1
|
Wellington NJ, Boųcas AP, Lagopoulos J, Quigley BL, Kuballa AV. Molecular pathways of ketamine: A systematic review of immediate and sustained effects on PTSD. Psychopharmacology (Berl) 2025; 242:1197-1243. [PMID: 40097854 DOI: 10.1007/s00213-025-06756-4] [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: 11/30/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025]
Abstract
RATIONALE Existing studies predominantly focus on the molecular and neurobiological mechanisms underlying Ketamine's acute treatment effects on post-traumatic stress disorder (PTSD). This emphasis has largely overlooked its sustained therapeutic effects, which hold significant potential for the development of targeted interventions. OBJECTIVES This systematic review examines the pharmacokinetic and pharmacodynamic effects of ketamine on PTSD, differentiating between immediate and sustained molecular effects. METHOD A comprehensive search across databases (Web of Science, Scopus, Global Health, PubMed) and grey literature yielded 317 articles, where 29 studies met the inclusion criteria. These studies included preclinical models and clinical trials, through neurotransmitter regulation, gene expression, synaptic plasticity, and neural pathways (PROSPERO ID: CRD42024582874). RESULTS We found accumulating evidence that the immediate effects of ketamine, which involve changes in GABA, glutamate, and glutamine levels, trigger the re-regulation of BDNF, enhancing synaptic plasticity via pathways such as TrkB and PSD-95. Other molecular influences also include c-Fos, GSK-3, HDAC, HCN1, and the modulation of hormones like CHR and ACTH, alongside immune responses (IL-6, IL-1β, TNF-α). Sustained effects arise from neurotransmitter remodulations and involve prolonged changes in gene expression. These include mTOR-mediated BDNF expression, alterations in GSK-3β, FkBP5, GFAP, ERK phosphorylation, and epigenetic modifications (DNMT3, MeCP2, H3K27me3, mir-132, mir-206, HDAC). CONCLUSION These molecular changes promote long-term synaptic stability and re-regulation in key brain regions, contributing to prolonged therapeutic benefits. Understanding the sustained molecular and epigenetic mechanisms behind ketamine's effects is critical for developing safe and effective personalised treatments, potentially leading to more effective recovery.
Collapse
Affiliation(s)
- Nathan J Wellington
- National PTSD Research Centre, Thompson Institute, University of the Sunshine Coast (UniSC), Birtinya, QLD, Australia.
- School of Health, UniSC, Sippy Downs, QLD, Australia.
- Centre for Bioinnovation, UniSC, Sippy Downs, QLD, Australia.
- Sunshine Coast Hospital and Health Service, Sunshine Coast Health Institute, Birtinya, QLD, Australia.
| | - Ana P Boųcas
- National PTSD Research Centre, Thompson Institute, University of the Sunshine Coast (UniSC), Birtinya, QLD, Australia
| | - Jim Lagopoulos
- Thompson Brain and Mind Healthcare, Maroochydore, QLD, Australia
| | - Bonnie L Quigley
- National PTSD Research Centre, Thompson Institute, University of the Sunshine Coast (UniSC), Birtinya, QLD, Australia
- Centre for Bioinnovation, UniSC, Sippy Downs, QLD, Australia
- Sunshine Coast Hospital and Health Service, Sunshine Coast Health Institute, Birtinya, QLD, Australia
| | - Anna V Kuballa
- School of Health, UniSC, Sippy Downs, QLD, Australia
- Centre for Bioinnovation, UniSC, Sippy Downs, QLD, Australia
- Sunshine Coast Hospital and Health Service, Sunshine Coast Health Institute, Birtinya, QLD, Australia
| |
Collapse
|
2
|
Liu L, Liu Y, Li R, Teng Y, Zhao S, Chen J, Li C, Hu X, Sun L. Identification of Critical Signature in Post-Traumatic Stress Disorder Using Bioinformatics Analysis and in Vitro Analyses. Brain Behav 2025; 15:e70243. [PMID: 39829117 PMCID: PMC11743987 DOI: 10.1002/brb3.70243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 10/07/2024] [Accepted: 12/14/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Post-traumatic stress disorder (PTSD) is a complex psychiatric condition that emerges following exposure to trauma and significantly affects daily functioning. Current research is focused on identifying effective treatments for PTSD. Advances in bioinformatics provide opportunities to elucidate the underlying mechanisms of PTSD. METHODS RNA sequencing (RNA-seq) datasets were retrieved from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were identified using GEO2R. Weighted gene co-expression network analysis (WGCNA) was employed to examine gene correlation patterns. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed for functional annotation and enrichment analysis, respectively. The MCODE plugin in Cytoscape software was utilized to analyze the protein-protein interaction (PPI) network. Anxiety and depression in a mice stress model were assessed using the open-field test (OFT), elevated plus maze test (EPMT), and forced swimming test (FST). Real-time quantitative PCR (qRT-PCR) was conducted to validate key genes in stress-exposed models. RESULTS A total of 157 common upregulated DEGs and 53 common downregulated DEGs were identified in the amygdala (AMY) and the hippocampus (HIP). Notably enriched pathways included neuroactive ligand-receptor interaction, mechanistic target of rapamycin (mTOR) signaling pathway, nicotine addiction, and dopaminergic synapse. The PPI network identified four hub genes, with key pathways associated with nicotine addiction and dopaminergic synapse. qRT-PCR validation confirmed that the expression trends of these four genes were consistent with microarray data. Behavioral tests (OFT, EMPT, and FST) revealed significant changes. CONCLUSION This study utilized bioinformatics and in vitro experiments to identify genes and pathways potentially crucial for PTSD development. Key genes were validated in a mouse model, providing insights into potential target genes for PTSD treatment.
