Document Type : Review Article

Authors

1 Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran

2 Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran

3 Research and Development Center, Shahid Beheshti Hospital, Qom University of Medical Sciences, Qom, Iran

4 1. Brain Mapping Research Center, Research Institute of Functional Neurosurgery, Shahid Beheshti University of Medical Sciences 2. Department of Internal Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran

5 School of Medicine, Tehran University of Medical Sciences, Tehran, Iran

6 1. Research Center for Psychiatry and Behavioral Sciences, Shiraz University of Medical Sciences, Shiraz, Iran 2. Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran

7 Universal Scientific Education and Research Network, Tehran, Iran

8 Trauma Research Center, Shiraz University of Medical Sciences, Shiraz, Iran

Abstract

Objectives: This systematic review explored gene expression and DNA methylation patterns to identify key pathways and molecular targets associated with post-traumatic stress disorder (PTSD), particularly its war-related subtype.
Methods: A comprehensive search of PubMed, Scopus, and Web of Science was conducted using keywords related to PTSD, gene expression, and DNA methylation. Studies published between 2000 to 2024 involving adult military personnel with confirmed PTSD based on the Diagnostic and Statistical Manual of Mental Disorders-5 (DSM-5) criteria were included. Animal studies, psychological interventions, and pharmacological research were excluded. Only cross-sectional, case-control, or cohort studies utilizing blood, saliva, or brain tissue samples were considered. Data from 28 studies were extracted using a predefined framework, focusing on population characteristics, study design, and identified hub genes.
Results: Key findings revealed the upregulation of immune-related genes (e.g., CCL4, NF-κB) and hypomethylation of inflammation-related genes. Downregulation of neurodevelopmental genes, such as Brain-Derived Neurotropic Factor (BDNF) and Down syndrome cell adhesion molecule (DSCAM), highlighted disruptions in synaptic plasticity. The identified pathways suggested potential biomarkers and therapeutic targets for precision medicine approaches.
Conclusion: This review highlighted the role of gene expression alterations in war-related PTSD. The identified genes might serve as candidates for personalized therapies. Further research is required to validate these findings and develop targeted interventions.

Keywords

Introduction

 

Post-traumatic stress disorder (PTSD) is a psychiatric disorder that develops as a result of exposure to life-threatening and traumatic events, such as war, natural disasters, violence, or accidents. Symptoms include intrusive thoughts, physiological changes, nightmares, and flashbacks [1, 2]. Among different subtypes of PTSD, combat-related trauma exhibits unique gene expression patterns, immune dysregulation, and molecular signatures. For example, Logue et al., [3] highlighted that the wound-healing module, which was downregulated in men exposed to combat trauma, differed from pathways activated in cases of interpersonal trauma.

The lifetime prevalence of PTSD in U.S. veterans was 6.9%, with rates as high as 20% among war veterans [4]. Factors contributing to the high prevalence of PTSD in this population included the nature of conflict, the duration of exposure to life-threatening situations, and the severity of injuries sustained during service [5]. Initial estimates suggested that 30% of Vietnam War veterans developed PTSD, but reanalysis indicated a lifetime prevalence of 19 [6]. A decade after the war, 28% of veterans with a history of combat exposure were diagnosed with PTSD. Recent studies highlighted a direct relationship between PTSD prevalence and combat exposure, with an average prevalence of 6% across population samples from various countries and services. The number of PTSD cases among veterans of the Afghanistan and Iraq wars has risen to over 20%, underscoring the need to understand the environmental and genetic mechanisms underlying this disorder [6].

Twins and heritability studies among military personnel showed that 30-70% of PTSD risk variability could be attributed to genetic factors depending on the type of trauma experienced [7, 8]. Advances in transcriptome and gene expression studies have provided new insights into PTSD, but the functional roles of these genetic changes remain poorly understood. Identifying the association between genetic variations and treatment outcomes could reveal novel therapeutic targets. Military personnel and soldiers, due to their frequent exposure to high-stress environments, represent a critical population for PTSD research. The epigenome can adapt to environmental influences through chemical modifications to proteins and chromatin, leading to long-lasting changes in gene expression and regulation. Most epigenetic studies on PTSD have primarily focused on DNA methylation, a key epigenetic process.

Recent hypothesis-free genome-wide, epigenome-wide, and transcriptomic studies have identified several genes associated with immune system functioning, highlighting their potential role in PTSD [9]. However, the specific gene pathways and mechanisms that determine vulnerability or resilience to PTSD following trauma remain largely unexplored [10].

This systematic review and computational analysis aimed to identify hub genes and key pathways involved in combat-related PTSD, with a focus on DNA methylation and gene expression. By deepening our understanding of the epigenetic and transcriptome processes underlying PTSD, this study aimed to identify actionable therapeutic targets and improve treatment outcomes for war veterans.

 

Materials and Methods

 

This systematic literature review was conducted in accordance with the Preferred Reporting Item for Systematic Reviews and Meta-Analyses (PRISMA) guidelines [11].

 

Search Strategy

A comprehensive and systematic search was performed across PubMed, Scopus, and Web of Science databases using the following keywords and Boolean operators:

((“Post-Traumatic Stress Disorder”[MeSH Terms] OR “PTSD” OR “Posttraumatic Stress Disorder” OR “Post Traumatic Stress Disorder”) AND (genetic association [MeSH Terms] OR polymorphism [MeSH Terms] OR genotype [MeSH Terms] OR “gene” OR “genetic” OR “epigenetic” OR “methylation” OR “genetic” OR “genogroup” OR “SNP” OR “single nucleotide polymorphism” OR “single-nucleotide polymorphism”)). All items were detailed in the search strategy figure (Figure 1)

 

Fig. 1. PRISMA flowchart of the selected studies

 

The search was restricted to studies published between 2000 and January 2024, as epigenetic research has gained prominence in the 21st century with the advent of modern assessment techniques. Prior to 2000, most studies relied on targeted candidate gene analysis using qPCR, often with limited sample sizes and lacking the advanced microarray technologies available today [12, 13].

 

Inclusion and Exclusion Criteria

This review included original research papers. The review articles were excluded due to their lack of primary data and subjective nature. Peer-reviewed studies were prioritized, while clinical trials were excluded because of the significant impact of interventions on genomic data. Animal studies were also excluded due to challenges in generalizing findings on gene expression and DNA methylation processes from animals to humans. Studies involving adult military personnel with confirmed PTSD, diagnosed according to standardized criteria, were included. Eligible study designs included cross-sectional, case-control, and both retrospective and prospective cohort studies. Participants with a history of substance abuse or traumatic brain injury (TBI) were excluded, as these factors could confound DNA methylation and gene expression patterns [14, 15]. The PRISMA flowchart detailing the study selection process is presented in Figure 1.

 

Confirmation of PTSD

PTSD diagnosis was confirmed using the Diagnostic and Statistical Manual of Mental Disorders-5 (DSM-5) criteria. Although comorbid psychological conditions such as depression and alcohol dependence often co-occur with PTSD, they were excluded from this review as they fall outside its scope. In studies examining multiple psychological outcomes, only findings related to PTSD were considered.

 

Genomic Outcomes

This review focused on gene expression and DNA methylation patterns. Other genomic or epigenetic pathways, such as genotyping, polymorphism research, histone modification, and epigenetic aging, were excluded. All tissue types used for DNA methylation analysis, including blood and saliva samples, were considered. However, studies examining epigenetic or gene expression changes resulting from psychological interventions or pharmacological treatments were excluded.

 

Study Selection and Data Extraction

Search results were exported into Rayyan, a systematic review tool, to streamline the selection process, manage duplicate articles, and maintain a record of the screening process [16]. The full texts of the identified articles were carefully assessed for adherence to the predefined inclusion criteria. Disagreements during the selection process were resolved through discussion among reviewers. Data were extracted using a predefined data extraction sheet in Microsoft Excel. The following data were extracted from the included studies: author, year of publication, study design, population characteristics, genetic findings, and identified hub genes. Extracted variables also included, but were not limited to these key elements. To assess the validity of the studies, the authors independently evaluated the risk of bias, focusing on the completeness of outcome data and addressing issues such as participant exclusions, attrition, and incomplete outcome data. This study adhered to ethical standards for the analysis of secondary data. No ethical approval was required, as the research utilized publicly available data.

