early life stressors (ELS). METHODS A total of 70 male veterans, who were deployed to a combat area during OIF/OEF/OND and who had been exposed to trauma during deployment, were included in the study. We applied a vertex-wise shape analysis of 3T MRI scans to measure indentation or expansion in hippocampal and amygdala shape. RESULTS Analyses showed a positive correlation between number of ELS and vertices in the right amygdala and the right hippocampus, as well as a positive correlation between PTSD symptom severity and right hippocampal vertices. There were no significant interactions between PTSD symptoms, ELS, and brain shape. DISCUSSION Results indicate a relationship between exposure to more childhood adversity and expansion in amygdala and hippocampal shape as well as between more severe PTSD symptoms and expansion in hippocampal shape. These findings may have important implications for the pathophysiology of trauma-related disorders.
Collapse
Affiliation(s)
- Ruth Klaming
- VA San Diego Healthcare System, University of California San Diego, San Diego, CA, USA; Department of Psychiatry, University of California San Diego, San Diego, CA, USA; Department of Psychiatry, VU University Medical Center, Amsterdam, The Netherlands.
| | - Andrea D Spadoni
- VA San Diego Healthcare System, University of California San Diego, San Diego, CA, USA; Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Dick J Veltman
- Department of Psychiatry, VU University Medical Center, Amsterdam, The Netherlands
| | - Alan N Simmons
- VA San Diego Healthcare System, University of California San Diego, San Diego, CA, USA; Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
10
|
RaiseAbdullahi P, Vafaei AA, Ghanbari A, Dadkhah M, Rashidy-Pour A. Time-dependent protective effects of morphine against behavioral and morphological deficits in an animal model of posttraumatic stress disorder. Behav Brain Res 2019; 364:19-28. [PMID: 30753875 DOI: 10.1016/j.bbr.2019.01.058] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/28/2019] [Accepted: 01/31/2019] [Indexed: 12/21/2022]
Abstract
Post-traumatic stress disorder (PTSD) arises after an individual has experienced a major traumatic event. Recent evidence suggests that acute morphine treatment may serve as a strategy to reduce PTSD development. In the present study, we investigated the time-dependent effects of morphine on behavioral and morphological deficits induced by the single prolonged stress (SPS), an experimental model of PTSD, in adult male rats. The rats were exposed to SPS (restraint for 2 h, forced swimming for 20 min, and ether anesthesia), and kept undistributed for 11 days. Morphine was injected immediately, 6, 12 and 24 h after SPS. Anxiety profile was evaluated using the elevated plus maze11 days after SPS. Then, animals were conditioned in a fear conditioning task and extinction training was performed on days 1, 2, 3, 4 and 11 after fear conditioning which followed by morphological assessments in the medial prefrontal cortex (mPFC). SPS rats showed increased anxiety levels and impaired contextual fear extinction retention. SPS also decreased dendritic length in the infra-limbic (IL) and dendritic spines in the IL and pre-limbic (PL) regions of the mPFC. Conversely, morphine treatment 6, 12 and 24 h but not immediately after SPS significantly improved anxiety-like behaviors, fear extinction, increased dendritic length, and spines in the mPFC. Morphine-induced much stronger response when injected 24 h after the SPS, and this effect was blocked by naloxone. Our findings show that morphine within a restricted time window selectively reversed the SPS-induced deficits in anxiety profile, fear extinction, and dendritic morphology in the mPFC. Finally, these findings suggest that the time point of morphine injection following a traumatic event is an important determinant of the full therapeutic effect of morphine against PTSD.
Collapse
Affiliation(s)
- Payman RaiseAbdullahi
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran; Laboratoryof Learning and Memory, Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Abbas Ali Vafaei
- Laboratoryof Learning and Memory, Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Ghanbari
- Laboratoryof Learning and Memory, Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Masoomeh Dadkhah
- Research Centers Development and Coordination Office, Deputy of Research& Technology, Ardabil University of Medical Sciences Ardabil, Iran
| | - Ali Rashidy-Pour
- Laboratoryof Learning and Memory, Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
11
|
Aten S, Page CE, Kalidindi A, Wheaton K, Niraula A, Godbout JP, Hoyt KR, Obrietan K. miR-132/212 is induced by stress and its dysregulation triggers anxiety-related behavior. Neuropharmacology 2019; 144:256-270. [PMID: 30342060 PMCID: PMC6823933 DOI: 10.1016/j.neuropharm.2018.10.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/25/2018] [Accepted: 10/14/2018] [Indexed: 02/08/2023]
Abstract
miR-132 and miR-212 are structurally-related microRNAs that are expressed from the same non-coding transcript. Accumulating evidence has shown that the dysregulation of these microRNAs contributes to aberrant neuronal plasticity and gene expression in the mammalian brain. Consistent with this, altered expression of miR-132 is associated with a number of affect-related psychiatric disorders. Here, we tested the functional contribution of the miR-132/212 locus to the development of stress-related and anxiety-like behaviors. Initially, we tested whether expression from the miR-132/212 locus is altered by stress-inducing paradigms. Using a 5-h acute-stress model, we show that both miR-132 and miR-212 are increased more than two-fold in the WT murine hippocampus and amygdala, whereas after a 15 day chronic-stress paradigm, expression of both miR-132 and miR-212 are upregulated more than two-fold within the amygdala but not in the hippocampus. Next, we used a tetracycline-inducible miR-132 overexpression mouse model and a miR-132/212 conditional knockout (cKO) mouse model to examine whether dysregulation of miR-132/212 expression alters basal anxiety-like behaviors. Interestingly, in both the miR-132 overexpression and cKO lines, significant increases in anxiety-like behaviors were detected. Importantly, suppression of transgenic miR-132 expression (via doxycycline administration) mitigated the anxiety-related behaviors. Further, expression of Sirt1 and Pten-two miR-132 target genes that have been implicated in the regulation of anxiety-were differentially regulated in the hippocampus and amygdala of miR-132/212 conditional knockout and miR-132 transgenic mice. Collectively, these data raise the prospect that miR-132 and miR-212 may play a key role in the modulation of stress responsivity and anxiety.
Collapse
Affiliation(s)
- Sydney Aten
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Chloe E Page
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Anisha Kalidindi
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Kelin Wheaton
- Division of Pharmaceutics and Pharmaceutical Chemistry, Ohio State University, Columbus, OH, USA
| | - Anzela Niraula
- Department of Neuroscience, Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Jon P Godbout
- Department of Neuroscience, Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Center for Brain and Spinal Cord Repair, Ohio State University, Columbus, OH, USA
| | - Kari R Hoyt
- Division of Pharmaceutics and Pharmaceutical Chemistry, Ohio State University, Columbus, OH, USA
| | - Karl Obrietan
- Department of Neuroscience, Ohio State University, Columbus, OH, USA.
| |
Collapse
|
12
|
Jia Y, Han Y, Wang X, Han F. Role of apoptosis in the Post-traumatic stress disorder model-single prolonged stressed rats. Psychoneuroendocrinology 2018; 95:97-105. [PMID: 29843020 DOI: 10.1016/j.psyneuen.2018.05.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/10/2018] [Accepted: 05/10/2018] [Indexed: 12/14/2022]
Abstract
Post-traumatic stress disorder (PTSD) is a stress-related mental disorder which occurs following exposure to traumatic events. A number of brain neuroimaging studies have revealed that PTSD patients have reduced volume and abnormal functions in the hippocampus and the amygdala. However, the pathogenesis of abnormalities in certain brain regions, as induced by PTSD, remains unclear. Recent studies, using the single prolonged stress (SPS) model, an animal model of PTSD, have found that abnormal apoptosis in certain brain regions, including the hippocampus, the amygdala, and the medial prefrontal cortex (mPFC); these areas are closely associated with emotion and cognition. In this review, we summarize the mechanism of apoptosis in SPS rats, including the endoplasmic reticulum (ER) and the mitochondria pathways. For the ER pathway, three individual pathways: PERK, IRE1, and ATF6 showed different roles on apoptosis and neuroprotection. Three key factors are thought to be involved in the mitochondrial pathway and PTSD-induced apoptosis: corticosteroid receptors, apoptosis-related factors, and anti-apoptosis factors. We have investigated the role of these factors and have attempted to identify which factors of the pathways are more focused towards neuronal protection, and which are more direct towards apoptosis. We also discussed the role of autophagy and the specific differences between autophagy and apoptosis in SPS rats. Finally, we discussed emerging researches related to anti-apoptosis treatment, including PERK inhibitors, IRE1 inhibitors, and metformin; collectively, these were exciting, but limited, This review provides a summary of the current understanding of apoptosis in SPS rats and the potential anti-apoptosis treatment strategies for PTSD.