Collapse
Affiliation(s)
- Lifen Liu
- School of PsychologyShandong Second Medical UniversityWeifangShandongPeople's Republic of China
| | - Yang Liu
- Department of Psychological Counseling CenterWeifang Mental Health Center, Weian Road, Weifang, ShandongChina
| | - Rui Li
- School of PsychologyShandong Second Medical UniversityWeifangShandongPeople's Republic of China
| | - Yue Teng
- Faculty of PsychologySouthwest University, ChongqingChina
| | - Shuyang Zhao
- Department of Bioscience and TechnologyShandong Second Medical UniversityWeifangShandongPeople's Republic of China
| | - Jinhong Chen
- College of Extended EducationShandong Second Medical UniversityWeifangShandongPeople's Republic of China
| | - Changjiang Li
- School of PsychologyShandong Second Medical UniversityWeifangShandongPeople's Republic of China
| | - Xinyu Hu
- CAS Key Laboratory of Mental Health, Institute of PsychologyChinese Academy of SciencesBeijingPeople's Republic of China
- Department of PsychologyUniversity of Chinese Academy of SciencesBeijingPeople's Republic of China
| | - Lin Sun
- School of PsychologyShandong Second Medical UniversityWeifangShandongPeople's Republic of China
- Management Committee of Shanting Economic Development ZoneZaozhuangShandongPeople's Republic of China
- Department of NeurosurgeryShanting District People's HospitalZaozhuangShandongPeople's Republic of China
| |
Collapse
|
3
|
Xie G, Qin Y, Wu N, Han X, Li J. Single-Nucleus Transcriptome Profiling from the Hippocampus of a PTSD Mouse Model and CBD-Treated Cohorts. Genes (Basel) 2024; 15:519. [PMID: 38674453 PMCID: PMC11050643 DOI: 10.3390/genes15040519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is the most common psychiatric disorder after a catastrophic event; however, the efficacious treatment options remain insufficient. Increasing evidence suggests that cannabidiol (CBD) exhibits optimal therapeutic effects for treating PTSD. To elucidate the cell-type-specific transcriptomic pathology of PTSD and the mechanisms of CBD against this disease, we conducted single-nucleus RNA sequencing (snRNA-seq) in the hippocampus of PTSD-modeled mice and CBD-treated cohorts. We constructed a mouse model by adding electric foot shocks following exposure to single prolonged stress (SPS+S) and tested the freezing time, anxiety-like behavior, and cognitive behavior. CBD was administrated before every behavioral test. The PTSD-modeled mice displayed behaviors resembling those of PTSD in all behavioral tests, and CBD treatment alleviated all of these PTSD-like behaviors (n = 8/group). Three mice with representative behavioral phenotypes were selected from each group for snRNA-seq 15 days after the SPS+S. We primarily focused on the excitatory neurons (ExNs) and inhibitory neurons (InNs), which accounted for 68.4% of the total cell annotations. A total of 88 differentially upregulated genes and 305 differentially downregulated genes were found in the PTSD mice, which were found to exhibit significant alterations in pathways and biological processes associated with fear response, synaptic communication, protein synthesis, oxidative phosphorylation, and oxidative stress response. A total of 63 overlapping genes in InNs were identified as key genes for CBD in the treatment of PTSD. Subsequent Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses revealed that the anti-PTSD effect of CBD was related to the regulation of protein synthesis, oxidative phosphorylation, oxidative stress response, and fear response. Furthermore, gene set enrichment analysis (GSEA) revealed that CBD also enhanced retrograde endocannabinoid signaling in ExNs, which was found to be suppressed in the PTSD group. Our research may provide a potential explanation for the pathogenesis of PTSD and facilitate the discovery of novel therapeutic targets for drug development. Moreover, it may shed light on the therapeutic mechanisms of CBD.