 

Results

 

The systematic review included 28 studies (n=2,677 cases and 4,901 controls) [3, 6, 8, 17-40] investigating gene expression and epigenetic changes in PTSD. The studies primarily employed case-control, cohort, or cross-sectional designs and involved diverse populations, including veterans, military personnel, and trauma-exposed civilians, with a predominance of male participants. Blood samples were the most commonly used biological material for gene expression and methylation analysis.

Among the included studies, 14 focused on DNA methylation, 9 on gene expression, and 6 on both. The study designs comprised 12 case-control studies, 4 cross-sectional studies, and 12 cohort studies. Geographically, 15 studies were conducted in the U.S., 3 in Australia, 3 in the Netherlands, 2 in Serbia, and 1 each in Canada, Germany, and South Korea. Veterans and controls were predominantly men, with some studies excluding female participants entirely. Participant ages ranged widely, with most individuals between 20 and 70 years. All studies utilized blood samples except for Montalvo-Ortiz et al., which examined saliva and postpartum brain samples [30].

The studies collectively highlighted the multifaceted nature of PTSD, driven by immune dysregulation, altered neuroplasticity, and accelerated aging. The identification of biomarkers and pathway-specific targets paved pathways for precision medicine approaches, highlighting the need for continued research to validate these findings and develop tailored interventions. A comprehensive Table 1 provides detailed characteristics of the included studies, including the country of study, sample sizes (cases and controls), source of biological samples, PTSD diagnostic criteria, differentially expressed genes (upregulated, downregulated, hypermethylated, or hypomethylated), key findings, and limitations. Specific focus was placed on the upregulation or downregulation of genes and their associated pathways, such as immune response and stress regulation, to identify patterns across studies. The extracted data included the proportion of hypomethylated and hypermethylated genes and their potential relevance to PTSD severity and progression.

 

 

Table 1. Summary of the main findings of the included studies.

Author / year (ref)

Study Type

Country

Members

Patients

Controls

Sample

Genes

Key Findings

All

Male

All

Male

Crombach A /  2024 [22]

Longitudinal study

Germany

Active-duty Burundian soldiers

191

All Male

NA

NA

Blood Sample

Hypermethylated

PLA2G4A PLA2G4B JMJD7 PLA2G1B

Hypomethylated

ALOX15

Differential methylation of genes in the linoleic acid metabolism pathway was significantly associated with PTSD symptom severity. This pathway’s involvement highlights potential epigenetic mechanisms mediating memory and stress responses.

Wani AH / 2024 [41]

Cohort

USA

Five diverse cohorts, including both civilian and military participants

314

(1.05) Total ratio

912

(1.05) Total ratio

 

Blood Sample

Hypomethylated

DYNC1H1 RAMP3 AP2B1

DAZAP2

Hypermethylated

CREBZF

 

Identified methylation markers such as FADS1 and GYTL1B associated with PTSD severity. DYNC1H1, RAMP3, AP2B1, and DAZAP2 were hypomethylated in PTSD patients, except for CREBZF, which was hypermethylated.

Montalvo-Ortiz/ 2022

[30]

Cohort

USA

Military veterans participated in the National Health and Resilience in Veterans Study (NHRVS)

Current PTSD: 34

All male

Current PTSD: 1101

All male

Saliva

Upregulated

DYNC1H1

RAMP3

AP2B1

DAZAP2

Downregulated

CREBZF

All genes were hypomethylated, except for CREBZF and, SENP7 in postpartum brain samples.

 

Lifetime PTSD: 65

All male

Lifetime PTSD: 1070

All male

Upregulated

GTF2IRD1

CD55

SENP7

Downregulated

SENP7

(Postmortem brain tissue from OFC)

Young/
2021

[42]

Case-Control

Australia

Australian Vietnam veterans

All: 159

Genome analysis: 48

All male

All: 140

Genome analysis: 48

All male

Whole blood

Upregulated

AIM2

Veterans with PTSD had significantly higher CRP levels and decreased AIM2 gene methylation compared to controls while no significant difference in CRP genotype counts.

R Yang /  2021

[6]

Cohort

USA

Combat trauma-exposed male veterans

83

All Male

83

All Male

Blood Sample

Hypermethylated

CST3

 PAI-1

TIMP-1

 

According to the study, men with PTSD who have experienced war trauma had higher AgeAccelGrim values, which suggests that their biological aging has accelerated. The PTSD group had significantly higher levels of four DNA methylation surrogate indicators, which are associated with age-related health issues like inflammation and cardiovascular diseases: cystatin C, PAI-1, TIMP1, and smoking pack-years.

Van der Wal/ 2020

[35]

Cohort

Netherlands

Dutch military

personnel deployed to Afghanistan for at least four months

All: 125/  Resilient: 74/  Recovering: 19/  Delayed onset: 32

All: 92.8%/  Resilient: 94.6%/  Recovering: 89.5%/  Delayed onset: 90.06%

--

--

Whole blood

Upregulated

DMR1

TUBA3FP

P2RX6

DMR2

EP300 miRNA 1281

(Delayed onset PTSD trajectory

compared to a resilient profile)

DMR7

Downregulated

DMR6

IMPA1

(a delayed onset PTSD trajectory compared to a recovering; one- and six-months post-deployment)

Fourteen genomic regions were identified in which PTSD symptom levels were associated with methylation changes over time (pre-deployment, one-, and six months post-deployment).

Wolf /  2020 [43]

Cross-sectional

USA

Non-Hispanic military veterans

    309

289/ 20

No control

No control

Blood Sample

Hypermethylated

KL

 

KL methylation inversely correlates with CRP, a marker of systemic inflammation.

CRP levels were used as a biomarker for inflammation-related biological aging.

The association between PTSD and CRP levels is mediated by KL methylation, highlighting its role in stress-related inflammation and aging.

Kang /  2019 [27]

Cohort

South Korea

South Korean veterans during the Vietnam War

123

All Male

116

All Male

Blood Sample

Hypermethylated

FKBP5 

 

The study found that the T allele of rs1360780 was associated with lower FKBP5 methylation levels, and the PTSD group showed significantly higher methylation than the non-PTSD group among veterans carrying the risk T allele. No difference was observed in methylation levels among veterans with the CC genotype. FKBP5 methylation levels were positively correlated with PTSD symptoms among T allele veterans.

Voisey J /  2019 [37]

Case-control

Australia

Male Vietnam veterans

48

All Male

48

All Male

Blood Sample

Hypermethyled

BDNF

PTSD was associated with increased BDNF methylation at three CpG sites, correlating with increased PTSD symptom severity. Active exercise was linked to lower methylation levels at these sites, suggesting exercise may modulate BDNF-related pathways and benefit PTSD treatment.

Heather L Rusch2019  / [31]

Case-control

USA

US military service members

39

All Male

27

All Male

Whole

blood

Upregulated

C5orf24

RBAK

CREBZF

CD69

PMAIP1

AGL

 ZNF644

ANKRD13C

ESCO1

ZCCHC10

All these genes are upregulated in participants with high intrusion symptoms.

Most downregulated genes are associated with symptom improvement. NF-kB was downregulated with symptom reduction.

Divya Mehta/  2018[44]

Cross-sectional

Australia

Australian Vietnam War veterans

 

48

All Male

48

All Male

Blood samples

Downregulated

MS4A15

MLPH

GUCA2A

LOC100128998

LOC642344

ARMC6

LOC402509

TMEM217

C22orf30

USP24

CHRAC1

LOC643402

Upregulated

LOC100131989

PCDHB9

Hypermethylated

C8orf37

EEF1A1

FAM40B

RPL14

RUNDC2C

SPTLC1

TMEM217

USP49

VPS41

49% of the differentially expressed genes also exhibited changes in DNA methylation. The study highlighted the enrichment of genes involved in immune response pathways, such as cytokine-cytokine receptor interaction, Jak-STAT signaling, and Toll-like receptor signaling.

Miller/
2018 [29]

Case-control

USA

US military veterans deployed on post-9/ 11 operations to Iraq and/ or Afghanistan

163

87.10%

123

90.20%

Whole blood

Upregulated

AIM2

The positive correlation between serum CRP was mediated by methylation at the AIM2 locus.

rs3091244, a functional SNP in the CRP promoter region moderated the association between lifetime trauma exposure and current PTSD severity.