Collapse
Affiliation(s)
- Yunbo Jia
- PTSD laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China
| | - Yunhe Han
- PTSD laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China
| | - Xinyue Wang
- PTSD laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China
| | - Fang Han
- PTSD laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
13
|
Lisieski MJ, Eagle AL, Conti AC, Liberzon I, Perrine SA. Single-Prolonged Stress: A Review of Two Decades of Progress in a Rodent Model of Post-traumatic Stress Disorder. Front Psychiatry 2018; 9:196. [PMID: 29867615 PMCID: PMC5962709 DOI: 10.3389/fpsyt.2018.00196] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 04/25/2018] [Indexed: 12/21/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a common, costly, and often debilitating psychiatric condition. However, the biological mechanisms underlying this disease are still largely unknown or poorly understood. Considerable evidence indicates that PTSD results from dysfunction in highly-conserved brain systems involved in stress, anxiety, fear, and reward. Pre-clinical models of traumatic stress exposure are critical in defining the neurobiological mechanisms of PTSD, which will ultimately aid in the development of new treatments for PTSD. Single prolonged stress (SPS) is a pre-clinical model that displays behavioral, molecular, and physiological alterations that recapitulate many of the same alterations observed in PTSD, illustrating its validity and giving it utility as a model for investigating post-traumatic adaptations and pre-trauma risk and protective factors. In this manuscript, we review the present state of research using the SPS model, with the goals of (1) describing the utility of the SPS model as a tool for investigating post-trauma adaptations, (2) relating findings using the SPS model to findings in patients with PTSD, and (3) indicating research gaps and strategies to address them in order to improve our understanding of the pathophysiology of PTSD.
Collapse
Affiliation(s)
- Michael J Lisieski
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Andrew L Eagle
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Alana C Conti
- Research and Development Service, John D. Dingell Veterans Affairs Medical Center, Detroit, MI, United States.,Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| | - Israel Liberzon
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States.,Mental Health Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, United States
| | - Shane A Perrine
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
14
|
Repeated shock stress facilitates basolateral amygdala synaptic plasticity through decreased cAMP-specific phosphodiesterase type IV (PDE4) expression. Brain Struct Funct 2017; 223:1731-1745. [PMID: 29204911 DOI: 10.1007/s00429-017-1575-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 11/07/2017] [Indexed: 10/18/2022]
Abstract
Previous studies have shown that exposure to stressful events can enhance fear memory and anxiety-like behavior as well as increase synaptic plasticity in the rat basolateral amygdala (BLA). We have evidence that repeated unpredictable shock stress (USS) elicits a long-lasting increase in anxiety-like behavior in rats, but the cellular mechanisms mediating this response remain unclear. Evidence from recent morphological studies suggests that alterations in the dendritic arbor or spine density of BLA principal neurons may underlie stress-induced anxiety behavior. Recently, we have shown that the induction of long-term potentiation (LTP) in BLA principal neurons is dependent on activation of postsynaptic D1 dopamine receptors and the subsequent activation of the cyclic adenosine 5'-monophosphate (cAMP)-protein kinase A (PKA) signaling cascade. Here, we have used in vitro whole-cell patch-clamp recording from BLA principal neurons to investigate the long-term consequences of USS on their morphological properties and synaptic plasticity. We provided evidence that the enhanced anxiety-like behavior in response to USS was not associated with any significant change in the morphological properties of BLA principal neurons, but was associated with a changed frequency dependence of synaptic plasticity, lowered LTP induction threshold, and reduced expression of phosphodiesterase type 4 enzymes (PDE4s). Furthermore, pharmacological inhibition of PDE4 activity with rolipram mimics the effects of chronic stress on LTP induction threshold and baseline startle. Our results provide the first evidence that stress both enhances anxiety-like behavior and facilitates synaptic plasticity in the amygdala through a common mechanism of PDE4-mediated disinhibition of cAMP-PKA signaling.
Collapse
|
15
|
Brain atrophy in the visual cortex and thalamus induced by severe stress in animal model. Sci Rep 2017; 7:12731. [PMID: 28986553 PMCID: PMC5630603 DOI: 10.1038/s41598-017-12917-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 09/12/2017] [Indexed: 12/28/2022] Open
Abstract
Psychological stress induces many diseases including post-traumatic stress disorder (PTSD); however, the causal relationship between stress and brain atrophy has not been clarified. Applying single-prolonged stress (SPS) to explore the global effect of severe stress, we performed brain magnetic resonance imaging (MRI) acquisition and Voxel-based morphometry (VBM). Significant atrophy was detected in the bilateral thalamus and right visual cortex. Fluorescent immunohistochemistry for Iba-1 as the marker of activated microglia indicates regional microglial activation as stress-reaction in these atrophic areas. These data certify the impact of severe psychological stress on the atrophy of the visual cortex and the thalamus. Unexpectedly, these results are similar to chronic neuropathic pain rather than PTSD clinical research. We believe that some sensitisation mechanism from severe stress-induced atrophy in the visual cortex and thalamus, and the functional defect of the visual system may be a potential therapeutic target for stress-related diseases.
Collapse
|
16
|
Goodman J, McIntyre CK. Impaired Spatial Memory and Enhanced Habit Memory in a Rat Model of Post-traumatic Stress Disorder. Front Pharmacol 2017; 8:663. [PMID: 29018340 PMCID: PMC5614977 DOI: 10.3389/fphar.2017.00663] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/06/2017] [Indexed: 01/25/2023] Open
Abstract
High levels of emotional arousal can impair spatial memory mediated by the hippocampus, and enhance stimulus-response (S-R) habit memory mediated by the dorsolateral striatum (DLS). The present study was conducted to determine whether these memory systems may be similarly affected in an animal model of post-traumatic stress disorder (PTSD). Sprague-Dawley rats were subjected to a “single-prolonged stress” (SPS) procedure and 1 week later received training in one of two distinct versions of the plus-maze: a hippocampus-dependent place learning task or a DLS-dependent response learning task. Results indicated that, relative to non-stressed control rats, SPS rats displayed slower acquisition in the place learning task and faster acquisition in the response learning task. In addition, extinction of place learning and response learning was impaired in rats exposed to SPS, relative to non-stressed controls. The influence of SPS on hippocampal spatial memory and DLS habit memory observed in the present study may be relevant to understanding some common features of PTSD, including hippocampal memory deficits, habit-like avoidance responses to trauma-related stimuli, and greater likelihood of developing drug addiction and alcoholism.
Collapse
Affiliation(s)
- Jarid Goodman
- School of Behavioral and Brain Sciences, University of Texas at Dallas, RichardsonTX, United States
| | - Christa K McIntyre
- School of Behavioral and Brain Sciences, University of Texas at Dallas, RichardsonTX, United States
| |
Collapse
|
17
|
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
|
18
|
Fatahi Z, Zibaii MI, Haghparast A. Effect of acute and subchronic stress on electrical activity of basolateral amygdala neurons in conditioned place preference paradigm: An electrophysiological study. Behav Brain Res 2017; 335:19-25. [DOI: 10.1016/j.bbr.2017.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/28/2017] [Accepted: 08/05/2017] [Indexed: 01/19/2023]
|
19
|
Lim SI, Song KH, Yoo CH, Woo DC, Choe BY. Decreased Glutamatergic Activity in the Frontal Cortex of Single Prolonged Stress Model: In vivo and Ex Vivo Proton MR Spectroscopy. Neurochem Res 2017; 42:2218-2229. [PMID: 28349360 DOI: 10.1007/s11064-017-2232-x] [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: 11/15/2016] [Revised: 03/02/2017] [Accepted: 03/10/2017] [Indexed: 01/01/2023]
Abstract
Single prolonged stress (SPS) is one of the preclinical models of posttraumatic stress disorder (PTSD) in humans. Not every traumatized person develops PTSD and the onset of the disease varies from months to many years after exposure to life-threatening events. The pathogenetic neurometabolites in PTSD have not been investigated to date, and could provide a means for therapeutic interventions. Therefore the present study aimed to evaluate neurochemical changes in the frontal cortex in the SPS model during time-dependent sensitization using in vivo and ex vivo proton magnetic spectroscopy (1H-MRS). Twenty-one male Sprague-Dawley rats (200-220 g) were randomly assigned into two groups (Control, n = 10; SPS, n = 11). SPS consists of three consecutive stressors (restraint, forced swimming, and ether exposure) followed by 7 days without disturbance. In vivo 1H-MRS scans were conducted at baseline, immediately after SPS, and 3 and 7 days after SPS to quantify time-dependent alterations in the frontal cortex. On day 7, all animals were sacrificed and ex vivo 1H-MRS was performed. After SPS exposure, the SPS group showed signs of excitatory activities (glutamate) and cellular membrane turnover (choline and total choline) for 7 days. After the time-sensitization period, the SPS group showed lower glutamate and creatine levels and higher choline and lactate levels than the control group. These results indicate that SPS induces sustained adaptation of glutamatergic neuronal activity in the frontal cortex. Therefore, we conclude that SPS-induced stress reduces glutamatergic metabolism in the frontal cortex.
