The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
ArticlesFull Access

Decoding Shared Versus Divergent Transcriptomic Signatures Across Cortico-Amygdala Circuitry in PTSD and Depressive Disorders

Abstract

Objective:

Posttraumatic stress disorder (PTSD) is a debilitating neuropsychiatric disease that is highly comorbid with major depressive disorder (MDD) and bipolar disorder. The overlap in symptoms is hypothesized to stem from partially shared genetics and underlying neurobiological mechanisms. To delineate conservation between transcriptional patterns across PTSD and MDD, the authors examined gene expression in the human cortex and amygdala in these disorders.

Methods:

RNA sequencing was performed in the postmortem brain of two prefrontal cortex regions and two amygdala regions from donors diagnosed with PTSD (N=107) or MDD (N=109) as well as from neurotypical donors (N=109).

Results:

The authors identified a limited number of differentially expressed genes (DEGs) specific to PTSD, with nearly all mapping to cortical versus amygdala regions. PTSD-specific DEGs were enriched in gene sets associated with downregulated immune-related pathways and microglia as well as with subpopulations of GABAergic inhibitory neurons. While a greater number of DEGs associated with MDD were identified, most overlapped with PTSD, and only a few were MDD specific. The authors used weighted gene coexpression network analysis as an orthogonal approach to confirm the observed cellular and molecular associations.

Conclusions:

These findings provide supporting evidence for involvement of decreased immune signaling and neuroinflammation in MDD and PTSD pathophysiology, and extend evidence that GABAergic neurons have functional significance in PTSD.

Posttraumatic stress disorder (PTSD) is a debilitating disorder that develops in a subset of individuals following trauma exposure. PTSD is highly comorbid with other mental health disorders (13); for example >50% of individuals with PTSD also have a diagnosis of major depressive disorder (MDD) (46), and the prevalence of PTSD among individuals with a bipolar disorder diagnosis is two to three times that of the general population (7, 8). PTSD is characterized by a unique set of clinical phenotypes but shares some diagnostic symptoms with depressive disorders. Comorbidity may arise from shared mechanistic underpinnings, including overlapping genetic heritability and common environmental risk factors, such as chronic stress and trauma exposure (4). However, the cellular and molecular mechanisms unique to PTSD versus those shared with MDD are not well understood. Here, we examined transcriptional patterns within and across PTSD and MDD in the human cortex and amygdala.

Aberrant activity in neural circuits that link amygdala and prefrontal cortical regions has been identified in individuals with PTSD as well as in animal models relevant for PTSD (911). The amygdala and prefrontal cortex are critical for emotional regulation, including the expression and extinction of fear, behavioral functions that are dysregulated in PTSD. Coordinated patterns of neural activity in cortico-amygdala circuits underlie functional connectivity between these regions, which controls fear and anxiety (1215). In accordance with human neuroimaging studies showing aberrant cortico-amygdala activity in PTSD (12, 1517), animal studies demonstrate that function in these circuits is strongly impacted by exposure to trauma, and experimentally manipulating neuronal activity or key cell signaling pathways in cortico-amygdala circuits impacts fear processing and anxiety (1821).

At the cellular level, deficits in inhibitory neurotransmission in cortico-amygdala circuits are associated with PTSD and depressive disorders (22, 23). Chronic stress and trauma exposure are hypothesized to impair inhibitory neuron function, impacting excitation-inhibition balance in cortico-amygdala circuits (24, 25). This is important because GABAergic inhibitory neurons in these circuits control neural activity and synaptic plasticity to regulate fear-related behaviors in animal models (26). However, how the molecular sequelae following chronic stress and trauma exposure impact inhibitory neuron function is not well understood. In addition to GABAergic inhibition, inflammation and immune signaling have emerged as potential contributors to PTSD and depressive disorders (2730). While inflammatory markers and genes related to immune signaling are altered in PTSD (3133), whether observed changes result from central versus peripheral immune signaling pathways, and whether they reflect increased risk or epiphenomena related to the pathophysiological sequelae of PTSD, is not clear.

Conducting the largest RNA sequencing study of PTSD in the human brain to date, we identified downregulation of microglia-related transcripts and immune-related coexpression modules in both the cortex and amygdala. We identified notable reductions in specific transcripts encoding neuromodulators that are associated with GABAergic neuron function, but there was also evidence for increased expression of transcripts associated with both excitatory and inhibitory neurons. Collectively, the findings contribute evidence supporting the involvement of immune signaling, neuroinflammation, and inhibitory neuron function in MDD and PTSD.

Methods

Detailed methods are available in the online supplement.

Postmortem human brains were donated through U.S. medical examiners’ offices at the time of autopsy, and a retrospective clinical diagnostic review was conducted on every brain to diagnose each donor into one of the three diagnosis groups (control, PTSD, MDD). Tissue was dissected from two subregions of the frontal cortex (the dorsolateral prefrontal cortex and the dorsal anterior cingulate cortex), and two subregions of the amygdala (the basolateral amygdala and the medial amygdala) under visual guidance. RNA was extracted and sequenced using Ribo-Zero Gold ribosomal RNA depletion on an Illumina HiSeq 3000. Raw sequencing reads were processed as previously described (34) to obtain gene counts relative to GENCODE release 25 (GRCh38.p7). Quality control, including sequencing quality and sample identity checks, resulted in 1,285 samples across 325 unique donors and four brain regions. We performed differential expression analyses within and across brain subregions using the voom function in the limma package for R (35), adjusting for clinical and technical covariates, as well as quality surrogate variables (qSVs) (36). These models account for donors from all three diagnosis groups to jointly estimate the effects of PTSD versus control, MDD versus control, and PTSD versus MDD. We performed RNAscope to validate cell type specificity of candidate DEGs. We defined sets of marginally significant (at p<0.005) genes, with and without enforcing directionality of effects (i.e., higher vs. lower expression in PTSD vs. control), and performed gene set and cell type enrichment analyses using the hypergeometric test. Lastly, we performed weighted gene coexpression network analysis (37) to assign genes to modules and assess the role of diagnosis on coexpressed gene sets.

Results

We generated deep bulk/homogenate RNA sequencing (RNA-seq) data from postmortem human tissue in two subregions of the frontal cortex (dorsolateral prefrontal cortex [dlPFC] and dorsal anterior cingulate cortex [dACC]) and two subregions of the amygdala (basolateral amygdala [BLA] and medial amygdala [MeA]) (see Tables S1–S3 in the online supplement) from neurotypical donors as well as donors with a singular diagnosis of PTSD or MDD, or PTSD comorbid with MDD or bipolar disorder (see section 1 in Supplementary Results and Table S2 in the online supplement). After extensive and rigorous quality control of RNA-seq data (see Supplementary Methods, Figure S1, and Table S4 in the online supplement), we performed differential expression and network analyses using 1,285 samples from 325 unique donors (Table 1) and across 26,020 jointly expressed genes (see section 2 in Supplementary Results in the online supplement).

TABLE 1. Demographic and RNA quality information for the subjects and associated brain tissue in this studya

CharacteristicControl (N=109)PTSD (N=107)MDD (N=109)PTSD vs. ControlPTSD vs. MDD
N%N%N%pp
Male8678.95450.560551.62E−050.586
European7669.79487.98880.73.01E−030.285
MeanSDMeanSDMeanSDpp
Age (years)49.615.140.811.345.914.51.93E−064.04E−03
RNA integrity number7.40.87.30.97.30.90.2390.957
Postmortem interval (hours)29.411.129.110.726.77.710.8220.0596
N%N%N%pp
Smoking1816.57973.67165.18.16E−180.187
Opioid use76.427166.46862.41.18E−210.572
Death by suicideNA2624.32522.9NA0.873
Drug-related deathNA7469.25045.9NA5.97E−04

aMDD=major depressive disorder; PTSD=posttraumatic stress disorder; NA=not applicable.