Boscarino/  2018

[20]

Case-control

Canada

Canadian Armed Forces infantry soldiers returning from deployment in Afghanistan

27

All male

58

All male

Whole blood

Upregulated

LRP8

GOLM1

LINC00943

LOC1001322 15

Downregulated

CYP2C8

PTSD symptoms were associated with increased expression of LRP8 and GOLM1 and decreased expression of CYP2C8 in peripheral blood samples.

Hammamieh/ 2017

[25]

Case-control

USA

Combat veterans from Operation Enduring Freedom (OEF)/

Operation Iraqi Freedom (OIF)

79 (48 in training set and 31 in  test set)

All male

 80 (51 in trainig test and 29 in testing test)

All male

Whole blood

Upregulated

DMRTA2

ELK1

GATA3

NFATC4

PTTG1IP

Downregulated

AKT1

BDNF

CNR1

CREB1

EFS

ETS-2

HES4

LHX1

MET

NR2E1

PAX5

PDGFB

PSD

TRERF1

Most CpG islands (84.5%) were hypermethylated in PTSD patients. 
Functional networks associated with PTSD include nervous system development, endocrine signaling, and somatic complications like inflammation and circadian rhythm dysregulation.

Bam M /  2017[17]

Case-control

USA

War veterans

8

All Male

4

All Male

Blood Sample

 Upregulated
JAK2
STAT1
IL23A
TGFB1

TGFB2
TGFB3
T-BET

Downregulated
AGO2
DCR1

Several miRNAs (specific miRNAs not listed)

Reduced expression of AGO2 and DCR1 in PTSD PBMCs leads to diminished miRNA biogenesis, contributing to elevated inflammation through dysregulated gene expression. STAT3 was identified as a regulator of AGO2 and DCR1, and its reduction further exacerbated the dysregulation.

Bam M /  2016[45]

Cross-sectional

USA

War veterans

miRNA microarray=8

RNA-Seq=5

All Male

4 for miRNA microarray; 5 for RNA-Seq

All Male

Blood Sample

Upregulated

GZMB

CXCL3

STAT4

Downregulated

MTRNR2L1

MMP25

CXCL8

G0S2

Hypermethylated

CSRNP1

Hypormethylated

12 genes had decreased DNA methylation

Dysregulated immune system pathways in PTSD were associated with altered miRNA expression and DNA methylation, highlighting their role in systemic inflammation observed in PTSD.

Boks/ 2016

[19]

Cohort

Netherlands

Dutch soldiers deployed to Afghanistan

1. High trauma high PTSD symptom: 32 
2. High trauma, low PTSD symptom: 29

3. Low trauma low PTSD symptoms: 32

4. All: 93

All male

--

--

Whole blood

Downregulated

SKA2

 

Decreases in SKA2 methylation post-deployment were associated with PTSD development. Increased SKA2 methylation was linked to exposure to traumatic stress without developing PTSD. 
SKA2 methylation predicted PTSD development, especially when combined with childhood trauma history.

Logue/

2015

[46]

Cohort

USA

Trauma-exposed white non-Hispanic veterans

115

All male

28

All male

Whole blood

Upregulated

TBC1D15

 

Downregulated

DSCAM

ATP6AP1L

NR3C1

BDNF TXNRD1

All but TBC1D15 had lower expression in PTSD. The most significant was DSCAM, a neurological gene expressed widely in the developing brain, amygdala, and hippocampus of the adult brain.

Biological Pathways Implicated

•              HIF: Related to cellular response to stress.

•              mTOR Implicated in cellular growth and metabolism.

•              Insulin Linked to metabolic effects

 

Sadeh/  2015

[33]

Cohort

USA

White non-Hispanic veterans from recent conflicts

200

Current PTSD: 116

91%

--

--

Whole blood

Downregulated

SKA2 (rs7208505)

Increased SKA2 methylation is associated with reduced cortical thickness in key prefrontal regions and higher PTSD symptom severity.

Pedro Guardado/  2015 [24]

Observational study

USA

Active duty U.S. military personnel

28

27/ 28

27

26/ 27

Blood samples

Upregulated

CCL4

 TLRs

 NF-κB

 MAPK

IL-1B

TNF

IL-6

DNMT3B HDAC6

 

Downregulated

GFI1

IGF1R

PER1

MGAT4A

TARP

GH

CDC14A, ZNF831

IL13

ITCH

HSPD1

SYNE2

Hypomethylated

DNMT3B

The study indicates dysregulation in genes related to innate immune responses, neuroendocrine functions, and NF-kappa B systems, which may inform future pharmaceutical interventions for PTSD.

Yehuda/  2015

[40]

Case-control

USA

Vietnam, Iraq, or Afghanistan veterans

61

All male

61

All male

Whole blood

Upregulated

NR3C1-1F

PTSD veterans exhibited lower NR3C1-1F promoter methylation compared to controls.

Lower methylation was correlated with higher glucocorticoid receptor sensitivity and altered neuroendocrine functions.

Daniel S /  2015[34]

Prospective cohort

USA

 

 

 

 

 

 

U.S. Marines

25

All Male

25

All Male

Blood Sample

 

Downregulated

GSTM1
GSTM2

C1orf50
F2R

TBC1D4

RPL10A AHNAK PINK1. MAGEA1 OLFML2A
NBPF3
EIF4B 
RP9

PSPH

A blood-based biomarker panel could predict PTSD with 80–90% accuracy, highlighting the role of immune-related genes and dysregulated oxidative stress pathways.

Rusiecki/ 2013

[32]

Case-control

USA

Army and Marines service members serving their first OEF/ OIF deployment

74

66.2%

74

66.2%

serum

upregulated

IGF2

IL16

IL8

 

Downregulated

IL18

H19

increased IL18, H19 methylation post-deployment.

 Stratified analyses revealed more pronounced differences in the adjusted means of pre-post H19 and IL18 methylation differences for cases versus controls among older service members, males, service members of white race, and those with shorter deployments (6–12 months).

Hollifield/ 2013

[26]

Case-control

USA

 Military personnel in Albuquerque with

 significant combat exposure in Iraq or Afghanistan

6

All male

11

N=7

Whole blood

Upregulated

TNFRSF10B

IL10RB

IL16

IL4R

Inflammatory disinhibition may be involved in combat-induced PTSD and may be responsible for the increased prevalence of inflammatory-related illnesses observed in PTSD.

Gordana Matić/  2013 [47]

Cross-sectional

Serbia

Male veterans who were recruited in Serbia

Current PTSD=113,

 With life-time PTSD=61

All Male

88

All Male

Blood Sample

 

Downregulated

FKBP5 STAT5B

Current PTSD patients exhibited reduced glucocorticoid hormone-binding potential and a diminished correlation between binding sites (Bmax) and hormone affinity (KD). In contrast, trauma controls had higher binding potential compared to current PTSD patients.

Glatt SJ /  2013[23]

Prospective cohort

USA

U.S. Marines

25

All Male

25

All Male

Blood Sample

Downregulated
SUV420H1
TMEM191A
RPL39
AGPHD1
RPL10A. CA13 

PARD6B

Gene expression profiles in peripheral blood pre-deployment can predict PTSD with up to 80% accuracy using exon-based biomarkers. Dysregulation in immune and inflammatory pathways was observed in individuals who later developed PTSD.

All genes decreased expression.

van Zuiden/ 2011[36]

Cohort

Netherlands

Male soldiers

35

All Male

413

All Male

Blood Sample

Downregulated

FKBP5

Upregulated

GILZ

SGK1

 GR

 

Pre-deployment GR pathway components are vulnerability factors for subsequent development of a high level of PTSD symptoms.

Downregulation of FKBP5 mRNA predicted an increased risk of developing a high level of PTSD symptoms.

Brkljačić 2010/  [21]

Case-control

Serbia

---

 

All=14

Current PTSD=7

Lifetime PTSD=7

All Male

All= 14 Current PTSD=7 Lifetime PTSD=7

All Male

Blood Sample

BA   

GAPDH

 B2M

 PolR2A

 

   

GAPDH, B2M, and BA as reference genes for accurate gene expression quantification in PBMCs from war veterans with and without PTSD.