Collapse
Affiliation(s)
- Song-I Lim
- Department of Biomedical Engineering, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Research Institute of Biomedical Engineering, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Kyu-Ho Song
- Department of Biomedical Engineering, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Research Institute of Biomedical Engineering, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Chi-Hyeon Yoo
- Department of Biomedical Engineering, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Research Institute of Biomedical Engineering, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Dong-Cheol Woo
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Bo-Young Choe
- Department of Biomedical Engineering, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea. .,Research Institute of Biomedical Engineering, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
20
|
Cacciaglia R, Nees F, Grimm O, Ridder S, Pohlack ST, Diener SJ, Liebscher C, Flor H. Trauma exposure relates to heightened stress, altered amygdala morphology and deficient extinction learning: Implications for psychopathology. Psychoneuroendocrinology 2017; 76:19-28. [PMID: 27871027 DOI: 10.1016/j.psyneuen.2016.11.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 11/06/2016] [Accepted: 11/10/2016] [Indexed: 12/13/2022]
Abstract
Stress exposure causes a structural reorganization in neurons of the amygdala. In particular, animal models have repeatedly shown that both acute and chronic stress induce neuronal hypertrophy and volumetric increase in the lateral and basolateral nuclei of amygdala. These effects are visible on the behavioral level, where stress enhances anxiety behaviors and provokes greater fear learning. We assessed stress and anxiety levels in a group of 18 healthy human trauma-exposed individuals (TR group) compared to 18 non-exposed matched controls (HC group), and related these measurements to amygdala volume. Traumas included unexpected adverse experiences such as vehicle accidents or sudden loss of a loved one. As a measure of aversive learning, we implemented a cued fear conditioning paradigm. Additionally, to provide a biological marker of chronic stress, we measured the sensitivity of the hypothalamus-pituitary-adrenal (HPA) axis using a dexamethasone suppression test. Compared to the HC, the TR group showed significantly higher levels of chronic stress, current stress and trait anxiety, as well as increased volume of the left amygdala. Specifically, we observed a focal enlargement in its lateral portion, in line with previous animal data. Compared to HC, the TR group also showed enhanced late acquisition of conditioned fear and deficient extinction learning, as well as salivary cortisol hypo-suppression to dexamethasone. Left amygdala volumes positively correlated with suppressed morning salivary cortisol. Our results indicate differences in trauma-exposed individuals which resemble those previously reported in animals exposed to stress and in patients with post-traumatic stress disorder and depression. These data provide new insights into the mechanisms through which traumatic stress might prompt vulnerability for psychopathology.
Collapse
Affiliation(s)
- Raffaele Cacciaglia
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany; Department of Psychiatry and Clinical Psychobiology, University of Barcelona, Passeig de la vall d'Hebron 171, Barcelona, Catalonia, Spain.
| | - Frauke Nees
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Oliver Grimm
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Frankfurt, Germany
| | - Stephanie Ridder
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Sebastian T Pohlack
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Slawomira J Diener
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Claudia Liebscher
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Herta Flor
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany.
| |
Collapse
|
21
|
Qi J, Chen C, Meng QX, Wu Y, Wu H, Zhao TB. Crosstalk between Activated Microglia and Neurons in the Spinal Dorsal Horn Contributes to Stress-induced Hyperalgesia. Sci Rep 2016; 6:39442. [PMID: 27995982 PMCID: PMC5171842 DOI: 10.1038/srep39442] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 11/21/2016] [Indexed: 12/30/2022] Open
Abstract
Stress has been shown to enhance pain sensitivity resulting in stress-induced hyperalgesia. However, the underlying mechanisms have yet to be elucidated. Using single-prolonged stress combined with Complete Freund's Adjuvant injection model, we explored the reciprocal regulatory relationship between neurons and microglia, which is critical for the maintenance of posttraumatic stress disorder (PTSD)-induced hyperalgesia. In our assay, significant mechanical allodynia was observed. Additionally, activated neurons in spinal dorsal horn were observed by analysis of Fos expression. And, microglia were also significantly activated with the presence of increased Iba-1 expression. Intrathecal administration of c-fos antisense oligodeoxynucleotides (ASO) or minocycline (a specific microglia inhibitor) attenuated mechanical allodynia. Moreover, intrathecal administration of c-fos ASO significantly suppressed the activation of neurons and microglia. Interestingly, inhibition of microglia activation by minocycline significantly suppressed the activation of both neurons and microglia in spinal dorsal horn. P38 inhibitor SB203580 suppressed IL-6 production, and inhibition of IL-6 receptor (IL-6R) activation by tocilizumab suppressed Fos expression. Together, our data suggest that the presence of a "crosstalk" between activated microglia and neurons in the spinal dorsal horn, which might contribute to the stress-induced hyperactivated state, leading to an increased pain sensitivity.
Collapse
Affiliation(s)
- Jian Qi
- Department of Spinal Cord Injury and Rehabilitation, The General Hospital of Jinan Military Command, Jinan, 250031, China
| | - Chen Chen
- Department of Pharmacy, The Second Hospital of Shandong University, Jinan, 250031, China
| | - Qing-Xi Meng
- Department of Spinal Cord Injury and Rehabilitation, The General Hospital of Jinan Military Command, Jinan, 250031, China
| | - Yan Wu
- Department of Neurobiology, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Haitao Wu
- Department of Neurobiology, Institute of Basic Medical Sciences, Beijing, 100850, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, 226001, China
| | - Ting-Bao Zhao
- Department of Spinal Cord Injury and Rehabilitation, The General Hospital of Jinan Military Command, Jinan, 250031, China
| |
Collapse
|
22
|
Knox D, Stanfield BR, Staib JM, David NP, Keller SM, DePietro T. Neural circuits via which single prolonged stress exposure leads to fear extinction retention deficits. ACTA ACUST UNITED AC 2016; 23:689-698. [PMID: 27918273 PMCID: PMC5110987 DOI: 10.1101/lm.043141.116] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/10/2016] [Indexed: 01/20/2023]
Abstract
Single prolonged stress (SPS) has been used to examine mechanisms via which stress exposure leads to post-traumatic stress disorder symptoms. SPS induces fear extinction retention deficits, but neural circuits critical for mediating these deficits are unknown. To address this gap, we examined the effect of SPS on neural activity in brain regions critical for extinction retention (i.e., fear extinction circuit). These were the ventral hippocampus (vHipp), dorsal hippocampus (dHipp), basolateral amygdala (BLA), prelimbic cortex (PL), and infralimbic cortex (IL). SPS or control rats were fear conditioned then subjected to extinction training and testing. Subsets of rats were euthanized after extinction training, extinction testing, or immediate removal from the housing colony (baseline condition) to assay c-Fos levels (measure of neural activity) in respective brain region. SPS induced extinction retention deficits. During extinction training SPS disrupted enhanced IL neural activity and inhibited BLA neural activity. SPS also disrupted inhibited BLA and vHipp neural activity during extinction testing. Statistical analyses suggested that SPS disrupted functional connectivity within the dHipp during extinction training and increased functional connectivity between the BLA and vHipp during extinction testing. Our findings suggest that SPS induces extinction retention deficits by disrupting both excitatory and inhibitory changes in neural activity within the fear extinction circuit and inducing changes in functional connectivity within the Hipp and BLA.