Collapse
Affiliation(s)
| | | | | | - Xiao Han
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (G.X.); (Y.Q.); (N.W.); (J.L.)
| | | |
Collapse
|
4
|
Wojick JA, Paranjapye A, Chiu JK, Mahmood M, Oswell C, Kimmey BA, Wooldridge LM, McCall NM, Han A, Ejoh LL, Chehimi SN, Crist RC, Reiner BC, Korb E, Corder G. A nociceptive amygdala-striatal pathway for chronic pain aversion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.12.579947. [PMID: 38405972 PMCID: PMC10888915 DOI: 10.1101/2024.02.12.579947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The basolateral amygdala (BLA) is essential for assigning positive or negative valence to sensory stimuli. Noxious stimuli that cause pain are encoded by an ensemble of nociceptive BLA projection neurons (BLAnoci ensemble). However, the role of the BLAnoci ensemble in mediating behavior changes and the molecular signatures and downstream targets distinguishing this ensemble remain poorly understood. Here, we show that the same BLAnoci ensemble neurons are required for both acute and chronic neuropathic pain behavior. Using single nucleus RNA-sequencing, we characterized the effect of acute and chronic pain on the BLA and identified enrichment for genes with known functions in axonal and synaptic organization and pain perception. We thus examined the brain-wide targets of the BLAnoci ensemble and uncovered a previously undescribed nociceptive hotspot of the nucleus accumbens shell (NAcSh) that mirrors the stability and specificity of the BLAnoci ensemble and is recruited in chronic pain. Notably, BLAnoci ensemble axons transmit acute and neuropathic nociceptive information to the NAcSh, highlighting this nociceptive amygdala-striatal circuit as a unique pathway for affective-motivational responses across pain states.
Collapse
Affiliation(s)
- Jessica A. Wojick
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Alekh Paranjapye
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Juliann K. Chiu
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Malaika Mahmood
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Corinna Oswell
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Blake A. Kimmey
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lisa M. Wooldridge
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nora M. McCall
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alan Han
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lindsay L. Ejoh
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samar Nasser Chehimi
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard C. Crist
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin C. Reiner
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erica Korb
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gregory Corder
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
5
|
Núñez-Rios DL, Martínez-Magaña JJ, Nagamatsu ST, Krystal JH, Martínez-González KG, Giusti-Rodríguez P, Montalvo-Ortiz JL. Cross-Species Convergence of Brain Transcriptomic and Epigenomic Findings in Posttraumatic Stress Disorder: A Systematic Review. Complex Psychiatry 2023; 9:100-118. [PMID: 37404872 PMCID: PMC10315001 DOI: 10.1159/000529536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 01/31/2023] [Indexed: 08/05/2023] Open
Abstract
Introduction Posttraumatic stress disorder (PTSD) is a complex multifactorial disorder influenced by the interaction of genetic and environmental factors. Analyses of epigenomic and transcriptomic modifications may help to dissect the biological factors underlying the gene-environment interplay in PTSD. To date, most human PTSD epigenetics studies have used peripheral tissue, and these findings have complex and poorly understood relationships to brain alterations. Studies examining brain tissue may help characterize the brain-specific transcriptomic and epigenomic profiles of PTSD. In this review, we compiled and integrated brain-specific molecular findings of PTSD from humans and animals. Methods A systematic literature search according to the PRISMA criteria was performed to identify transcriptomic and epigenomic studies of PTSD, focusing on brain tissue from human postmortem samples or animal-stress paradigms. Results Gene- and pathway-level convergence analyses revealed PTSD-dysregulated genes and biological pathways across brain regions and species. A total of 243 genes converged across species, with 17 of them significantly enriched for PTSD. Chemical synaptic transmission and signaling by G-protein-coupled receptors were consistently enriched across omics and species. Discussion Our findings point out dysregulated genes highly replicated across PTSD studies in humans and animal models and suggest a potential role for the corticotropin-releasing hormone/orexin pathway in PTSD's pathophysiology. Further, we highlight current knowledge gaps and limitations and recommend future directions to address them.
Collapse
Affiliation(s)
- Diana Leandra Núñez-Rios
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center for Posttraumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | - José Jaime Martínez-Magaña
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center for Posttraumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | - Sheila Tiemi Nagamatsu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center for Posttraumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | - John H. Krystal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center for Posttraumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | | | - Paola Giusti-Rodríguez
- Department of Psychiatry, University of Florida College of Medicine, Gainesville, FL, USA
| | - Janitza L. Montalvo-Ortiz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center for Posttraumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| |
Collapse
|
6
|
Zhou D, Long C, Shao Y, Li F, Sun W, Zheng Z, Wang X, Huang Y, Pan F, Chen G, Guo Y, Huang Y. Integrated Metabolomics and Proteomics Analysis of Urine in a Mouse Model of Posttraumatic Stress Disorder. Front Neurosci 2022; 16:828382. [PMID: 35360173 PMCID: PMC8963102 DOI: 10.3389/fnins.2022.828382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/23/2022] [Indexed: 11/23/2022] Open
Abstract
Posttraumatic stress disorder (PTSD) is a serious stress disorder that occurs in individuals who have experienced major traumatic events. The underlying pathological mechanisms of PTSD are complex, and the related predisposing factors are still not fully understood. In this study, label-free quantitative proteomics and untargeted metabolomics were used to comprehensively characterize changes in a PTSD mice model. Differential expression analysis showed that 12 metabolites and 27 proteins were significantly differentially expressed between the two groups. Bioinformatics analysis revealed that the differentiated proteins were mostly enriched in: small molecule binding, transporter activity, extracellular region, extracellular space, endopeptidase activity, zymogen activation, hydrolase activity, proteolysis, peptidase activity, sodium channel regulator activity. The differentially expressed metabolites were mainly enriched in Pyrimidine metabolism, D-Glutamine and D-glutamate metabolism, Alanine, aspartate and glutamate metabolism, Arginine biosynthesis, Glutathione metabolism, Arginine, and proline metabolism. These results expand the existing understanding of the molecular basis of the pathogenesis and progression of PTSD, and also suggest a new direction for potential therapeutic targets of PTSD. Therefore, the combination of urine proteomics and metabolomics explores a new approach for the study of the underlying pathological mechanisms of PTSD.