CRP C-Reactive Protein. CpG Cytosine-phosphate-Guanine. miRNA  MicroRNA.  OFC Orbitofrontal Cortex. NHRVS  National Health and Resilience in Veterans Study. OEF Operation Enduring Freedom. OIF Operation Iraqi Freedom. PTSD Post-Traumatic Stress Disorder.PBMCs  Peripheral Blood Mononuclear Cells. SNP  Single Nucleotide Polymorphism.

 

The key findings include:

 

  1. Alterations in Gene Expression

Studies reported significant gene expression changes associated with PTSD, highlighting dysregulation in immune, inflammatory, and neurodevelopmental processes:  

 

  1. Immune Response and Inflammation

Mehta et al., [8] identified 60 differentially expressed genes (DEGs), with enrichment in the cytokine-cytokine receptor interaction and Janus kinase-signal transducer and activator of transcription (JAK/STAT) signaling pathways. Guardado et al., reported significant upregulation of C-C motif chemokine ligand 4 (CCL4) with a fold change of 3.39, along with other inflammatory mediators such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), tumor necrosis factor (TNF), and interleukin (IL-6). Rusch et al., [31] found that cluster of differentiation (CD69) was upregulated in individuals with severe PTSD symptoms, further implicating immune activation. Rusiecki et al., demonstrated the upregulation of  IL-8 and  IL-16, which were associated with neuroinflammation and act as chemoattractants for immune cells expressing surface  CD4 molecules, respectively [32]. In contrast, they reported downregulation of IL-8, which induced interferon-gamma (IFN-γ) and facilitated its passage through the blood-brain barrier. Logue and colleagues observed lower expression of thioredoxin reductase 1 (TXDR1), which induced an anti-oxidant response [3]. Hollifield et al., reported the upregulation of several immune-related genes, including TNFRSF10B and IL-4R, both of which were involved in pro-inflammatory regulation [26].

 

  1. Neuroplasticity and Neurodevelopment  

Logue et al., observed reduced expression of genes such as Down syndrome cell adhesion molecule (DSCAM) and Brain-Derived Neurotropic Factor (BDNF), both of which are critical for neural development and synaptic plasticity [3]. Matić et al., [23] and Zuiden et al., [36] reported decreased expression of FKBP5, consistent with hypothalamic pituitary adrenal (HPA) axis dysregulation and increased stress vulnerability. Increased glucocorticoid receptor (GR) numbers [36] further indicated heightened glucocorticoid receptor sensitivity. Consistently, Hollifield et al., [26] demonstrated upregulation of IL16, which was regulated by GR.

 

  1. Pro-inflammatory Markers

Bam et al., [17] identified elevated levels of pro-inflammatory genes such as Janus kinase 2 (JAK2) and Signal Transducer And Activator Of Transcription 1 (STAT1). Meanwhile, downregulation of microRNA (miRNA) biogenesis genes, such as Argonaute RISC Catalytic Component 2 (AGO2) and Decoy Receptor 1 (DCR1), suggested disrupted post-transcriptional regulation.  

 

  1. Epigenetic Modifications

Epigenetic alterations, particularly DNA methylation, were consistently associated with PTSD.  

  1. Methylation Patterns

Hypomethylation of genes such as Doublesex and Mab-3-related Transcription Factor A2 (DMRTA2) and ELK1 disrupted synaptic and immune functions. Similarly, Wani et al., [38] identified methylation markers such as FADS1 and GYTL1B associated with PTSD severity. DYNC1H1, RAMP3, AP2B1, and DAZAP2 were hypomethylated in PTSD patients, while CREBZF, was hypermethylated [30]. Gene SUMO Specific Peptidase 7 (SENP7), involved in stress response in T-cells, showed a hypomethylation pattern in the saliva sample of patients with lifetime PTSD; however, the postpartum brain tissue sample showed downregulation [30]. Veterans with PTSD exhibited decreased methylation of AIM2, which correlated with increased C-reactive protein (CRP) levels and indicated inflammatory responses [28, 29]. Methylation of TUBA3FP, P2RX6, and IMPA1 was linked to delayed-onset PTSD, suggesting roles in memory and cognition-related pathways [35]. In PTSD patients, Hammamieh et al., [25] reported hypermethylation in 2,401 genes, consisting of 84.5% of DNA sequences in which cytosine and guanine were linked by phosphate (CpG islands), including genes such as AKT Serine/Threonine Kinase 1 (AKT1), BDNF, Cannabinoid receptor 1 (CNR1), and CAMP Responsive Element Binding Protein 1 (CREB1), which were involved in the nervous system and inflammation pathways [25]. The decreased methylation of Spindle and Kinetochore Associated Complex Subunit 2 (SKA2) was associated with PTSD development, particularly in individuals with a history of trauma in childhood [19, 33]. The NR3C1 promoter in PTSD patients had lower methylation, with increased sensitivity of glucocorticoid receptors and disturbed neuroendocrine functions [40]. Increased methylation of IL-18 and H19 was seen in PTSD patients after deployment, highlighting the importance of immune regulation and stress response [32]. Furthermore, Boscarino et al., found hypomethylation of LRP8, and GOLM1, which mediated synaptic plasticity and memory formation in PTSD patients [20].

 

  1. Exercise-induced Methylation Changes 

Voisey et al., [37] highlighted the role of physical activity in reducing BDNF methylation levels, suggesting exercise as a potential modulator of PTSD-related epigenetic changes.  

 

  1. Biological Pathways

Disruption of critical pathways underscored the complexity of PTSD pathophysiology:  

 

Stress and Immune Pathways 

Dysregulation in glucocorticoid signaling pathways was evident, with altered expression of FKBP Prolyl Isomerase-5 (FKBP-5) and GR [23, 36, 40]. The NR3C1 (glucocorticoid receptor) gene, which encodes the glucocorticoid receptor, was associated with glucocorticoid sensitivity and neuroendocrine responses. Hypomethylation of the NR3C1 promoter was associated with altered stress responses and PTSD symptoms [3, 40].

The SKA2 gene was found to regulate cortisol feedback inhibition through the Hypothalamic-Pituitary-Adrenal (HPA) axis, with methylation changes influencing stress susceptibility and PTSD development [19, 33]. Hyper- and hypomethylation of genes such as IL-8, IL-16, and IL-18 reflect dysregulated immune responses in PTSD, with these interleukins playing roles in inflammation and neuroinflammation [25, 28, 29, 32]. AIM2 methylation changes were linked to increased systemic inflammation and innate immune activation, suggesting its role in PTSD pathogenesis [28, 29].

 

  1. Linoleic Acid Metabolism

Crombach et al., linked altered methylation in linoleic acid metabolism genes to PTSD, suggesting its impacts on memory, immune regulation, and stress resilience [22].  

 

  1. Pro-inflammatory Pathways 

Bam et al., [17] emphasized the upregulation of JAK/STAT, IL-23A, and Tumor Growth Factor-β (TGF-β) pathways, reinforcing their role in PTSD-associated inflammation.  

 

Discussion 

 

This review of 28 studies explored gene expression and epigenetic changes, particularly DNA methylation, in combat-related PTSD, highlighting immune dysregulation, altered neuroplasticity, potential biomarkers, and therapeutic targets.

 

  1. Immunity dysregulation and Inflammation

Immune dysregulation in PTSD could lead to chronic stress responses, increased inflammation, and altered cytokine activity, all of which significantly contribute to the pathophysiology of PTSD [48, 49]. Analyses of immunological markers showed elevated plasma levels of pro-inflammatory cytokines, such as IFN-γ, IL-6, TNF-α, and IL-17, as well as increased levels of immune-stimulating Th1 cells and inflammatory Th17 cells in PTSD patients, indicating a pro-inflammatory-state-and-impaired immune balance [50]. Changes in the methylation levels of some gene promoters (e.g., IL-12b, and IFN-γ) in peripheral blood monocytes can cause a rise in inflammatory cytokines (e.g., IL-12) in PTSD patients [18]. Traumatic events might also weaken the immune system by altering gene expression [48]. Inflammatory processes increase susceptibility to psychological stress, thereby elevating the risk of developing PTSD [48, 49, 51]. Dysregulated immunological pathways, including the overexpression of pro-inflammatory genes (e.g., CCL4, NF-κB, and TNF), highlighted the significant role of systemic inflammation in PTSD [31]. The findings of this study suggested that alterations in DNA methylation and the activity of genes such as CD55, DAZAP2, AHRR, CDC42BPB, DOCK2, EP300, and P2RX6 could influence immune responses and inflammatory processes, increasing an individual’s susceptibility to psychological stress, and ultimately increased the risk of PTSD. Therefore, a detailed understanding of these pathways and their interactions was crucial for developing new diagnostic and therapeutic approaches for PTSD.