Collapse
Affiliation(s)
- Dayan Knox
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware 19716, USA
| | - Briana R Stanfield
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, New Jersey 07102, USA
| | - Jennifer M Staib
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware 19716, USA
| | - Nina P David
- School of Public Policy and Administration, University of Delaware, Newark, Delaware 19716, USA
| | - Samantha M Keller
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware 19716, USA
| | - Thomas DePietro
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware 19716, USA
| |
Collapse
|
23
|
Schmeltzer SN, Herman JP, Sah R. Neuropeptide Y (NPY) and posttraumatic stress disorder (PTSD): A translational update. Exp Neurol 2016; 284:196-210. [PMID: 27377319 PMCID: PMC8375392 DOI: 10.1016/j.expneurol.2016.06.020] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 06/15/2016] [Accepted: 06/20/2016] [Indexed: 12/12/2022]
Abstract
Posttraumatic stress disorder (PTSD) is a trauma-evoked syndrome, with variable prevalence within the human population due to individual differences in coping and resiliency. In this review, we discuss evidence supporting the relevance of neuropeptide Y (NPY), a stress regulatory transmitter in PTSD. We consolidate findings from preclinical, clinical, and translational studies of NPY that are of relevance to PTSD with an attempt to provide a current update of this area of research. NPY is abundantly expressed in forebrain limbic and brainstem areas that regulate stress and emotional behaviors. Studies in rodents demonstrate a role for NPY in stress responses, anxiety, fear, and autonomic regulation, all relevant to PTSD symptomology. Genetic studies support an association of NPY polymorphisms with stress coping and affect. Importantly, cerebrospinal fluid (CSF) measurements in combat veterans provide direct evidence of NPY association with PTSD diagnosis and symptomology. In addition, NPY involvement in pain, depression, addiction, and metabolism may be relevant to comorbidities associated with PTSD. Collectively, the literature supports the relevance of NPY to PTSD pathophysiology, although knowledge gaps remain. The NPY system is an attractive target in terms of understanding the physiological basis of PTSD as well as treatment of the disorder.
Collapse
Affiliation(s)
- Sarah N Schmeltzer
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States
| | - James P Herman
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States
| | - Renu Sah
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States; VA Medical Center, Cincinnati, OH, 45220, United States.
| |
Collapse
|
24
|
Gastrodin reversed the traumatic stress-induced depressed-like symptoms in rats. J Nat Med 2016; 70:749-59. [PMID: 27417451 DOI: 10.1007/s11418-016-1010-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/12/2016] [Indexed: 10/21/2022]
Abstract
Exposure to severe stress can lead to the development of neuropsychiatric disorders such as depression and post-traumatic stress disorder (PTSD) in at-risk individuals. Gastrodin (GAS), a primary constituent of an Oriental herbal medicine, has been shown to effectively treat various mood disorders. Thus, the present study aimed to determine whether GAS would ameliorate stress-associated depression-like behaviors in a rat model of single prolonged stress (SPS)-induced PTSD. Following the SPS procedure, rats received intraperitoneal administration of GAS (20, 50, or 100 mg/kg) once daily for 2 weeks. Subsequently, the rats performed the forced swimming test, and norepinephrine (NE) levels in the hippocampus were measured. Daily GAS (100 mg/kg) significantly reversed depression-like behaviors and restored SPS-induced increases in hippocampal NE concentrations as well as tyrosine hydroxylase expression in the locus coeruleus. Furthermore, the administration of GAS attenuated SPS-induced decreases in the hypothalamic expression of neuropeptide Y and the hippocampal mRNA expression of brain-derived neurotrophic factor. These findings indicate that GAS possesses antidepressant effects in the PTSD and may be an effective herbal preparation for the treatment of PTSD.
Collapse
|
25
|
Hoffman JR, Ostfeld I, Kaplan Z, Zohar J, Cohen H. Exercise Enhances the Behavioral Responses to Acute Stress in an Animal Model of PTSD. Med Sci Sports Exerc 2016; 47:2043-52. [PMID: 25699481 DOI: 10.1249/mss.0000000000000642] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION This study examined the effects of endurance exercise on the behavioral response to stress and patterns of brain-derived neurotrophic factor (BDNF), neuropeptide Y (NPY), and δ-opioid receptor (phospho-DOR) expression in the hippocampus. METHODS Animals ran on a treadmill at 15 m·min, 5 min·d gradually increasing to 20 min·d, 5 d·wk for 6 wk. After training, one group of animals was exposed to a predator scent stress (PSS) protocol for 10 min. Outcome measurements included behavior in an elevated plus-maze (EPM) and acoustic startle response (ASR) 7 d after exposure to stress. Immunohistochemical technique was used to detect the expression of the BDNF, NPY, and phospho-DOR in the hippocampus 8 d after exposure. RESULTS Sedentary animals exposed to PSS were observed to have a greater incidence of extreme behavior responses including higher anxiety, less total activity in the EPM, and greater amplitude in the ASR than unexposed and/or trained animals. Exercise-trained animals exposed to PSS developed a resiliency to the stress, reflected by significantly greater total activity in the EPM, reduced anxiety, and reduced ASR compared to the sedentary, exposed animals. Exercise in the absence of stress significantly elevated the expression of BDNF and phospho-DOR, whereas exposure to PSS resulted in a significant decline in the expression of NPY, BDNF, and phospho-DOR. Trained animals that were exposed maintained expression of BDNF, NPY, and phospho-DOR in most subregions of the hippocampus. CONCLUSION Results indicated that endurance training provided a mechanism to promote resilience and/or recovery from stress. In addition, exercise increased expression of BDNF, NPY, and DOR signaling in the hippocampus that was associated with the greater resiliency seen in the trained animals.
Collapse
Affiliation(s)
- Jay R Hoffman
- 1Institute of Exercise Physiology and Wellness, Sport and Exercise Science, University of Central Florida, Orlando, FL; 2Israel Defense Force, Medical Corps, Tel Hashomer, ISRAEL; 3Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Faculty of Health Sciences, Division of Psychiatry, Ben-Gurion University of the Negev, Beer-Sheva, ISRAEL; and 4The State of Israel Ministry of Health, The Chaim Sheba Medical Center, Division of Psychiatry, Ramat-Gan, Israel, Sackler Medical School, Tel-Aviv University, ISRAEL
| | | | | | | | | |
Collapse
|
26
|
Stress-Induced Increases in Levels of Caspases in the Prefrontal Cortex in a Rat Model of PTSD. NEUROPHYSIOLOGY+ 2016. [DOI: 10.1007/s11062-016-9563-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
27
|
Behavioral changes over time in post-traumatic stress disorder: Insights from a rat model of single prolonged stress. Behav Processes 2016; 124:123-9. [DOI: 10.1016/j.beproc.2016.01.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 12/30/2015] [Accepted: 01/01/2016] [Indexed: 12/11/2022]
|
28
|
Leitermann RJ, Rostkowski AB, Urban JH. Neuropeptide Y input to the rat basolateral amygdala complex and modulation by conditioned fear. J Comp Neurol 2016; 524:2418-39. [PMID: 26779765 DOI: 10.1002/cne.23960] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 12/30/2015] [Accepted: 01/05/2016] [Indexed: 12/14/2022]
Abstract
Within the basolateral amygdaloid complex (BLA), neuropeptide Y (NPY) buffers against protracted anxiety and fear. Although the importance of NPY's actions in the BLA is well documented, little is known about the source(s) of NPY fibers to this region. The current studies identified sources of NPY projections to the BLA by using a combination of anatomical and neurochemical approaches. NPY innervation of the BLA was assessed in rats by examining the degree of NPY coexpression within interneurons or catecholaminergic fibers with somatostatin and tyrosine hydroxylase (TH) or dopamine β-hydroxylase (DβH), respectively. Numerous NPY(+) /somatostatin(+) and NPY(+) /somatostatin(-) fibers were observed, suggesting at least two populations of NPY fibers within the BLA. No colocalization was noted between NPY and TH or DβH immunoreactivities. Additionally, Fluorogold (FG) retrograde tracing with immunohistochemistry was used to identify the precise origin of NPY projections to the BLA. FG(+) /NPY(+) cells were identified within the amygdalostriatal transition area (AStr) and stria terminalis and scattered throughout the bed nucleus of the stria terminalis. The subpopulation of NPY neurons in the AStr also coexpressed somatostatin. Subjecting animals to a conditioned fear paradigm increased NPY gene expression within the AStr, whereas no changes were observed within the BLA or stria terminalis. Overall, these studies identified limbic regions associated with stress circuits providing NPY input to the BLA and demonstrated that a unique NPY projection from the AStr may participate in the regulation of conditioned fear. J. Comp. Neurol. 524:2418-2439, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Randy J Leitermann
- Department of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - Amanda B Rostkowski
- Department of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - Janice H Urban
- Department of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| |
Collapse
|
29
|
Dendritic Spines in Depression: What We Learned from Animal Models. Neural Plast 2016; 2016:8056370. [PMID: 26881133 PMCID: PMC4736982 DOI: 10.1155/2016/8056370] [Citation(s) in RCA: 304] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/26/2015] [Indexed: 02/07/2023] Open
Abstract
Depression, a severe psychiatric disorder, has been studied for decades, but the underlying mechanisms still remain largely unknown. Depression is closely associated with alterations in dendritic spine morphology and spine density. Therefore, understanding dendritic spines is vital for uncovering the mechanisms underlying depression. Several chronic stress models, including chronic restraint stress (CRS), chronic unpredictable mild stress (CUMS), and chronic social defeat stress (CSDS), have been used to recapitulate depression-like behaviors in rodents and study the underlying mechanisms. In comparison with CRS, CUMS overcomes the stress habituation and has been widely used to model depression-like behaviors. CSDS is one of the most frequently used models for depression, but it is limited to the study of male mice. Generally, chronic stress causes dendritic atrophy and spine loss in the neurons of the hippocampus and prefrontal cortex. Meanwhile, neurons of the amygdala and nucleus accumbens exhibit an increase in spine density. These alterations induced by chronic stress are often accompanied by depression-like behaviors. However, the underlying mechanisms are poorly understood. This review summarizes our current understanding of the chronic stress-induced remodeling of dendritic spines in the hippocampus, prefrontal cortex, orbitofrontal cortex, amygdala, and nucleus accumbens and also discusses the putative underlying mechanisms.
