Next Article in Journal
High Arterial Lactate Levels after Hepatic Resection Are Associated with Low Oxygen Delivery and Predict Severe Postoperative Complications
Next Article in Special Issue
Expression Profile of mRNAs and miRNAs Related to the Oxidative-Stress Phenomenon in the Ishikawa Cell Line Treated Either Cisplatin or Salinomycin
Previous Article in Journal
Signaling Pathways Regulating the Expression of the Glioblastoma Invasion Factor TENM1
Previous Article in Special Issue
Variances in the Expression of mRNAs and miRNAs Related to the Histaminergic System in Endometrioid Endometrial Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Central and Peripheral Immune Dysregulation in Posttraumatic Stress Disorder: Convergent Multi-Omics Evidence

by
Diana L. Núñez-Rios
1,2,
José J. Martínez-Magaña
1,2,
Sheila T. Nagamatsu
1,2,
Diego E. Andrade-Brito
1,2,
Diego A. Forero
3,
Carlos A. Orozco-Castaño
3 and
Janitza L. Montalvo-Ortiz
1,2,*
1
Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
2
VA CT Healthcare Center, West Haven, CT 06516, USA
3
Health and Sport Sciences Research Group, School of Health and Sport Sciences, Fundación Universitaria del Área Andina, Bogotá 110231, Colombia
*
Author to whom correspondence should be addressed.
Submission received: 2 April 2022 / Revised: 29 April 2022 / Accepted: 4 May 2022 / Published: 10 May 2022
(This article belongs to the Special Issue Epigenetic Regulation of the Immune System)

Abstract

:
Posttraumatic stress disorder (PTSD) is a chronic and multifactorial disorder with a prevalence ranging between 6–10% in the general population and ~35% in individuals with high lifetime trauma exposure. Growing evidence indicates that the immune system may contribute to the etiology of PTSD, suggesting the inflammatory dysregulation as a hallmark feature of PTSD. However, the potential interplay between the central and peripheral immune system, as well as the biological mechanisms underlying this dysregulation remain poorly understood. The activation of the HPA axis after trauma exposure and the subsequent activation of the inflammatory system mediated by glucocorticoids is the most common mechanism that orchestrates an exacerbated immunological response in PTSD. Recent high-throughput analyses in peripheral and brain tissue from both humans with and animal models of PTSD have found that changes in gene regulation via epigenetic alterations may participate in the impaired inflammatory signaling in PTSD. The goal of this review is to assess the role of the inflammatory system in PTSD across tissue and species, with a particular focus on the genomics, transcriptomics, epigenomics, and proteomics domains. We conducted an integrative multi-omics approach identifying TNF (Tumor Necrosis Factor) signaling, interleukins, chemokines, Toll-like receptors and glucocorticoids among the common dysregulated pathways in both central and peripheral immune systems in PTSD and propose potential novel drug targets for PTSD treatment.

1. Introduction

Posttraumatic stress disorder (PTSD) is a chronic and multifactorial disorder with a prevalence ranging between 6–10% in the general population, and 35% among individuals with high-lifetime trauma exposure (e.g., combat veterans) [1,2,3]. PTSD can develop after experiencing traumatic events; however, not all individuals exposed to trauma are diagnosed with PTSD, and this is known as resilience [4]. That differential response to trauma unmasks the existence of biological processes contributing to PTSD risk [1,2,3,5]. Growing evidence indicates that the immune system may contribute to the development, maintenance, and clinical outcomes of PTSD, establishing the dysregulation of the immune system as a hallmark feature of PTSD [6,7]. By examining 65 published studies in PTSD, one study summarized a comprehensive list of dysregulated inflammatory factors reported in the blood and cerebrospinal fluid samples of PTSD patients [7]. The dysregulation of immune cells, the human leukocyte antigen (HLA), and immune-related genes in PTSD also support the important role of the immune system in PTSD [6]. While the immune dysregulation in PTSD is a well-established finding in the literature, the differences between the central and peripheral immune system, its crosstalk, and the biological mechanisms underlying this dysregulation remain poorly understood.
The role of the hypothalamic-pituitary-adrenal (HPA) axis on the immune response is one of the most described physiological mechanisms in PTSD, impacting and linking both the central and peripheral systems. Briefly, after exposure to a traumatic event, the HPA axis is activated and induces a release of glucocorticoid hormones (GCs) [8,9], which, in turn, can activate glucocorticoid receptors (GR) and subsequently regulate the gene expression of inflammatory molecules, such as cytokines and the nuclear factor kappa B (NF-κB) [8,10]. In the periphery, GCs can regulate the response of innate immune cells, including the monocytes, macrophages, and dendritic cells [10]. In the brain, abnormal levels of GCs can impact the regulatory feedback of the HPA axis (known as glucocorticoid resistance), followed by an increased neuroinflammatory response, glial dysfunction and synaptic loss [9,11,12,13,14,15]. An animal model study found that pro-inflammatory cytokines in central and peripheral tissues are released after activation of central fear circuits and suggests that neuroinflammation can promote peripheral inflammation, migration of peripheral monocytes to the brain and the subsequent increased activity of microglia [16]. Neuroinflammatory processes orchestrated by central and peripheral systems [17] are mediated by microglia, brain cells also related to disturbances in fear memory in animal models [18,19] and PTSD [20]. These studies support an interplay between central and peripheral immune systems in PTSD and highlight the utility of animal models to shed light on the immunological mechanisms implicated in PTSD [21,22,23,24].
In recent years, high-throughput methods have facilitated genomic, transcriptomic, epigenomic, and proteomic evaluation to identify the mechanisms of dysregulated pathways in both central and peripheral tissues from humans with PTSD, as well as animal models of PTSD [2,25,26]. The noticeable dysregulation of the immune system in PTSD has also been also supported at different regulatory levels. For instance, the C-reactive protein (CRP) that participates in the activation of the complement system is frequently elevated in individuals with PTSD and is related with symptom severity [27,28,29,30]. At the genomic level, genetic variants such as rs1130864, rs3091244, rs1205 and rs2794520 identified through genome wide association studies (GWAS) have been associated with variable levels of CRP in individuals with PTSD [6,27,30]. Another immunoregulatory molecule reported with multi-omic dysregulation is FKBP5 (FKBP Prolyl Isomerase 5, which mediates the GR translocation to the nucleus) with the associated genetic risk variants [31], alterations in DNA methylation patterns [32] and dysregulated expression in different brain regions [2,33,34].
This systematic review summarizes the recent evidence of multi-omics studies to dissect the role of the central and peripheral immune dysregulation in PTSD. We highlight convergent inflammatory factors across tissues and species and examine whether gene regulatory mechanisms may orchestrate the system-wide immune dysregulation in PTSD. Furthermore, we use computational approaches to explore immunoregulatory targets as potential PTSD treatments.

2. Literature Search

The literature search in the PubMed electronic database was conducted in December 2021 to capture full-text studies that investigated the role of the immune system in central and peripheral tissues of individuals with or animal models of PTSD. To reduce the bias in the selection of studies, we followed the Preferred Reporting Items for Systematic Reviews analysis and Metal Analysis (PRISMA) criteria [35]. We found 77 full-text records using the following keywords and Boolean operators: (“Immune” OR “Inflammatory” OR “immunological”) AND (“PTSD” OR “Posttraumatic Stress Disorder” OR “Post-traumatic Stress Disorder”) AND (“Transcriptome” OR “Transcriptomic” OR “Epigenome” OR “Epigenomic” OR “Bisulfite” OR “Array” OR “RNA-Seq” OR “EWAS” OR “epigenome-wide association”). To carry out the records screening, our inclusion criteria were as follows: (1) full-text articles published in the last 10 years, (2) studies using genome-wide approaches and (3) studies using human samples or wild-type and drug-free animal models of PTSD. For this last criteria, wild-type and drug-free animal were selected as a criterion to reduce noise in the transcriptional/epigenetic results related to PTSD. Furthermore, among the animal stress paradigms, we selected studies utilizing physical stressors, such as immobilization, electric shock, predator stress, single prolonged stress and stress-enhanced fear learning. These stress paradigms have been shown to induce fear memory in animals and mimic PTSD-like behaviors (referred here as animal models of PTSD) [21,22,23,36]. Records screening is represented in Figure 1.

3. Results

3.1. Genetic Variants Associated with PTSD Targeting Immune-Related Genes

PTSD has an estimated heritability of ~49%, of which only 6–20% is explained by common variants [3,37,38]. GWAS studies have reported loci associated with an increased risk of developing PTSD, some of these mapping to immune-related genes [3,37,39,40]. In 2015, a GWAS study in 3494 males with European and African ancestries from military cohorts identified significant single nucleotide polymorphisms (SNPs) in two genes related to immune response, JAK1 and FASLG [41]. In a targeted study focusing on the Human Leukocyte Antigen (HLA) genes in an African American cohort (n = 429), five SNPs were identified as mapping to B, C, DRB1, DQA1, DQB1, and the DPB1 immune-related genes associated with PTSD. This study also examined gene expression modules of PTSD through weighted gene co-expression network analysis (WCGNA) and identified immune-related enriched pathways [42]. Another GWAS replicated the association of the HLA-B gene with PTSD, a gene essential in the antigen presentation to lymphocytes [3,43]. In the same year,. another group studied rare genetic variations in 707 adolescents exposed to a tornado who met PTSD criteria, and identified the immune system in the gene network pathway analysis [44]. By performing a variant calling analysis on an RNA-seq dataset in a small cohort of infantry soldiers (n = 85, 27 with PTSD symptoms), a study revealed a possible role of tumor necrosis factor (TNF) in PTSD based on the identified genes with high impact mutations observed in solders with PTSD symptoms [45]. Another study assessing (CRP) in 286 U.S. military veterans of post-9/11 conflicts reported an association between the rs3091244 SNP, lifetime trauma exposure, and PTSD severity [30].
Recent large-scale GWAS of PTSD from consortia efforts have also identified genome-wide significant SNPs located in immune-related genes. A study of 146,660 European Americans and 19,983 African Americans individuals from the Million Veteran Program (MVP) cohort assessing reexperiencing PTSD symptoms identified a significant association in the RAB27B gene, which plays a role in inflammatory response [39]. PACRG was later identified in 250,000 participants from the MVP of European and African ancestries [40]. PARK2 was reported assessing the Psychiatric Genomics Consortium PTSD (PGC-PTSD) Freeze 2 dataset (PGC2) and replicating their results in the MVP cohort [3]. Both PARCG and PARK2 genes are part of the parkin complex that participate in critical autophagic immune processes [3,40,46].

3.2. Transcriptomic and Epigenomic Dysregulation of Immune-Related Genes in PTSD

Studies have found that the dysregulation of immune-related genes, such as IL-1A and FKBP5, may be mediated by epigenetic mechanisms which, in turn, may result in up- or down-regulation of gene expression and the subsequent immune signaling dysfunction [2,27,29,31,33,34,47,48]. Here, we reviewed genome-wide transcriptomic and epigenomic studies of PTSD across tissues and species. We identified four studies in brain samples, one in human brains and three in animal models of PTSD [25,49,50,51,52]. From 22 studies in peripheral samples, 21 in human samples [6,45,50,51,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72], and one in animal models of PTSD [50,51], only one study evaluated both peripheral and brain samples in a rodent model of PTSD and reported the immune dysregulated genes across tissues [50,51]. Twelve studies indicated the immune-related genes identified in their cohorts [25,49,50,51,52,54,58,59,61,68,71]. Seven studies published the whole list of dysregulated genes [45,55,57,60,62,69,73] from which immune-related genes were filtered out through GO enrichment analysis using STRING [74] and Metascape [75] resources. We retained all genes involved in immune signaling and conducted a comprehensive convergent analysis across peripheral and brain tissues in both human and animal models of PTSD.