Methylation of CD55 and DAZAP2 could alter the balance of cytokine activity and the regulation of immune responses. For instance, hypermethylation in the CD55 gene might result in inflammation increased by reducing its regulatory role in complement inhibition [30]. DAZAP2 plays a key role in immune signaling pathways, such as NF-κB and JAK/STAT, regulating inflammation and immune responses, and modulating the function of immune cells, such as macrophages, T cells, and B cells [52]. Hypermethylation of the DAZAP2 promoter regions decreased its expression in individuals with combat-related PTSD, leading to immune dysregulation and chronic inflammation [30, 38]. Systemic inflammation resulting from DAZAP2 alterations could elevate levels of CRP, a marker of chronic inflammation in PTSD [38]. Long-term inflammation might exacerbate PTSD-related neurological disorders, such as memory problems and anxiety [8]. Therefore, since this gene plays a role in modulating the immune system in different ways, it can be a therapeutic target for PTSD. However, the lack of extensive studies directly investigating DAZAP2 changes in PTSD, particularly in combat veterans, and the complexity of its associated pathways and interaction with other genes highlighted the necessity for future research to explore its potential therapeutic targets. 

The association between the immune system and PTSD was supported by the discovery of three CpGs (cg05575921, cg21161138, and cg23576855) in AHRR [39, 53], a gene involved in immunomodulation, including T lymphocyte regulation, B cell maturation, and the activity of macrophage, dendritic cell, and neutrophils. Downregulation of AHRR could cause excessive immune response and cytokine release, such as IL-6 and TNF-α, which were consistent with the onset and exacerbation of PTSD symptoms [54]. Hypermethylation of the CDC42BPB gene, which is involved in cytoskeletal rearrangement, cell migration, and neurodevelopment, was linked to PTSD and associated with depressive symptoms [55] and increased CRP levels [4], suggesting inflammation as a key component of PTSD [38, 56]. Hypomethylation of the AIM2 gene, involved in innate immunity, was also associated with increased CRP levels in veterans with PTSD [28]. Future research should investigate CDC42BPB and AIM2 methylation as mediators in the relationship between PTSD and inflammation, as well as the role of CRP as a diagnostic marker and potential therapeutic target. The upregulation of IL-10RB, IL-16, and IL-4R genes, which play crucial roles in immune modulation, leads to inflammatory disinhibition and immune dysregulation in combat-induced PTSD [26]. IL-10RB controls mucosal immune tolerance and anti-inflammatory macrophage activity [57], while IL-4R stimulates cell proliferation, tissue regeneration, and neurological functions [58]. IL-16 regulates T-cell growth, activation, and motility [59]. Dysregulation of these immunological signaling pathways is a hallmark of PTSD, exacerbating neuroinflammation, and anxiety-like behaviors [26].

 

  1. Pro-inflammatory markers

TBX21 and STAT4 play crucial roles in regulating T cell activity. TBX21 controls the transcription of interferon-gamma (IFN-γ), a pro-inflammatory gene upregulated in PTSD. Furthermore, the expression of pro-inflammatory cytokines, such as CCL4, CCL5, CXCL1, CXCL2, CXCL3, CXCL6, and CXCL8 is generally altered in PTSD. These findings suggested that shifts in T cell biology are a primary driver of the underlying inflammation observed in PTSD [17].

Several potential biomarkers and immune therapeutic implications emerged from the studies. For instance, Guardado et al., [24] and Mehta et al., [8] highlighted the upregulation of immune-related genes such as CCL4 and CD69 as diagnostic markers, while inflammatory markers such as CCL4 and NF-κB point to immune-modulating therapies. Due to the high costs of measuring gene expression, downstream molecules such as microRNAs (miRNAs) or proteins encoded by these genes might serve as practical diagnostic biomarkers and therapeutic options. For instance, hsa-miR-193a-5p and hsa-miR-125a, which target the pro-inflammatory cytokines interleukin-12B (IL12B) and interferon-gamma (IFNG), respectively, were downregulated in PTSD [17, 18]. Additionally, Bam et al., identified several miRNAs that were predicted to target TBX-21 and STAT4, key genes in the Th cell differentiation pathway [17]. Therefore, these miRNAs represent potential diagnostic or therapeutic biomarkers for veterans with PTSD.

However, implementing these diagnostic and therapeutic methods presents certain challenges. One of the major limitations of anti-inflammatory and anti-immune therapies in PTSD patients is the heterogeneity in molecular pathways and immune responses, which renders standard treatment approaches ineffective for all individuals. The complex interference and interactions of inflammatory pathways, such as NF-κB and JAK/STAT, along with epigenetic changes such as methylation of genes including DAZAP2, CD55, and AIM2, complicates the development of targeted therapies. In addition, many potential drugs, such as TNF-α inhibitors or methylation-regulating agents, have significant systemic side effects that limit their widespread use. Therefore, the development of personalized therapies based on gene expression patterns, DNA methylation, and other genetic and epigenetic changes may offer more effective treatment options.

 

  1. Stress pathways

Traumatic events can impair the regulation of the sympathetic adrenaline-medulla (SAM) and HPA axes, leading to abnormal circadian cortisol secretion rhythms. Cortisol, a key endocrine regulator in immune responses, plays a critical role in immune dysfunction. Chronic stress, such as that experienced by combat veterans, could cause resistance to the glucocorticoid receptor (GCR), resulting in chronic inflammation and increasing susceptibility to physical diseases and PTSD [48].

The NR3C1 gene encodes the glucocorticoid receptor, which is essential for HPA axis function and cortisol regulation. Under normal conditions, high cortisol levels signal the brain to stop further cortisol production, maintaining homeostasis [60]. In PTSD, this feedback system might be compromised due to decreased expression or altered methylation of NR3C1, leading to prolonged or heightened stress hormone activity. This imbalance might exacerbate the persistent hyperactivation or hypoactivation of the HPA axis, both of which are linked to PTSD pathophysiology.

Given its role in cortisol regulation, NR3C1 is a strong candidate for research on stress-related biomarkers [61]. Methylation patterns in NR3C1 may enable early identification of PTSD risk or onset, particularly in trauma-exposed populations [62].

The CREBZF gene, involved in regulating the HPA axis, might exhibit increased methylation in its promoter regions, suppressing its expression and leading to increased stress sensitivity [63, 64]. Changes in stress response-related genes, such as glucocorticoid-regulating genes, FKBP5, have been documented in both cross-sectional and longitudinal investigations [27, 31]. Dysregulation of stress responses, linked to the FKBP5 gene, influences PTSD risk and treatment responsiveness. Genetic and epigenetic alteration in FKBP5 contributes to PTSD pathophysiology, making it a potential therapeutic target or biomarker for monitoring treatment response [27].

 

  1. Neuroplasticity and Neurodevelopment

Memory and fear play a central role in PTSD [65], as the condition is characterized by persistent traumatic memories and dysregulation of fear responses. Altered memory processing exacerbates the intrusive symptoms of PTSD, while hyperactive fear recall and impaired fear extinction contribute to hyperarousal and avoidance behaviors [66]. Disruptions in neurodevelopmental processes and synaptic plasticity hinder the brain’s resilience and recovery after trauma, affecting neural connectivity and regulating fear and memory circuits. Disruptions contribute to the persistence of PTSD symptoms and may be linked to genetic and epigenetic changes in key neural circuits.