Collapse
|
30
|
Lee B, Sur B, Cho SG, Yeom M, Shim I, Lee H, Hahm DH. Ginsenoside Rb1 rescues anxiety-like responses in a rat model of post-traumatic stress disorder. J Nat Med 2015; 70:133-44. [PMID: 26611866 DOI: 10.1007/s11418-015-0943-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 10/01/2015] [Indexed: 01/11/2023]
Abstract
Single prolonged stress (SPS), a rat model of post-traumatic stress disorder (PTSD), induces alterations in the hypothalamic-pituitary-adrenal axis. Korean red ginseng, whose major active component is ginsenoside Rb1 (GRb1), is one of the widely used traditional anxiolytics. However, the efficacy of GRb1 in alleviating PTSD-associated anxiety-like abnormalities has not been investigated. The present study used several behavioral tests to examine the effects of GRb1 on symptoms of anxiety in rats after SPS exposure and on the central noradrenergic system. Male Sprague-Dawley rats received GRb1 (10 or 30 mg/kg, i.p., once daily) during 14 days of SPS. Daily GRb1 (30 mg/kg) administration significantly increased the number and duration of open-arm visits in the elevated plus maze (EPM) test, reduced the anxiety index, increased the risk assessment, reduced grooming behaviors in the EPM test, and increased the total number of line crossings of an open field after SPS. The higher dose of GRb1 also blocked SPS-induced decreases in hypothalamic neuropeptide Y expression, increases in locus coeruleus tyrosine hydroxylase expression, and decreases in hippocampal mRNA expression of brain-derived neurotrophic factor. These findings suggest that GRb1 has anxiolytic-like effects on both behavioral and biochemical symptoms similar to those observed in patients with 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, 130-701, Republic of Korea.
| | - Bongjun Sur
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Seong-Guk Cho
- The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea
| | - Mijung Yeom
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Insop Shim
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 130-701, Republic of Korea.,The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea
| | - Hyejung Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 130-701, 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, 130-701, Republic of Korea. .,The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea.
| |
Collapse
|
31
|
Saur L, Baptista PPA, Bagatini PB, Neves LT, de Oliveira RM, Vaz SP, Ferreira K, Machado SA, Mestriner RG, Xavier LL. Experimental Post-traumatic Stress Disorder Decreases Astrocyte Density and Changes Astrocytic Polarity in the CA1 Hippocampus of Male Rats. Neurochem Res 2015; 41:892-904. [DOI: 10.1007/s11064-015-1770-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/20/2015] [Accepted: 11/11/2015] [Indexed: 12/16/2022]
|
32
|
Wilson MA, Grillo CA, Fadel JR, Reagan LP. Stress as a one-armed bandit: Differential effects of stress paradigms on the morphology, neurochemistry and behavior in the rodent amygdala. Neurobiol Stress 2015; 1:195-208. [PMID: 26844236 PMCID: PMC4721288 DOI: 10.1016/j.ynstr.2015.06.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 06/04/2015] [Accepted: 06/05/2015] [Indexed: 12/12/2022] Open
Abstract
Neuroplasticity may be defined as the ability of the central nervous system (CNS) to respond to changes in the internal and external environment and it is well established that some stimuli have the ability to facilitate or impair neuroplasticity depending on the pre-existing milieu. A classic example of a stimulus that can both facilitate and impair neuroplasticity is stress. Indeed, the ability of CNS to respond to acute stress is often dependent upon the prior stress history of the individual. While responses to acute stress are often viewed as adaptive in nature, stress reactivity in subjects with prior chronic stress experiences are often linked to neuropsychiatric disorders, including major depressive disorder, post-traumatic stress disorder (PTSD) and anxiety. In rodent studies, chronic stress exposure produces structural and functional alterations in the hippocampus and medial prefrontal cortex that are consistent across different types of stress paradigms. Conversely, the amygdala appears to exhibit differential structural and functional responses to stress that are dependent on a variety of factors, including the type of stressor performed and the duration of the stress paradigm. This is most evident in output measures including morphological analysis of amygdala neurons, measurement of glutamatergic tone in amygdalar subdivisions and the analysis of amygdala-centric behaviors. Accordingly, this review will provide an overview of the effects of stress on the structural and functional plasticity of the rodent amygdala, especially in relation to the differential effects of repeated or chronic stress paradigms on dendritic architecture, neurochemistry of the glutamatergic system and behavior.
Collapse
Affiliation(s)
- Marlene A. Wilson
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
- WJB Dorn Veterans Affairs Medical Center, Columbia, SC, USA
| | - Claudia A. Grillo
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Jim R. Fadel
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Lawrence P. Reagan
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
- WJB Dorn Veterans Affairs Medical Center, Columbia, SC, USA
- Corresponding author. Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Road, D40, Columbia, SC 29208, USA.
| |
Collapse
|
33
|
Marks W, Fenton E, Guskjolen A, Kalynchuk L. The effect of chronic corticosterone on fear learning and memory depends on dose and the testing protocol. Neuroscience 2015; 289:324-33. [DOI: 10.1016/j.neuroscience.2015.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 12/04/2014] [Accepted: 01/07/2015] [Indexed: 01/15/2023]
|
34
|
|
35
|
Distinctive hippocampal and amygdalar cytoarchitectural changes underlie specific patterns of behavioral disruption following stress exposure in an animal model of PTSD. Eur Neuropsychopharmacol 2014; 24:1925-44. [PMID: 25451698 DOI: 10.1016/j.euroneuro.2014.09.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 07/14/2014] [Accepted: 09/24/2014] [Indexed: 01/01/2023]
Abstract
Alterations in cytoarchitecture and molecular signaling have been observed in adaptive and maladaptive responses to stress and presumably underlie the physiological and behavioral changes observed. The relationship between behavioral responses to stress exposure and changes in cytoarchitecture of subregions of the hippocampus and amygdala was investigated in an animal model of PTSD. Behaviors in elevated plus-maze and acoustic startle response tests were assessed in rats 7 days after exposure to predator scent stress. Brains were harvested 24h later. Neurons from CA1, CA3, and dentate gyrus subregions and basolateral amygdala were reconstructed and subjected to Sholl analysis and spine density estimation. Glucocorticoid receptor, brain-derived neurotrophic factor, phospho-NR1-Ser-889, phospho-GluR1-Ser-845, phospho-calcium/calmodulin dependent protein kinase II-Thy-286, post-synaptic density protein 95 and phospho-CREB-Ser-133 were evaluated in the hippocampus. Data were analyzed by retrospective classification of individual rats into three behavioral response groups. The extent and distribution of changes in the morphology of hippocampal and amygdalar dendrites was significantly associated with stress-induced behavioral response classification. Extreme (PTSD-like) behavioral disruption was associated with extensive neuronal retraction in the hippocampus and proliferation in the amygdala. Neither structure displayed such changes in minimal behavioral responders. Partial behavioral response was associated with identical changes in the hippocampus only. Patterns of change in requisite molecular signaling genes and endophenotypic markers corresponded to the structural and behavioral responses. The extent and distribution of changes in the cytoarchitecture of hippocampal and amygdalar subregions is directly related to the pattern of behavioral response of the individual to stress exposure.