3.2.1. Peripheral Immune Dysregulation

  • Transcriptomic findings
Only one animal model of PTSD study has evaluated gene expression changes at the genome-wide level. This transcriptomic study examined the effects of aggressor exposure at different points (1, 10, and 42 days after exposure). The authors evaluated the hippocampus, amygdala, and medial prefrontal cortex (mPFC) in a 2015 study [50], and the hemibrain, blood and spleen samples in a follow-up 2017 study [51]. The identified DEGs across tissues and different time points evaluated were enriched in inflammatory pathways such as activation of leukocytes, adhesion of immune cells, cytokine activity, interleukins and (NF-κB) signaling [51]. These findings suggest a sustained neuroinflammatory response in PTSD [51].
In human samples, we identified 245 immune-related genes differentially expressed in PTSD. A research group conducted three transcriptomic studies on blood samples in 2017, 2019 and 2021. In the first study, they evaluated 324 World Trade Center responders and reported 448 differentially expressed genes (FDR < 0.05). Further, PBRM1, PIK3CA, MED1, MAP3K1, JAK2, CEBPB, CD163, MED14, NCOA3, KAT2B, NFAT5, AKT1, PPP3CB, NCOR1 and FKBP5 genes were nominally enriched in the glucocorticoid receptor signaling pathway [57]. In the second study, they conducted a transcriptomic analysis on unsorted blood cells as well as cell-type specific transcriptomic analysis in 39 World Trade Center responders (20 PTSD cases and 19 controls). They identified both cell-type specific and common DEGs across all cell types. In B cells, ABCA6, ANEP, CHN2, IL-17RB, IL-7R, KIAA1217, TBC1D4, TESK2, and TGFBI were differentially expressed in PTSD. PIK3AP1 and SMPDL3A were identified only in CD4T cells; SMAD1 and TLR5 in CD8T cells; and HSF5, IPO5, REST, SLC38A1, and SLC44A2 in monocyte cells. FKBP5 and PI4KAP1 were upregulated across all cell types evaluated [68]. In a recent transcriptomic study assessing PTSD symptom-level data in 226 World Trade Center responders, they identified expression modules associated with inflammatory processes, such as neutrophil activation and the interferon signaling pathway [69].
A peripheral immune dysregulation in PTSD was also indicated by three transcriptomic studies from another research group. In the first study, dysregulation of the innate immune system and interferon signaling was identified in 188 U.S. Marines exposed to conflict zones [59]. On the same dataset, a deconvolution analysis performed to identify hub transcription factors orchestrating gene network functions (master regulators), indicated that TNFAIP3, TRAFD1 and PML may act as master regulators affecting the immune signaling in PTSD [58]. Furthermore, a follow-up study integrated information of seven types of traumas from five independent PTSD blood transcriptome studies [60]. By using an integrative co-expression network analysis, they confirmed a dysregulation of the immune response and cytokine signaling across trauma types in PTSD [60].
The IL-6 is a cytokine with pro-inflammatory and anti-inflammatory pleiotropic effects in the immune system [76,77,78], and elevated serum levels of IL-6 have been reported in individuals with PTSD [79]. Transcriptomic changes related to varying serum levels of IL-6 were assessed by evaluating differential expression in Japanese civilian women with PTSD having high (n = 16) or normal (n = 16) IL-6 levels and health controls (n = 16) [54]. Women with PTSD and elevated levels of IL-6 showed differential expression of IL-4, IL-18R1, IL-28RA, DEFA3, DEFA4, FCGR1A, FCGR1B, CEACAM8 and LTF, and an enrichment of the inflammatory signaling pathway. In contrast, women with PTSD and normal IL-6 levels showed DEGs enriched for neurotransmission and nervous system development [54]. These findings suggest a key role of immunological factor IL-6 in PTSD.
Differential expression of immune-related genes, particularly those involved in Th cell differentiation, was also indicated in blood samples of eight PTSD veterans compared with four healthy controls [61,72]. In a follow-up study, they showed that differential expression of WNT10B may be mediated by epigenetic changes (See the ‘Epigenomic’ section) [72]. Innate immune response, cytokine-cytokine receptor interaction, Jak-STAT and Toll-like receptor were additional enriched pathways reported in transcriptomic studies comparing veterans with and without PTSD [62,73]. Another study comparing military service members with high and low symptoms severity found a significant differential expression only in PTSD patients with high intrusion symptoms and an upregulation of the immune response related to the NF-κB hub. It suggested an implication of this pathway in intrusion symptoms of PTSD [55]. Taken together, findings from peripheral transcriptomic studies support a role of the immune dysregulation on PTSD and suggests cell-type specificity effects. Further, it provides evidence on how changes in gene expression may be mediated by epigenetic modifications.
  • Epigenomic findings
Epigenetics are chemical modifications at the DNA that influence the chromatin configuration and, subsequently, gene transcription processes. DNA methylation (DNAm) is one of the most common epigenetic modifications studied, defined as the addition of a methyl group in the 5′ position of a cytosine ring (5-methyl cytosine, 5mC). This mechanism is mainly associated with transcriptional repression when the 5mC occurs at the promoter region [2,55,79,80,81,82]. Studies in the last decade have evaluated epigenetic changes associated with PTSD at the genome-wide level through epigenome-wide association studies (EWAS). In our literature search, we identified six records examining peripheral samples of individuals with PTSD and evaluating blood and saliva samples.
One of the first EWAS of PTSD conducted in 100 trauma-exposed civilians (23 with PTSD) from the Detroit Neighborhood Health Study (DNHS) assessed methylation and non-methylation (unmethylated) and reported the unmethylation of LTA4H, CXCL8, AQP9, TREM1, F8, CCL1, PYDC1, KLRG1, IFI35, CD1D, CD2, NLRP12, GBP1, IFI16, LST1, PTPN22, TLR1, TLR3, CMKLR1, STAP1 and SLAMF7 in individuals with PTSD diagnosis, all immune-related genes. Another early EWAS study of PTSD (n = 25 cases and 25 controls) showed increased global DNA methylation in PTSD subjects with significant differential methylation in immune-related genes such as ANXA2 and TLR8 [64]. Epigenomic changes impacting immune signaling in PTSD were also found in two additional studies, but gene-level data was not provided [80,83]. Another meta-analysis from the PGC PTSD EWAS Workgroup (n = 545) identified cg19577098 covering the HGS gene, part of the endosome complex and related to interleukin 6 (IL-6) and Tumor Necrosis Factor alpha (TNF-α) signaling [69]. DNA methylation at cg10636246 targeting the Absent in Melanoma 2 (AIM2) gene was related to lower serum CRP levels and immune response in PTSD [30]. A large EWAS of PTSD performed by the PGC PTSD EWAS Workgroup in blood samples from ten military and civilian cohorts (n = 1896) reported four genome-wide significant CpG sites in the AHRR, a gene involved in the kynurenine metabolism and immune response [70]. In saliva, an EWAS of current and lifetime PTSD in 1135 male veterans from the National Health and Resilience Study cohort from our group identified the immune-related genes DYNC1H1 and AP2B1 (current PTSD), and CD55 (lifetime PTSD) [80,83].
EWAS studies of PTSD have also utilized a longitudinal study design evaluating pre- and post-deployment in male veteran cohorts, all examining blood tissue. By evaluating 429 subjects from three male military cohorts (US Marine Resiliency Study (MRS), US Army Study to Assess Risk (STARRS), and Resilience in Servicemembers (Army STARRS), a study reported an association of CpG in the immune-related gene F2R with both pre- and post-deployment PTSD symptoms severity [65]. A significant differentially methylated region (DMR) in HEXDC was associated with post-deployment in a longitudinal meta-EWAS of PTSD in 226 individuals from these three male veteran cohorts [72]. HEXDC is related to immune signaling in PTSD based on the association of this gene with rheumatoid arthritis [72,84]. Another significant PTSD DMR targeted the human leukocyte antigen complex proteins HLA-DPB1, HLA-DBP1, and HLA-DRB1 genes which are involved in immune processes and play a role in synaptic plasticity, learning memory and stress reactivity [72].
In addition to DNA methylation, histones modifications (e.g., methylation or acetylation) and micro-RNAs (inducing the degradation of mRNA) can also impact gene expression and have been associated with psychiatric disorders [85,86,87], including PTSD [85,88]. The role of these epigenetic mechanisms in the immune signaling and PTSD was demonstrated by Bam 2020 in blood samples of eight PTSD veterans [71]. They found that the expression of the immune-related gene WNT10B may be mediated by increased H3K4me3 (tri-methylation at the 4th lysine residue) at the promoter region and a downregulation of the miRNA hsa-miR-7113-5p [71].
An integrative multi-omic analysis can help disentangle the relationship between epigenetic modifications and gene expression and identify markers and mechanisms involved in complex psychiatric disorders [89,90,91,92,93]. To assess the convergent transcriptomic and epigenomic changes that may mediate the peripheral immune dysregulation in PTSD, we conducted an integrative multi-omic analysis across species. For transcriptomic studies, we found 69 immune-relate DEGs identified in more than one independent study (Supplementary Table S1). Transcriptional regulators, such as STAT (Signal Transducer and Activator of Transcription), IRF (interferon regulatory factors), Toll-like receptors (TLRs), and C1q proteins are well-known protein families involved in the innate and adaptive immune system [94,95,96,97]. Here, we identified the gene families from these immune proteins among the 69 convergent DEGs across species: Interleukins (IL-10, IL-16, IL-1RN, IL-2, and IL-6), interferon regulatory factors (IRF3 and IRF9), STATs proteins (STAT1, STAT2 and STAT4) and C1q proteins (C1QA and C1QB). When evaluating the overlap between the epigenomic and transcriptomic findings, we identified eight convergent genes: AQP9, WNT10B, GBP1, IFI35, NLRP12, CXCL8, TLR1 and TLR3. Our multi-omic exploration integrating epigenomic and transcriptomic findings in peripheral tissues helped identify convergent mechanisms for the immune dysregulation in PTSD and reveal potential therapeutic targets. In the next section, we review transcriptomic and epigenomic changes in brain samples of PTSD.

3.2.2. Central Immune Dysregulation

  • Transcriptomic findings
A total of 415 unique DEGs were identified across the four brain transcriptomic studies examined in this review: three DEGs in human samples and 412 DEGs in animal models.
In animal models of PTSD, mitochondrial expression analysis in the amygdala identified 17 dysregulated genes related to inflammatory signaling [52]. Furthermore, another study evaluating transcriptional dysregulation across central and peripheral tissues in an animal model of PTSD reported DEGs enriched for inflammatory pathways, such as the activation of leukocytes, adhesion of immune cells, cytokine activity, interleukins and NF-κB signaling [50,51]. In our convergent analysis, we identified 12 convergent DEGs across the studies using rodent models of PTSD: ADORA2A, ADORA3, ADRBK1, AKT1, ALS2, ANGPT2, ANXA2, AP3B1, APOE, APP, ARRB2 [49,50,51] and GNB4 [50,51,52], which were related with immune signaling in PTSD. In humans, a transcriptomic study of advanced epigenetic age in PTSD examined the motor cortex, ventromedial, and dorsolateral prefrontal cortex (dlPFC) of 97 postmortem brains from the VA National PTSD Brain Bank [80]. Epigenetic age is a biological age indicator based on DNAm predictors of age-related morbidity and mortality of higher accuracy than chronological age [81]. This study identified BMPER (related to glucocorticoid pathway), ALB (oxidative stress), and CCL19 (immune response) immune-related DEGs. Using a targeted approach, reduced expression of IL-1A was observed in the prefrontal cortex (PFC) of individuals with PTSD [82].
When evaluating the convergence between human and animal models of PTSD studies, we identified two immune-related DEGs: IL-1A [50,51,82] and TNFRSF14 [29,50,51]. The pro-inflammatory interleukin 1 alpha (IL-1A) initiates immune response processes in the brain when it is released from the microglia [83]. IL-1A then (like IL-1b) activate the IL-1R1 signaling which, in turn, may trigger the downstream nuclear factor-kappa B (NFκB) and mitogen-activated protein (MAP) kinase pathways, both involved in innate and adaptative immune responses [84,85]. TNF receptor superfamily member 14 (TNFRSF14), a family member of the TNFRSF, is released by glial cells in the brain to trigger the production of proinflammatory cytokines and mediate neuroinflammation and cell death [86].
We extended our convergence analysis to evaluate if the immune-related genes reported in brain samples of animal models of PTSD were also identified in bulk-transcriptomic analyses in post-mortem brains of individuals with PTSD from the VA National PTSD Brain Bank [2,25]. We identified BLNK, C1RL, CCL5, CFH, DOCK2, FOXO3, INPP5D, ITGAX, NFIL3, TNFRSF13C, FKBP5, CDK2, EMP1, ADAM12, TIMELESS and KIF5A as convergent immune-related DEGs across species [2,25,49,50,51]. Chemokine (C-C motif) ligand 5 (CCL5) participates in the peripheral immune response, stimulates migration of phagocytes across the BBB (Blood Brain Barrier) during neuroinflammation, and can act as a pro-inflammatory chemokine in the brain impacting metabolic and neurotrophic processes [87,88]. Dedicator of cytokinesis 2 (DOCK2) is a microglia marker that regulates neuroinflammatory processes related to neurodegenerative disorders, such as Alzheimer’s disease [89,90]. FOXO3 (Transcription factor forkhead box O-3) is a transcription factor that regulates the expression of IL-10 anti-inflammatory cytokine in innate immune cells like macrophages and dendritic cells [91,92]. Another convergent DEG across species is the stress-related FKBP5 gene. FKBP5 participates in the glucocorticoid (cortisol) inflammatory response to stress by mediating the GR translocation to the nucleus [93,94].
An interplay between inflammation, glial cells, and glutamate has been proposed in psychiatric disorders, suggesting that a dysregulated immune response in the brain may stimulate glial cells to release glutamate. The increased concentration of glutamate in the extra synaptic space may promote the aberrant activation of the ionotropic and glutamate receptor, which could result in synaptic dysfunction and loss [17,95,96]. In this review, microglia markers such as DOCK2, as well as neuronal ionotropic and glutamate markers such as GRIN2B (Glutamate Ionotropic Receptor NMDA Type Subunit 2B) and SCN8A (sodium voltage-gated channel alpha subunit 1) were observed as dysregulated in PTSD. These findings suggest that epigenetic regulation in PTSD may also impact the interplay between inflammation, glial cells, and glutamate and induce synaptic loss in the brain.
  • Epigenomic findings
In animal models of PTSD, a study using the social-stress model evaluated both transcriptomic and methylomic changes in brain, blood, and spleen tissues; it also showed the differential methylation of promoter regions of genes related to inflammation in the hemibrain (gene-level data not provided) [50,51]. Malan-Muller evaluated long noncoding RNAs (lncRNAs) in the hypothalamus and reported that mRNAs translated to 13 fear extinction-related proteins, which were predicted targets of the associated lncRNAs to PTSD [49]. In humans, no PTSD epigenomic studies have been conducted in brain tissue. A targeted epigenetic age study performed in 117 postmortem motor cortex samples from the VA National PTSD Brain Bank reported an interaction between rs9315202 SNP and the decreased expression of klotho (KL) longevity gene [97]. The gene product of KL is known to control the brain-immune system interface in the choroid plexus and to regulate autophagy in Alzheimer’s disease [98,99]. Evidently, more research is needed to identify brain-specific epigenomic alterations in PTSD.
Our research group has recently conducted a parallel profiling of neuronal-specific DNA methylation (5mC) and hydroxymethylation (5hmC) in the human postmortem orbitofrontal cortex (OFC) region from individuals with PTSD and controls collected at the VA National PTSD Brain Bank. We identified PTSD-associated significant CpG sites in immune-related genes: IL-7R, CD34, CD8A and C1QL1 genes with differential 5mC; and IL-4R, IL-15, IL-21-AS1, TLR5 and TNFSF14 with differential 5hmC [Unpublished data]. The CD8A and IL-15 genes were differentially expressed immune-related genes across central and peripheral tissues reported in animal models studies [50,51]. Our human neuronal-specific epigenomic results replicate findings observed in a rodent model of PTSD with cross-tissue overlap and suggest that epigenetic mechanisms may regulate gene expression patterns of immune-related genes.