The DYNC1H1, BDNF, and AKT1 genes, play crucial roles in axon guidance, hippocampal development, and neurogenesis [29, 32, 34]. The DYNC1H1 gene regulates axonal transport, synaptic regeneration, and plasticity and its methylation can affect memory processes [64]. The AP2B1 gene, involved in endocytosis, under the influence of methylation changes, may experience reduced neuronal transmission efficiency due to methylation changes [64, 67]. The NR2E1 and CREB1 genes are involved in fear response, while BDNF and AKT1 regulate fear memory, and CNR1 and ETS-2 are associated with fear-related risk [25]. The BDNF gene methylation is related to neurogenesis and memory [3, 25, 28]. The DSCAM gene plays a significant role in neural development, contributing to dendritic patterning, self-avoidance, axon guidance, and synapse formation [68]. Downregulation of DSCAM in PTSD may impair neural network integrity and plasticity, leading to neurotransmitter release, cognitive deficits, and emotional dysregulations [3, 69]. BDNF, a protein, activates the phospholipase C -gamma, PI3K, and MAPK/ERK pathways, which are crucial for neurogenesis and synaptic plasticity [70]. Downregulation of these pathways correlated with reduced resilience and impaired recovery in PTSD, potentially leading to cognitive impairment. 

LRP8 (ApoER2) is crucial for lysosomal breakdown and signal transmission. Its upregulation has been linked to cognitive decline, anxiety, disrupted nest construction, disturbed circadian rhythms, and altered stress responses in PTSD, suggesting its role in maladaptive neuronal development under chronic stress [71, 72]. ATP6AP1L, which facilitates lysosomal function and cellular energy production, may influence energy-dependent neural processes [73]. Deficiency in ATP6AP1 results in immunodeficiency, hepatopathy, and cognitive impairment [74]. Downregulation of ATP6AP1L may disrupt energy-dependent neural processes and increase susceptibility to PTSD due to its involvement in the glucocorticoid receptor pathway and neural responses to stress[3].

The CNR1 and BDNF genes are involved in dopamine and serotonin signaling, while CREB1 and HES4 genes are implicated in the cortisol hormone network [29]. CYP2C8, a key enzyme in the metabolism of neuroactive steroids and stress-regulating compounds [75], is involved in individuals with PTSD, potentially hindering the synthesis of neurosteroids that regulate anxiety and fear response [20].

These networks and genes represent epigenetic changes that can serve as biomarkers for improved diagnosis and targeted treatment of PTSD. For example, BDNF plays a role in the pathogenesis of PTSD through various pathways and represents a potential therapeutic target. In the reviewed studies, Crombach et al., [22] and Voisey et al., [37] highlighted the therapeutic potential of interventions targeting epigenetic markers such as BDNF methylation. Voisey et al., [37] underscored the role of exercise and lifestyle interventions in reducing BNDF methylation and improving neural plasticity, offering a non-invasive treatment option. Hammamieh et al., [25] and Wolf et al., [39] advocated for integrating genetic and epigenetic markers into personalized treatment plans, leveraging tools such as Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and other gene-editing tools to modulate key pathways. 

Studies indicated that miRNAs that target genes such as DYNC1H1, BDNF, and GOLM1 are emerging as promising biomarkers for therapeutic monitoring and early diagnosis in cancer research [76-78]. These miRNAs may also prove valuable for the diagnosis and treatment of PTSD. Assessments of pathways at the protein level, such as the BDNF-PI3K-MAPK/ERK pathway, could serve as useful instruments for monitoring the development of PTSD progression [79].

Although this study provided valuable insights, several limitations must be acknowledged. First, much of the research focused on war veterans, which might limit the generalizability of the findings to other traumatized populations, such as civilians or children. Additionally, the use of different tissue sources, such as blood and saliva, might yield different results due to tissue-specific methylation patterns. Many studies focused on peripheral blood for methylation and gene expression analysis, but it remained unclear whether these findings apply to the brain or other relevant tissues.

Additionally, a large number of studies employed cross-sectional designs, making it difficult to track longitudinal epigenetic changes associated with PTSD or establish causal relationships.

Fully understanding gene-environment dynamics remains challenging due to insufficient research on the complex interplay between genetic predispositions, environmental factors, and trauma exposure. Furthermore, while promising biomarkers have been identified, their therapeutic significance is limited until confirmed in broader and more diverse populations. Methodological heterogeneity, including variations in tissue sources, analytical techniques, and control populations, further complicates direct comparisons between studies. This variability underscores the need for consistent methodologies in future research.

Gene expression and epigenetic changes, particularly DNA methylation, play a critical role in the pathophysiology of war-related PTSD. These alterations disrupt immune system pathways, inflammation, neuroplasticity, and stress-related mechanisms. Changes in neuroplasticity and memory-related gene expression, such as BDNF, DSCAM, and CREB1, could lead to impairments in learning processes, and fear extinction, along with cognitive and emotional symptoms characteristic of combat-related PTSD.

These findings highlighted the potential to target the immune and neurological pathways related to PTSD, enabling the design of more effective interventions and personalized treatments based on an individual’s gene expression and epigenetic profile.

Future research should focus on validating these findings in larger and more diverse populations, exploring the intricate interplay of molecular pathways, and identifying novel biomarkers and therapeutic targets. These efforts will pave the way for developing more sensitive diagnostic tools and effective therapeutic interventions for PTSD, particularly its war-related subtype.

 

Declaration

 

Ethics approval and consent to participate: Not applicable.

 

Consent for publication: Not applicable.

 

Conflict of Interest: The authors declared that they had no competing interests related to this work.

 

Funding: This research received no specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

 

Authors’ Contribution: AHR: Conceptualization, methodology, and original draft preparation; SZ: Conceptualization, methodology, and original draft preparation; KJ: Conceptualization, methodology, and original draft preparation; MMB: Data analysis, review, editing, Supervision, and critical revisions; OB: Data analysis, review, editing, Supervision, and critical revisions; GA: Data analysis, review, editing, Supervision and critical revisions; PH: Data analysis, review, editing, Supervision and critical revisions; SP: Conceptualization, methodology, and original draft preparation.