TABLE 1. Demographic and RNA quality information for the subjects and associated brain tissue in this studya

Enlarge table

Expression Differences Related to PTSD Diagnosis

We first explored the gene expression effects of PTSD diagnosis versus neurotypical control donors. We identified 41 PTSD differentially expressed genes (DEGs) in cortex (Figure 1A) and one PTSD DEG in amygdala (Figure 1B) at genome-wide significance (FDR<0.05), while a more liberal threshold of FDR<0.1 identified an additional 78 genes in cortex (and no additional genes in amygdala). We highlight several representative DEGs in PTSD versus neurotypical control donors in cortex, including decreased expression of CORT, which is expressed in a subpopulation of GABAergic inhibitory neurons (38) (Figure 1C); increased expression of the histone deacetylase HDAC4 (Figure 1D); and increased expression of SPRED1, which encodes a protein involved in the Ras/MAPK signaling pathway (Figure 1E). In amygdala, a single gene was consistently downregulated in PTSD versus neurotypical control donors across both subregions—CRHBP (Figure 1F), the gene encoding corticotropin-releasing hormone binding protein, which is an antagonist of the stress hormone corticotropin-releasing hormone (39). Overall, cortical regions showed more association with PTSD than amygdala subregions, and the observed expression differences were largely consistent across subregions of the cortex, with only five genes showing marginal interaction (at p<0.01) between PTSD diagnosis and cortical subregion (NRSN1, PHF20L1, RP11-505E24.2, OXLD1, and CARD8-AS1), and CRHBP only showing modest interaction between PTSD and amygdala subregions (p=0.037).

FIGURE 1.

FIGURE 1. Differential gene expression associated with PTSD compared with neurotypical control subjectsa

aVolcano plots for cortex (panel A) and amygdala (panel B) subregion-combined data set. p values were calculated using linear mixed-effects modeling; the horizontal dashed line indicates the p value that controls a false discovery rate (FDR) <0.05. Positive log2 fold changes indicate higher expression in the PTSD group compared with the neurotypical control group, and negative log2 fold changes indicate lower expression in the PTSD group. Example differentially expressed genes include CORT, HDAC4, SPRED1, and CRHBP (panels C–F), with “adjusted” expression on the y-axis (regressing out unwanted technical and clinical confounders, preserving group and region effects; see Supplementary Methods in the online supplement). BLA=basolateral amygdala; dACC=dorsal anterior cingulate cortex; dlPFC=dorsolateral prefrontal cortex; MeA=medial amygdala; MDD=major depressive disorder; PTSD=posttraumatic stress disorder.

We next performed secondary analyses within each of the four subregions (dlPFC, dACC, BLA, and MeA) to identify additional DEGs associated with PTSD diagnosis. Differential expression statistics were highly correlated with the combined subregion analyses, with the cortical associations driven predominantly by dACC, and the amygdala associations driven primarily by BLA (see Figure S2 in the online supplement). The cortical subregions again showed more PTSD DEGs, with 16 genes in the dACC (see Figure S3A in the online supplement) and one gene in the dlPFC (see Figure S3B in the online supplement) (and no genes in amygdala subregions) at genome-wide significance (FDR<0.05). Using a more liberal cutoff of FDR<0.1, we identified 74 unique genes across the cortical subregions (72 genes in dACC and three genes in dlPFC; one gene was shared: AC124804.1, a novel transcript, antisense to SDK2) and 18 unique genes across amygdala subregions (three genes in BLA and 16 in MeA; one gene was shared: CORT). Joint analysis of all data identified 117 genes with consistent PTSD versus control effects across all four subregions at FDR<0.05 (and with 276 genes at FDR<0.1) (see Figure S4 in the online supplement), further highlighting the similar effects of PTSD across multiple brain regions. Interestingly, these cross-region results were best represented by the amygdala (predominantly BLA), and not the cortex, even though the cortex had more DEGs when considered alone (see Figure S2 in the online supplement). A comprehensive list of all differential expression statistics for all expressed genes and all statistical models is presented in Data S1 in the online supplement.

We next used a series of sensitivity analyses to determine the robustness of our differential expression model by specifically interrogating the role of potential confounders and risk factors. Specifically, we tested a series of additional potential variables (including antidepressant treatment and presence of opioids via toxicology) for attenuating the DEGs identified above in each brain region. Overall, subsequently adjusting our models for these variables had minimal effects on differential expression signals across all expressed genes, including those identified as DEGs (see Figure S5 in the online supplement). We further examined the role of sex on our identified DEGs using sex-specific analyses and found that subsets of DEGs were more strongly explained by effects within a single sex (see Figures S6 and S7 and Table S5 in the online supplement). The identified DEGs mostly confirmed results from a recent study that used a cohort of partially overlapping subjects, with upwards of 80% of expressed genes being directionally consistent (see section 2 of Supplemental Results and Figure S7A in the online supplement) (40). Lastly, we assessed the effects of combat, comparing the 25 combat-exposed donors with PTSD to the 82 PTSD donors without combat exposure within each brain region, and found DEGs exclusively in the MeA (three genes at FDR<0.05, 29 at FDR<0.1, and 116 at FDR<0.2) (see Table S6 in the online supplement).

Taken together, these analyses identified robust sets of differentially expressed genes associated with PTSD that are not a result of association with substance abuse or mood disorder diagnoses.

Gene Sets and Cell Types Associated With PTSD

We next performed gene set and pathway enrichment analyses to identify biological and molecular functions associated with PTSD within and across brain regions. To facilitate these analyses, we used more liberal significance thresholds to define PTSD DEGs (marginal p<0.005 rather than FDR control) and directionality, and tested for enrichment among DEGs more highly and more lowly expressed in PTSD subjects compared with neurotypical control subjects. Overall, genes associated with PTSD showed the strongest enrichment for immune-related gene sets and pathways in both the cortex and the amygdala (Figure 2A; see also Table S7 in the online supplement), largely driven by decreased expression of genes in donors with PTSD compared with control subjects. Interrogating PTSD differences within subregions further identified unique molecular associations. For example, the MeA and dlPFC each showed decreased expression of genes associated with receptor ligand activity (that were further marginally significant in other regions). Interestingly, dlPFC associations were driven by eight genes (CORT, CSF1, SST, OSTN, CXCL10, CXCL11, GDF9, and CCL3) and MeA associations by 10 genes (CORT, TNFSF10, CXCL11, SFRP2, OSGIN2, OGN, IGF2, CTF1, CCL5, and TTR), with only two genes in common (CORT and CXCL11), highlighting the convergence of molecular functions across brain regions.

FIGURE 2.

FIGURE 2. Molecular and cellular enrichments for genes associated with PTSD compared with neurotypical control subjectsa

aGene set enrichment (panel A) and cell-specific enrichment (panel B) analyses for genes more highly expressed (“up”) or more lowly expressed (“down”) in PTSD donors compared with neurotypical donors. Color indicates −log10(p). BLA=basolateral amygdala; dACC=dorsal anterior cingulate cortex; dlPFC=dorsolateral prefrontal cortex; MeA=medial amygdala; PTSD=posttraumatic stress disorder.

We next used cell type-specific enrichment analysis (CSEA) (41) to identify cell types that preferentially express these sets of differentially expressed genes. We found consistent enrichment of cortistatin-expressing GABAergic inhibitory neurons (“Ctx.cort”) and immune cells (“Ctx.etv1_ts88”) among genes where expression was decreased in donors with PTSD compared with neurotypical control subjects. Stronger enrichments were observed in the amygdala, particularly the BLA, compared with the cortex (Figure 2B; see also Table S8 in the online supplement). For example, using a specificity threshold of 0.01 and the BLA, immune cell enrichments were driven by decreased expression of FERMT3, CRHBP, FOLR2, PTGS1, SLCO1C1, P2RY13, and GLT8D2 (odds ratio=8.9, p=2.94e−5), and cortistatin-positive inhibitory neuron enrichments were driven by decreased expression of NPY, CORT, CRHBP, DLL3, NXPH2, and SST (odds ratio=23.7, p=5.9e−7).