Collapse
Affiliation(s)
- Daxue Zhou
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Chengyan Long
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Yan Shao
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Fei Li
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Wei Sun
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Zihan Zheng
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Xiaoyang Wang
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Yiwei Huang
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Feng Pan
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Gang Chen
- Biomedical Analysis Center, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
- Chongqing Key Laboratory of Cytomics, Chongqing, China
- *Correspondence: Gang Chen,
| | - Yanlei Guo
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Yanlei Guo,
| | - Yi Huang
- Biomedical Analysis Center, Army Medical University, Chongqing, China
- Yi Huang,
| |
Collapse
|
7
|
Katrinli S, Maihofer AX, Wani AH, Pfeiffer JR, Ketema E, Ratanatharathorn A, Baker DG, Boks MP, Geuze E, Kessler RC, Risbrough VB, Rutten BPF, Stein MB, Ursano RJ, Vermetten E, Logue MW, Nievergelt CM, Smith AK, Uddin M. Epigenome-wide meta-analysis of PTSD symptom severity in three military cohorts implicates DNA methylation changes in genes involved in immune system and oxidative stress. Mol Psychiatry 2022; 27:1720-1728. [PMID: 34992238 PMCID: PMC9106882 DOI: 10.1038/s41380-021-01398-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/13/2021] [Accepted: 11/23/2021] [Indexed: 11/08/2022]
Abstract
Epigenetic factors modify the effects of environmental factors on biological outcomes. Identification of epigenetic changes that associate with PTSD is therefore a crucial step in deciphering mechanisms of risk and resilience. In this study, our goal is to identify epigenetic signatures associated with PTSD symptom severity (PTSS) and changes in PTSS over time, using whole blood DNA methylation (DNAm) data (MethylationEPIC BeadChip) of military personnel prior to and following combat deployment. A total of 429 subjects (858 samples across 2 time points) from three male military cohorts were included in the analyses. We conducted two different meta-analyses to answer two different scientific questions: one to identify a DNAm profile of PTSS using a random effects model including both time points for each subject, and the other to identify a DNAm profile of change in PTSS conditioned on pre-deployment DNAm. Four CpGs near four genes (F2R, CNPY2, BAIAP2L1, and TBXAS1) and 88 differentially methylated regions (DMRs) were associated with PTSS. Change in PTSS after deployment was associated with 15 DMRs, of those 2 DMRs near OTUD5 and ELF4 were also associated with PTSS. Notably, three PTSS-associated CpGs near F2R, BAIAP2L1 and TBXAS1 also showed nominal evidence of association with change in PTSS. This study, which identifies PTSD-associated changes in genes involved in oxidative stress and immune system, provides novel evidence that epigenetic differences are associated with PTSS.
Collapse
Affiliation(s)
- Seyma Katrinli
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
| | - Adam X Maihofer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Agaz H Wani
- Genomics Program, College of Public Health, University of South Florida, Tampa, FL, USA
| | - John R Pfeiffer
- Department of Psychology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA
| | - Elizabeth Ketema
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | | | - Dewleen G Baker
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Veterans Affairs Center of Excellence for Stress and Mental Health, San Diego, CA, USA
| | - Marco P Boks
- Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Elbert Geuze
- Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
- Brain Research and Innovation Centre, Netherlands Ministry of Defence, Utrecht, The Netherlands
| | - Ronald C Kessler
- Department of Health Care Policy, Harvard Medical School, Boston, MA, USA
| | - Victoria B Risbrough
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs Center of Excellence for Stress and Mental Health, San Diego, CA, USA
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, Maastricht University Medical Centre, School for Mental Health and Neuroscience, Maastricht, The Netherlands
| | - Murray B Stein
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- School of Public Health, University of California San Diego, La Jolla, CA, USA
| | - Robert J Ursano
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University School of Medicine, Bethesda, MD, USA
| | - Eric Vermetten
- Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Psychiatry, Leiden University Medical Center, ZA, Leiden, The Netherlands
- Research Center, Netherlands Defense Department, UT, AA Utrecht, The Netherlands
- Arq Psychotrauma Expert Group, XE, Diemen, The Netherlands
| | - Mark W Logue
- National Center for PTSD, Behavioral Science Division at VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Caroline M Nievergelt