  1. Compean E, Hamner M. Posttraumatic stress disorder with secondary psychotic features (PTSD-SP): Diagnostic and treatment challenges. Prog Neuropsychopharmacol Biol Psychiatry. 2019;88:265-75.
  2. Hori H, Kim Y. Inflammation and post-traumatic stress disorder. Psychiatry Clin Neurosci. 2019;73(4):143-53.
  3. Logue MW, Amstadter AB, Baker DG, Duncan L, Koenen KC, Liberzon I, et al. The Psychiatric Genomics Consortium Posttraumatic Stress Disorder Workgroup: Posttraumatic Stress Disorder Enters the Age of Large-Scale Genomic Collaboration. Neuropsychopharmacology. 2015;40(10):2287-97.
  4. Ligthart S, Marzi C, Aslibekyan S, Mendelson MM, Conneely KN, Tanaka T, et al. DNA methylation signatures of chronic low-grade inflammation are associated with complex diseases. Genome Biol. 2016;17(1):255.
  5. Kozarić-Kovacić D, Borovecki A. Prevalence of psychotic comorbidity in combat-related post-traumatic stress disorder. Mil Med. 2005;170(3):223-6.
  6. Yang R, Gautam A, Getnet D, Daigle BJ, Miller S, Misganaw B, et al. Epigenetic biotypes of post-traumatic stress disorder in war-zone exposed veteran and active duty males. Mol Psychiatry. 2021;26(8):4300-14. doi: 10.1038/s41380-020-00966-2.
  7. Huckins LM, Chatzinakos C, Breen MS, Hartmann J, Klengel T, da Silva Almeida AC, et al. Analysis of Genetically Regulated Gene Expression Identifies a Prefrontal PTSD Gene, SNRNP35, Specific to Military Cohorts. Cell Rep. 2020;31(9):107716.
  8. Breen MS, Tylee DS, Maihofer AX, Neylan TC, Mehta D, Binder EB, et al. PTSD Blood Transcriptome Mega-Analysis: Shared Inflammatory Pathways across Biological Sex and Modes of Trauma. Neuropsychopharmacology. 2018;43(3):469-81.
  9. Daskalakis NP, Rijal CM, King C, Huckins LM, Ressler KJ. Recent Genetics and Epigenetics Approaches to PTSD. Curr Psychiatry Rep. 2018;20(5):30.
  10. Banerjee SB, Morrison FG, Ressler KJ. Genetic approaches for the study of PTSD: Advances and challenges. Neurosci Lett. 2017;649:139-46.
  11. Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6(7):e1000097.
  12. Avramopoulos D. Genetics of psychiatric disorders methods: molecular approaches. Psychiatr Clin North Am. 2010;33(1):1-13.
  13. Felsenfeld G. A brief history of epigenetics. Cold Spring Harb Perspect Biol. 2014;6(1).
  14. Lehrmann E, Freed WJ. Transcriptional correlates of human substance use. Ann N Y Acad Sci. 2008;1139:34-42.
  15. Dressler J, Vemuganti R. Apoptosis and gene expression after TBI. Leg Med (Tokyo). 2009;11 Suppl 1:S54-5.
  16. Ouzzani M, Hammady H, Fedorowicz Z, Elmagarmid A. Rayyan—a web and mobile app for systematic reviews. Systematic Reviews. 2016;5(1):210.
  17. Bam M, Yang X, Zumbrun EE, Ginsberg JP, Leyden Q, Zhang J, et al. Decreased AGO2 and DCR1 in PBMCs from War Veterans with PTSD leads to diminished miRNA resulting in elevated inflammation. Transl Psychiatry. 2017;7(8):e1222.
  18. Bam M, Yang X, Zhou J, Ginsberg JP, Leyden Q, Nagarkatti PS, et al. Evidence for Epigenetic Regulation of Pro-Inflammatory Cytokines, Interleukin-12 and Interferon Gamma, in Peripheral Blood Mononuclear Cells from PTSD Patients. J Neuroimmune Pharmacol. 2016;11(1):168-81.
  19. Boks MP, Rutten BPF, Geuze E, Houtepen LC, Vermetten E, Kaminsky Z, et al. SKA2 Methylation is Involved in Cortisol Stress Reactivity and Predicts the Development of Post-Traumatic Stress Disorder (PTSD) After Military Deployment. Neuropsychopharmacology. 2016;41(5):1350-6.
  20. Boscarino C, Nalpathamkalam T, Pellecchia G, Li W, Thiruvahindrapuram B, Merico D. Using Next-Generation Sequencing Transcriptomics To Determine Markers of Post-traumatic Symptoms: Preliminary Findings from a Post-deployment Cohort of Soldiers. G3 (Bethesda). 2019;9(2):463-71.
  21. Brkljacić J, Tanić N, Milutinović DV, Elaković I, Jovanović SM, Perisić T, et al. Validation of endogenous controls for gene expression studies in peripheral lymphocytes from war veterans with and without PTSD. BMC Mol Biol. 2010;11:26.
  22. Crombach A, Rukundo-Zeller AC, Vukojevic V, Nandi C, Bambonye M, de Quervain DJF, et al. Differential methylation of linoleic acid pathway genes is associated with PTSD symptoms – a longitudinal study with Burundian soldiers returning from a war zone. Translational Psychiatry. 2024;14(1):32.
  23. Glatt SJ, Tylee DS, Chandler SD, Pazol J, Nievergelt CM, Woelk CH, et al. Blood-based gene-expression predictors of PTSD risk and resilience among deployed marines: a pilot study. Am J Med Genet B Neuropsychiatr Genet. 2013;162b(4):313-26.
  24. Guardado P, Olivera A, Rusch HL, Roy M, Martin C, Lejbman N, et al. 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.
  25. Hammamieh R, Chakraborty N, Gautam A, Muhie S, Yang R, Donohue D, et al. Whole-genome DNA methylation status associated with clinical PTSD measures of OIF/OEF veterans. Transl Psychiatry. 2017;7(7):e1169.
  26. Hollifield M, Moore D, Yount G. Gene expression analysis in combat veterans with and without post‑traumatic stress disorder. Mol Med Rep. 2013;8(1):238-44
  27. Kang JI, Kim TY, Choi JH, So HS, Kim SJ. Allele-specific DNA methylation level of FKBP5 is associated with post-traumatic stress disorder. Psychoneuroendocrinology. 2019;103:1-7.
  28. Al Jowf GI, Snijders C, Rutten BPF, de Nijs L, Eijssen LMT. The Molecular Biology of Susceptibility to Post-Traumatic Stress Disorder: Highlights of Epigenetics and Epigenomics. Int J Mol Sci. 2021;22(19).
  29. Miller MW, Maniates H, Wolf EJ, Logue MW, Schichman SA, Stone A, et al. CRP polymorphisms and DNA methylation of the AIM2 gene influence associations between trauma exposure, PTSD, and C-reactive protein. Brain Behav Immun. 2018;67:194-202.
  30. Montalvo-Ortiz JL, Gelernter J, Cheng Z, Girgenti MJ, Xu K, Zhang X, et al. Epigenome-wide association study of posttraumatic stress disorder identifies novel loci in US military veterans. Translational psychiatry. 2022;12(1):65.
  31. Rusch HL, Robinson J, Yun S, Osier ND, Martin C, Brewin CR, et al. 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;80:904-8.
  32. Rusiecki JA, Byrne C, Galdzicki Z, Srikantan V, Chen L, Poulin M, et al. PTSD and DNA Methylation in Select Immune Function Gene Promoter Regions: A Repeated Measures Case-Control Study of U.S. Military Service Members. Front Psychiatry. 2013;4:56.
  33. Sadeh N, Spielberg JM, Logue MW, Wolf EJ, Smith AK, Lusk J, et al. SKA2 methylation is associated with decreased prefrontal cortical thickness and greater PTSD severity among trauma-exposed veterans. Molecular Psychiatry. 2016;21(3):357-63.
  34. Tylee DS, Chandler SD, Nievergelt CM, Liu X, Pazol J, Woelk CH, et al. Blood-based gene-expression biomarkers of post-traumatic stress disorder among deployed marines: A pilot study. Psychoneuroendocrinology. 2015;51:472-94.
  35. van der Wal SJ, Maihofer AX, Vinkers CH, Smith AK, Nievergelt CM, Cobb DO, et al. Associations between the development of PTSD symptoms and longitudinal changes in the DNA methylome of deployed military servicemen: A comparison with polygenic risk scores. Compr Psychoneuroendocrinol. 2020;4:100018.
  36. van Zuiden M, Geuze E, Willemen HL, Vermetten E, Maas M, Amarouchi K, et al. Glucocorticoid receptor pathway components predict posttraumatic stress disorder symptom development: a prospective study. Biol Psychiatry. 2012;71(4):309-16.
  37. Voisey J, Lawford B, Bruenig D, Harvey W, Morris CP, Young RM, et al. Differential BDNF methylation in combat exposed veterans and the association with exercise. Gene. 2019;698:107-12.
  38. Katrinli S, Wani AH, Maihofer AX, Ratanatharathorn A, Daskalakis NP, Montalvo-Ortiz J, et al. Epigenome-wide association studies identify novel DNA methylation sites associated with PTSD: A meta-analysis of 23 military and civilian cohorts. medRxiv. 2024.
  39. Logue MW, Miller MW, Wolf EJ, Huber BR, Morrison FG, Zhou Z, et al. An epigenome-wide association study of posttraumatic stress disorder in US veterans implicates several new DNA methylation loci. Clin Epigenetics. 2020;12(1):46.