3.3. Inflammatory Proteins Reported in PTSD

Both anti- and pro-inflammatory proteins have been implicated in PTSD [100,101,102,103]. Recent reviews (each including more than 50 studies) have supported an increase of pro-inflammatory cytokines and the reduction of anti-inflammatory signals in individuals with PTSD [7,104]. For instance, IL-1b, IL-6, and TNF-α are common cytokines increased in blood samples of individuals with PTSD [81,105,106,107,108,109,110]. Lower levels of plasma IL-4 levels in individuals with PTSD were consistent with changes observed at the epigenetic level [65]. Furthermore, a variation of the immune cell proportion (based on the ratio of CD4 helper/inducer cells and CD8 cytotoxic/suppressor cells) may also be implicated in the inflammatory dysregulation of PTSD. For instance, increased atypical NK (Natural Killer), reduction of regulatory T cells [111,112,113,114,115,116,117] and increased monocytes [118] cells have been reported in peripheral samples of PTSD. A study examining astroglia and neurotrophic markers in the plasma of 20 PTSD veterans and 20 age-matched healthy control veterans reported low levels of neurotrophic factors (BDNF and NGF-β) as well as increased levels of glial fibrillary acidic protein (GFAP), TNF-α and IL-6 in PTSD veterans. Plasma levels of matrix metalloproteinases MMP2 and MMP9, which play a role in the maintenance of the BBB integrity, are elevated in individuals with PTSD [119]. In the cerebrospinal fluid, a dysregulation of pro-inflammatory and anti-inflammatory cytokines was indicated in individuals with PTSD [120]. After the intramuscular administration of capsaicin, a compound with anti-inflammatory properties [121], an increase of IL-1B and delayed increase of IL-10 was observed in individuals with PTSD [120].
In the brain, a recent neuroimaging study using positron emission tomography (PET) evaluated the microglia marker translocator protein (TSPO) tracked by the [11C]PBR28 probe in the prefrontal-limbic region of 23 individuals with PTSD and 26 healthy controls. TSPO availability in the prefrontal-limbic region was lower in PTSD cases and negatively associated with PTSD symptom severity. Further, PTSD cases showed higher CRP levels in blood samples. These results suggest a deficient neuroprotective function and peripheral immune activation in PTSD [29]. This study evaluating central and peripheral samples confirmed the association of system-wide CRP levels with PTSD, previously reported in other studies [29].
Taken together, studies evaluating the inflammatory dysregulation of PTSD at the protein level show an impaired homeostasis of the central and peripheral immunological response in the pathophysiology of PTSD. Moreover, it supports the notion that these alterations at the protein level maybe mediated by changes in gene expression resulting from epigenetic modifications of immune-related genes.

3.4. PTSD, Comorbidities, and the Immune Response

PTSD is often comorbid with other mental and physical health disorders [122,123], possibly due to shared mechanisms of immune dysregulation. A PTSD transcriptomic study evaluating the effects of Body Mass Index (BMI) and sex [2,124] found that IL-1B is differentially expressed in PTSD males with high BMI, implicating IL-1B in the comorbidity of obesity and metabolic syndromes in PTSD [2,124,125]. Peripheral inflammation, mainly driven by IL-1β, TNF-α and IL-6, have been related to the disturbance of metabolic processes in individuals with schizophrenia (including insulin resistance, hepatic inflammation, and obesity) [126]. TNF-α and IL-6 have also been connected to PTSD [105,106,107,108,109,110], which suggest that these cytokines may simultaneously participate in PTSD comorbidities. Furthermore, PTSD GWAS studies, which have identified genetic variants mapping to immune-related genes, have also shown a genetic correlation between PTSD and asthma (rg = 0.49, P = 0.0002), a respiratory disease linked to the immune system and inflammation [3]. Cognitive impairment and dementia have also been related to PTSD [127,128,129,130,131,132]. In a proteomic analysis, NCAN, BCAN, CTSS, MSR1, MDGA1, and CPA2 were reported as upregulated in individuals with PTSD and comorbid mild cognitive impairment [133]. Of these proteins, MSR1 and CTSS are also upregulated in a blood transcriptomic study of individuals with comorbid PTSD and Alzheimer’s disease [60,134,135,136,137]. These findings suggest that dysregulation of the immune system may be a shared mechanism of PTSD and concomitant mental and physical disorder comorbidities, and this highlights novel therapeutical interventions targeting the immune signaling for the treatment of PTSD, as well as associated comorbidities [126,138,139].

3.5. Deciphering Systemic Immune Response in PTSD Mediated by Epigenetic and Transcriptomic Changes

Integrative multi-omics analyses are an excellent strategy to identify and prioritize molecular markers that can help inform disease risk, prognosis, and treatment interventions [140,141,142]. Further, a comprehensive evaluation of multi-omics domains in a dimensional manner reveals convergent biological pathways across multiple layers and assesses their interactions at the molecular and circuit levels [140,143,144]. In this review, we show that cross-species studies at the genomic, epigenomic, transcriptomic, and proteomic level support a system-wide dysregulation of the immune signaling in PTSD [7,138,145]. Here, we conducted an integrative multi-omics analysis of the literature in PTSD across the central and peripheral systems in both human and animal models, which revealed 84 convergent immune-related genes (Supplementary Table S2).
Annexin A2 (ANXA2) was upregulated in the dlPFC of human postmortem brains [2] and a DEG in animal models of PTSD including the hypothalamus [49], hippocampus, amygdala, mPFC, hemibrain, blood, and spleen [50,51]. ANXA2 is expressed on the surface of several innate immune cells such as dendritic cells, macrophages, and monocytes; and its anti-inflammatory and pro-inflammatory activities vary among acute and chronic stages of inflammation [146]. ANXA2 facilitates wound healing orchestrating matrix remodeling, membrane repair, angiogenesis, vesicle fusion, and cytoskeletal organization [147]. Adenosine A2a receptor (ADORA2A) is also a dysregulated gene in the hypothalamus [51] and the hippocampus, amygdala, mPFC, hemibrain, blood and spleen [50,51] from animal models of PTSD. ADORA2A was also identified in our neuronal-specific epigenomic analysis in post-mortem orbitofrontal samples of individuals with PTSD [Unpublished data]. The gene product of ADORA2A is a receptor involved in the modulation of the response of microglia and astrocytes during neuro-inflammation [148].
The TNF signaling was a convergent dysregulated pathway across all approaches (central and peripheral tissues, with epigenomic and transcriptomic dysregulation and human and animal models of PTSD). At the protein level, TNF-α was increased in plasma samples of individuals with PTSD [119]. At the transcriptomic level, TNFRSF14 and LTB (also known as tumor necrosis factor C) were convergent downregulated genes across transcriptomic analyses performed in peripheral tissues of both species. Furthermore, TNFRSF14 and TSPOAP1 had a decreased gene expression in females with PTSD [2]. At the epigenomic level, in our ongoing neuronal-specific study, we identified a hyper hydroxymethylated CHH site in the intergenic region between CD70 and TNFSF14 genes (chr19:6614775), which has a predicted transcription factor binding site to ZNF135. These multi-omic findings suggest an important role of the TNF pathway in PTSD.
Based on the protein-protein interaction analysis of convergent genes in the central and peripheral tissues using STRING [74], significant enriched pathways (FDR < 0.05) include cytokine-cytokine receptor interaction (OSM, IL-2, IL-1RN, TNFRSF13C, CXCL3, CCR1, CXCL10, TGFBR1, IL-6, IL-10, CXCL2, IL-12RB1, and CCL5), NF-kappa B signaling pathway (BLNK, PRKCQ, ZAP70, TNFRSF13C, CXCL3, BCL2L1, TLR4, MYD88, and CXCL2), Toll-like receptor signaling pathway (CXCL10, TLR1, TLR4, IL-6, TLR6, MYD88, PIK3R1, and CCL5), JAK-STAT signaling pathway (OSM, IL-2, BCL2L1, GRB2, IL-6, IL-10, PIK3R1, and IL-12RB1), Natural killer cell mediated cytotoxicity (ZAP70, GRB2, NFATC2, and PIK3R1), and Th17 cell differentiation (IL-2, PRKCQ, ZAP70, TGFBR1, NFATC2, IL-6, and IL-12RB1).
Brain-specific protein markers were dysregulated in the plasma of individuals with PTSD [119]. Brain cell-type specific gene expression patterns of human and murine RNA expression data sets were recently reported showing the top 1000 cell type-specific genes from astrocytes, oligodendrocytes, microglia, neuron and endothelial cells [149]. Taking advantage of a recent study reporting brain cell type markers and based on 84 convergent genes recovered in this review, we explored if brain cell type specific genes may be affected by epigenomic and transcriptomic changes in PTSD across central and peripheral systems in human and animal models. We found that ~50% of convergent genes were enriched for cell type markers in the brain. CXCL3 is enriched in astrocyte cells; PRKCQ, SEMA4D, LPAR1 and SHC4 in oligodendrocytes cells. Also, C1QA, C1QB, CCR1, IL-10, IL-1RN, NFATC2, OSM, PLXDC2, TGFBR1, TLR1, TLR4, AKAP13, MILR1, TLR6, TMEM106A, BLNK, DOCK2, TNFRSF13C, ITGAX, INPP5D, CTSS, GRB2 and CHSY1 were enriched for microglial cells, CXCL2, IL-6, LTF, IFI6, EPSTI1, CDK2, ANXA2, CFH, EMP1, COL1A1, CYP1B1, COL4A1, IGFBP4, COL6A2 and RCSD1 for endothelial cells, and WIPF3 for neuronal cells [149]. By using flow-sorted PTSD transcriptomic data from the blood [68], we evaluated whether these convergent genes enriched for brain cell type markers are also differentially expressed in peripheral cells such as monocytes, CD4, and CD8 cells. PXK, TAOK1 and SET (enriched for oligodendrocyte cells), and CNOT1 and NCKAP1L (enriched for microglial cells) were PTSD DEGs reported in monocytes. Furthermore, FGD4, PLXDC2, AKAP13 and CD163 (enriched for microglial cells) were DEGs in CD4/CD8 cells [68].
The multiomic convergence of the immune-related genes summarized in this review support a system-wide dysregulation of the immune signaling in PTSD and reinforce the immunological systemic dysregulation as a hallmark of PTSD [7,138,145]. Innate immunity plays an important role in the adaptative response to physiological or psychological factors provoking the release of both anti and pro-inflammatory molecules which are mediated by macrophage, mast cells, and polymorphonuclear leucocytes in peripheral tissues, as well as microglia in the brain [150,151,152]. Innate immune response upon cellular stress may be activated by damage-associated molecular patterns (DAMPs), endogenous molecules that are released by injured tissues. Toll-like receptors (TLRs, one of the cross-tissue convergent pathways identified here) recognize DAMPs which, in turn, activate the NF-κB pathway (another convergent pathway across tissues identified here) and induce cytokines releasing [151,153,154]. A homeostatic immune signaling in both peripheral and central tissues is necessary to maintain its protective function and reduce the risk of developing PTSD [152,155]. In contrast, a maladaptive immune response, possibly mediated by glucocorticoids [9,11,12,13,14,15], may have the opposite effect, reflected by the activation of microglia, neuronal death, accelerated neurodegenerative processes, as well as a disruption of the BBB structure allowing peripheral immune cells into the brain [152,156,157].
The intercommunicative nature of immune signaling across central and peripheral tissues is observed in healthy brains where IL-6 and IL-1 (released by glial cells) are able to activate the immune signaling in both brain and periphery [150]. In PTSD, a study evaluating microglial markers in the brain and CRP in peripheral tissues suggested that inflammatory suppression in the brain is related to increased peripheral inflammatory response [29]. In contrast, an animal model study evaluating acute and chronic stress responses in both central and peripheral tissues suggests a sustained systemic inflammatory response in PTSD resulting in tissue damage in peripheral tissues and the inhibition of synaptic plasticity and neurogenesis in the brain [50,51].
Some of the identified convergent molecules in this review support the hypothesis of an increased peripheral inflammation in PTSD. The transcriptomic dysregulation of immune cells (i.e., monocytes, CD4T, CD8T and B lymphocytes) in PTSD involves the upregulation of FKBP5 and PI4KAP1 across all peripheral cell types [68]. IL-6, with both anti and pro-inflammatory functions, has been a consistent finding at multiple levels in the increased peripheral inflammation of PTSD [7,16]. At the transcript level, IL-6 was reported as upregulated in two transcriptomic studies, one conducted in blood samples of PTSD individuals [62] and one in animal models examining both central and peripheral tissues [50,51]. At the epigenetic level, no studies have reported differential methylation of IL-6 in PTSD. At the protein level, IL-6 was increased in blood samples of PTSD [119]. Elevated serum levels of IL-6 in women with PTSD was related to dysregulation of gene expression of other inflammatory molecules such as IL-4 and IL-18R1 [54]. Additional PTSD peripheral studies of pro-inflammatory molecules have shown increased levels of TNF-α in PTSD veterans [119], an upregulation of IL-1, IL-1B, IL-8, IL-4R, IL-16 and NF-κB in military personnel [62], increased levels of CRP associated with PTSD symptom severity [29], as well as an association between the CRP polymorphism (rs3091244)and lifetime trauma exposure and PTSD severity and the association between rs1205 and rs2794520 with PTSD severity and serum CRP levels relation [6,30].
In brain tissue, animal model studies show an increase of IL-6 related to PTSD [16]. The upregulation of IL-6 across central and peripheral tissues has only been reported in an animal model study [50,51]. In the human brain, IL-6 is known to be involved in neurogenesis and oligodendrogenesis during early postnatal developmental stages [158,159]. However, in PTSD, no studies examined in this review show IL-6 dysregulation in the brain. A recent human study supports brain inflammatory suppression in PTSD based on low brain availability of TSPO, and decreased expression of TNFRSF14 and TSPOAP1 in the postmortem brains of a female subgroup with PTSD [29]. A downregulation of these TNFRSF14 and TSPOAP1 genes has also been found in an animal model study [50,51]. In addition, reduced expression of the proinflammatory IL-1A was observed in the PFC of individuals with PTSD [82] and in an animal model [50,51]. FKBP5 and NR3C1, both anti-inflammatory molecules [156], were downregulated in the medial mPFC and amygdala of animal models, respectively [50,51]. In contrast, a study in postmortem dlPFC samples of individuals with PTSD found an upregulation of FKBP5 [2]. Similarly, upregulation of FKBP5 in the amygdala was reported in another animal model study of PTSD-like behavior [160].
Overall, the studies reviewed here reinforce the hypothesis of an increased peripheral inflammation and a suppressed neuroinflammation in PTSD. However, given the somewhat inconsistent findings in the literature, more research is needed to better characterize the immune dysregulation of PTSD in the brain. Several limitations of the current literature include the lack of evaluation of varying trauma types as well as the duration of trauma exposure. It is important to also note the high heterogeneity of the PTSD’s phenotypic definition used across the human studies, from using case-control binary traits to symptom-level continuous traits. In the animal work, there is an inherent limitation in mimicking socio-cultural environmental exposures observed in individuals with PTSD [24] and recapitulating the complex clinical outcomes of PTSD and related comorbidities. Lastly, studies are needed to directly evaluate the role and possible interplay of anti- and pro-inflammatory mechanisms in the PTSD immune dysregulation.
In conclusion, the convergent cross-tissue immune-related genes and pathways identified here in both human and animal models support a systemic immune dysregulation in PTSD and a potential crosstalk between the central and peripheral systems (Figure 2).