We further confirmed enrichment of PTSD DEGs related to immune signaling and inhibitory neurons using snRNA-seq data generated in the human brain from amygdala and dlPFC (42) (see Table S9 in the online supplement). For amygdala DEGs where expression was lower in individuals with PTSD compared with neurotypical control subjects, we found strong enrichment within microglial populations identified in human amygdala (42). These enrichments for DEGs with lower expression in PTSD were strongest in a combined subregion analysis (odds ratio=7.1, p=1.8e−23), but results were driven by the BLA (odds ratio=3.0, p=9.7e−7), with no significant enrichment in the MeA (p=0.17). These more lowly expressed DEGs were also enriched in T-cells at the subregion level (p=2.7e−5), with these results driven by the MeA (p=8.6e−3). Unlike with CSEA, we found evidence for inhibitory neuron enrichments among DEGs with higher expression in PTSD, particularly in the MeA (Inhib_C: odds ratio=2.6, p=2e−5; Inhib_F: odds ratio=3.6, p=1.8e−9). However, this discrepancy could arise as a result of low expression levels in the snRNA-seq data of some genes that drove enrichments in the CSEA analysis. Analogous enrichment analyses using snRNA-seq data on cortical cell types in human brain similarly showed strong enrichment with PTSD DEGs. Using our snRNA-seq data from the dlPFC (42), we found similar strong microglial cell enrichments among DEGs with decreased expression in PTSD (microglia: p=7.7e−22; macrophage: p=1.8e−15), whereas DEGs with increased expression in PTSD were enriched in neuronal populations (Excit_A: p=7.1e−6; Excit_E: p=3.1e−7; Inhib_B: p=1.1e−6; Inhib_D: p=1.1e−6) (42). Using snRNA-seq data from a second study of human prefrontal and cingulate cortices (43), we found that DEGs with increased expression in PTSD were most enriched in a somatostatin (SST)–expressing inhibitory neuron population (IN-SST: p=5.3e−5), whereas DEGs decreased in PTSD were enriched for microglial (p=4.5e−24) and endothelial (p=7.1e−6) populations. Finally, other snRNA-seq data from human prefrontal cortex (44) showed that DEGs with increased expression in PTSD were associated with Ast0, Ex12, In0, In1, In6, In7, and In9 populations, while DEGs with decreased expression in PTSD were associated with Ast2, End1, Mic0, Mic1, Mic2, and Mic3 populations (see Table S9 in the online supplement). Given the limitations of snRNA-seq for detecting relatively rare cell populations, we used an RNAscope single-molecule fluorescence in situ hybridization approach (see Supplementary Methods in the online supplement) in BLA and dlPFC tissue derived from independent neurotypical donors to better understand coexpression of PTSD DEGs associated with inhibitory neurons. For RNAscope analysis, we targeted expression of PTSD DEGs: CORT, SST, and CRHBP (see Data S1 in the online supplement), as well as GAD2 as a cell marker of inhibitory GABAergic cells (Figure 3B). We compared expression levels of these genes across nuclei and found high correlations (Figure 3B), with the highest between CORT and SST (ρ=0.72, p=8.7e−84) and the lowest between GAD2 and CRHBP (ρ=0.166, p=2.1e−13). Almost all SST-positive neurons coexpressed CORT, whereas less than half of CORT-positive neurons coexpressed SST. The top amygdala DEG—CRHBP—showed coexpression with both CORT and GAD2 across many regions of interest in both brain regions (45, 46).

FIGURE 3.

FIGURE 3. Single-molecule fluorescence in situ hybridization validation of inhibitory neuron coexpressiona

aPanel A is a representative image of coexpressing region-of-interest/nucleus across multichannel image. Panel B presents pairwise coexpression plots among four target genes, where axes indicate the number of post-lipofuscin-masked segmented transcript dots (on the log2 scale).

Gene Expression Comparisons Between PTSD and MDD

We next incorporated existing bulk RNA-seq data from MDD donors to better understand the gene expression differences unique to PTSD. We first compared donors with MDD to neurotypical control subjects among the broader cortical and amygdala brain regions, and again identified a larger number of differentially expressed genes in the cortex (182 genes at FDR<0.05, 352 genes at FDR<0.1) (Table 2) compared with the amygdala (zero genes at FDR<0.05, one gene at FDR<0.1). These differences were driven by the dACC (249 genes at FDR<0.1) compared with the dlPFC (two genes at FDR<0.1), similar to PTSD effects. There were similarly increased MDD differences in the MeA (16 genes at FDR<0.05, 32 genes at FDR<0.1) and no differences in BLA when stratifying the amygdala into subregions. Genes with decreased expression in MDD donors compared with neurotypical control subjects showed analogous enrichment of immune-related processes in the cortex using both gene set enrichment analysis (see Table S10 in the online supplement) and CSEA (“Ctx.etv1_ts88” cell type; see Table S11 in the online supplement). CSEA results related to cortistatin-positive neurons were attenuated in MDD compared with PTSD, particularly in the amygdala (best p value, 0.01).

TABLE 2. Number of differentially expressed genes in each data set/brain region at two false discovery rate cutoffsa

PTSD vs. ControlMDD vs. Control
Data SetFDR<0.05FDR<0.1FDR<0.05FDR<0.1
Cortex41119182352
Dorsal anterior cingulate cortex167467249
Dorsolateral prefrontal cortex1312
Amygdala1101
Basolateral amygdala0300
Medial amygdala0161634
Joint11727655192

aFDR=false discovery rate; MDD=major depressive disorder; PTSD=posttraumatic stress disorder.

TABLE 2. Number of differentially expressed genes in each data set/brain region at two false discovery rate cutoffsa

Enlarge table

Globally, there was high concordance between PTSD and MDD effects on gene expression (Figure 4A; see also Figure S10 in the online supplement; ρ range, 0.647–0.695), with highly overlapping DEGs at marginal significance in each brain region or subregion (all Fisher p values <1.72e−46). While global effects were correlated and significant genes were overlapping, there was nevertheless variation among significantly differentially expressed genes across the two disorders. For example, among the genes marginally associated with MDD in each subregion, only one-quarter were significantly differentially expressed when comparing PTSD to control subjects (each at p<0.005), and among the genes that were marginally associated with PTSD, only one-third of genes in cortical regions and one-quarter of genes in amygdala regions showed similar marginal association in MDD.

FIGURE 4.

FIGURE 4. Contrasting PTSD and MDD effects on gene expressiona

aPanel A is a scatterplot comparing the t-statistics for MDD versus control differential expression effects (y-axis) against PTSD versus control effects (x-axis). Colors indicate marginal significance at p<0.005 for PTSD (red), MDD (blue), or both (purple). Panel B is a volcano plot directly comparing the PTSD and MDD groups to each other. The horizontal dotted line indicates marginal p<0.005. Panel C presents gene set enrichment analyses for genes differentially expressed between PTSD and MDD, stratified by directionality. BLA=basolateral amygdala; dACC=dorsal anterior cingulate cortex; dlPFC=dorsolateral prefrontal cortex; MeA=medial amygdala; MDD=major depressive disorder; PTSD=posttraumatic stress disorder.

We therefore directly compared expression between PTSD and MDD donors to better partition these differences across diagnoses (see Supplementary Methods in the online supplement), and identified only a limited number of differentially expressed genes (at FDR<0.1) (Figure 4B). Specifically, we saw increased expression of KCNC1, FAM234B, and RASD2 and decreased expression of CH507-513H4.4 in PTSD versus MDD in the cortex, decreased expression of LMCD1 in PTSD in the MeA, and decreased expression of DNAH11 in PTSD in the dlPFC. In the cortex, marginally significant genes that were more highly expressed in PTSD compared with MDD (at p<0.005) were associated with neuronal processes and synapses (both inhibitory and excitatory), whereas marginally significant genes with decreased expression in PTSD compared with MDD in the amygdala were associated with neuronal migration and PI3K signaling (Figure 4C; see also Table S12 in the online supplement). There were no enrichments for the immune-related gene sets for these disorder-specific contrasts, suggesting that decreased expression of immune processes and/or microglia involvement were shared across both disorders relative to neurotypical individuals (see Table S13 in the online supplement). These results together suggest largely similar transcriptomic changes in PTSD and MDD compared with neurotypical donors.

We then used RNA deconvolution to better determine whether microglia or neurons were more or less prevalent in PTSD and MDD donors (see Supplementary Methods in the online supplement) (47). While the proportion of microglia and neuron RNAs differed by brain region (see Figures S11A,B in the online supplement), there were no differences between diagnoses for either cell type (microglia: PTSD, p=0.84; MDD, p=0.59; neurons: PTSD, p=0.632; MDD, p=0.649). The RNA fractions across all evaluated cell types also were strongly associated with the qSVs used to control for latent heterogeneity—in line with our previous work (34)—suggesting that our DEGs were not confounded by tissue composition (see Figure S11C in the online supplement).