Collapse
|
36
|
Novel antidepressant-like activity of caffeic Acid phenethyl ester is mediated by enhanced glucocorticoid receptor function in the hippocampus. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:646039. [PMID: 25477995 PMCID: PMC4248557 DOI: 10.1155/2014/646039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/16/2014] [Accepted: 09/17/2014] [Indexed: 11/17/2022]
Abstract
Caffeic acid phenethyl ester (CAPE) is an active component of propolis that has a variety of potential pharmacological effects. Although we previously demonstrated that propolis has antidepressant-like activity, the effect of CAPE on this activity remains unknown. The present study assessed whether treatment with CAPE (5, 10, and 20 µmol/kg for 21 days) has an antidepressant-like effect in mice subjected to chronic unpredictable stress via tail suspension (TST) and forced swim (FST) tests. CAPE administration induced behaviors consistent with an antidepressant effect, evidenced by decreased immobility in the TST and FST independent of any effect on serum corticosterone secretion. Western blots, conducted subsequent to behavioral assessment, revealed that CAPE significantly decreased glucocorticoid receptor phosphorylation at S234 (pGR(S234)), resulting in an increased pGR(S220/S234) ratio. We also observed negative correlations between pGR(S220)/(S234) and p38 mitogen-activated protein kinase (p38MAPK) phosphorylation, which was decreased by CAPE treatment. These findings suggest that CAPE treatment exerts an antidepressant-like effect via downregulation of p38MAPK phosphorylation, thereby contributing to enhanced GR function.
Collapse
|
37
|
Lee B, Sur B, Yeom M, Shim I, Lee H, Hahm DH. L-tetrahydropalmatine ameliorates development of anxiety and depression-related symptoms induced by single prolonged stress in rats. Biomol Ther (Seoul) 2014; 22:213-22. [PMID: 25009702 PMCID: PMC4060081 DOI: 10.4062/biomolther.2014.032] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 04/14/2014] [Accepted: 05/07/2014] [Indexed: 11/07/2022] Open
Abstract
Abnormal adaptation of the stress-response system following traumatic stress can lead to alterations in the hypothalamic-pituitary-adrenal (HPA) axis that may contribute to the development of post-traumatic stress disorder (PTSD). The present study used several behavioral tests to investigate the anxiolytic-like and antidepressant activity of L-tetrahydropalmatine (L-THP) in an experimental rat model of anxiety and depression induced by single prolonged stress (SPS), an animal model of PTSD. Male rats were treated intraperitoneally (i.p.) with vehicle or varied doses of THP 30 min prior to SPS for 8 consecutive days. Daily THP (50 mg/kg) administration significantly increased the number and duration of open arm visits in the elevated plus maze (EPM) test, reduced the anxiety index, increased the risk assessment, and increased the number of head dips over the borders of the open arms after SPS. THP was also associated with increased time spent at the center of the open field, reduced grooming behaviors in the EPM test, and reduced time spent immobile in the forced swimming test (FST). It also blocked the decrease in neuropeptide Y (NPY) and the increase in corticotrophin-releasing factor (CRF) expression in the hypothalamus. This is the first study to determine that THP exerts pronounced anxiolytic-like and antidepressant effects on the development of the behavioral and biochemical symptoms associated with PTSD, indicating its prophylactic potential. Thus, THP reversed several behavioral impairments triggered by the traumatic stress of SPS and is a potential non-invasive therapeutic intervention for PTSD.
Collapse
Affiliation(s)
- Bombi Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Bongjun Sur
- The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Mijung Yeom
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Insop Shim
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea ; The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Hyejung Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea ; The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Dae-Hyun Hahm
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea ; The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| |
Collapse
|
38
|
Han F, Ding J, Shi Y. Expression of amygdala mineralocorticoid receptor and glucocorticoid receptor in the single-prolonged stress rats. BMC Neurosci 2014; 15:77. [PMID: 24947040 PMCID: PMC4074391 DOI: 10.1186/1471-2202-15-77] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/09/2014] [Indexed: 01/10/2023] Open
Abstract
Background Post-traumatic stress disorder (PTSD) is an anxious disorder associated with low levels of corticosterone and enhanced negative feedback of the hypothalamic–pituitary–adrenal (HPA) axis. Previous studies showed that the amygdala not only has an excitatory effect on the HPA axis but also plays a key role in fear-related behaviors. Coticosterone exert actions through binding to the mineralocorticoid (MR) and glucocorticoid receptor (GR), which are abundant in the amygdala. In our previous study, down-regulation of MR and GR in the hippocampus of PTSD rats was found. But the roles of MR and GR in the amygdala of PTSD rats is incompletely understood. Results wistar rats were divided into 1 d, 7 d, 14 d groups after single prolonged stress (SPS) and control group. SPS is a reliable animal model of PTSD. Open field test (OF) and elevated plus maze tests (EPM) were performed to examine fear-related behaviors. Morphological changes of the ultrastructure of the amygdala neurons were assessed by transmission electron microscopy (TEM). Dual-immunofluorescence histochemistry was used to determined subcellular distribution and colocalization of MR- and GR-ir. Protein and mRNA of MR and GR was examined by western blotting and RT-PCR. OF and EPM showed enhanced fear in SPS rats. Abnormal neuronal morphology was discovered in the amygdala of SPS rats. The expression of MR- and GR-ir intensity, mRNA and protein within the amygdala decreased after SPS at 1 day, and then gradually recovered by 14 days, although the degree of decrease and recovery were different amongst techniques. We found no change in the MR/GR ratio at 3 levels of the amygdala. But more cytoplasmic distribution and decreased colocalization of MR- and GR-ir were observed in the amygdala after 7 days of SPS. Conclusion These data suggest that change of MR and GR in the amygdala are involved in the mechanisms of fear in PTSD.
Collapse
Affiliation(s)
| | | | - Yuxiu Shi
- PTSD lab, Department of Histology and Embryology, Institute of pathology and Pathophysiology, China Medical University, Shenyang 110001, China.
| |
Collapse
|
39
|
Pagliaccio D, Luby JL, Bogdan R, Agrawal A, Gaffrey MS, Belden AC, Botteron KN, Harms MP, Barch DM. Stress-system genes and life stress predict cortisol levels and amygdala and hippocampal volumes in children. Neuropsychopharmacology 2014; 39:1245-53. [PMID: 24304824 PMCID: PMC3957120 DOI: 10.1038/npp.2013.327] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 10/25/2013] [Accepted: 11/14/2013] [Indexed: 02/08/2023]
Abstract
Depression has been linked to increased cortisol reactivity and differences in limbic brain volumes, yet the mechanisms underlying these alterations are unclear. One main hypothesis is that stress causes these effects. This is supported by animal studies showing that chronic stress or glucocorticoid administration can lead to alterations in hippocampal and amygdala structures. Relatedly, life stress is cited as one of the major risk factors for depression and candidate gene studies have related variation in stress-system genes to increased prevalence and severity of depression. The present study tested the hypothesis that genetic profile scores combining variance across 10 single nucleotide polymorphisms from four stress-system genes (CRHR1, NR3C2, NR3C1, and FKBP5) and early life stress would predict increases in cortisol levels during laboratory stressors in 120 preschool-age children (3-5 years old), as well as hippocampal and amygdala volumes assessed with MRI in these same children at school age (7-12 years old). We found that stress-system genetic profile scores positively predicted cortisol levels while the number of stressful/traumatic life events experienced by 3-5 years old negatively predicted cortisol levels. The interaction of genetic profile scores and early life stress predicted left hippocampal and left amygdala volumes. Cortisol partially mediated the effects of genetic variation and life stress on limbic brain volumes, particularly on left amygdala volume. These results suggest that stress-related genetic and early environmental factors contribute to variation in stress cortisol reactivity and limbic brain volumes in children, phenotypes associated with depression in adulthood.