3.6. Drug Development

Substantial evidence supports an important role of a systemic immune dysregulation in PTSD, opening new avenues of drug development for treatment interventions. A previous study showed that immune-related genes, specifically the pro-inflammatory macrophage imbalance (M1/M2 ratio), can serve as a biomarker of response to psychotherapy in the treatment of individuals with PTSD [161]. Therapeutical properties of anti-inflammatory drugs have been also evaluated in animal models of PTSD. For instance, minocycline, an antibiotic with anti-inflammatory properties, reduced the level of IL-1, IL-6 and TNF-α and ameliorated anxiety behaviors after trauma exposure [162]. Here, we performed a drug repurposing analysis using the Drug-Gene Interaction Database (DGIbd) [163] to identify druggable gene targets as potential therapeutic pharmacological treatments [164]. The DGIbd analyzes drug-gene interactions and assigns an interaction score for each pair of gene-drug pairs to help prioritize gene-level data. We conducted a drug repurposing analysis for the 84 identified convergent genes across tissues and species in PTSD (applying query score 5 [163]. We found that convergent genes enriched in Th17 differentiation in the protein-protein interaction analysis (mentioned before)could be a possible target mechanism for drug action. In this analysis, we identified drug targets for genes involved in three main pathways: the toll-like receptor signaling pathway (CXCL10, IL-6, and TLR4), Th17 cell differentiation (ZAP10, IL-6, and TGFBR1), and cytokine-cytokine receptor interaction (CCR1, CXCL10, IL-6, TGFBR1) (Supplementary Table S5). Drug discoveries are based on their target on monoclonal antibodies for the inhibition of immune proteins, including siltuximab, olokizumab, and clazakizumab (IL-6), vactosertib (TGFBR1), and petesicatib, and odanacatib (Cathepsin S). More research is needed to evaluate the effects of these drugs in the treatment of PTSD.

3.7. Limitations, Future Directions, and Concluding Remarks

A growing body of research in humans and animal models supports the dysregulation of the immune system as an important mechanism in PTSD. Here, we reviewed the recent PTSD literature of the genomic, epigenomic, transcriptomic, and proteomic studies to identify the dysregulated immune factors in the central and peripheral systems of animal models and humans implicated in PTSD. We reveal a system-wide immune dysregulation on PTSD and characterize a potential crosstalk between the central and peripheral immune systems. However, the immunological dysregulation in PTSD can be highly heterogeneous, acting on both pro-inflammatory and anti-inflammatory signaling pathways. This heterogeneity may be driven by differences in the duration, type, and intensity of the trauma, but also by potential cell-type specificity with different pathophysiological functions in the acute and long-lasting effects of trauma response which is essential for the development and maintenance of PTSD. Of note, PTSD is a complex multifactorial disorder with high phenotypical heterogeneity. Research aimed to disentangle the role of the immune system in the different domains or symptoms of PTSD is greatly needed. Furthermore, we also highlight the need to study the role of the immune system in the brain in the context of PTSD, particularly in humans. This review sheds new light into the convergent roles of the central and peripheral immune dysregulation in PTSD and identifies potential novel drug targets for treatment development.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/biomedicines10051107/s1, Table S1: convergent genes in peripheral tissues; Table S2: convergent genes in central peripheral tissues; Table S3: Drug development results; Table S4: List of selected genes stratified by brain and peripheral samples; Table S5: Drug development results.

Author Contributions

D.L.N.-R.: Conceptualization, investigation, methodology and writing—original draft preparation. J.J.M.-M.: investigation, methodology and writing—review and editing. S.T.N.: investigation, methodology and writing—review and editing, D.E.A.-B.: investigation, methodology and writing—review and editing, D.A.F.: writing—review and editing, C.A.O.-C.: writing—review and editing. J.L.M.-O.: Conceptualization, writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work is supported by the Department of Veterans Affairs via 1IK2CX002095-01A1 (JLMO), NIDA R21DA050160 (JLMO), and the U.S. Department of Veterans Affairs National Center for Posttraumatic Stress Disorder.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare that they have no conflict of interest.

Abbreviations

5mC5-methyl cytosine
BBBBrain Blood Barrier
CRPC-reactive protein
DAMPsDamage-associated molecular patterns
DNAmDNA methylation
dlPFCDorsolateral prefrontal cortex
EWASEpigenome Wide Association Studies
GWASGenome Wide Association Studies
GCsglucocorticoid hormones
GRglucocorticoid receptors
HPAhypothalamic-pituitary-adrenal
ILInterleukin
mPFCMedial prefrontal cortex
MVPMillion Veteran Program
PTSDPosttraumatic stress disorder
PFCPrefrontal cortex
PGC-PTSDPsychiatric Genomics Consortium PTSD
TLRsToll-like receptors
TNFTumor necrosis factor