Lastly, we performed weighted gene coexpression analyses (WGCNA) to better understand network-level gene expression differences between PTSD and MDD (see section 4 of Supplementary Results in the online supplement). This analysis identified a total of 156 modules across six WGCNA runs (regions: cortex, amygdala; subregions: dACC, dlPFC, MeA, BLA; see Table S14 in the online supplement), of which 35 were enriched for PTSD DEGs (N=22) or MDD DEGs (N=22; nine overlapping) (Table 3; see also Table S15 in the online supplement). In the cortex and its subregions, the strongest disorder-related module (Cortex.ME7) related to regulation of cell activation, a broad category encompassing many immune processes, associated with both PTSD (p=1.6e−25) and MDD (p=3.3e−126) DEGs, with its eigengene further associated with these diagnoses at the subject level (PTSD, p=2.9e−4; MDD, p=8.4e−6). The strongest disorder-related module in the amygdala (Amygdala.ME2) was specifically enriched with PTSD DEGs (p=2.97e−23), with its eigengene further associated with PTSD compared with control subjects (p=0.005). Sensitivity analyses for other potential confounders, including combat, childhood maltreatment, and toxicology-determined smoking, SSRI antidepressant use, and opioid use, showed minor effects on the WGCNA eigengene associations with PTSD or MDD diagnosis. These variables themselves had weak associations with only a few eigengenes (see section 4 of Supplementary Results in the online supplement). These analyses further highlight biological processes associated with PTSD and MDD using convergent approaches to traditional gene set enrichments of DEGs.

TABLE 3. Module-level associations with PTSD and MDDa

Module_NamenumGenesDEG EnrichmentEigengene AssociationGene Ontology (BP)Cellular Enrichment
PTSDMDDPTSDMDDDescriptionpClassp
Cortex_ME21,3604.40E−034.66E−111.69E−012.58E−05Synapse organization3.49E−14Excit_A5.7E−192
Cortex_ME76201.57E−253.29E−1262.85E−048.45E−06Regulation of cell activation1.68E−42Micro<1E−300
Cortex_ME181253.61E−012.22E−033.03E−015.81E−04Modulation of chemical synaptic transmission6.21E−07Excit_B2.1E−52
Cortex_ME30567.94E−036.65E−013.86E−012.14E−01Regulation of neurotransmitter receptor activity8.82E−05Inhib_A6.4E−14
Cortex_ME31551.70E−101.88E−013.22E−022.42E−01Learning or memory1.37E−04Inhib_B2.7E−03
Cortex_ME34357.27E−031.00E+008.39E−023.82E−01Response to cAMP1.55E−08Astro1.4E−08
dlPFC_ME31,1043.53E−011.12E−031.53E−012.78E−03Regulation of synaptic plasticity6.77E−12Excit_E4.7E−67
dlPFC_ME65027.77E−058.26E−022.35E−036.92E−03Regulation of leukocyte activation5.16E−41Micro<1E−300
dlPFC_ME112996.12E−011.89E−061.93E−026.93E−04Meiotic chromosome separation1.87E−04Inhib_E4.9E−07
dlPFC_ME201172.71E−154.44E−029.14E−033.23E−01UDP-N-acetylglucosamine metabolic process1.36E−04Tcell2.4E−03
dlPFC_ME22982.37E−036.32E−011.38E−018.25E−01Response to estradiol4.15E−04Excit_B1.4E−25
dlPFC_ME24841.61E−042.69E−035.41E−027.92E−02Membrane depolarization during action potential7.82E−07Inhib_D1.1E−21
dACC_ME31,3931.55E−031.61E−231.01E−021.84E−08Modulation of chemical synaptic transmission2.95E−22Excit_A6.9E−263
dACC_ME57473.60E−032.70E−062.20E−021.10E−03Forebrain development6.78E−10Excit_B3.6E−172
dACC_ME77022.66E−033.28E−841.26E−032.03E−05Lymphocyte activation3.57E−42Micro<1E−300
dACC_ME95991.42E−086.02E−014.79E−014.68E−02Modulation of chemical synaptic transmission2.01E−09Excit_B3.1E−42
dACC_ME121971.03E−016.02E−048.96E−011.11E−01Heart development1.48E−06Astro4.7E−121
dACC_ME131776.56E−031.00E+006.39E−018.99E−02Negative regulation of translation9.03E−05Tcell5.8E−07
Amygdala_ME11,0351.90E−053.66E−042.49E−013.99E−02Myelination2.73E−13Oligo<1E−300
Amygdala_ME29042.97E−232.59E−034.70E−031.15E−01Lymphocyte activation1.70E−38Micro<1E−300
Amygdala_ME46961.90E−013.67E−052.56E−011.12E−01Modulation of chemical synaptic transmission5.27E−26Excit_B5.2E−169
Amygdala_ME19479.15E−027.72E−037.88E−035.43E−03Extracellular matrix constituent secretion3.05E−04Inhib_D7.3E−06
Amygdala_ME21398.55E−032.86E−011.49E−021.15E−02Regulation of system process3.77E−03Inhib_B3.8E−06
Amygdala_ME24322.94E−012.60E−033.17E−011.09E−01Formation of quadruple SL/U4/U5/U6 snRNP4.84E−07Astro_A1.7E−01
MeA_ME34319.88E−125.72E−014.09E−025.13E−01Modulation of chemical synaptic transmission7.17E−18Inhib_D8.3E−147
MeA_ME43753.63E−013.22E−042.60E−014.32E−02Modulation of chemical synaptic transmission1.71E−28Excit_A1.1E−159
MeA_ME52811.11E−013.82E−036.05E−025.99E−03Regulation of ion transmembrane transport1.21E−07Inhib_C2.9E−69
MeA_ME71535.00E−032.78E−061.53E−017.61E−02Extracellular structure organization2.98E−10Astro_B1.2E−90
MeA_ME9866.34E−017.33E−044.35E−012.20E−03Ameboidal-type cell migration1.45E−04Excit_A3.9E−31
MeA_ME10853.21E−011.65E−051.01E−014.65E−04Regulation of membrane potential7.35E−06Inhib_F8.3E−27
MeA_ME13637.22E−212.34E−265.51E−037.62E−04Extracellular matrix organization1.36E−18Mural3.0E−36
BLA_ME21,3003.25E−095.09E−011.32E−029.98E−02Lymphocyte activation1.81E−30Micro<1E−300
BLA_ME121994.08E−036.60E−022.52E−021.37E−01Homophilic cell adhesion via plasma membrane adhesion molecules4.41E−06Astro_A3.1E−92
BLA_ME131762.36E−076.47E−013.85E−035.21E−01Locomotory behavior1.98E−06Inhib_D2.6E−68
BLA_ME20375.31E−021.13E−086.37E−021.17E−02Spliceosomal tri-snRNP complex assembly9.98E−08Astro_A1.1E−01

aFisher’s exact test enrichment for differentially expressed genes (DEGs) (at p<0.005) in PTSD (PTSD_Pval) and MDD (MDD_Pval) among module gene membership. Eigengene subject-level associations with PTSD versus control (PTSD_p) and MDD versus control (MDD_p). The top Gene Ontology biological process is shown for module gene membership (GOBP_Description) with corresponding p value (GOBP_pvalue). MDD=major depressive disorder; PTSD=posttraumatic stress disorder.

TABLE 3. Module-level associations with PTSD and MDDa

Enlarge table

Discussion

The goal of this study was to identify shared versus divergent in transcriptional patterns within and across PTSD and MDD in the prefrontal cortex and amygdala. We identified a limited number of DEGs specific to PTSD, with nearly all mapping to cortex versus amygdala. PTSD-specific DEGs were enriched in gene sets associated with immune-related pathways and microglia and with subpopulations of GABAergic inhibitory neurons. While we identified a greater number of DEGs associated with MDD, most overlapped with PTSD, and only a few were MDD specific. These findings provide supporting evidence for involvement of immune signaling and neuroinflammation in MDD and PTSD pathophysiology and extend evidence that GABAergic neurons have functional significance in PTSD.