Collapse
Affiliation(s)
- David Pagliaccio
- Program in Neuroscience, Washington University in St Louis, St Louis, MO, USA,The Program in Neuroscience, Washington University in St Louis, Campus Box 1125, One Brookings Drive, St Louis, MO 63130, USA, Tel: +9146450103, Fax: +913149358790, E-mail: or
| | - Joan L Luby
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Ryan Bogdan
- Program in Neuroscience, Washington University in St Louis, St Louis, MO, USA,Department of Psychology, Washington University in St Louis, St Louis, MO, USA
| | - Arpana Agrawal
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Michael S Gaffrey
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Andrew C Belden
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Kelly N Botteron
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA,Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Michael P Harms
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Deanna M Barch
- Program in Neuroscience, Washington University in St Louis, St Louis, MO, USA,Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA,Department of Psychology, Washington University in St Louis, St Louis, MO, USA,Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| |
Collapse
|
40
|
Stress, anxiety, and dendritic spines: What are the connections? Neuroscience 2013; 251:108-19. [DOI: 10.1016/j.neuroscience.2012.04.021] [Citation(s) in RCA: 195] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Revised: 04/10/2012] [Accepted: 04/11/2012] [Indexed: 01/11/2023]
|
41
|
Chao L, Weiner M, Neylan T. Regional cerebral volumes in veterans with current versus remitted posttraumatic stress disorder. Psychiatry Res 2013; 213:193-201. [PMID: 23816189 DOI: 10.1016/j.pscychresns.2013.03.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 02/21/2013] [Accepted: 03/13/2013] [Indexed: 11/28/2022]
Abstract
We previously reported that hippocampal volume was associated with current, but not lifetime posttraumatic stress disorder (PTSD) symptom severity. In the present study, we test the hypothesis that like the hippocampus, the volumes of other brain regions previously implicated in PTSD, are also negatively related to current, but not lifetime PTSD symptom severity. One hundred ninety-one veterans underwent structural magnetic resonance imaging (MRI) on a 4T scanner. Seventy-five veterans were trauma unexposed, 43 were trauma exposed without PTSD, 39 were trauma exposed with current PTSD, and 34 were trauma exposed veterans with remitted PTSD. Hippocampal, amygdala, rostral and caudal anterior cingulate, insula, and corpus callosum volumes, quantified with Freesurfer version 4.5, were analyzed by group using multivariate analysis of covariance. Veterans with PTSD had smaller hippocampal, caudal anterior cingulate, insula, and corpus callosum volumes than the unexposed controls (p≤0.009); smaller hippocampal, caudal anterior cingulate, insula (p≤0.009) and marginally smaller corpus callosum (p=0.06) than veterans with remitted PTSD; and smaller hippocampal and caudal anterior cingulate volumes than veterans without PTSD (p≤0.04). In contrast, there was no significant volume differences between veterans with remitted PTSD compared to those without PTSD or unexposed controls. The finding that current but not lifetime PTSD accounts for the volumes of multiple brain regions suggests that either smaller brain volume is a vulnerability factor that impedes recovery from PTSD or that recovery from PTSD is accompanied by a wide-spread restoration of brain tissue.
Collapse
Affiliation(s)
- Linda Chao
- Department of Radiology, University of California, San Francisco 94121, USA.
| | | | | |
Collapse
|
42
|
FGF2 blocks PTSD symptoms via an astrocyte-based mechanism. Behav Brain Res 2013; 256:472-80. [PMID: 24013012 DOI: 10.1016/j.bbr.2013.08.048] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 08/23/2013] [Accepted: 08/28/2013] [Indexed: 12/20/2022]
Abstract
Although posttraumatic stress disorder (PTSD) is characterized by traumatic memories or experiences and increased arousal, which can be partly alleviated by antidepressants, the underlying cellular mechanisms are not fully understood. As emerging studies have focused on the critical role of astrocytes in pathological mood disorders, we hypothesized that several 'astrocyte-related' mechanisms underlying PTSD exist. In the present study, using the single prolonged stress (SPS) model, we investigated the effects of intraperitoneal FGF2 on SPS-induced PTSD behavior response as well as the astrocytic activation after FGF2 administration in SPS rats. Behavioral data showed that intraperitoneal FGF2 inhibited SPS-induced hyperarousal and anxiety behavior; however, immunohistochemistry showed that SPS-induced astrocytic inhibition was activated by intraperitoneal FGF2. Quantitative western blotting showed that intraperitoneal FGF2 up-regulated glial fibrillary acidic protein (GFAP), but not NeuN, expression in the hippocampus. We suggest that intraperitoneal FGF2 could block the SPS-induced fear response and anxiety behavior in PTSD via astrocyte-based but not neuron-based mechanisms.
Collapse
|
43
|
Ganon-Elazar E, Akirav I. Cannabinoids and traumatic stress modulation of contextual fear extinction and GR expression in the amygdala-hippocampal-prefrontal circuit. Psychoneuroendocrinology 2013; 38:1675-87. [PMID: 23433741 DOI: 10.1016/j.psyneuen.2013.01.014] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 01/02/2013] [Accepted: 01/30/2013] [Indexed: 11/19/2022]
Abstract
Considerable evidence suggests that cannabinoids modulate the behavioral and physiological response to stressful events. We have recently shown that activating the cannabinoid system using the CB1/CB2 receptor agonist WIN55,212-2 (WIN) in proximity to exposure to single-prolonged stress (SPS), a rat model of emotional trauma, prevented the stress-induced enhancement of acoustic startle response, the impairment in avoidance extinction and the enhanced negative feedback on the hypothalamic-pituitary-adrenal (HPA) axis (Ganon-Elazar and Akirav, 2012). Some of the effects were found to be mediated by CB1 receptors in the basolateral amygdala (BLA). Here we examined whether cannabinoid receptor activation in a putative brain circuit that includes the BLA, hippocampus and prefrontal cortex (PFC), could prevent the effects of traumatic stress on contextual fear extinction and alterations in glucocorticoid receptor (GR) protein levels. We found that: (i) SPS impaired contextual fear extinction tested one week after trauma exposure and that WIN prevented the stress-induced impairment of extinction when microinjected immediately after trauma exposure into the BLA or hippocampus (5 μg), but not when microinjected into the PFC, (ii) the ameliorating effects of WIN on contextual extinction were prevented by blocking GRs in the BLA and hippocampus, and (iii) SPS up regulated GRs in the BLA, PFC and hippocampus and systemic WIN administration (0.5 mg/kg) after trauma exposure normalized GR levels in the BLA and hippocampus, but not in the PFC. Cannabinoid receptor activation in the aftermath of trauma exposure may regulate the emotional response to the trauma and prevent stress-induced impairment of extinction and GR up regulation through the mediation of CB1 receptors in the BLA and hippocampus. Taken together, the findings suggest that the interaction between the cannabinoid and glucocorticoid systems is crucial in the modulation of emotional trauma.
Collapse
MESH Headings
- Amygdala/drug effects
- Amygdala/physiopathology
- Animals
- Benzoxazines/pharmacology
- Cannabinoids/pharmacology
- Electroshock
- Extinction, Psychological/drug effects
- Extinction, Psychological/physiology
- Fear/drug effects
- Fear/physiology
- Freezing Reaction, Cataleptic/drug effects
- Freezing Reaction, Cataleptic/physiology
- Hippocampus/drug effects
- Hippocampus/physiopathology
- Hypothalamo-Hypophyseal System/physiopathology
- Male
- Microinjections
- Mifepristone/pharmacology
- Models, Psychological
- Morpholines/pharmacology
- Naphthalenes/pharmacology
- Piperidines/pharmacology
- Pituitary-Adrenal System/physiopathology
- Prefrontal Cortex/drug effects
- Prefrontal Cortex/physiopathology
- Pyrazoles/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptor, Cannabinoid, CB1/drug effects
- Receptor, Cannabinoid, CB1/physiology
- Receptor, Cannabinoid, CB2/drug effects
- Receptor, Cannabinoid, CB2/physiology
- Receptors, Glucocorticoid/antagonists & inhibitors
- Receptors, Glucocorticoid/biosynthesis
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/physiology
- Stress, Psychological/physiopathology
Collapse
Affiliation(s)
- Eti Ganon-Elazar
- Department of Psychology, University of Haifa, Haifa 31905, Israel
| | | |
Collapse
|
44
|
Maroun M, Ioannides PJ, Bergman KL, Kavushansky A, Holmes A, Wellman CL. Fear extinction deficits following acute stress associate with increased spine density and dendritic retraction in basolateral amygdala neurons. Eur J Neurosci 2013; 38:2611-20. [PMID: 23714419 PMCID: PMC3773716 DOI: 10.1111/ejn.12259] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 04/16/2013] [Accepted: 04/18/2013] [Indexed: 11/28/2022]
Abstract
Stress-sensitive psychopathologies such as post-traumatic stress disorder are characterized by deficits in fear extinction and dysfunction of corticolimbic circuits mediating extinction. Chronic stress facilitates fear conditioning, impairs extinction, and produces dendritic proliferation in the basolateral amygdala (BLA), a critical site of plasticity for extinction. Acute stress impairs extinction, alters plasticity in the medial prefrontal cortex-to-BLA circuit, and causes dendritic retraction in the medial prefrontal cortex. Here, we examined extinction learning and basolateral amygdala pyramidal neuron morphology in adult male rats following a single elevated platform stress. Acute stress impaired extinction acquisition and memory, and produced dendritic retraction and increased mushroom spine density in basolateral amygdala neurons in the right hemisphere. Unexpectedly, irrespective of stress, rats that underwent fear and extinction testing showed basolateral amygdala dendritic retraction and altered spine density relative to non-conditioned rats, particularly in the left hemisphere. Thus, extinction deficits produced by acute stress are associated with increased spine density and dendritic retraction in basolateral amygdala pyramidal neurons. Furthermore, the finding that conditioning and extinction as such was sufficient to alter basolateral amygdala morphology and spine density illustrates the sensitivity of basolateral amygdala morphology to behavioral manipulation. These findings may have implications for elucidating the role of the amygdala in the pathophysiology of stress-related disorders.