References

  1. Girgenti, M.J.; Duman, R.S. Transcriptome Alterations in Posttraumatic Stress Disorder. Biol. Psychiatry 2017, 83, 840–848. [Google Scholar] [CrossRef] [PubMed]
  2. Girgenti, M.J.; Wang, J.; Ji, D.; Cruz, D.A.; Stein, M.B.; Gelernter, J.; Young, K.A.; Huber, B.R.; Williamson, D.E.; Friedman, M.J.; et al. Transcriptomic organization of the human brain in post-traumatic stress disorder. Nat. Neurosci. 2021, 24, 24–33. [Google Scholar] [CrossRef] [PubMed]
  3. Nievergelt, C.M.; Maihofer, A.X.; Klengel, T.; Atkinson, E.G.; Chen, C.-Y.; Choi, K.W.; Coleman, J.R.I.; Dalvie, S.; Duncan, L.E.; Gelernter, J.; et al. International meta-analysis of PTSD genome-wide association studies identifies sex- and ancestry-specific genetic risk loci. Nat. Commun. 2019, 10, 4558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. American Psychiatric Association (APA). Diagnostic and Statistical Manual of Mental Disorders, 5th ed.; American Psychiatric Association: Arlington, VA, USA, 2013. [Google Scholar] [CrossRef]
  5. Wolf, E.J.; Miller, M.; Sullivan, D.R.; Amstadter, A.B.; Mitchell, K.S.; Goldberg, J.; Magruder, K.M. A classical twin study of PTSD symptoms and resilience: Evidence for a single spectrum of vulnerability to traumatic stress. Depress. Anxiety 2017, 35, 132–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Katrinli, S.; Smith, A.K. Immune system regulation and role of the human leukocyte antigen in posttraumatic stress disorder. Neurobiol. Stress 2021, 15, 100366. [Google Scholar] [CrossRef] [PubMed]
  7. Sun, Y.; Qu, Y.; Zhu, J. The Relationship between Inflammation and Post-traumatic Stress Disorder. Front. Psychiatry 2021, 12, 707543. [Google Scholar] [CrossRef]
  8. Merkulov, V.M.; Merkulova, T.I.; Bondar, N.P. Mechanisms of brain glucocorticoid resistance in stress-induced psychopathologies. Biochemistry 2017, 82, 351–365. [Google Scholar] [CrossRef]
  9. Bellavance, M.A.; Rivest, S. The HPA—Immune axis and the immunomodulatory actions of glucocorticoids in the brain. Front. Immunol. 2014, 5, 136. [Google Scholar] [CrossRef] [Green Version]
  10. Tapp, Z.M.; Godbout, J.P.; Kokiko-Cochran, O.N. A Tilted Axis: Maladaptive Inflammation and HPA Axis Dysfunction Contribute to Consequences of TBI. Front. Neurol. 2019, 10, 345. [Google Scholar] [CrossRef]
  11. Silverman, M.N.; Sternberg, E.M. Glucocorticoid regulation of inflammation and its behavioural and metabolic correlates: From HPA axis to glucocorticoid receptor dysfunction. Ann. N. Y. Acad. Sci. 2012, 1261, 55–63. [Google Scholar] [CrossRef]
  12. Salvador, E.; Shityakov, S.; Förster, C. Glucocorticoids and endothelial cell barrier function. Cell Tissue Res. 2013, 355, 597–605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Joëls, M. Corticosteroids and the brain. J. Endocrinol. 2018, 238, R121–R130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Merlo, D.; Cuchillo-Ibañez, I.; Parlato, R.; Rammes, G. DNA Damage, Neurodegeneration, and Synaptic Plasticity. Neural Plast. 2016, 2016, 1206840. [Google Scholar] [CrossRef] [PubMed]
  15. Kertser, A.; Baruch, K.; Deczkowska, A.; Weiner, A.; Croese, T.; Kenigsbuch, M.; Cooper, I.; Tsoory, M.; Ben-Hamo, S.; Amit, I.; et al. Corticosteroid signaling at the brain-immune interface impedes coping with severe psychological stress. Sci. Adv. 2019, 5, eaav4111. [Google Scholar] [CrossRef] [Green Version]
  16. Deslauriers, J.; Powell, S.B.; Risbrough, V.B. Immune signaling mechanisms of PTSD risk and symptom development: Insights from animal models. Curr. Opin. Behav. Sci. 2017, 14, 123–132. [Google Scholar] [CrossRef] [Green Version]
  17. Lurie, D.I. An Integrative Approach to Neuroinflammation in Psychiatric disorders and Neuropathic Pain. J. Exp. Neurosci. 2018, 12. [Google Scholar] [CrossRef] [Green Version]
  18. Enomoto, S.; Kato, T.A. Involvement of microglia in disturbed fear memory regulation: Possible microglial contribution to the pathophysiology of posttraumatic stress disorder. Neurochem. Int. 2020, 142, 104921. [Google Scholar] [CrossRef]
  19. Li, S.; Liao, Y.; Dong, Y.; Li, X.; Li, J.; Cheng, Y.; Cheng, J.; Yuan, Z. Microglial deletion and inhibition alleviate behavior of post-traumatic stress disorder in mice. J. Neuroinflamm. 2021, 18, 7. [Google Scholar] [CrossRef]
  20. Rahimian, R.; Wakid, M.; O’Leary, L.A.; Mechawar, N. The emerging tale of microglia in psychiatric disorders. Neurosci. Biobehav. Rev. 2021, 131, 1–29. [Google Scholar] [CrossRef]
  21. Richter-Levin, G.; Stork, O.; Schmidt, M.V. Animal models of PTSD: A challenge to be met. Mol. Psychiatry 2018, 24, 1135–1156. [Google Scholar] [CrossRef] [Green Version]
  22. Verbitsky, A.; Dopfel, D.; Zhang, N. Rodent models of post-traumatic stress disorder: Behavioral assessment. Transl. Psychiatry 2020, 10, 132. [Google Scholar] [CrossRef] [PubMed]
  23. Zoladz, P.R.; Diamond, D.M. Predator-based psychosocial stress animal model of PTSD: Preclinical assessment of traumatic stress at cognitive, hormonal, pharmacological, cardiovascular and epigenetic levels of analysis. Exp. Neurol. 2016, 284, 211–219. [Google Scholar] [CrossRef] [PubMed]
  24. Girgenti, M.J.; Pothula, S.; Newton, S.S. Stress and Its Impact on the Transcriptome. Biol. Psychiatry 2021, 90, 102–108. [Google Scholar] [CrossRef] [PubMed]
  25. Wolf, E.J.; Zhao, X.; Hawn, S.E.; Morrison, F.G.; Zhou, Z.; Fein-Schaffer, D.; Huber, B.; Miller, M.W.; Logue, M.W. Gene expression correlates of advanced epigenetic age and psychopathology in postmortem cortical tissue. Neurobiol. Stress 2021, 15, 100371. [Google Scholar] [CrossRef]
  26. Logue, M.W.; Zhou, Z.; Morrison, F.G.; Wolf, E.J.; Daskalakis, N.P.; Chatzinakos, C.; Georgiadis, F.; Labadorf, A.T.; Girgenti, M.J.; Young, K.A.; et al. Gene expression in the dorsolateral and ventromedial prefrontal cortices implicates immune-related gene networks in PTSD. Neurobiol. Stress 2021, 15, 100398. [Google Scholar] [CrossRef]
  27. Michopoulos, V.; Rothbaum, A.O.; Jovanovic, T.; Almli, L.M.; Bradley, B.; Rothbaum, B.O.; Gillespie, C.F.; Ressler, K.J. Association of CRP Genetic Variation and CRP Level with Elevated PTSD Symptoms and Physiological Responses in a Civilian Population With High Levels of Trauma. Am. J. Psychiatry 2015, 172, 353–362. [Google Scholar] [CrossRef] [Green Version]
  28. Eraly, S.A.; Nievergelt, C.; Maihofer, A.X.; Barkauskas, D.A.; Biswas, N.; Agorastos, A.; O’Connor, D.T.; Baker, D.G. Assessment of Plasma C-Reactive Protein as a Biomarker of Posttraumatic Stress Disorder Risk. JAMA Psychiatry 2014, 71, 423–431. [Google Scholar] [CrossRef]
  29. Bhatt, S.; Traumatic Stress Brain Study Group; Hillmer, A.T.; Girgenti, M.J.; Rusowicz, A.; Kapinos, M.; Nabulsi, N.; Huang, Y.; Matuskey, D.; Angarita, G.A.; et al. PTSD is associated with neuroimmune suppression: Evidence from PET imaging and postmortem transcriptomic studies. Nat. Commun. 2020, 11, 2360. [Google Scholar] [CrossRef]
  30. Miller, M.; Maniates, H.; Wolf, E.; Logue, M.; Schichman, S.; Stone, A.; Milberg, W.; McGlinchey, R. CRP polymorphisms and DNA methylation of the AIM2 gene influence associations between trauma exposure, PTSD, and C-reactive protein. Brain, Behav. Immun. 2017, 67, 194–202. [Google Scholar] [CrossRef]
  31. Kang, J.I.; Kim, T.Y.; Choi, J.H.; So, H.S.; Kim, S.J. Allele-specific DNA methylation level of FKBP5 is associated with post-traumatic stress disorder. Psychoneuroendocrinology 2019, 103, 1–7. [Google Scholar] [CrossRef]
  32. Bishop, J.R.; Lee, A.M.; Mills, L.J.; Thuras, P.D.; Eum, S.; Clancy, D.; Erbes, C.R.; Polusny, M.A.; Lamberty, G.J.; Lim, K.O. Methylation of FKBP5 and SLC6A4 in Relation to Treatment Response to Mindfulness Based Stress Reduction for Posttraumatic Stress Disorder. Front. Psychiatry 2018, 9, 418. [Google Scholar] [CrossRef]
  33. Young, K.A.; Thompson, P.M.; Cruz, D.A.; Williamson, D.; Selemon, L.D. BA11 FKBP5 expression levels correlate with dendritic spine density in postmortem PTSD and controls. Neurobiol. Stress 2015, 2, 67–72. [Google Scholar] [CrossRef] [Green Version]
  34. Holmes, S.E.; Girgenti, M.J.; Davis, M.T.; Pietrzak, R.H.; DellaGioia, N.; Nabulsi, N.; Matuskey, D.; Southwick, S.; Duman, R.S.; Carson, R.E.; et al. Altered metabotropic glutamate receptor 5 markers in PTSD: In vivo and postmortem evidence. Proc. Natl. Acad. Sci. USA 2017, 114, 8390–8395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, 105906. [Google Scholar] [CrossRef]
  36. Borghans, B. Animal models for posttraumatic stress disorder: An overview of what is used in research. World J. Psychiatry 2015, 5, 387–396. [Google Scholar] [CrossRef] [PubMed]
  37. Mundy, S.S.; Foss, S.L.W.; Poulsen, S.; Hjorthøj, C.; Carlsson, J. Sex differences in trauma exposure and symptomatology in trauma-affected refugees. Psychiatry Res. 2020, 293, 113445. [Google Scholar] [CrossRef] [PubMed]
  38. Mundy, J.; Hübel, C.; Gelernter, J.; Levey, D.; Murray, R.M.; Skelton, M.; Stein, M.B.; Vassos, E.; Breen, G.; Coleman, J.R.I. Psychological trauma and the genetic overlap between posttraumatic stress disorder and major depressive disorder. Psychol. Med. 2021, 51, 1–10. [Google Scholar] [CrossRef]
  39. Gelernter, J.; Sun, N.; Polimanti, R.; Pietrzak, R.; Levey, D.; Bryois, J.; Lu, Q.; Hu, Y.; Li, B.; Radhakrishnan, K.; et al. Genome-wide association study of post-traumatic stress disorder reexperiencing symptoms in >165,000 US veterans. Nat. Neurosci. 2019, 22, 1394–1401. [Google Scholar] [CrossRef] [PubMed]
  40. Stein, M.B.; Levey, D.F.; Cheng, Z.; Wendt, F.R.; Harrington, K.; Pathak, G.A.; Cho, K.; Quaden, R.; Radhakrishnan, K.; Girgenti, M.J.; et al. Genome-wide association analyses of post-traumatic stress disorder and its symptom subdomains in the Million Veteran Program. Nat. Genet. 2021, 53, 174–184. [Google Scholar] [CrossRef] [PubMed]
  41. Nievergelt, C.M.; Maihofer, A.X.; Mustapic, M.; Yurgil, K.A.; Schork, N.J.; Miller, M.; Logue, M.W.; Geyer, M.A.; Risbrough, V.B.; O’Connor, D.T.; et al. Genomic predictors of combat stress vulnerability and resilience in U.S. Marines: A genome-wide association study across multiple ancestries implicates PRTFDC1 as a potential PTSD gene. Psychoneuroendocrinology 2015, 51, 459–471. [Google Scholar] [CrossRef] [Green Version]
  42. Katrinli, S.; Lori, A.; Kilaru, V.; Carter, S.; Powers, A.; Gillespie, C.; Wingo, A.P.; Michopoulos, V.; Jovanovic, T.; Ressler, K.J.; et al. Association of HLA locus alleles with posttraumatic stress disorder. Brain Behav. Immun. 2019, 81, 655–658. [Google Scholar] [CrossRef] [PubMed]
  43. Yarzabek, B.; Zaitouna, A.J.; Olson, E.; Silva, G.N.; Geng, J.; Geretz, A.; Thomas, R.; Krishnakumar, S.; Ramon, D.S.; Raghavan, M. Variations in HLA-B cell surface expression, half-life and extracellular antigen receptivity. eLife 2018, 7, e34961. [Google Scholar] [CrossRef] [PubMed]
  44. Sheerin, C.M.; Vladimirov, V.; Williamson, V.; Bountress, K.; Danielson, C.K.; Ruggiero, K.; Amstadter, A.B. A preliminary investigation of rare variants associated with genetic risk for PTSD in a natural disaster-exposed adolescent sample. Eur. J. Psychotraumatol. 2019, 10, 1688935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Sragovich, S.; Gershovits, M.; Lam, J.C.; Li, V.O.; Gozes, I. Putative Blood Somatic Mutations in Post-Traumatic Stress Disorder-Symptomatic Soldiers: High Impact of Cytoskeletal and Inflammatory Proteins. J. Alzheimer’s Dis. 2021, 79, 1723–1734. [Google Scholar] [CrossRef] [PubMed]
  46. Levine, B.; Mizushima, N.; Virgin, H.W. Autophagy in immunity and inflammation. Nature 2011, 469, 323–335. [Google Scholar] [CrossRef]
  47. Miller, O.; Shakespeare-Finch, J.; Bruenig, D.; Mehta, D. DNA methylation of NR3C1 and FKBP5 is associated with posttraumatic stress disorder, posttraumatic growth, and resilience. Psychol. Trauma 2020, 12, 750–755. [Google Scholar] [CrossRef] [PubMed]
  48. Morrison, F.G.; Miller, M.W.; Logue, M.W.; Assef, M.; Wolf, E. DNA methylation correlates of PTSD: Recent findings and technical challenges. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2019, 90, 223–234. [Google Scholar] [CrossRef]
  49. Malan-Müller, S.; De Souza, V.B.; Daniels, W.M.; Seedat, S.; Robinson, M.; Hemmings, S.M. Shedding Light on the Transcriptomic Dark Matter in Biological Psychiatry: Role of Long Noncoding RNAs in D-cycloserine-Induced Fear Extinction in Posttraumatic Stress Disorder. OMICS J. Integr. Biol. 2020, 24, 352–369. [Google Scholar] [CrossRef]
  50. Muhie, S.; Gautam, A.; Meyerhoff, J.; Chakraborty, N.; Hammamieh, R.; Jett, M. Brain transcriptome profiles in mouse model simulating features of post-traumatic stress disorder. Mol. Brain 2015, 8, 14. [Google Scholar] [CrossRef] [Green Version]