Decreased expression of genes included in immune-related Gene Ontology sets were associated with PTSD diagnosis in both cortical and amygdala brain regions (Figure 3A). CSEA using mouse cell-specific markers and snRNA-seq data from human brain demonstrated enrichment of these DEGs, with decreased expression in PTSD among microglia profiles (41, 42). Genes with decreased expression in MDD donors compared with neurotypical control subjects showed analogous enrichment of immune-related processes using both gene set enrichment analysis and CSEA, and there were no enrichments for the immune and microglia-related genes when contrasting PTSD and MDD, suggesting that decreased expression of immune processes and microglia involvement are not specific to PTSD. The downward direction of dysregulation was somewhat surprising, considering that higher pre-trauma levels of C-reactive protein (a marker of blood inflammation) have been reported to predict elevated PTSD symptoms after trauma (48). Furthermore, elevated levels of selected markers of low-grade blood inflammation have been reported in a meta-analysis of PTSD studies (49). However, over time, and with repeated exposure to chronic stress and trauma, immune function may become dysregulated in a myriad of ways, with neuronal, glial, and peripheral systems attempting to compensate for immune activation and increased inflammation (33, 5052). While decreased expression of the microglial immune transcriptome and/or reductions in microglial cell ratios due to chronic immune dysregulation are possible explanations for the present data, we noted that many of the genes included in the associated immune Gene Ontology sets encode proteins with known immunosuppressive activity. This could also explain the somewhat paradoxical finding of decreased expression of immune-related genes. For example, in the immune-related regulation of cell activity category, we identified 13 member PTSD DEGs (IL1RL2, DPP4, IGFBP2, TGFBR2, TAC1, MDK, CD4, PTPN6, TESPA1, IGF1, ITGAM, TYROBP, and ITGB2), of which seven have potential immunosuppressive activity (DPP4, TGFBR2, CD4, IGF1, ITGAM, TYROBP, and ITGB2) (53). These observations do not support a high level of microglial immune activation in chronic PTSD or MDD in cortex or amygdala, but they do suggest dysregulation or possibly a compensatory response to stress.

We observed downregulation of CORT mRNA across all four subregions in individuals with PTSD. CORT encodes the secreted neuropeptide cortistatin, which is expressed in the cerebral cortex, hippocampus, and amygdala in a subset of GABAergic neurons (38, 54). Loss of cortistatin cells in mice causes spontaneous seizures, demonstrating that these cells provide strong inhibitory control (55, 56). In the rodent, cells expressing cortistatin constitute a subset of SST-expressing neurons (55), and in human brain we confirmed coexpression of CORT with GABAergic inhibitory neuron markers (GAD2, SST, and CRHBP). Decreased CORT and SST expression were previously reported in amygdala of female postmortem human brain donors with MDD (57), and our CSEA analyses showed enrichment of genes differentially expressed in PTSD in cortistatin-expressing cells. We also identified enrichment of PTSD DEGs with specific inhibitory neuron clusters from snRNA-seq data in human amygdala that have been associated with anxiety and HPA axis function (42, 58). WGCNA further implicated inhibitory neuron function, in line with the gene set enrichment results applied directly to PTSD DEGs. Decreased expression of CORT, SST, and CRHBP mRNA provides additional support for the hypothesis that GABAergic neuron dysfunction is mechanistically associated with PTSD (22). Strong evidence implicates GABAergic neurons in controlling fear-related behaviors in preclinical animal models relevant for PTSD and other trauma-related disorders by controlling neural activity and synaptic plasticity in cortico-amygdala circuits (23). For example, firing of excitatory cells that project from the BLA to the frontal cortex is under tight regulation by local GABAergic inhibitory neurons (25), which provides negative feedback regulation of the BLA to control both the expression and extinction of fear (26, 59). Strong evidence links somatostatin signaling and SST-positive cells in cortico-amygdala circuits with threat perception and fear memory processing (6064). CRHBP, which we showed to be coexpressed with CORT and SST in GABAergic neurons in the human brain, was the only gene consistently downregulated in PTSD versus neurotypical control subjects across both subregions of the amygdala. CRHBP encodes corticotropin-releasing hormone binding protein, which sequesters and antagonizes CRH signaling (39). The robust DEG signal for CRHBP is interesting given many studies implicating the stress hormones corticotropin-releasing hormone (CRH) and cortisol in PTSD (6568). CRH activates the release of adrenocorticotropic hormone (ACTH), which stimulates production of cortisol to control the body’s response to stress and trauma, which is important given that stress is a leading risk factor for the development of PTSD (69). Additional genetic support for a critical role of the stress axis in PTSD comes from recent genome-wide association study associations of CRHR1 (70), which encodes the primary CRH receptor—CRF1—and CRH binds to CRF1 to mediate the behavioral and endocrine responses to stress exposure.

We further ran a number of analyses to better identify gene expression differences that were selectively associated with PTSD but not MDD. In general, we identified more DEGs for MDD than PTSD, particularly in the cortex (which were primarily driven by the dACC). However, gene expression differences were highly concordant between the two diagnoses, with most highly significant DEGs showing the same directionality of effects (i.e., log2 fold changes) in both diagnoses. Marginally selective between-diagnosis DEG gene sets included more highly expressed glutamatergic synapse-related DEGs in PTSD cortex (driven by the dACC) and more highly expressed neuronal activity-related DEGs in MDD amygdala (driven by the BLA). Differences between the two diagnoses were more prominent in WGCNA analyses, where seven potentially overlapping modules showed PTSD-specific enrichment (Cortex_ME31, dlPFC_ME20, dACC_ME9, Amygdala_ME2, MeA_ME3, BLA_ME2, BLA_ME13) and five modules showed MDD-specific enrichment (dlPFC_ME11, dACC_ME3, dACC_ME7, MeA_ME9, BLA_ME20).

DEGs from a recent RNA-seq study of human postmortem PTSD tissue by Girgenti et al. (40), which used a partially overlapping set of donors (see below), provide support for top DEGs identified here. For example, within our combined cortical PTSD analyses (see Figure 1), six of the seven most robustly affected transcripts comparing PTSD and control subjects (CORT, HDAC4, CRHBP, ADAMTS2, FBXO9, and APOC1) were directionally consistent and at least marginally significant in this previous data set. These genes further showed decreased expression in MDD versus control subjects in the present study, with at least marginal significance, suggesting that these particular findings may be related to shared pathophysiological changes accompanying PTSD and MDD. A key upregulated gene identified in the Girgenti et al. study, ELK1, was significantly upregulated in both cortical regions in the present study, and SST, identified as robustly downregulated in several regions of the cortex in the Girgenti et al. study, was in the top 10 of all downregulated transcripts in both dlPFC and dACC here. ADAMTS2, the second highest upregulated DEG in the combined cortical sample, was the top upregulated gene in the dACC and the third most upregulated in the dlPFC in the Girgenti et al. study. HDAC4, a top DEG, has been associated previously with both PTSD and rodent models of PTSD (71, 72). The Girgenti et al. study also identified enrichment of downregulated PTSD-associated DEGs using CSEA that were related to GABAergic neurons and their molecular functions.

In addition to these common elements, the present results extend previous findings from Girgenti et al. (40) in several key areas. First, this study extended the search for differential gene expression beyond the cortex and into the amygdala, a relatively understudied brain area in postmortem human brain research with high relevance to PTSD. Second, we provide compelling evidence implicating decreased expression of immune-related genes and associated processes in PTSD and MDD compared with neurotypical control subjects. This is an important observation because it runs counter to most expectations for immune response directionality. We further refined these cellular enrichments in GABAergic neurons more specifically to CORT-positive interneurons, which we subsequently validated with RNAscope. The cell type analyses in the present study provide direct evidence of these enrichments by interrogating DEGs directly against cell type-specific genes from both human and mouse studies, complementing the indirect strategy taken by Girgenti et al. of first identifying genes in discrete coexpressed modules and then associating those genes with both PTSD DEGs and cell type-specific genes separately (such that different genes captured the cell type vs. PTSD signal in the same module). Third, we believe our larger sample size (more than twice as large in all diagnostic groups), obtained from a single postmortem brain collection under identical sample ascertainment and inclusion criteria, refined several of the clinical associations identified by Girgenti et al. We identified more similarities than differences between PTSD and MDD and replicated this finding across both amygdala and cortical subregions, with far less sex-specific diagnosis-associated signal than discussed in the earlier study. Contributing to both the similarities and differences between the two studies was the fact that 77 donors were shared across the studies, although the two studies used different hemispheres, independent dissections and RNA extractions, and different data analysis pipelines. Over half (53.8%) of the donors in the Girgenti et al. study overlapped with one-quarter (23.7%) of the donors in the present study.