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Veterans Affairs Center of Excellence for Stress and Mental Health, San Diego, CA, USA
| | - Alicia K Smith
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Monica Uddin
- Genomics Program, College of Public Health, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
8
|
Interleukine-17 Modulates Neurogenesis and Behavior Following Exposure to Trauma in Mice. Cells 2022; 11:cells11030343. [PMID: 35159158 PMCID: PMC8834196 DOI: 10.3390/cells11030343] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/11/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a psychiatric disorder accompanied by deficits in cognitive and social skills. Adult hippocampal neurogenesis is a lifelong phenomenon, with new neurons being formed in the granular cell layer of the dentate gyrus. Impaired neurogenesis is associated with multiple behavioral disorders including Alzheimer's disease and schizophrenia. PTSD patients often present hippocampal atrophy and animal models clearly present impaired neurogenesis. Previous studies on PTSD patients demonstrated elevated levels of Th17 cells and plasma levels of the pro-inflammatory cytokine interleukin-17A (IL-17A). Since IL-17A can impair neurogenesis in mice, we thus hypothesized that decreasing the serum levels of IL-17A will increase hippocampal neurogenesis and alleviate symptoms in a murine model of PTSD. Surprisingly, our results showed that attempting to neutralize IL-17A with an antibody resulted in increased serum levels of IL-17A, while targeting IL-23, the upstream regulator of IL-17, did lower the levels of IL-17A in trauma-exposed mice. As expected, increased levels of serum IL-17A (in anti-IL-17A treated mice) resulted in impaired neurogenesis, reflected by reduced number of proliferating Ki67+ neural progenitors and newly formed DCX+ neurons, which was correlated with increased expression of Hes1. Nevertheless, increased maturation was noted by the expression of Slit2 and Ache. In contrast, treatment with anti-IL-23 indeed resulted in increased neurogenesis. Behaviorally, both treatments did not affect trauma-related freezing behavior but did affect trauma-related social deficits. Unexpectedly, increased levels of serum IL-17A (in anti-IL-17A treated mice) prevented social deficits in trauma-exposed mice while anti-IL-23 exacerbated these deficits. We thus conclude that IL-17 is involved in regulating neurogenesis following exposure to stress but may be important in maintaining social behavior.
Collapse
|
9
|
Ou G, Li Q, Zhu L, Zhang Y, Liu Y, Li X, Du L, Jin Y. Intranasal Hydrogel of Armodafinil Hydroxypropyl-β-Cyclodextrin Inclusion Complex for the Treatment of Post-Traumatic Stress Disorder. Saudi Pharm J 2022; 30:265-282. [PMID: 35498223 PMCID: PMC9051980 DOI: 10.1016/j.jsps.2022.01.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 01/12/2022] [Indexed: 11/30/2022] Open
Abstract
Armodafinil inclusion complex (AIC) hydrogel was prepared and evaluated for its therapeutic effect on Post-traumatic Stress Disorder (PTSD). After computer simulation and physicochemical property investigation, the AIC was formed by lyophilization of armodafinil with ethanol as solvent and hydroxypropyl-beta-cyclodextrin (HP-β-CD) aqueous solution, in which the molar ratio of armodafinil and HP-β-CD was 1–1. The AIC encapsulation efficiency (EE) was (90.98 ± 3.72)% and loading efficiency (LE) was (13.95 ± 0.47)% and it increased the solubility of armodafinil in aqueous solution to 21 times. AIC hydrogel was prepared by adding AIC to methylcellulose (MC) hydrogels (3.33% w/v), and its higher drug release amount and slower release rate were testified by the in-vitro release assay and the rheological test. The mucosa irritation of AIC hydrogel was also evaluated. Healthy group, Model group, Sertraline group with 30 mg/kg sertraline gavage, AIC Hydrogel group with 20 mg/kg AIC hydrogel intranasal administration and AIC Aqueous Solution group with 20 mg/kg AIC aqueous solution gavage were set up for the treatment of mice with PTSD generated from foot shock method. Based on freezing response test in fear-conditioning box and open field test, compared with other groups, PTSD mice in AIC Hydrogel group showed significant improvement in behavioral parameters after 11 days of continuous drug administration and 5 days of drug withdrawal. After sacrifice, the plasma CORT level of PTSD mice in AIC Hydrogel group was elevated compared to Model group. Besides, the western blot (WB) of hippocampal brain-derived neurotrophic factor (BDNF) and amygdala dopamine transporter (DAT) immunohistochemistry sections indicated that AIC hydrogel had a protective effect on the brain tissue of PTSD mice. The brain targeting of intranasal administration was evaluated by fluorescence imaging characteristics of Cy7 hydrogel in the nasal route of drug administration, pharmacokinetics and in-vivo distribution of armodafinil. In short, AIC hydrogel is a promising formulation for the treatment of PTSD based on its high brain delivery and anti-PTSD effect.
Collapse
Affiliation(s)
- Ge Ou
- Medical School of Chinese PLA, Beijing 100853, China
- Pharmacy Department, Chinese PLA General Hospital, Beijing 100853, China
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Qian Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lin Zhu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yuanyuan Zhang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yijing Liu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xin Li
- Pharmacy Department, Chinese PLA General Hospital, Beijing 100853, China
- Corresponding authors at: Pharmacy Department, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China (X. Li). Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China (L. Du).
| | - Lina Du
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Corresponding authors at: Pharmacy Department, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China (X. Li). Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China (L. Du).
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| |
Collapse
|
10
|
Duan J, Li W, Li W, Liu Q, Tian M, Chen C, Zhang L, Zhang M. Quantitative Proteomics Analysis of Susceptibility and Resilience to Stress in a Rat model of PTSD. Behav Brain Res 2021; 415:113509. [PMID: 34358573 DOI: 10.1016/j.bbr.2021.113509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/31/2021] [Accepted: 08/01/2021] [Indexed: 11/16/2022]
Abstract
Posttraumatic stress disorder (PTSD) is a prevalent psychiatric disorder and sometimes deadly consequence of exposure to severe psychological trauma. However, there has been little known about the definitive molecular changes involved in determining vulnerability to PTSD. In the current study, we used proteomics to quantify protein changes in the hippocampus of foot shocks rats. A total of 6151 proteins were quantified and 97 proteins were significantly differentially expressed. The protein-protein interaction (PPI) analysis showed that oxidation-reduction process and glutathione homeostasis may be the potential key progress of being vulnerable to PTSD. The Gene Ontology analysis revealed enriched GO terms in the protein groups of Susceptible group vs Control group rats for glutathione binding,oligopeptide binding,modified amino acid binding,and glutathione transferase activity for their molecular functions (MF) and in the process of cellular response to toxic substance,xenobiotic metabolic process, urea metabolic process, and response to drug for the biological process (BP).SIGNIFICANCE:In recent years, there has been a growing interest in mental illness associated with trauma exposure. We found that stress susceptibility was associated with increased expression of arginase 1 indicated as a potential treatment target. Our results also proposed that carbonic anhydrases 3 could be a biomarker for the development of PTSD. This research helps to explain the potential molecular mechanism in PTSD and supply a new method for ameliorating PTSD.
Collapse
Affiliation(s)
- Jiao Duan
- Department of Anesthesiology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Wenjun Li
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, China
| | - Weiyan Li
- Department of Anesthesiology, Jinling Hospital, the first School of Clinical Medicine, Southern Medical University, China
| | - Qingzhen Liu
- Department of Anesthesiology, Jinling Hospital, the first School of Clinical Medicine, Southern Medical University, China
| | - Mi Tian
- Department of Anesthesiology, Jinling Hospital, the first School of Clinical Medicine, Southern Medical University, China
| | - Chunlong Chen
- Department of Anesthesiology, Jinling Hospital, the first School of Clinical Medicine, Southern Medical University, China
| | - Lidong Zhang
- Department of Anesthesiology, Jinling Hospital, the first School of Clinical Medicine, Southern Medical University, China
| | - Minhao Zhang
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, China.
| |
Collapse
|
11
|
BDNF Protein and BDNF mRNA Expression of the Medial Prefrontal Cortex, Amygdala, and Hippocampus during Situational Reminder in the PTSD Animal Model. Behav Neurol 2021; 2021:6657716. [PMID: 33763156 PMCID: PMC7964114 DOI: 10.1155/2021/6657716] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/09/2021] [Accepted: 02/22/2021] [Indexed: 11/17/2022] Open
Abstract
Whether BDNF protein and BDNF mRNA expression of the medial prefrontal cortex (mPFC; cingulated cortex area 1 (Cg1), prelimbic cortex (PrL), and infralimbic cortex (IL)), amygdala, and hippocampus (CA1, CA2, CA3, and dentate gyrus (DG)) was involved in fear of posttraumatic stress disorder (PTSD) during the situational reminder of traumatic memory remains uncertain. Footshock rats experienced an inescapable footshock (3 mA, 10 s), and later we have measured fear behavior for 2 min in the footshock environment on the situational reminder phase. In the final retrieval of situational reminder, BDNF protein and mRNA levels were measured. The results showed that higher BDNF expression occurred in the Cg1, PrL, and amygdala. Lower BDNF expression occurred in the IL, CA1, CA2, CA3, and DG. BDNF mRNA levels were higher in the mPFC and amygdala but lower in the hippocampus. The neural connection analysis showed that BDNF protein and BDNF mRNA exhibited weak connections among the mPFC, amygdala, and hippocampus during situational reminders. The present data did not support the previous viewpoint in neuroimaging research that the mPFC and hippocampus revealed hypoactivity and the amygdala exhibited hyperactivity for PTSD symptoms. These findings should be discussed with the previous evidence and provide clinical implications for PTSD.