  51. Muhie, S.; Gautam, A.; Chakraborty, N.; Hoke, A.; Meyerhoff, J.; Hammamieh, R.; Jett, M. Molecular indicators of stress-induced neuroinflammation in a mouse model simulating features of post-traumatic stress disorder. Transl. Psychiatry 2017, 7, e1135. [Google Scholar] [CrossRef] [Green Version]
  52. Li, H.; Li, X.; Smerin, S.E.; Zhang, L.; Jia, M.; Xing, G.; Su, Y.A.; Wen, J.; Benedek, D.; Ursano, R. Mitochondrial Gene Expression Profiles and Metabolic Pathways in the Amygdala Associated with Exaggerated Fear in an Animal Model of PTSD. Front. Neurol. 2014, 5, 164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Snijders, C.; PGC PTSD EWAS Consortium; Maihofer, A.X.; Ratanatharathorn, A.; Baker, D.G.; Boks, M.P.; Geuze, E.; Jain, S.; Kessler, R.C.; Pishva, E.; et al. Longitudinal epigenome-wide association studies of three male military cohorts reveal multiple CpG sites associated with post-traumatic stress disorder. Clin. Epigenetics 2020, 12, 11. [Google Scholar] [CrossRef] [PubMed]
  54. Hori, H.; Yoshida, F.; Itoh, M.; Lin, M.; Niwa, M.; Ino, K.; Imai, R.; Ogawa, S.; Matsui, M.; Kamo, T.; et al. Proinflammatory status-stratified blood transcriptome profiling of civilian women with PTSD. Psychoneuroendocrinology 2020, 111, 104491. [Google Scholar] [CrossRef]
  55. Rusch, H.L.; Robinson, J.; Yun, S.; Osier, N.D.; Martin, C.; Brewin, C.R.; Gill, J.M. Gene expression differences in PTSD are uniquely related to the intrusion symptom cluster: A transcriptome-wide analysis in military service members. Brain Behav. Immun. 2019, 80, 904–908. [Google Scholar] [CrossRef] [PubMed]
  56. Mehta, D.D.; Bruenig, D.; Carrillo-Roa, T.; Lawford, B.; Harvey, W.; Morris, C.P.; Smith, A.K.; Binder, E.B.; Young, R.; Voisey, J. Genomewide DNA methylation analysis in combat veterans reveals a novel locus for PTSD. Acta Psychiatr. Scand. 2017, 136, 493–505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Kuan, P.-F.; Waszczuk, M.A.; Kotov, R.; Clouston, S.; Yang, X.; Singh, P.K.; Glenn, S.T.; Gomez, E.C.; Wang, J.; Bromet, E.; et al. Gene expression associated with PTSD in World Trade Center responders: An RNA sequencing study. Transl. Psychiatry 2017, 7, 1297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Torshizi, A.D.; Wang, K. Deconvolution of Transcriptional Networks in Post-Traumatic Stress Disorder Uncovers Master Regulators Driving Innate Immune System Function. Sci. Rep. 2017, 7, 14486. [Google Scholar] [CrossRef] [Green Version]
  59. Breen, M.S.; Maihofer, A.X.; Glatt, S.; Tylee, D.S.; Chandler, S.D.; Tsuang, M.T.; Risbrough, V.B.; Baker, D.G.; O’Connor, D.T.; Nievergelt, C.M.; et al. Gene networks specific for innate immunity define post-traumatic stress disorder. Mol. Psychiatry 2015, 20, 1538–1545. [Google Scholar] [CrossRef] [Green Version]
  60. Breen, M.S.; Tylee, D.S.; Maihofer, A.X.; Neylan, T.C.; Mehta, D.; Binder, E.B.; Chandler, S.D.; Hess, J.L.; Kremen, W.S.; Risbrough, V.B.; et al. PTSD Blood Transcriptome Mega-Analysis: Shared Inflammatory Pathways across Biological Sex and Modes of Trauma. Neuropsychopharmacology 2018, 43, 469–481. [Google Scholar] [CrossRef] [Green Version]
  61. Bam, M.; Yang, X.; Zumbrun, E.E.; Zhong, Y.; Zhou, J.; Ginsberg, J.P.; Leyden, Q.; Zhang, J.; Nagarkatti, P.S.; Nagarkatti, M. Dysregulated immune system networks in war veterans with PTSD is an outcome of altered miRNA expression and DNA methylation. Sci. Rep. 2016, 6, 31209. [Google Scholar] [CrossRef]
  62. Guardado, P.; Olivera, A.; Rusch, H.L.; Roy, M.; Martin, C.; Lejbman, N.; Lee, H.; Gill, J.M. Altered gene expression of the innate immune, neuroendocrine, and nuclear factor-kappa B (NF-κB) systems is associated with posttraumatic stress disorder in military personnel. J. Anxiety Disord. 2016, 38, 9–20. [Google Scholar] [CrossRef] [PubMed]
  63. Montalvo-Ortiz, J.L.; Gelernter, J.; Cheng, Z.; Girgenti, M.J.; Xu, K.; Zhang, X.; Gopalan, S.; Zhou, H.; Duman, R.S.; Southwick, S.M.; et al. Epigenome-wide association study of posttraumatic stress disorder identifies novel loci in U.S. military veterans. Transl. Psychiatry 2022, 12, 65. [Google Scholar] [CrossRef] [PubMed]
  64. Uddin, M.; Aiello, A.E.; Wildman, D.E.; Koenen, K.C.; Pawelec, G.; Santos, R.D.L.; Goldmann, E.; Galea, S. Epigenetic and immune function profiles associated with posttraumatic stress disorder. Proc. Natl. Acad. Sci. USA 2010, 107, 9470–9475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Smith, A.K.; Conneely, K.N.; Kilaru, V.; Mercer, K.B.; Weiss, T.E.; Bradley, B.; Tang, Y.; Gillespie, C.; Cubells, J.F.; Ressler, K.J. Differential immune system DNA methylation and cytokine regulation in post-traumatic stress disorder. Am. J. Med Genet. Part B Neuropsychiatr. Genet. 2011, 156, 700–708. [Google Scholar] [CrossRef] [Green Version]
  66. Katrinli, S.; Maihofer, A.X.; Wani, A.H.; Pfeiffer, J.R.; Ketema, E.; Ratanatharathorn, A.; Baker, D.G.; Boks, M.P.; Geuze, E.; Kessler, R.C.; et al. 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, in press. [Google Scholar] [CrossRef]
  67. Kuan, P.-F.; Waszczuk, M.A.; Kotov, R.; Marsit, C.; Guffanti, G.; Gonzalez, A.; Yang, X.; Koenen, K.; Bromet, E.; Luft, B.J. An epigenome-wide DNA methylation study of PTSD and depression in World Trade Center responders. Transl. Psychiatry 2017, 7, e1158. [Google Scholar] [CrossRef]
  68. Kuan, P.-F.; Yang, X.; Clouston, S.; Ren, X.; Kotov, R.; Waszczuk, M.; Singh, P.K.; Glenn, S.T.; Gomez, E.C.; Wang, J.; et al. Cell type-specific gene expression patterns associated with posttraumatic stress disorder in World Trade Center responders. Transl. Psychiatry 2019, 9, 1. [Google Scholar] [CrossRef]
  69. Kuan, P.-F.; Yang, X.; Ren, X.; Che, C.; Waszczuk, M.; Kotov, R.; Clouston, S.; Singh, P.K.; Glenn, S.T.; Gomez, E.C.; et al. Mapping the transcriptomics landscape of post-traumatic stress disorder symptom dimensions in World Trade Center responders. Transl. Psychiatry 2021, 11, 310. [Google Scholar] [CrossRef]
  70. Uddin, M.; Ratanatharathorn, A.; Armstrong, D.; Kuan, P.-F.; Aiello, A.E.; Bromet, E.J.; Galea, S.; Koenen, K.C.; Luft, B.; Ressler, K.J.; et al. Epigenetic meta-analysis across three civilian cohorts identifies NRG1 and HGS as blood-based biomarkers for post-traumatic stress disorder. Epigenomics 2018, 10, 1585–1601. [Google Scholar] [CrossRef]
  71. Smith, A.K.; INTRuST Clinical Consortium; Ratanatharathorn, A.; Maihofer, A.X.; Naviaux, R.K.; Aiello, A.E.; Amstadter, A.B.; Ashley-Koch, A.E.; Baker, D.G.; Beckham, J.C.; et al. Epigenome-wide meta-analysis of PTSD across 10 military and civilian cohorts identifies methylation changes in AHRR. Nat. Commun. 2020, 11, 5965. [Google Scholar] [CrossRef]
  72. Bam, M.; Yang, X.; Busbee, B.P.; Aiello, A.E.; Uddin, M.; Ginsberg, J.P.; Galea, S.; Nagarkatti, P.S.; Nagarkatti, M. Increased H3K4me3 methylation and decreased miR-7113-5p expression lead to enhanced Wnt/β-catenin signaling in immune cells from PTSD patients leading to inflammatory phenotype. Mol. Med. 2020, 26, 110. [Google Scholar] [CrossRef] [PubMed]
  73. Mehta, D.; Voisey, J.; Bruenig, D.; Harvey, W.; Morris, C.P.; Lawford, B.; Young, R. Transcriptome analysis reveals novel genes and immune networks dysregulated in veterans with PTSD. Brain, Behav. Immun. 2018, 74, 133–142. [Google Scholar] [CrossRef] [PubMed]
  74. Szklarczyk, D.; Gable, A.L.; Lyon, D.; Junge, A.; Wyder, S.; Huerta-Cepas, J.; Simonovic, M.; Doncheva, N.T.; Morris, J.H.; Bork, P.; et al. STRING v11: Protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019, 47, D607–D613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Zhou, Y.; Zhou, B.; Pache, L.; Chang, M.; Khodabakhshi, A.H.; Tanaseichuk, O.; Benner, C.; Chanda, S.K. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 2019, 10, 1523. [Google Scholar] [CrossRef] [PubMed]
  76. Fuster, J.J.; Walsh, K. The Good, the Bad, and the Ugly of interleukin-6 signaling. EMBO J. 2014, 33, 1425–1427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Tanaka, T.; Narazaki, M.; Kishimoto, T. IL-6 in Inflammation, Immunity, and Disease. Cold Spring Harb. Perspect. Biol. 2014, 6, a016295. [Google Scholar] [CrossRef]
  78. Gabay, C. Interleukin-6 and chronic inflammation. Arthritis Res. Ther. 2006, 8, S3. [Google Scholar] [CrossRef] [Green Version]
  79. Passos, I.C.; Vasconcelos-Moreno, M.P.; Costa, L.G.; Kunz, M.; Brietzke, E.; Quevedo, J.; Salum, G.; Magalhães, P.V.; Kapczinski, F.; Kauer-Sant’Anna, M. Inflammatory markers in post-traumatic stress disorder: A systematic review, meta-analysis, and meta-regression. Lancet Psychiatry 2015, 2, 1002–1012. [Google Scholar] [CrossRef]
  80. Friedman, M.J.; The Traumatic Stress Brain Research Group; Huber, B.R.; Brady, C.B.; Ursano, R.J.; Benedek, D.M.; Kowall, N.W.; McKee, A.C. VA’s National PTSD Brain Bank: A National Resource for Research. Curr. Psychiatry Rep. 2017, 19, 73. [Google Scholar] [CrossRef]
  81. Kuan, P.-F.; Ren, X.; Clouston, S.; Yang, X.; Jonas, K.; Kotov, R.; Bromet, E.; Luft, B.J. PTSD is associated with accelerated transcriptional aging in World Trade Center responders. Transl. Psychiatry 2021, 11, 311. [Google Scholar] [CrossRef]
  82. Morrison, F.G.; Miller, M.W.; Wolf, E.J.; Logue, M.W.; Maniates, H.; Kwasnik, D.; Cherry, J.D.; Svirsky, S.; Restaino, A.; Hildebrandt, A.; et al. Reduced interleukin 1A gene expression in the dorsolateral prefrontal cortex of individuals with PTSD and depression. Neurosci. Lett. 2018, 692, 204–209. [Google Scholar] [CrossRef] [PubMed]
  83. Dinarello, C.A. Overview of the interleukin-1 family of ligands and receptors. Semin. Immunol. 2013, 25, 389–393. [Google Scholar] [CrossRef] [PubMed]
  84. Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef] [Green Version]
  85. Shaftel, S.S.; Griffin, W.S.T.; O’Banion, M.K. The role of interleukin-1 in neuroinflammation and Alzheimer disease: An evolving perspective. J. Neuroinflamm. 2008, 5, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Lee, W.-H.; Seo, D.; Lim, S.-G.; Suk, K. Reverse Signaling of Tumor Necrosis Factor Superfamily Proteins in Macrophages and Microglia: Superfamily Portrait in the Neuroimmune Interface. Front. Immunol. 2019, 10, 262. [Google Scholar] [CrossRef] [Green Version]
  87. Ubogu, E.E.; Callahan, M.K.; Tucky, B.H.; Ransohoff, R.M. Determinants of CCL5-driven mononuclear cell migration across the blood–brain barrier. Implications for therapeutically modulating neuroinflammation. J. Neuroimmunol. 2006, 179, 132–144. [Google Scholar] [CrossRef]
  88. Lanfranco, M.F.; Mocchetti, I.; Burns, M.P.; Villapol, S. Glial- and Neuronal-Specific Expression of CCL5 mRNA in the Rat Brain. Front. Neuroanat. 2018, 11, 137. [Google Scholar] [CrossRef] [Green Version]
  89. Cimino, P.J.; Sokal, I.; Leverenz, J.; Fukui, Y.; Montine, T.J. DOCK2 Is a Microglial Specific Regulator of Central Nervous System Innate Immunity Found in Normal and Alzheimer’s Disease Brain. Am. J. Pathol. 2009, 175, 1622–1630. [Google Scholar] [CrossRef] [Green Version]
  90. Shi, L. Dock protein family in brain development and neurological disease. Commun. Integr. Biol. 2013, 6, e26839. [Google Scholar] [CrossRef] [Green Version]
  91. Dejean, A.S.; Beisner, D.R.; Ch’En, I.L.; Kerdiles, Y.; Babour, A.; Arden, K.C.; Castrillon, D.H.; DePinho, R.; Hedrick, S.M. Transcription factor Foxo3 controls the magnitude of T cell immune responses by modulating the function of dendritic cells. Nat. Immunol. 2009, 10, 504–513. [Google Scholar] [CrossRef] [Green Version]
  92. Bouzeyen, R.; Haoues, M.; Barbouche, M.-R.; Singh, R.; Essafi, M. FOXO3 Transcription Factor Regulates IL-10 Expression in Mycobacteria-Infected Macrophages, Tuning Their Polarization and the Subsequent Adaptive Immune Response. Front. Immunol. 2019, 10, 2922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Matosin, N.; Arloth, J.; Martinelli, S.; Czamara, D.; Maitra, M.; Halldorsdottir, T.; Cruceanu, C.; Kaul, D.; Gassen, N.C.; Hafner, K.; et al. Brain expressed FKBP5 delineates a therapeutic subtype of severe mental illness. bioRxiv 2021. [Google Scholar] [CrossRef]
  94. Criado-Marrero, M.; Smith, T.M.; Gould, L.A.; Kim, S.; Penny, H.J.; Sun, Z.; Gulick, D.; Dickey, C.A.; Blair, L.J. FKBP5 and early life stress affect the hippocampus by an age-dependent mechanism. Brain, Behav. Immun. Health 2020, 9, 100143. [Google Scholar] [CrossRef] [PubMed]
  95. Najjar, S.; Pearlman, D.M.; Alper, K.; Najjar, A.; Devinsky, O. Neuroinflammation and psychiatric illness. J. Neuroinflamm. 2013, 10, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Haroon, E.; Miller, A.H.; Sanacora, G. Inflammation, Glutamate, and Glia: A Trio of Trouble in Mood Disorders. Neuropsychopharmacology 2017, 42, 193–215. [Google Scholar] [CrossRef]