Collapse
Affiliation(s)
- Mouna Maroun
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel.
| | | | | | | | | | | |
Collapse
|
45
|
He YQ, Chen Q, Ji L, Wang ZG, Bai ZH, Stephens RL, Yang M. PKCγ receptor mediates visceral nociception and hyperalgesia following exposure to PTSD-like stress in the spinal cord of rats. Mol Pain 2013; 9:35. [PMID: 23837410 PMCID: PMC3751645 DOI: 10.1186/1744-8069-9-35] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 07/04/2013] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Clinical studies indicate that patients with post-traumatic stress disorder (PTSD) frequently share comorbidity with numerous chronic pain conditions. However, the sustained effects of PTSD-like stress over time on visceral nociception and hyperalgesia have been rarely studied, and the underlying mechanisms of stress-induced modulation of visceral hyperalgesia remain elusive. The purpose of this study was to investigate the characterization of visceral nociception and hyperalgesia over time in rats exposed to PTSD-like stress, and to explore the potential role of protein kinase C gamma (PKCγ) in mediating visceral hyperalgesia following exposure to PTSD-like stress. RESULTS On day 1, the rats exposed to single-prolonged stress (SPS, an established animal model for PTSD) exhibited an analgesic response and its visceromotor response (VMR) to graded colorectal distention (CRD) at 40 and 60 mmHg was reduced compared with the control group (all P < 0.05). On day 6, the VMR returned to the baseline value. However, as early as 7 days after SPS, VMR dramatically increased compared with its baseline value and that in the controls (all P < 0.001) and this increase persisted for 28 days, with the peak on day 9. Abdominal withdrawal reflex (AWR) scores were higher in SPS rats than in controls on days 7, 9, 14, 21 and 28 (all P < 0.001). Intrathecal administration of GF109203X (an inhibitor of PKC gamma), attenuated the SPS-induced increase in both VMR and AWR scores on days 7, 14, 21 and 28 (all P < 0.05). PKCγ protein expression determined by immunofluorescence was reduced in the spinal cord within 3 days after the exposure to SPS (P < 0.01), which returned to normal levels between days 4 and 6, and significantly increased from day 7, and this increase was maintained on days 14, 21, and 28 (all P < 0.001), with the peak on day 9. In addition, Western blotting showed a consistent trend in the changes of PKCγ protein expression. CONCLUSIONS The modified SPS alters visceral sensitivity to CRD, and contributes to the maintenance of visceral hyperalgesia, which is associated with enhanced PKCγ expression in the spinal cord. Functional blockade of the PKCγ receptors attenuates SPS-induced visceral hyperalgesia. Thus, the present study identifies a specific molecular mechanism for visceral hyperalgesia which may pave the way for novel therapeutic strategies for PTSD-like conditions.
Collapse
|
46
|
Stress-induced lipocalin-2 controls dendritic spine formation and neuronal activity in the amygdala. PLoS One 2013; 8:e61046. [PMID: 23593384 PMCID: PMC3621903 DOI: 10.1371/journal.pone.0061046] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 03/05/2013] [Indexed: 12/11/2022] Open
Abstract
Behavioural adaptation to psychological stress is dependent on neuronal plasticity and dysfunction at this cellular level may underlie the pathogenesis of affective disorders such as depression and post-traumatic stress disorder. Taking advantage of genome-wide microarray assay, we performed detailed studies of stress-affected transcripts in the amygdala – an area which forms part of the innate fear circuit in mammals. Having previously demonstrated the role of lipocalin-2 (Lcn-2) in promoting stress-induced changes in dendritic spine morphology/function and neuronal excitability in the mouse hippocampus, we show here that the Lcn-2 gene is one of the most highly upregulated transcripts detected by microarray analysis in the amygdala after acute restraint-induced psychological stress. This is associated with increased Lcn-2 protein synthesis, which is found on immunohistochemistry to be predominantly localised to neurons. Stress-naïve Lcn-2−/− mice show a higher spine density in the basolateral amygdala and a 2-fold higher rate of neuronal firing rate compared to wild-type mice. Unlike their wild-type counterparts, Lcn-2−/− mice did not show an increase in dendritic spine density in response to stress but did show a distinct pattern of spine morphology. Thus, amygdala-specific neuronal responses to Lcn-2 may represent a mechanism for behavioural adaptation to psychological stress.
Collapse
|
47
|
Serotonergic innervation and serotonin receptor expression of NPY-producing neurons in the rat lateral and basolateral amygdaloid nuclei. Brain Struct Funct 2012; 218:421-35. [PMID: 22527118 PMCID: PMC3580143 DOI: 10.1007/s00429-012-0406-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Accepted: 03/13/2012] [Indexed: 01/28/2023]
Abstract
Pharmacobehavioral studies in experimental animals, and imaging studies in humans, indicate that serotonergic transmission in the amygdala plays a key role in emotional processing, especially for anxiety-related stimuli. The lateral and basolateral amygdaloid nuclei receive a dense serotonergic innervation in all species studied to date. We investigated interrelations between serotonergic afferents and neuropeptide Y (NPY)-producing neurons, which are a subpopulation of inhibitory interneurons in the rat lateral and basolateral nuclei with particularly strong anxiolytic properties. Dual light microscopic immunolabeling showed numerous appositions of serotonergic afferents on NPY-immunoreactive somata. Using electron microscopy, direct membrane appositions and synaptic contacts between serotonin-containing axon terminals and NPY-immunoreactive cellular profiles were unequivocally established. Double in situ hybridization documented that more than 50 %, and about 30–40 % of NPY mRNA-producing neurons, co-expressed inhibitory 5-HT1A and excitatory 5-HT2C mRNA receptor subtype mRNA, respectively, in both nuclei with no gender differences. Triple in situ hybridization showed that individual NPY mRNA-producing interneurons co-express both 5-HT1A and 5-HT2C mRNAs. Co-expression of NPY and 5-HT3 mRNA was not observed. The results demonstrate that serotonergic afferents provide substantial innervation of NPY-producing neurons in the rat lateral and basolateral amygdaloid nuclei. Studies of serotonin receptor subtype co-expression indicate a differential impact of the serotonergic innervation on this small, but important, population of anxiolytic interneurons, and provide the basis for future studies of the circuitry underlying serotonergic modulation of emotional stimulus processing in the amygdala.
Collapse
|
48
|
Brain–spinal cord neural circuits controlling male sexual function and behavior. Neurosci Res 2012; 72:103-16. [DOI: 10.1016/j.neures.2011.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 10/14/2011] [Accepted: 10/25/2011] [Indexed: 01/10/2023]
|
49
|
Liberzon I, Knox D. Expanding our understanding of neurobiological mechanisms of resilience by using animal models. Neuropsychopharmacology 2012; 37:317-8. [PMID: 22157946 PMCID: PMC3242323 DOI: 10.1038/npp.2011.259] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Israel Liberzon
- Department of Psychiatry, University of Michigan, Ann Arbor, MI 48109-5765, USA.
| | - Dayan Knox
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
50
|
Ultrahigh voltage electron microscopy links neuroanatomy and neuroscience/neuroendocrinology. ANATOMY RESEARCH INTERNATIONAL 2011; 2012:948704. [PMID: 22567316 PMCID: PMC3335508 DOI: 10.1155/2012/948704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 09/15/2011] [Indexed: 11/18/2022]
Abstract
The three-dimensional (3D) analysis of anatomical ultrastructures is extremely important in most fields of biological research. Although it is very difficult to perform 3D image analysis on exact serial sets of ultrathin sections, 3D reconstruction from serial ultrathin sections can generally be used to obtain 3D information. However, this technique can only be applied to small areas of a specimen because of technical and physical difficulties. We used ultrahigh voltage electron microscopy (UHVEM) to overcome these difficulties and to study the chemical neuroanatomy of 3D ultrastructures. This methodology, which links UHVEM and light microscopy, is a useful and powerful tool for studying molecular and/or chemical neuroanatomy at the ultrastructural level.
Collapse
|