Therefore, it might seem counterintuitive that we identified many fewer DEGs in this much larger study, particularly with overlapping donors. We believe these differences can be accounted for by our more conservative statistical analyses, including the modeling of both diagnostic groups in a single statistical model against the neurotypical group, which further accounted for robust observed and latent confounders. The differential expression models in the Girgenti et al. study (40) only adjusted for age, RNA integrity number, postmortem interval, and race, and did not account for sequencing-derived RNA quality metrics and other latent confounders, which can greatly increase false positive rates in human postmortem brain gene expression studies (36). For example, this less comprehensive statistical model applied to our larger data set resulted in 1,243 DEGs in the dlPFC, 1,719 DEGs in the dACC, 1,813 DEGs in the BLA, and 10,283 DEGs in the MeA for PTSD at FDR<0.05, which is many more genes than obtained with our more conservative approach. There has been some debate regarding the optimal methods of latent variable correction in these types of postmortem studies, including the potential for “overcorrection” (73). A major analytic element of the present investigation was the use of quality surrogate variable (qSV) analysis to identify and correct for expressed sequences that are particularly prone to degradation in human postmortem brain (36). The qSVs utilized here were defined from the top 1,000 degradation-susceptible expressed regions generated from independent time course experiments. Dropping the qSVs from our main analyses resulted in 209 DEGs in the dlPFC, 43 DEGs in the dACC, 62 DEGs in the BLA, and 1,054 DEGs in the MeA (at FDR<0.05) for PTSD in the present data set. It is, however, possible that if sex or disease-associated interactions related to gene transcript degradation exist, use of qSVs may have limited the emergence of these genes as DEGs and contributed to the differences between the present findings and those discussed in the Girgenti et al. study (40). Similarly, in MDD, the most prominent previous report of differential gene expression (74) actually identified no DEGs when correcting for multiple testing via the FDR (from the supplementary tables included with that study), making it difficult to assess replication of our DEGs using previously published data sets. While these issues may seem rather nuanced, they nevertheless have important consequences for identifying DEGs in human postmortem RNA-seq data sets and require careful consideration in past and future work.

Limitations of this work include potential underrepresentation of female donors in the neurotypical control group (21.1% female) compared with the case group (∼50% female). While we adjusted for sex in differential expression analyses, secondary analyses did suggest some potential differences in diagnosis effects across sex. Our cohort included donors with only a PTSD diagnosis or only an MDD diagnosis, as well as PTSD donors with a comorbid MDD or bipolar disorder diagnosis. However, we did not have subjects with only a bipolar disorder diagnosis. Furthermore, as is common in most psychiatric postmortem human studies, psychotropic medications, substance use, smoking, and suicide were more common in the MDD and PTSD groups, and further work will be needed to investigate their potential influence on brain gene expression patterns.

In summary, these analyses of the largest postmortem brain cohort of patients with PTSD and MDD to date highlight microglia and other immune cell types as having potential functional significance in PTSD, and provide additional evidence for dysregulated neuroinflammation and neuroimmune signaling in MDD and PTSD pathophysiology.

Lieber Institute for Brain Development, Baltimore (Jaffe, Tao, Page, Maynard, Pattie, Nguyen, Deep-Soboslay, Bharadwaj, Shin, Hyde, Martinowich, Kleinman); Department of Neuroscience (Jaffe, Martinowich), Department of Genetic Medicine, McKusick-Nathans Institute of Genetic Medicine (Jaffe), and Department of Psychiatry and Behavioral Sciences (Jaffe, Hyde, Martinowich, Kleinman), Johns Hopkins University School of Medicine, Baltimore; Center for Computational Biology, Johns Hopkins University, Baltimore (Jaffe); Department of Biostatistics and Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore (Jaffe); Department of Psychiatry and Behavioral Sciences, Texas A&M College of Medicine, Bryan, Tex., Department of Veterans Affairs, VISN 17 Center of Excellence for Research on Returning War Veterans, Waco, Tex., Central Texas Veterans Health Care System, Temple, Tex., and Baylor Scott & White Psychiatry, Temple, Tex. (Young); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Hanover, N.H. (Friedman); National Center for PTSD, U.S. Department of Veterans Affairs (Friedman); Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, N.C. (Williamson); Durham VA Health Care System, Durham, N.C. (Williamson); Department of Neurology, Johns Hopkins School of Medicine, Baltimore (Hyde).
Send correspondence to Dr. Martinowich () and Dr. Kleinman ().

Supported by the Lieber Institute for Brain Development and contract VA-241-17-C-0138 from the U.S. Department of Veterans Affairs.

The views expressed here are those of the authors and do not necessarily reflect the position or policy of the U.S. Department of Veterans Affairs or the U.S. government.

Data availability: All code and figures associated with this manuscript are available through GitHub: https://github.com/LieberInstitute/LIBD_VA_PTSD_RNAseq_4Region. All raw and processed data may be requested via the PTSD Brain Bank Resource Request process described on the “For Investigators” tab of the following page: https://www.research.va.gov/programs/tissue_banking/ptsd/default.cfm.

Dr. Jaffe is now employed by Neumora Therapeutics. The other authors report no financial relationships with commercial interests.

The authors express their gratitude to their colleagues whose tireless efforts have led to the donation of postmortem tissue to advance these studies: the Office of the Chief Medical Examiner of the District of Columbia, the Office of the Chief Medical Examiner for Northern Virginia (Fairfax), the Office of the Chief Medical Examiner of the State of Maryland (Baltimore), the Office of the Chief Medical Examiner for Kalamazoo County (Kalamazoo, Mich.), the University of North Dakota School of Medicine Department of Pathology, Forensic Pathology Center (Grand Forks), and the Santa Clara County Office of the Chief Medical Examiner (San Jose, Calif.). This work was supported with resources and use of facilities at the VA Connecticut Health Care System, West Haven; the Central Texas Veterans Health Care System, Temple, Tex.; the Durham VA Healthcare System, Durham, N.C.; the VA San Diego Healthcare System, La Jolla, Calif.; the VA Boston Healthcare System, Boston; and the National Center for PTSD, U.S. Department of Veterans Affairs. The authors acknowledge the contributions of Amy Deep-Soboslay and Llewellyn B. Bigelow, M.D., for their diagnostic expertise and Dr. Daniel R. Weinberger for providing constructive commentary and editing of the manuscript. Finally, the authors are indebted to the generosity of the families of the decedents, who donated the brain tissue used in these studies.

References

1. Armenta RF, Walter KH, Geronimo-Hara TR, et al.: Longitudinal trajectories of comorbid PTSD and depression symptoms among US service members and veterans. BMC Psychiatry 2019; 19:396Crossref, MedlineGoogle Scholar

2. Walter KH, Levine JA, Highfill-McRoy RM, et al.: Prevalence of posttraumatic stress disorder and psychological comorbidities among US active duty service members, 2006–2013. J Trauma Stress 2018; 31:837–844Crossref, MedlineGoogle Scholar

3. Kang B, Xu H, McConnell ES: Neurocognitive and psychiatric comorbidities of posttraumatic stress disorder among older veterans: a systematic review. Int J Geriatr Psychiatry 2019; 34:522–538Crossref, MedlineGoogle Scholar

4. Flory JD, Yehuda R: Comorbidity between post-traumatic stress disorder and major depressive disorder: alternative explanations and treatment considerations. Dialogues Clin Neurosci 2015; 17:141–150Crossref, MedlineGoogle Scholar

5. Kessler RC, Sonnega A, Bromet E, et al.: Posttraumatic stress disorder in the National Comorbidity Survey. Arch Gen Psychiatry 1995; 52:1048–1060Crossref, MedlineGoogle Scholar

6. Rytwinski NK, Scur MD, Feeny NC, et al.: The co-occurrence of major depressive disorder among individuals with posttraumatic stress disorder: a meta-analysis. J Trauma Stress 2013; 26:299–309Crossref, MedlineGoogle Scholar

7. Otto MW, Perlman CA, Wernicke R, et al.: Posttraumatic stress disorder in patients with bipolar disorder: a review of prevalence, correlates, and treatment strategies. Bipolar Disord 2004; 6:470–479Crossref, MedlineGoogle Scholar

8. Neria Y, Olfson M, Gameroff MJ, et al.: Trauma exposure and posttraumatic stress disorder among primary care patients with bipolar spectrum disorder. Bipolar Disord 2008; 10:503–510Crossref, MedlineGoogle Scholar

9. Fenster RJ, Lebois LAM, Ressler KJ, et al.: Brain circuit dysfunction in post-traumatic stress disorder: from mouse to man. Nat Rev Neurosci 2018; 19:535–551Crossref, MedlineGoogle Scholar

10. Berretta S: Cortico-amygdala circuits: role in the conditioned stress response. Stress 2005; 8:221–232Crossref, MedlineGoogle Scholar

11. Lobo I, de Oliveira L, David IA, et al.: The neurobiology of posttraumatic stress disorder: dysfunction in the prefrontal-amygdala circuit? Psychol Neurosci 2011; 4:191–203CrossrefGoogle Scholar

12. Brown VM, LaBar KS, Haswell CC, et al.: Altered resting-state functional connectivity of basolateral and centromedial amygdala complexes in posttraumatic stress disorder. Neuropsychopharmacology 2014; 39:351–359Crossref, MedlineGoogle Scholar

13. Zhu X, Helpman L, Papini S, et al.: Altered resting state functional connectivity of fear and reward circuitry in comorbid PTSD and major depression. Depress Anxiety 2017; 34:641–650Crossref, MedlineGoogle Scholar

14. Chen AC, Etkin A: Hippocampal network connectivity and activation differentiates post-traumatic stress disorder from generalized anxiety disorder. Neuropsychopharmacology 2013; 38:1889–1898Crossref, MedlineGoogle Scholar

15. Rauch SL, Shin LM, Phelps EA: Neurocircuitry models of posttraumatic stress disorder and extinction: human neuroimaging research–past, present, and future. Biol Psychiatry 2006; 60:376–382Crossref, MedlineGoogle Scholar

16. Stevens JS, Jovanovic T, Fani N, et al.: Disrupted amygdala-prefrontal functional connectivity in civilian women with posttraumatic stress disorder. J Psychiatr Res 2013; 47:1469–1478Crossref, MedlineGoogle Scholar

17. Sun D, Gold AL, Swanson AL, et al.: Threat-induced anxiety during goal pursuit disrupts amygdala-prefrontal cortex connectivity in posttraumatic stress disorder. Transl Psychiatry 2020; 10:61Crossref, MedlineGoogle Scholar

18. Likhtik E, Stujenske JM, Topiwala MA, et al.: Prefrontal entrainment of amygdala activity signals safety in learned fear and innate anxiety. Nat Neurosci 2014; 17:106–113Crossref, MedlineGoogle Scholar

19. Kumar S, Hultman R, Hughes D, et al.: Prefrontal cortex reactivity underlies trait vulnerability to chronic social defeat stress. Nat Commun 2014; 5:4537Crossref, MedlineGoogle Scholar

20. Bukalo O, Pinard CR, Silverstein S, et al.: Prefrontal inputs to the amygdala instruct fear extinction memory formation. Sci Adv 2015; 1:e1500251Crossref, MedlineGoogle Scholar

21. Sotres-Bayon F, Sierra-Mercado D, Pardilla-Delgado E, et al.: Gating of fear in prelimbic cortex by hippocampal and amygdala inputs. Neuron 2012; 76:804–812Crossref, MedlineGoogle Scholar

22. Fogaça MV, Duman RS: Cortical GABAergic dysfunction in stress and depression: new insights for therapeutic interventions. Front Cell Neurosci 2019; 13:87Crossref, MedlineGoogle Scholar

23. Fang Q, Li Z, Huang G-D, et al.: Traumatic stress produces distinct activations of GABAergic and glutamatergic neurons in amygdala. Front Neurosci 2018; 12:387Crossref, MedlineGoogle Scholar

24. Tovote P, Fadok JP, Lüthi A: Neuronal circuits for fear and anxiety. Nat Rev Neurosci 2015; 16:317–331Crossref, MedlineGoogle Scholar

25. Ghosal S, Hare B, Duman RS: Prefrontal cortex GABAergic deficits and circuit dysfunction in the pathophysiology and treatment of chronic stress and depression. Curr Opin Behav Sci 2017; 14:1–8Crossref, MedlineGoogle Scholar

26. Krabbe S, Gründemann J, Lüthi A: Amygdala inhibitory circuits regulate associative fear conditioning. Biol Psychiatry 2018; 83:800–809Crossref, MedlineGoogle Scholar

27. Yu B, Becnel J, Zerfaoui M, et al.: Serotonin 5-hydroxytryptamine(2A) receptor activation suppresses tumor necrosis factor-alpha-induced inflammation with extraordinary potency. J Pharmacol Exp Ther 2008; 327:316–323Crossref, MedlineGoogle Scholar

28. Lori A, Maddox SA, Sharma S, et al.: Dynamic patterns of threat-associated gene expression in the amygdala and blood. Front Psychiatry 2018; 9:778Crossref, MedlineGoogle Scholar

29. Règue M, Poilbout C, Martin V, et al.: Increased 5-HT2C receptor editing predisposes to PTSD-like behaviors and alters BDNF and cytokines signaling. Transl Psychiatry 2019; 9:100Crossref, MedlineGoogle Scholar

30. Young MB, Howell LL, Hopkins L, et al.: A peripheral immune response to remembering trauma contributes to the maintenance of fear memory in mice. Psychoneuroendocrinology 2018; 94:143–151Crossref, MedlineGoogle Scholar

31. Bhatt S, Hillmer AT, Girgenti MJ, et al.: PTSD is associated with neuroimmune suppression: evidence from PET imaging and postmortem transcriptomic studies. Nat Commun 2020; 11:2360Crossref, MedlineGoogle Scholar

32. Breen MS, Tylee DS, Maihofer AX, et al.: PTSD blood transcriptome mega-analysis: shared inflammatory pathways across biological sex and modes of trauma. Neuropsychopharmacology 2018; 43:469–481Crossref, MedlineGoogle Scholar

33. Michopoulos V, Powers A, Gillespie CF, et al.: Inflammation in fear- and anxiety-based disorders: PTSD, GAD, and beyond. Neuropsychopharmacology 2017; 42:254–270Crossref, MedlineGoogle Scholar

34. Collado-Torres L, Burke EE, Peterson A, et al.: Regional heterogeneity in gene expression, regulation, and coherence in the frontal cortex and hippocampus across development and schizophrenia. Neuron 2019; 103:203–216.e8Crossref, MedlineGoogle Scholar

35. Law CW, Chen Y, Shi W, et al.: voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol 2014; 15:R29Crossref, MedlineGoogle Scholar

36. Jaffe AE, Tao R, Norris AL, et al.: qSVA framework for RNA quality correction in differential expression analysis. Proc Natl Acad Sci USA 2017; 114:7130–7135Crossref, MedlineGoogle Scholar

37. Langfelder P, Horvath S: WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 2008; 9:559Crossref, MedlineGoogle Scholar

38. de Lecea L, del Rio JA, Criado JR, et al.: Cortistatin is expressed in a distinct subset of cortical interneurons. J Neurosci 1997; 17:5868–5880Crossref, MedlineGoogle Scholar

39. Laryea G, Arnett MG, Muglia LJ: Behavioral studies and genetic alterations in corticotropin-releasing hormone (CRH) neurocircuitry: insights into human psychiatric disorders. Behav Sci (Basel) 2012; 2:135–171Crossref, MedlineGoogle Scholar

40. Girgenti MJ, Wang J, Ji D, et al.: Transcriptomic organization of the human brain in post-traumatic stress disorder. Nat Neurosci 2021; 24:24–33Crossref, MedlineGoogle Scholar

41. Xu X, Wells AB, O’Brien DR, et al.: Cell type-specific expression analysis to identify putative cellular mechanisms for neurogenetic disorders. J Neurosci 2014; 34:1420–1431Crossref, MedlineGoogle Scholar

42. Tran MN, Maynard KR, Spangler A, et al.: Single-nucleus transcriptome analysis reveals cell-type-specific molecular signatures across reward circuitry in the human brain. Neuron 2021; 109:3088–3103.e5Crossref, MedlineGoogle Scholar

43. Velmeshev D, Schirmer L, Jung D, et al.: Single-cell genomics identifies cell type-specific molecular changes in autism. Science 2019; 364:685–689Crossref, MedlineGoogle Scholar

44. Mathys H, Davila-Velderrain J, Peng Z, et al.: Single-cell transcriptomic analysis of Alzheimer’s disease. Nature 2019; 570:332–337Crossref, MedlineGoogle Scholar

45. Ketchesin KD, Huang NS, Seasholtz AF: Cell type-specific expression of corticotropin-releasing hormone-binding protein in GABAergic interneurons in the prefrontal cortex. Front Neuroanat 2017; 11:90Crossref, MedlineGoogle Scholar

46. Calakos KC, Blackman D, Schulz AM, et al.: Distribution of type I corticotropin-releasing factor (CRF1) receptors on GABAergic neurons within the basolateral amygdala. Synapse 2017; 71:10CrossrefGoogle Scholar

47. Burke EE, Chenoweth JG, Shin JH, et al.: Dissecting transcriptomic signatures of neuronal differentiation and maturation using iPSCs. Nat Commun 2019; 11:462CrossrefGoogle Scholar

48. Eraly SA, Nievergelt CM, Maihofer AX, et al.: Assessment of plasma C-reactive protein as a biomarker of posttraumatic stress disorder risk. JAMA Psychiatry 2014; 71:423–431Crossref, MedlineGoogle Scholar

49. Passos IC, Vasconcelos-Moreno MP, Costa LG, et al.: Inflammatory markers in post-traumatic stress disorder: a systematic review, meta-analysis, and meta-regression. Lancet Psychiatry 2015; 2:1002–1012Crossref, MedlineGoogle Scholar

50. Haroon E, Raison CL, Miller AH: Psychoneuroimmunology meets neuropsychopharmacology: translational implications of the impact of inflammation on behavior. Neuropsychopharmacology 2012; 37:137–162Crossref, MedlineGoogle Scholar

51. Ménard C, Pfau ML, Hodes GE, et al.: Immune and neuroendocrine mechanisms of stress vulnerability and resilience. Neuropsychopharmacology 2017; 42:62–80Crossref, MedlineGoogle Scholar

52. Deslauriers J, Powell S, Risbrough VB: Immune signaling mechanisms of PTSD risk and symptom development: insights from animal models. Curr Opin Behav Sci 2017; 14:123–132Crossref, MedlineGoogle Scholar

53. Liu Y, He M, Wang D, et al.: HisgAtlas 1.0: a human immunosuppression gene database. Database (Oxford) 2017; 2017:bax094Crossref, MedlineGoogle Scholar

54. de Lecea L: Cortistatin: functions in the central nervous system. Mol Cell Endocrinol 2008; 286:88–95Crossref, MedlineGoogle Scholar

55. Hill JL, Jimenez DV, Mai Y, et al.: Cortistatin-expressing interneurons require TrkB signaling to suppress neural hyper-excitability. Brain Struct Funct 2019; 224:471–483Crossref, MedlineGoogle Scholar

56. Maynard KR, Kardian A, Hill JL, et al.: TrkB signaling influences gene expression in cortistatin-expressing interneurons. eNeuro 2020; 7:ENEURO.0310-19.2019CrossrefGoogle Scholar

57. Guilloux J-P, Douillard-Guilloux G, Kota R, et al.: Molecular evidence for BDNF- and GABA-related dysfunctions in the amygdala of female subjects with major depression. Mol Psychiatry 2012; 17:1130–1142Crossref, MedlineGoogle Scholar

58. Babaev O, Piletti Chatain C, Krueger-Burg D: Inhibition in the amygdala anxiety circuitry. Exp Mol Med 2018; 50:1–16Crossref, MedlineGoogle Scholar

59. Janak PH, Tye KM: From circuits to behaviour in the amygdala. Nature 2015; 517:284–292Crossref, MedlineGoogle Scholar

60. Cummings KA, Clem RL: Prefrontal somatostatin interneurons encode fear memory. Nat Neurosci 2020; 23:61–74Crossref, MedlineGoogle Scholar

61. Koppensteiner P, Von Itter R, Melani R, et al.: Diminished fear extinction in adolescents is associated with an altered somatostatin interneuron-mediated inhibition in the infralimbic cortex. Biol Psychiatry 2019; 86:682–692Crossref, MedlineGoogle Scholar

62. Xu H, Liu L, Tian Y, et al.: A disinhibitory microcircuit mediates conditioned social fear in the prefrontal cortex. Neuron 2019; 102:668–682.e5Crossref, MedlineGoogle Scholar

63. Sun Y, Qian L, Xu L, et al.: Somatostatin neurons in the central amygdala mediate anxiety by disinhibition of the central sublenticular extended amygdala. Mol Psychiatry (Online ahead of print, October 1, 2020)Google Scholar

64. Yu K, Garcia da Silva P, Albeanu DF, et al.: Central amygdala somatostatin neurons gate passive and active defensive behaviors. J Neurosci 2016; 36:6488–6496Crossref, MedlineGoogle Scholar

65. Tyrka AR, Price LH, Gelernter J, et al.: Interaction of childhood maltreatment with the corticotropin-releasing hormone receptor gene: effects on hypothalamic-pituitary-adrenal axis reactivity. Biol Psychiatry 2009; 66:681–685Crossref, MedlineGoogle Scholar

66. Dedic N, Kühne C, Jakovcevski M, et al.: Chronic CRH depletion from GABAergic, long-range projection neurons in the extended amygdala reduces dopamine release and increases anxiety. Nat Neurosci 2018; 21:803–807Crossref, MedlineGoogle Scholar

67. Risbrough VB, Stein MB: Role of corticotropin releasing factor in anxiety disorders: a translational research perspective. Horm Behav 2006; 50:550–561Crossref, MedlineGoogle Scholar

68. Toth M, Flandreau EI, Deslauriers J, et al.: Overexpression of forebrain CRH during early life increases trauma susceptibility in adulthood. Neuropsychopharmacology 2016; 41:1681–1690Crossref, MedlineGoogle Scholar

69. Gradus J: Epidemiology of PTSD. National Center for PTSD, US Department of Veterans Affairs, 2016Google Scholar

70. Gelernter J, Sun N, Polimanti R, et al.: Genome-wide association study of post-traumatic stress disorder reexperiencing symptoms in >165,000 US veterans. Nat Neurosci 2019; 22:1394–1401Crossref, MedlineGoogle Scholar

71. Maddox SA, Kilaru V, Shin J, et al.: Estrogen-dependent association of HDAC4 with fear in female mice and women with PTSD. Mol Psychiatry 2018; 23:658–665Crossref, MedlineGoogle Scholar

72. Chaby LE, Sadik N, Burson NA, et al.: Repeated stress exposure in mid-adolescence attenuates behavioral, noradrenergic, and epigenetic effects of trauma-like stress in early adult male rats. Sci Rep 2020; 10:17935Crossref, MedlineGoogle Scholar

73. Gandal MJ, Zhang P, Hadjimichael E, et al.: Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia, and bipolar disorder. Science 2018; 362:eaat8127Crossref, MedlineGoogle Scholar

74. Labonté B, Engmann O, Purushothaman I, et al.: Sex-specific transcriptional signatures in human depression. Nat Med 2017; 23:1102–1111Crossref, MedlineGoogle Scholar