  97. Wolf, E.J.; Traumatic Stress Brain Research Group; Chen, C.-D.; Zhao, X.; Zhou, Z.; Morrison, F.G.; Daskalakis, N.P.; Stone, A.; Schichman, S.; Grenier, J.G.; et al. Klotho, PTSD, and advanced epigenetic age in cortical tissue. Neuropsychopharmacology 2021, 46, 721–730. [Google Scholar] [CrossRef]
  98. Zhou, H.; Pu, S.; Zhou, H.; Guo, Y. Klotho as Potential Autophagy Regulator and Therapeutic Target. Front. Pharmacol. 2021, 12, 755366. [Google Scholar] [CrossRef]
  99. Zhu, L.; Stein, L.R.; Kim, D.; Ho, K.; Yu, G.-Q.; Zhan, L.; Larsson, T.E.; Mucke, L. Klotho controls the brain–immune system interface in the choroid plexus. Proc. Natl. Acad. Sci. USA 2018, 115, E11388–E11396. [Google Scholar] [CrossRef] [Green Version]
  100. Kotas, M.E.; Medzhitov, R. Homeostasis, Inflammation, and Disease Susceptibility. Cell 2015, 160, 816–827. [Google Scholar] [CrossRef] [Green Version]
  101. Toft, H.; Bramness, J.G.; Lien, L.; Abebe, D.S.; Wampold, B.E.; Tilden, T.; Hestad, K.; Neupane, S.P. PTSD patients show increasing cytokine levels during treatment despite reduced psychological distress. Neuropsychiatr. Dis. Treat. 2018, 14, 2367–2378. [Google Scholar] [CrossRef] [Green Version]
  102. Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W.; et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef] [PubMed]
  103. Zhang, L.; Team, B.; Hu, X.-Z.; Li, X.; Chen, Z.; Benedek, D.M.; Fullerton, C.S.; Wynn, G.; Ursano, R.J.; Naifeh, J.A. Potential chemokine biomarkers associated with PTSD onset, risk and resilience as well as stress responses in US military service members. Transl. Psychiatry 2020, 10, 31. [Google Scholar] [CrossRef] [PubMed]
  104. Kim, T.D.; Lee, S.; Yoon, S. Inflammation in Post-Traumatic Stress Disorder (PTSD): A Review of Potential Correlates of PTSD with a Neurological Perspective. Antioxidants 2020, 9, 107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Gill, J.M.; Saligan, L.; Woods, S.; Page, G. PTSD is Associated With an Excess of Inflammatory Immune Activities. Perspect. Psychiatr. Care 2009, 45, 262–277. [Google Scholar] [CrossRef] [PubMed]
  106. Dowlati, Y.; Herrmann, N.; Swardfager, W.; Liu, H.; Sham, L.; Reim, E.K.; Lanctôt, K.L. A Meta-Analysis of Cytokines in Major Depression. Biol. Psychiatry 2010, 67, 446–457. [Google Scholar] [CrossRef]
  107. Wohleb, E.S.; McKim, D.B.; Sheridan, J.F.; Godbout, J.P. Monocyte trafficking to the brain with stress and inflammation: A novel axis of immune-to-brain communication that influences mood and behavior. Front. Neurosci. 2015, 8, 447. [Google Scholar] [CrossRef]
  108. Maes, M.; Meltzer, H.Y.; Bosmans, E.; Bergmans, R.; Vandoolaeghe, E.; Ranjan, R.; Desnyder, R. Increased plasma concentrations of interleukin-6, soluble interleukin-6, soluble interleukin-2 and transferrin receptor in major depression. J. Affect. Disord. 1995, 34, 301–309. [Google Scholar] [CrossRef]
  109. Maes, M.; Bosmans, E.; De Jongh, R.; Kenis, G.; Vandoolaeghe, E.; Neels, H. Increased serum IL-6 and IL-1 receptor antagonist concentrations in major depression and treatment resistant depression. Cytokine 1997, 9, 853–858. [Google Scholar] [CrossRef]
  110. Hodes, G.E.; Pfau, M.L.; Leboeuf, M.; Golden, S.A.; Christoffel, D.J.; Bregman, D.; Rebusi, N.; Heshmati, M.; Aleyasin, H.; Warren, B.L.; et al. Individual differences in the peripheral immune system promote resilience versus susceptibility to social stress. Proc. Natl. Acad. Sci. USA 2014, 111, 16136–16141. [Google Scholar] [CrossRef] [Green Version]
  111. Cohen, H.; Ziv, Y.; Cardon, M.; Kaplan, Z.; Matar, M.A.; Gidron, Y.; Schwartz, M.; Kipnis, J. Maladaptation to mental stress mitigated by the adaptive immune system via depletion of naturally occurring regulatory CD4+CD25+ cells. J. Neurobiol. 2006, 66, 552–563. [Google Scholar] [CrossRef]
  112. Jergović, M.; Bendelja, K.; Vidović, A.; Savić, A.; Vojvoda, V.; Aberle, N.; Rabatić, S.; Jovanovic, T.; Sabioncello, A. Patients with posttraumatic stress disorder exhibit an altered phenotype of regulatory T cells. Allergy, Asthma Clin. Immunol. 2014, 10, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Inoue-Sakurai, C.; Maruyama, S.; Morimoto, K. Posttraumatic Stress and Lifestyles Are Associated with Natural Killer Cell Activity in Victims of the Hanshin-Awaji Earthquake in Japan. Prev. Med. 2000, 31, 467–473. [Google Scholar] [CrossRef] [PubMed]
  114. Vidović, A.; Vilibić, M.; Sabioncello, A.; Gotovac, K.; Rabatić, S.; Folnegović-Smalc, V.; Dekaris, D. Circulating lymphocyte subsets, natural killer cell cytotoxicity, and components of hypothalamic-pituitary-adrenal axis in Croatian war veterans with posttraumatic stress disorder: Cross-sectional study. Croat. Med. J. 2007, 48, 198–206. [Google Scholar] [PubMed]
  115. Bersani, F.S.; Wolkowitz, O.M.; Milush, J.M.; Sinclair, E.; Eppling, L.; Aschbacher, K.; Lindqvist, D.; Yehuda, R.; Flory, J.; Bierer, L.M.; et al. A population of atypical CD56−CD16+ natural killer cells is expanded in PTSD and is associated with symptom severity. Brain Behav. Immun. 2016, 56, 264–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Aiello, A.E.; Dowd, J.B.; Jayabalasingham, B.; Feinstein, L.; Uddin, M.; Simanek, A.M.; Cheng, C.K.; Galea, S.; Wildman, D.E.; Koenen, K.; et al. PTSD is associated with an increase in aged T cell phenotypes in adults living in Detroit. Psychoneuroendocrinology 2016, 67, 133–141. [Google Scholar] [CrossRef] [Green Version]
  117. Gotovac, K.; Vidović, A.; Vukušić, H.; Krčmar, T.; Sabioncello, A.; Rabatić, S.; Dekaris, D. Natural killer cell cytotoxicity and lymphocyte perforin expression in veterans with posttraumatic stress disorder. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2010, 34, 597–604. [Google Scholar] [CrossRef]
  118. Kim, G.S.; Smith, A.K.; Xue, F.; Michopoulos, V.; Lori, A.; Armstrong, D.L.; Aiello, A.E.; Koenen, K.C.; Galea, S.; Wildman, D.E.; et al. Methylomic profiles reveal sex-specific differences in leukocyte composition associated with post-traumatic stress disorder. Brain Behav. Immun. 2019, 81, 280–291. [Google Scholar] [CrossRef]
  119. Brahmajothi, M.V.; Abou-Donia, M.B. PTSD Susceptibility and Challenges: Pathophysiological Consequences of Behavioral Symptoms. Mil. Med. 2020, 185, 279–285. [Google Scholar] [CrossRef] [Green Version]
  120. Lerman, I.; Davis, B.A.; Bertram, T.M.; Proudfoot, J.; Hauger, R.L.; Coe, C.L.; Patel, P.M.; Baker, D.G. Posttraumatic stress disorder influences the nociceptive and intrathecal cytokine response to a painful stimulus in combat veterans. Psychoneuroendocrinology 2016, 73, 99–108. [Google Scholar] [CrossRef] [Green Version]
  121. Fattori, V.; Hohmann, M.S.N.; Rossaneis, A.C.; Pinho-Ribeiro, F.A.; Verri, W.A. Capsaicin: Current Understanding of Its Mechanisms and Therapy of Pain and Other Pre-Clinical and Clinical Uses. Molecules 2016, 21, 844. [Google Scholar] [CrossRef] [Green Version]
  122. Pietrzak, R.H.; Goldstein, R.; Southwick, S.M.; Grant, B.F. Prevalence and Axis I comorbidity of full and partial posttraumatic stress disorder in the United States: Results from Wave 2 of the National Epidemiologic Survey on Alcohol and Related Conditions. J. Anxiety Disord. 2011, 25, 456–465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Cavalcanti-Ribeiro, P.; Andrade-Nascimento, M.; Morais-De-Jesus, M.; De Medeiros, G.M.; de Oliveira, R.D.; Conceicao, J.O.; Rocha, M.F.; Miranda-Scippa, A.; Koenen, K.C.; Quarantini, L.C. Post-traumatic stress disorder as a comorbidity: Impact on disease outcomes. Expert Rev. Neurother. 2012, 12, 1023–1037. [Google Scholar] [CrossRef] [PubMed]
  124. Stone, L.A.; Girgenti, M.J.; Wang, J.; Ji, D.; Zhao, H.; Krystal, J.H.; Duman, R.S. Cortical Transcriptomic Alterations in Association With Appetitive Neuropeptides and Body Mass Index in Posttraumatic Stress Disorder. Int. J. Neuropsychopharmacol. 2021, 24, 118–129. [Google Scholar] [CrossRef] [PubMed]
  125. Van den Berk-Clark, C.; Secrest, S.; Walls, J.; Hallberg, E.; Lustman, P.J.; Schneider, F.D.; Scherrer, J.F. Association between posttraumatic stress disorder and lack of exercise, poor diet, obesity, and co-occuring smoking: A systematic review and meta-analysis. Health Psychol. 2018, 37, 407–416. [Google Scholar] [CrossRef] [PubMed]
  126. Borovcanin, M.M.; Jovanovic, I.; Radosavljevic, G.; Pantic, J.; Minic Janicijevic, S.; Arsenijevic, N.; Lukic, M.L. Interleukin-6 in Schizophrenia—Is There a Therapeutic Relevance? Front. Psychiatry 2017, 8, 221. [Google Scholar] [CrossRef] [PubMed]
  127. Blom, E.S.; Giedraitis, V.; Zetterberg, H.; Fukumoto, H.; Blennow, K.; Hyman, B.T.; Irizarry, M.C.; Wahlund, L.-O.; Lannfelt, L.; Ingelsson, M. Rapid Progression from Mild Cognitive Impairment to Alzheimer’s Disease in Subjects with Elevated Levels of Tau in Cerebrospinal Fluid and the APOE ε4/ε4 Genotype. Dement. Geriatr. Cogn. Disord. 2009, 27, 458–464. [Google Scholar] [CrossRef]
  128. Nordberg, A.; Carter, S.F.; Rinne, J.; Drzezga, A.; Brooks, D.J.; Vandenberghe, R.; Perani, D.; Forsberg, A.; Långström, B.; Scheinin, N.; et al. A European multicentre PET study of fibrillar amyloid in Alzheimer’s disease. Eur. J. Pediatr. 2013, 40, 104–114. [Google Scholar] [CrossRef] [Green Version]
  129. Masters, C.L.; Bateman, R.; Blennow, K.; Rowe, C.C.; Sperling, R.A.; Cummings, J.L. Alzheimer’s disease. Nat. Rev. Dis. Prim. 2015, 1, 15056. [Google Scholar] [CrossRef]
  130. Rowe, C.C.; Bourgeat, P.; Ellis, K.A.; Brown, B.; Lim, Y.Y.; Mulligan, R.; Jones, G.; Maruff, P.; Woodward, M.; Price, R.; et al. Predicting Alzheimer disease with β-amyloid imaging: Results from the Australian imaging, biomarkers, and lifestyle study of ageing. Ann. Neurol. 2013, 74, 905–913. [Google Scholar] [CrossRef]
  131. Diminich, E.D.; Clouston, S.A.P.; Kranidis, A.; Kritikos, M.; Kotov, R.; Kuan, P.; Carr, M.; Bromet, E.J.; Luft, B.J. Chronic Posttraumatic Stress Disorder and Comorbid Cognitive and Physical Impairments in World Trade Center Responders. J. Trauma. Stress 2021, 34, 616–627. [Google Scholar] [CrossRef]
  132. Bonanni, L.; Franciotti, R.; Martinotti, G.; Vellante, F.; Flacco, M.E.; Di Giannantonio, M.; Thomas, A.; Onofrj, M. Post Traumatic Stress Disorder Heralding the Onset of Semantic Frontotemporal Dementia. J. Alzheimers Dis. 2018, 63, 203–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Kuan, P.-F.; Clouston, S.; Yang, X.; Kotov, R.; Bromet, E.; Luft, B.J. Molecular linkage between post-traumatic stress disorder and cognitive impairment: A targeted proteomics study of World Trade Center responders. Transl. Psychiatry 2020, 10, 269. [Google Scholar] [CrossRef] [PubMed]
  134. Frenkel, D.; Wilkinson, K.; Zhao, L.; Hickman, S.E.; Means, T.K.; Puckett, L.; Farfara, D.; Kingery, N.D.; Weiner, H.L.; El Khoury, J. Scara1 deficiency impairs clearance of soluble amyloid-β by mononuclear phagocytes and accelerates Alzheimer’s-like disease progression. Nat. Commun. 2013, 4, 2030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Dean, K.R.; The PTSD Systems Biology Consortium; Hammamieh, R.; Mellon, S.H.; Abu-Amara, D.; Flory, J.D.; Guffanti, G.; Wang, K.; Daigle, B.J., Jr.; Gautam, A.; et al. Multi-omic biomarker identification and validation for diagnosing warzone-related post-traumatic stress disorder. Mol. Psychiatry 2020, 25, 3337–3349. [Google Scholar] [CrossRef]
  136. Wu, Z.; Ni, J.; Liu, Y.; Teeling, J.; Takayama, F.; Collcutt, A.; Ibbett, P.; Nakanishi, H. Cathepsin B plays a critical role in inducing Alzheimer’s disease-like phenotypes following chronic systemic exposure to lipopolysaccharide from Porphyromonas gingivalis in mice. Brain Behav. Immun. 2017, 65, 350–361. [Google Scholar] [CrossRef]
  137. Grupe, A.; Abraham, R.; Li, Y.; Rowland, C.; Hollingworth, P.; Morgan, A.; Jehu, L.; Segurado, R.; Stone, D.; Schadt, E.; et al. Evidence for novel susceptibility genes for late-onset Alzheimer’s disease from a genome-wide association study of putative functional variants. Hum. Mol. Genet. 2007, 16, 865–873. [Google Scholar] [CrossRef]
  138. Wang, Z.; Caughron, B.; Young, M.R.I. Posttraumatic Stress Disorder: An Immunological Disorder? Front. Psychiatry 2017, 8, 222. [Google Scholar] [CrossRef] [Green Version]
  139. Michopoulos, V.; Vester, A.; Neigh, G. Posttraumatic stress disorder: A metabolic disorder in disguise? Exp. Neurol. 2016, 284, 220–229. [Google Scholar] [CrossRef] [Green Version]
  140. Mokhtari, A.; Porte, B.; Belzeaux, R.; Etain, B.; Ibrahim, E.C.; Marie-Claire, C.; Lutz, P.-E.; Delahaye-Duriez, A. The molecular pathophysiology of mood disorders: From the analysis of single molecular layers to multi-omic integration. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2022, 116, 110520. [Google Scholar] [CrossRef]