Collapse
|
12
|
Basolateral Amygdala but Not Medial Prefrontal Cortex Contributes to Chronic Fluoxetine Treatments for PTSD Symptoms in Mice. Behav Neurol 2020; 2020:8875087. [PMID: 33299494 PMCID: PMC7710423 DOI: 10.1155/2020/8875087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/16/2020] [Accepted: 11/02/2020] [Indexed: 11/17/2022] Open
Abstract
Do chronic fluoxetine treatments reduced footshock-induced posttraumatic stress disorder (PTSD) symptoms, including fear and comorbid depression, in the situational reminder phase? Moreover, are the subareas of the medial prefrontal cortex (mPFC), including the cingulate cortex 1 (Cg1), prelimbic cortex (PrL), infralimbic cortex (IL), and basolateral amygdala (BLA), involved in the fluoxetine amelioration of PTSD symptoms? These two crucial issues were addressed in the present study. All mice were injected with chronic fluoxetine or normal saline treatments for the adaptation (14 days), footshock fear conditioning (1 day), and situational reminder (3 days) phases. After adaptation, the mice were subjected to footshock (2 mA, 10 seconds) or nonfootshock and stayed 2 min in a footshock box for 2 min for fear conditioning. Later, they were placed in the footshock box for 2 min in the situational reminder phase. In the final session of the situational reminder phase, a forced swimming test (FST) and immunohistochemical staining were conducted. The results indicated that footshock induced fear and comorbid depression. Meanwhile, chronic fluoxetine treatments reduced fear and depression behaviors. The Cg1, PrL, IL, and BLA were seemingly to increase c-Fos expression after footshock-induced PTSD symptoms in the situational reminder phase. The fluoxetine treatments reduced only the BLA's c-Fos expression. The findings suggest that BLA contributes to the fluoxetine amelioration of PTSD symptoms; however, the mPFC, including the Cg1, PrL, and IL, did not mediate PTSD symptoms' amelioration stemming from fluoxetine. The present data might help us to further understand the neural mechanism of fluoxetine treatments in PTSD symptoms.
Collapse
|
13
|
Sullivan KE, Kendrick RM, Cembrowski MS. Elucidating memory in the brain via single-cell transcriptomics. J Neurochem 2020; 157:982-992. [PMID: 33230878 DOI: 10.1111/jnc.15250] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 01/17/2023]
Abstract
Elucidating the neural mechanisms of memory in the brain is a central goal of neuroscience. Here, we discuss modern-day transcriptomics methodologies, and how they are well-poised to revolutionize our insight into memory mechanisms at unprecedented resolution and throughput. Focusing on the hippocampus and amygdala, two regions extensively examined in memory research, we show how single-cell transcriptomics technologies have been leveraged to understand the naïve state of these brain regions. Building upon this foundation, we show that these technologies can be applied to single-trial learning paradigms to comprehensively identify molecules and cells that participate in the encoding and retrieval of memory. Transcriptomics also provides an opportunity to understand the cell-type organization of the human hippocampus and amygdala, and due to conservation of these brain regions between humans and rodents, to infer behavioral and causal contributions in the human brain by leveraging rodent cell-type homologies and interventions. Ultimately, such transcriptomic technologies are poised to usher in a qualitatively novel understanding of memory in the brain.
Collapse
Affiliation(s)
- Kaitlin E Sullivan
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Rennie M Kendrick
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Mark S Cembrowski
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada.,Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, Canada.,Institute of Applied Mathematics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
14
|
Blaze J, Choi I, Wang Z, Umali M, Mendelev N, Tschiffely AE, Ahlers ST, Elder GA, Ge Y, Haghighi F. Blast-Related Mild TBI Alters Anxiety-Like Behavior and Transcriptional Signatures in the Rat Amygdala. Front Behav Neurosci 2020; 14:160. [PMID: 33192359 PMCID: PMC7604767 DOI: 10.3389/fnbeh.2020.00160] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/11/2020] [Indexed: 12/21/2022] Open
Abstract
The short and long-term neurological and psychological consequences of traumatic brain injury (TBI), and especially mild TBI (mTBI) are of immense interest to the Veteran community. mTBI is a common and detrimental result of combat exposure and results in various deleterious outcomes, including mood and anxiety disorders, cognitive deficits, and post-traumatic stress disorder (PTSD). In the current study, we aimed to further define the behavioral and molecular effects of blast-related mTBI using a well-established (3 × 75 kPa, one per day on three consecutive days) repeated blast overpressure (rBOP) model in rats. We exposed adult male rats to the rBOP procedure and conducted behavioral tests for anxiety and fear conditioning at 1-1.5 months (sub-acute) or 12-13 months (chronic) following blast exposure. We also used next-generation sequencing to measure transcriptome-wide gene expression in the amygdala of sham and blast-exposed animals at the sub-acute and chronic time points. Results showed that blast-exposed animals exhibited an anxiety-like phenotype at the sub-acute timepoint but this phenotype was diminished by the chronic time point. Conversely, gene expression analysis at both sub-acute and chronic timepoints demonstrated a large treatment by timepoint interaction such that the most differentially expressed genes were present in the blast-exposed animals at the chronic time point, which also corresponded to a Bdnf-centric gene network. Overall, the current study identified changes in the amygdalar transcriptome and anxiety-related phenotypic outcomes dependent on both blast exposure and aging, which may play a role in the long-term pathological consequences of mTBI.
Collapse
Affiliation(s)
- Jennifer Blaze
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Inbae Choi
- Research and Development Service, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
| | - Zhaoyu Wang
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Michelle Umali
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Natalia Mendelev
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Anna E Tschiffely
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, MD, United States
| | - Stephen T Ahlers
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, MD, United States
| | - Gregory A Elder
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Neurology Service, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
| | - Yongchao Ge
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Fatemeh Haghighi
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Research and Development Service, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
15
|
Zhang K, Wang L, Li G, Cao C, Fang R, Liu P, Luo S, Zhang X. Correlation between hypothalamic-pituitary-adrenal axis gene polymorphisms and posttraumatic stress disorder symptoms. Horm Behav 2020; 117:104604. [PMID: 31655035 DOI: 10.1016/j.yhbeh.2019.104604] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 09/20/2019] [Accepted: 09/21/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND The hypothalamic-pituitary-adrenal (HPA) axis is the main neuroendocrine system that controls stress responses, including fear learning. To further understand the correlation between the HPA axis and stress- and fear-related symptoms in humans, the current study investigated the relationship between HPA axis gene polymorphisms and a stress- and fear-related disorder, posttraumatic stress disorder (PTSD). This is the first study that systematically investigates the correlations between HPA axis genes and distinct PTSD symptom clusters. METHODS Participants included 1132 Chinese earthquake survivors (772 women and 360 men). PTSD symptoms were measured by the PTSD Checklist for DSM-5 (PCL-5), and the severity (total symptoms) and symptom clusters were calculated according to the hybrid seven-factor model of DSM-5 PTSD. We genotyped eight single nucleotide polymorphisms (SNPs) of three HPA axis genes, including FKBP5, CRHR1 and CRHR2. RESULTS The main effects of the CRHR2 SNP rs2267715 were associated with PTSD severity (P = 0.0035) and all PTSD symptom clusters except dysphoric arousal (P ranging from 0.0011 to 0.048). In women, a gene-environment interaction (G × E) effect of FKBP5 (rs3800373 × trauma exposure) was correlated with PTSD severity (P = 0.038), externalizing behaviors, anxious arousal and dysphoric arousal symptoms (P ranging from 0.014 to 0.028); the G × E effect of CRHR1 (rs4458044 × trauma exposure) was associated with anxious arousal symptoms (P = 0.016). In men, a gene-gene interaction (G × G) effect of FKBP5-CRHR1 (rs9470080 × rs4458044) was associated with PTSD severity (P = 0.0091), intrusion, negative affect, externalizing behaviors and anxious arousal (P ranging 0.012-0.049). CONCLUSION Our results systematically revealed that the main effects and G × E and G × G effects of some genetic polymorphisms of HPA axis genes are involved in the severity and distinct symptom clusters of PTSD.
Collapse
Affiliation(s)
- Kunlin Zhang
- Laboratory for Traumatic Stress Studies and Center for Genetics and BioMedical Informatics Research, CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Wang
- Laboratory for Traumatic Stress Studies and Center for Genetics and BioMedical Informatics Research, CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Gen Li
- Laboratory for Traumatic Stress Studies and Center for Genetics and BioMedical Informatics Research, CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chengqi Cao
- Laboratory for Traumatic Stress Studies and Center for Genetics and BioMedical Informatics Research, CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; Shenzhen Key Laboratory of Affective and Social Cognitive Science, Shenzhen University, Shenzhen 518060, China
| | - Ruojiao Fang
- Laboratory for Traumatic Stress Studies and Center for Genetics and BioMedical Informatics Research, CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ping Liu
- People's Hospital of Deyang City, Deyang, Sichuan 618000, China
| | - Shu Luo
- People's Hospital of Deyang City, Deyang, Sichuan 618000, China
| | - Xiangyang Zhang
- Laboratory for Traumatic Stress Studies and Center for Genetics and BioMedical Informatics Research, CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
16
|
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
|
17
|
Tanaka M, Li H, Zhang X, Singh J, Dalgard CL, Wilkerson M, Zhang Y. Correction to: Region- and time-dependent gene regulation in the amygdala and anterior cingulate cortex of a PTSD-like mouse model. Mol Brain 2019; 12:47. [PMID: 31072367 PMCID: PMC6509754 DOI: 10.1186/s13041-019-0470-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 04/29/2019] [Indexed: 12/04/2022] Open
Affiliation(s)
- Mikiei Tanaka
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA
| | - Hongyun Li
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA
| | - Xijun Zhang
- Collaborative Health Initiative Research Program (CHIRP), Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA
| | - Jatinder Singh
- Collaborative Health Initiative Research Program (CHIRP), Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA
| | - Clifton L Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA.,Collaborative Health Initiative Research Program (CHIRP), Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA
| | - Matthew Wilkerson
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA.,Collaborative Health Initiative Research Program (CHIRP), Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA
| | - Yumin Zhang
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA. .,Collaborative Health Initiative Research Program (CHIRP), Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA.
| |
Collapse
|