  40. Yehuda R, Flory JD, Bierer LM, Henn-Haase C, Lehrner A, Desarnaud F, et al. Lower methylation of glucocorticoid receptor gene promoter 1F in peripheral blood of veterans with posttraumatic stress disorder. Biol Psychiatry. 2015;77(4):356-64.
  41. Wani A, Katrinli S, Zhao X, Daskalakis N, Zannas A, Aiello A, et al. Blood-based DNA methylation and exposure risk scores predict PTSD with high accuracy in military and civilian cohorts. Res Sq. 2024.
  42. Mc DYR, Lawford B, Mellor R, Morris CP, Voisey J. Investigation of C-reactive protein and AIM2 methylation as a marker for PTSD in Australian Vietnam veterans. Gene. 2021;803:145898.
  43. Wolf EJ, Logue MW, Zhao X, Daskalakis NP, Morrison FG, Escarfulleri S, et al. PTSD and the klotho longevity gene: Evaluation of longitudinal effects on inflammation via DNA methylation. Psychoneuroendocrinology. 2020;117:104656.
  44. Mehta D, Voisey J, Bruenig D, Harvey W, Morris CP, Lawford B, et al. Transcriptome analysis reveals novel genes and immune networks dysregulated in veterans with PTSD. Brain Behav Immun. 2018;74:133-42.
  45. Bam M, Yang X, Zumbrun EE, Zhong Y, Zhou J, Ginsberg JP, et al. Dysregulated immune system networks in war veterans with PTSD is an outcome of altered miRNA expression and DNA methylation. Scientific Reports. 2016;6(1):31209.
  46. Logue MW, Smith AK, Baldwin C, Wolf EJ, Guffanti G, Ratanatharathorn A, et al. An analysis of gene expression in PTSD implicates genes involved in the glucocorticoid receptor pathway and neural responses to stress. Psychoneuroendocrinology. 2015;57:1-13.
  47. Matić G, Milutinović DV, Nestorov J, Elaković I, Jovanović SM, Perišić T, et al. Lymphocyte glucocorticoid receptor expression level and hormone-binding properties differ between war trauma-exposed men with and without PTSD. Prog Neuropsychopharmacol Biol Psychiatry. 2013;43:238-45.
  48. Sun Y, Qu Y, Zhu J. The Relationship Between Inflammation and Post-traumatic Stress Disorder. Frontiers in Psychiatry. 2021;12.
  49. Katrinli S, Oliveira NCS, Felger JC, Michopoulos V, Smith AK. The role of the immune system in posttraumatic stress disorder. Transl Psychiatry. 2022;12(1):313.
  50. Cohen S, Janicki-Deverts D, Doyle WJ, Miller GE, Frank E, Rabin BS, et al. Chronic stress, glucocorticoid receptor resistance, inflammation, and disease risk. Proc Natl Acad Sci U S A. 2012;109(16):5995-9.
  51. Lauten TH, Natour T, Case AJ. Innate and adaptive immune system consequences of post-traumatic stress disorder. Autonomic Neuroscience. 2024;252:103159.
  52. Yan Z, Gibson SA, Buckley JA, Qin H, Benveniste EN. Role of the JAK/STAT signaling pathway in regulation of innate immunity in neuroinflammatory diseases. Clin Immunol. 2018;189:4-13.
  53. Smith AK, Ratanatharathorn A, Maihofer AX, Naviaux RK, Aiello AE, Amstadter AB, et al. Epigenome-wide meta-analysis of PTSD across 10 military and civilian cohorts identifies methylation changes in AHRR. Nat Commun. 2020;11(1):5965.
  54. Hao N, Whitelaw ML. The emerging roles of AhR in physiology and immunity. Biochem Pharmacol. 2013;86(5):561-70.
  55. Story Jovanova O, Nedeljkovic I, Spieler D, Walker RM, Liu C, Luciano M, et al. DNA Methylation Signatures of Depressive Symptoms in Middle-aged and Elderly Persons: Meta-analysis of Multiethnic Epigenome-wide Studies. JAMA Psychiatry. 2018;75(9):949-59.
  56. Secting the genetic association of C-reactive protein with PTSD, traumatic events, and social support. Neuropsychopharmacology. 2021;46(6):1071-7.
  57. Shouval DS, Biswas A, Goettel JA, McCann K, Conaway E, Redhu NS, et al. Interleukin-10 receptor signaling in innate immune cells regulates mucosal immune tolerance and anti-inflammatory macrophage function. Immunity. 2014;40(5):706-19.
  58. Kedong H, Wang D, Sagaram M, An HS, Chee A. Anti-inflammatory effects of interleukin-4 on intervertebral disc cells. Spine J. 2020;20(1):60-8.
  59. Richmond J, Tuzova M, Cruikshank W, Center D. Regulation of cellular processes by interleukin-16 in homeostasis and cancer. J Cell Physiol. 2014;229(2):139-47.
  60. Bakusic J, Ghosh M, Polli A, Bekaert B, Schaufeli W, Claes S, et al. Role of NR3C1 and SLC6A4 methylation in the HPA axis regulation in burnout. J Affect Disord. 2021;295:505-12.
  61. Iftimovici A, Kebir O, He Q, Jay TM, Rouleau GA, Krebs MO, et al. Stress, Cortisol and NR3C1 in At-Risk Individuals for Psychosis: A Mendelian Randomization Study. Front Psychiatry. 2020;11:680.
  62. Wilker S, Vukojevic V, Schneider A, Pfeiffer A, Inerle S, Pauly M, et al. Epigenetics of traumatic stress: The association of NR3C1 methylation and posttraumatic stress disorder symptom changes in response to narrative exposure therapy. Transl Psychiatry. 2023;13(1):14.
  63. Danan D, Todder D, Zohar J, Cohen H. Is PTSD-Phenotype Associated with HPA-Axis Sensitivity? Feedback Inhibition and Other Modulating Factors of Glucocorticoid Signaling Dynamics. Int J Mol Sci. 2021;22(11).
  64. Montalvo-Ortiz JL, Gelernter J, Cheng Z, Girgenti MJ, Xu K, Zhang X, et al. Epigenome-wide association study of posttraumatic stress disorder identifies novel loci in U.S. military veterans. Translational Psychiatry. 2022;12(1):65.
  65. Gonzalez P, Martinez KG. The role of stress and fear in the development of mental disorders. Psychiatr Clin North Am. 2014;37(4):535-46.
  66. Careaga MBL, Girardi CEN, Suchecki D. Understanding posttraumatic stress disorder through fear conditioning, extinction and reconsolidation. Neuroscience & Biobehavioral Reviews. 2016;71:48-57.
  67. Cestari V, Rossi-Arnaud C, Saraulli D, Costanzi M. The MAP(K) of fear: From memory consolidation to memory extinction. Brain Research Bulletin. 2014;105:8-16.
  68. Zhu K, Xu Y, Liu J, Xu Q, Ye H. Down syndrome cell adhesion molecule and its functions in neural development. Neurosci Bull. 2011;27(1):45-52.
  69. Stachowicz K. The role of DSCAM in the regulation of synaptic plasticity: possible involvement in neuropsychiatric disorders. Acta Neurobiol Exp (Wars). 2018;78(3):210-9.
  70. Numakawa T, Odaka H, Adachi N. Actions of Brain-Derived Neurotrophin Factor in the Neurogenesis and Neuronal Function, and Its Involvement in the Pathophysiology of Brain Diseases. Int J Mol Sci. 2018;19(11).
  71. Johnson LA, Zuloaga DG, Bidiman E, Marzulla T, Weber S, Wahbeh H, et al. ApoE2 Exaggerates PTSD-Related Behavioral, Cognitive, and Neuroendocrine Alterations. Neuropsychopharmacology. 2015;40(10):2443-53.
  72. Xiao X, Yu H, Li J, Wang L, Li L, Chang H, et al. Further evidence for the association between LRP8 and schizophrenia. Schizophrenia Research. 2020;215:499-505.
  73. Tian Y, Gao M, Huang L, Zhou H, Wang J. ATP6AP1 is a potential prognostic biomarker and is associated with iron metabolism in breast cancer. Frontiers in Genetics. 2022;13:958290.
  74. Jansen EJR, Timal S, Ryan M, Ashikov A, van Scherpenzeel M, Graham LA, et al. ATP6AP1 deficiency causes an immunodeficiency with hepatopathy, cognitive impairment and abnormal protein glycosylation. Nature Communications. 2016;7(1):11600.
  75. Daskalopoulos EP, Malliou F, Rentesi G, Marselos M, Lang MA, Konstandi M. Stress is a critical player in CYP3A, CYP2C, and CYP2D regulation: role of adrenergic receptor signaling pathways. Am J Physiol Endocrinol Metab. 2012;303(1):E40-54.
  76. Kojima S, Enokida H, Yoshino H, Itesako T, Chiyomaru T, Kinoshita T, et al. The tumor-suppressive microRNA-143/145 cluster inhibits cell migration and invasion by targeting GOLM1 in prostate cancer. J Hum Genet. 2014;59(2):78-87.
  77. Ma L, Guo H, Zhao Y, Liu Z, Wang C, Bu J, et al. Liquid biopsy in cancer current: status, challenges and future prospects. Signal Transduct Target Ther. 2024;9(1):336.
  78. Condrat CE, Thompson DC, Barbu MG, Bugnar OL, Boboc A, Cretoiu D, et al. miRNAs as Biomarkers in Disease: Latest Findings Regarding Their Role in Diagnosis and Prognosis. Cells. 2020;9(2).
  79. Zhu Z, Huang X, Du M, Wu C, Fu J, Tan W, et al. Recent advances in the role of miRNAs in post-traumatic stress disorder and traumatic brain injury. Mol Psychiatry. 2023;28(7):2630-44.