  141. Filiou, M.D.; Teplytska, L.; Nussbaumer, M.; Otte, D.-M.; Zimmer, A.; Turck, C.W. Multi-Omics Analysis Reveals Myelin, Presynaptic and Nicotinate Alterations in the Hippocampus of G72/G30 Transgenic Mice. J. Pers. Med. 2022, 12, 244. [Google Scholar] [CrossRef]
  142. Pathak, G.A.; Singh, K.; Wendt, F.R.; Fleming, T.W.; Overstreet, C.; Koller, D.; Tylee, D.S.; De Angelis, F.; Mendoza, B.C.; Levey, D.F.; et al. Genetically regulated multi-omics study for symptom clusters of posttraumatic stress disorder highlights pleiotropy with hematologic and cardio-metabolic traits. Mol. Psychiatry 2022, in press. [Google Scholar] [CrossRef] [PubMed]
  143. Oron, O.; Elliott, E. Delineating the Common Biological Pathways Perturbed by ASD’s Genetic Etiology: Lessons from Network-Based Studies. Int. J. Mol. Sci. 2017, 18, 828. [Google Scholar] [CrossRef] [PubMed]
  144. Ma, C.; Gu, C.; Huo, Y.; Li, X.; Luo, X.-J. The integrated landscape of causal genes and pathways in schizophrenia. Transl. Psychiatry 2018, 8, 67. [Google Scholar] [CrossRef] [PubMed]
  145. Neigh, G.N.; Ali, F.F. Co-morbidity of PTSD and immune system dysfunction: Opportunities for treatment. Curr. Opin. Pharmacol. 2016, 29, 104–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Dallacasagrande, V.; Hajjar, K.A. Annexin A2 in Inflammation and Host Defense. Cells 2020, 9, 1499. [Google Scholar] [CrossRef] [PubMed]
  147. Lim, H.I.; Hajjar, K.A. Annexin A2 in Fibrinolysis, Inflammation and Fibrosis. Int. J. Mol. Sci. 2021, 22, 6836. [Google Scholar] [CrossRef]
  148. Hou, X.; Li, Y.; Huang, Y.; Zhao, H.; Gui, L. Adenosine Receptor A1-A2a Heteromers Regulate EAAT2 Expression and Glutamate Uptake via YY1-Induced Repression of PPAR γ Transcription. PPAR Res. 2020, 2020, 2410264. [Google Scholar] [CrossRef] [Green Version]
  149. McKenzie, A.T.; Wang, M.; Hauberg, M.E.; Fullard, J.F.; Kozlenkov, A.; Keenan, A.; Hurd, Y.L.; Dracheva, S.; Casaccia, P.; Roussos, P.; et al. Brain Cell Type Specific Gene Expression and Co-expression Network Architectures. Sci. Rep. 2018, 8, 8868. [Google Scholar] [CrossRef] [Green Version]
  150. Besedovsky, H.O.; del Rey, A. Central and Peripheral Cytokines Mediate Immune-Brain Connectivity. Neurochem. Res. 2011, 36, 1–6. [Google Scholar] [CrossRef]
  151. Schwartz, M.; Kipnis, J.; Rivest, S.; Prat, A. How Do Immune Cells Support and Shape the Brain in Health, Disease, and Aging? J. Neurosci. 2013, 33, 17587–17596. [Google Scholar] [CrossRef] [Green Version]
  152. Novellino, F.; Saccà, V.; Donato, A.; Zaffino, P.; Spadea, M.F.; Vismara, M.; Arcidiacono, B.; Malara, N.; Presta, I.; Donato, G. Innate Immunity: A Common Denominator between Neurodegenerative and Neuropsychiatric Diseases. Int. J. Mol. Sci. 2020, 21, 1115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Gong, T.; Liu, L.; Jiang, W.; Zhou, R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat. Rev. Immunol. 2020, 20, 95–112. [Google Scholar] [CrossRef] [PubMed]
  154. Roh, J.S.; Sohn, D.H. Damage-Associated Molecular Patterns in Inflammatory Diseases. Immune Netw. 2018, 18, e27. [Google Scholar] [CrossRef] [PubMed]
  155. Jones, K.A.; Thomsen, C. The role of the innate immune system in psychiatric disorders. Mol. Cell. Neurosci. 2013, 53, 52–62. [Google Scholar] [CrossRef]
  156. Zass, L.J.; Hart, S.A.; Seedat, S.; Hemmings, S.M.J.; Malan-Müller, S. Neuroinflammatory genes associated with post-traumatic stress disorder. Psychiatr. Genet. 2017, 27, 1–16. [Google Scholar] [CrossRef]
  157. Kempuraj, D.; Thangavel, R.; Selvakumar, G.P.; Zaheer, S.; Ahmed, M.E.; Raikwar, S.P.; Zahoor, H.; Saeed, D.; Natteru, P.A.; Iyer, S.; et al. Brain and Peripheral Atypical Inflammatory Mediators Potentiate Neuroinflammation and Neurodegeneration. Front. Cell. Neurosci. 2017, 11, 216. [Google Scholar] [CrossRef]
  158. Shigemoto-Mogami, Y.; Hoshikawa, K.; Goldman, J.E.; Sekino, Y.; Sato, K. Microglia Enhance Neurogenesis and Oligodendrogenesis in the Early Postnatal Subventricular Zone. J. Neurosci. 2014, 34, 2231–2243. [Google Scholar] [CrossRef] [Green Version]
  159. Lenz, K.M.; Nelson, L. Microglia and Beyond: Innate Immune Cells As Regulators of Brain Development and Behavioral Function. Front. Immunol. 2018, 9, 698. [Google Scholar] [CrossRef] [Green Version]
  160. Lori, A.; Maddox, S.A.; Sharma, S.; Andero, R.; Ressler, K.J.; Smith, A.K. Dynamic Patterns of Threat-Associated Gene Expression in the Amygdala and Blood. Front. Psychiatry 2019, 9, 778. [Google Scholar] [CrossRef]
  161. Aschbacher, K.; Cole, S.; Hagan, M.; Rivera, L.; Baccarella, A.; Wolkowitz, O.M.; Lieberman, A.F.; Bush, N.R. An immunogenomic phenotype predicting behavioral treatment response: Toward precision psychiatry for mothers and children with trauma exposure. Brain Behav. Immun. 2022, 99, 350–362. [Google Scholar] [CrossRef]
  162. Levkovitz, Y.; Fenchel, D.; Kaplan, Z.; Zohar, J.; Cohen, H. Early post-stressor intervention with minocycline, a second-generation tetracycline, attenuates post-traumatic stress response in an animal model of PTSD. Eur. Neuropsychopharmacol. 2015, 25, 124–132. [Google Scholar] [CrossRef] [PubMed]
  163. Freshour, S.L.; Kiwala, S.; Cotto, K.C.; Coffman, A.C.; McMichael, J.F.; Song, J.J.; Griffith, M.; Griffith, O.L.; Wagner, A.H. Integration of the Drug–Gene Interaction Database (DGIdb 4.0) with open crowdsource efforts. Nucleic Acids Res. 2021, 49, D1144–D1151. [Google Scholar] [CrossRef] [PubMed]
  164. Pushpakom, S.; Iorio, F.; Eyers, P.A.; Escott, K.J.; Hopper, S.; Wells, A.; Doig, A.; Guilliams, T.; Latimer, J.; McNamee, C.; et al. Drug repurposing: Progress, challenges and recommendations. Nat. Rev. Drug Discov. 2019, 18, 41–58. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (a) Search and selection procedure of the literature on the immune system in PTSD. By screening by title and abstract, we selected a total of five papers conducted on brain tissues and 27 in peripheral tissues across species. These studies assessed immune-related markers at distinct levels: genetic variants conferring risk to PTSD, transcriptomic and epigenomic changes that occurred after traumatic exposure or associated with trauma exposure or PTSD, and the dysregulation of inflammatory proteins related to the pathophysiology of PTSD. (b) After the selection of immune-related genes reported in the studies, we explored the convergence across species in both central and peripheral tissues in terms of regulatory level (5mC: methylation and DEG: Differentially Expressed Genes). We then explored the convergence between central and peripheral tissues and conducted a GO enrichment analysis, brain cell-type enrichment analysis and drug repurposing analysis to identify potential markers involved in the systemic immune dysregulation in PTSD as well as potential treatments.
Figure 1. (a) Search and selection procedure of the literature on the immune system in PTSD. By screening by title and abstract, we selected a total of five papers conducted on brain tissues and 27 in peripheral tissues across species. These studies assessed immune-related markers at distinct levels: genetic variants conferring risk to PTSD, transcriptomic and epigenomic changes that occurred after traumatic exposure or associated with trauma exposure or PTSD, and the dysregulation of inflammatory proteins related to the pathophysiology of PTSD. (b) After the selection of immune-related genes reported in the studies, we explored the convergence across species in both central and peripheral tissues in terms of regulatory level (5mC: methylation and DEG: Differentially Expressed Genes). We then explored the convergence between central and peripheral tissues and conducted a GO enrichment analysis, brain cell-type enrichment analysis and drug repurposing analysis to identify potential markers involved in the systemic immune dysregulation in PTSD as well as potential treatments.
Biomedicines 10 01107 g001
Figure 2. Depicting systematic immune response in PTSD. The immune signaling pathway is dysregulated in PTSD at multiple regulatory levels. Recent whole genome analysis in PTSD conducted on both human and animal models of PTSD have demonstrated that epigenomic and transcriptomic changes may mediate the development, maintenance, and clinical presentation of PTSD. HPA axis releasing ACHT stimulates cortisol (human (GC) glucocorticoids) production which, in turn, also mediates gene expression and exacerbates immune response in the periphery and brain. In this review, 84 genes that participate in immune signaling processes were commonly reported as differentially expressed or methylated genes in central (brain) and peripheral (such as blood and saliva) tissues across species. ~50% of those convergent genes correspond to brain-cell type specific markers such as astrocytes and microglia, which are discriminated by color. Upon inflammatory response, the blood brain barrier (BBB) may be affected and allow a higher migration of molecules crossing the brain and periphery. Thus, based on the literature review, we propose the existence of common regulatory markers able to cross BBB, impact the expression of those 84 convergent genes and orchestrate systemic immune dysregulation in PTSD through important immunological pathways such as cytokine-cytokine interaction, TNF and NF-κB/JAK/STAT signaling. These common regulatory markers may correspond to damage-associated molecular patterns (DAMPs) which are released from damaged cells and activate the innate immune response. By using drug repurposing analysis, we also suggest that Th17 differentiation may be a possible target mechanism for drug action, as well as Toll-like receptor signaling and cytokine-cytokine receptor interaction, for the treatment of PTSD.
Figure 2. Depicting systematic immune response in PTSD. The immune signaling pathway is dysregulated in PTSD at multiple regulatory levels. Recent whole genome analysis in PTSD conducted on both human and animal models of PTSD have demonstrated that epigenomic and transcriptomic changes may mediate the development, maintenance, and clinical presentation of PTSD. HPA axis releasing ACHT stimulates cortisol (human (GC) glucocorticoids) production which, in turn, also mediates gene expression and exacerbates immune response in the periphery and brain. In this review, 84 genes that participate in immune signaling processes were commonly reported as differentially expressed or methylated genes in central (brain) and peripheral (such as blood and saliva) tissues across species. ~50% of those convergent genes correspond to brain-cell type specific markers such as astrocytes and microglia, which are discriminated by color. Upon inflammatory response, the blood brain barrier (BBB) may be affected and allow a higher migration of molecules crossing the brain and periphery. Thus, based on the literature review, we propose the existence of common regulatory markers able to cross BBB, impact the expression of those 84 convergent genes and orchestrate systemic immune dysregulation in PTSD through important immunological pathways such as cytokine-cytokine interaction, TNF and NF-κB/JAK/STAT signaling. These common regulatory markers may correspond to damage-associated molecular patterns (DAMPs) which are released from damaged cells and activate the innate immune response. By using drug repurposing analysis, we also suggest that Th17 differentiation may be a possible target mechanism for drug action, as well as Toll-like receptor signaling and cytokine-cytokine receptor interaction, for the treatment of PTSD.
Biomedicines 10 01107 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Núñez-Rios, D.L.; Martínez-Magaña, J.J.; Nagamatsu, S.T.; Andrade-Brito, D.E.; Forero, D.A.; Orozco-Castaño, C.A.; Montalvo-Ortiz, J.L. Central and Peripheral Immune Dysregulation in Posttraumatic Stress Disorder: Convergent Multi-Omics Evidence. Biomedicines 2022, 10, 1107. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10051107

AMA Style

Núñez-Rios DL, Martínez-Magaña JJ, Nagamatsu ST, Andrade-Brito DE, Forero DA, Orozco-Castaño CA, Montalvo-Ortiz JL. Central and Peripheral Immune Dysregulation in Posttraumatic Stress Disorder: Convergent Multi-Omics Evidence. Biomedicines. 2022; 10(5):1107. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10051107

Chicago/Turabian Style

Núñez-Rios, Diana L., José J. Martínez-Magaña, Sheila T. Nagamatsu, Diego E. Andrade-Brito, Diego A. Forero, Carlos A. Orozco-Castaño, and Janitza L. Montalvo-Ortiz. 2022. "Central and Peripheral Immune Dysregulation in Posttraumatic Stress Disorder: Convergent Multi-Omics Evidence" Biomedicines 10, no. 5: 1107. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10051107

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop