Abstract Objective: Septic shock is the severe complication of sepsis, with a high mortality. The inflammatory response regulates the immune status and mediates the progression of septic shock. In this study, we aim to identify the key immune-related genes (IRGs) of septic shock and explore their potential mechanism. Methods: Gene expression profiles of septic shock blood samples and normal whole blood samples were retrieved from the Gene Expression Omnibus (GEO) and Genotype-Tissue Expression Portal (GTEx). The differential expression genes (DEGs) and septic shock-specific immune-related genes (SSSIRGs) were evaluated and identified, along with the immune components by “cell type identification by estimating relative subsets of RNA transcripts (CIBERSORT, version x)” algorithm. Additionally, in order to explore the key regulatory network, the relationship among SSSIRGs, upstream transcription factors (TFs), and downstream signaling pathways were also identified by Gene Set Variation Analysis (GSVA) and co-expression analysis. Moreover, the Connectivity Map (CMap) analysis was applied to find bioactive small molecules against the members of regulation network while Chromatin Immunoprecipitation sequencing (ChIP-seq) and Assay for Targeting Accessible-Chromatin with high-throughput sequencing (ATAC-seq) data were used to validate the regulation mechanism of the network. Results: A total of 14,843 DEGs were found between 63 septic shock blood samples and 337 normal whole blood samples. Then, we identified septic shock-specific 839 IRGs as the intersection of DEGs and IRGs. Moreover, we uncovered the regulatory networks based on co-expression analysis and found 28 co-expression interaction pairs. In the regulation network, protein phosphatase 3, catalytic subunit, alpha isozyme (PPP3CA) may regulate late estrogen response, glycolysis and TNFα signaling via NFκB and HLA; Kirsten rat sarcoma viral oncogene homolog (KRAS) may be related to late estrogen response and HLA; and Toll-like receptor 8 (TLR8) may be associated with TNFα signaling via NFκB. And the regulation mechanisms between TFs and IRGs (TLR8, PPP3CA, and KRAS) were validated by ChIP-seq and ATAC-seq. Conclusion: Our data identify three SSSIRGs (TLR8, PPP3CA, and KRAS) as candidate therapeutic targets for septic shock and provide constructed regulatory networks in septic shock to explore its potential mechanism. Keywords: septic shock, immune-related genes, TLR8, PPP3CA, KRAS Introduction Septic shock is the most severe complication of a severe microbial infection (sepsis), with a short treatment time-window and a high death rate ([33]Sharawy and Lehmann, 2015). Pathogenetically, pathogens invade microorganisms; induce intracellular events in immune, epithelial, and endothelial cells (ECs); and trigger an uncontrolled systemic inflammatory response ([34]Annane et al., 2005). In the process, the inflammatory response associated with “cytokine storm” leads to the damages to host tissues and organs, while the anti-inflammatory response reprograms the immune microenvironment and regulates the immune status ([35]Russell and Walley, 2010). Thus, the regulation and dysregulation of immune microenvironment may play important roles in the development, progression, and prognosis of septic shock. Current therapeutic strategies often focus on controlling the infection source by antibiotic therapy and restoring hemodynamic homeostasis by norepinephrine ([36]Kumar, 2014). In addition, septic shock patients may also benefit from some drugs such as corticosteroids or activated protein C ([37]Seam, 2008). Despite unceasing advances in management, septic shock still represents a major healthcare problem worldwide. Based on the crucial roles of immune microenvironment in septic shock, we suppose that targeting septic shock-specific immune-related genes (SSSIRGs) may be novel therapeutic options ([38]Walley et al., 2014; [39]Francois et al., 2018). In this study, in order to identify the SSSIRGs, the public databases were searched, and the RNA profiles of blood samples in septic shock and healthy patients were downloaded. The differentially expressed SSSIRGs were identified between them. In addition, the algorithm “cell type identification by estimating relative subsets of RNA transcripts (CIBERSORT)” was used to evaluate the cell fraction. Moreover, we also constructed regulatory networks based on upstream transcription factors (TFs), SSSIRGs, immune cells, and downstream signaling pathways to decipher the potential mechanism of septic shock and identify candidate targets. Materials and Methods Data Extraction and Differential Expression Analysis The gene expression profiles of 63 septic shock blood samples and 337 normal whole blood samples were downloaded from the Gene Expression Omnibus (GEO^[40]1) ([41]Braga et al., 2019) and the Genotype-Tissue Expression Portal ([42]GTEx, 2015^[43]2), respectively. The gene expression profiles were retrieved in formats of HTseq-counts and were normalized to Transcripts Per Million (TPM) based on gene length. Clinical data including demographics (age and gender) and blood collection time points (T1: within 16 h of ICU admission; T2: 48 h after study enrollment; T3: on day 7 from ICU admission or before discharge from the ICU) were also extracted from the database. Samples and patients with incomplete clinical information were excluded, and conformers who met the inclusion and exclusion criteria were extracted for further analysis. In order to reduce the error caused by the different length and sequencing depth of each single gene in the sequencing process, firstly, raw counts (RCs) of RNA-seq data obtained from GTEx and [44]GSE131411 were quantified, respectively, which were then standardized using voom function in limma (Linear Models for Microarray Analysis) package. To eliminate the batch effect, these two batches of RNA-seq data were corrected using the normalizeBetweenArrays function in the limma package and then merged for differential expression analysis ([45]Smyth, 2004). Differential expression genes (DEGs) between 63 septic shock blood samples and 337 normal whole blood samples were determined using the Edge R package ([46]Smyth, 2004). The standard to define the DEGs was | log[2] Fold Change (FC)| > 1.0 along with False Discovery Rate (FDR) < 0.05. Besides, the Gene Oncology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) items of enrichment analysis were performed to explore the biological processes (BPs), cellular components (CCs), molecular functions (MFs), and KEGG pathways that septic shock-specific DEGs enriched. Identification of Septic Shock-Specific Immune-Related Genes in Whole Blood A total of 2,498 immune-related genes (IRGs) were retrieved from the ImmPort database^[47]3 and Molecular Signatures Database (MSigDB) v7.1,^[48]4 respectively ([49]Liberzon et al., 2015). The intersection of the 2,498 IRGs and DEGs was filtered by further non-parametric test (Kruskal–Wallis H test) to identify SSSIRGs. Identification of Immune Components Potentially Regulated by Septic Shock-Specific Immune-Related Genes The “CIBERSORT, version x” algorithm was applied to estimate the fraction of 22 types of immune cell in each whole blood sample ([50]Newman et al., 2019). The non-parametric test (Mann–Whitney U test) was conducted to illuminate the different immune cell components between septic shock blood samples and normal whole blood samples. In addition, the fraction of 29 immunomodulating gene sets was quantified using single-sample Gene Set Enrichment Analysis (ssGSEA) ([51]Barbie et al., 2009). Correlation analysis was applied to SSSIRGs, 22 immune cells, and 29 immune gene sets. The pairwise interactions between SSSIRGs and immune cells (| correlation coefficient| > 0.500, p-value < 0.001) along with SSSIRGs and immune gene sets (| correlation coefficient| > 0.600, p < 0.001) were identified as immune components potentially regulated by SSSIRGs. Identification of Key Transcription Factors and Pathways of the Shock-Specific Immune-Related Genes Fifty hallmark gene sets (signaling pathways) and the 318 TFs were retrieved from the MSigDB v7.1 (see text footnote 4) and Cistrome database,^[52]5 respectively ([53]Liberzon et al., 2015; [54]Zheng et al., 2019). Differential expression and correlation analyses were conducted to determine upstream TFs, while hallmark pathways of SSSIRGs were absolutely quantified by Gene Set Variation Analysis (GSVA) ([55]Hanzelmann et al., 2013). Next, the correlation analysis was applied to key SSSIRGs, TFs, and aforementioned signaling pathways. Pairwise interactions between TFs and SSSIRGs (| correlation coefficient| > 0.900, p < 0.001), SSSIRGs, and the 50 hallmark pathways (| correlation coefficient| > 0.600, p < 0.001) were extracted for further analysis. All above significant interaction pairs were integrated to construct the regulation network among SSSIRGs and their upstream TFs and downstream pathways/immune cells/immune gene sets. Integrated Analysis of Clinical Correlation Based on Length of Hospital Stay In order to better understand and validate the relationship between SSSIRGs and prognosis of patients with septic shock, patients were categorized into three groups (T1–T3) based on blood sample collection time points (T1: within 16 h of ICU admission; T2: 48 h after study enrollment; T3: on day 7 from ICU admission or before discharge from the ICU). Further differential expression analysis based on different time points was carried out, respectively, using the edge R package. In addition, gene sets over-representation analysis (GSORA) was performed to evaluate the fraction of genes in tested gene clusters ([56]Pomyen et al., 2015). In this study, 54 septic shock-related gene sets downloaded from MSigDB were divided in nine subclusters on the basis of function features, which included C1–C8 and the hallmark pathway cluster (H) ([57]Liberzon et al., 2015). In addition, ORA was carried out to determine the enrichment level of these gene sets.^[58]6 Based on the key SSSIRGs obtained previously and different lengths of hospital stay (LOS) of septic shock patients, non-parametric test and clinical correlation analysis were performed, and the intersection of the results from these two analyses which were statistically significant was extracted for further analysis. Specifically, LOS served as an important sign in clinical judgment of the severity of septic shock. Connectivity Map Analysis Here, we applied the Connectivity Map (CMap, build 02) to find potential inhibitors that may target key SSSIRGs. In total, 6,100 gene expression cases covering 1,309 drugs were obtained from the CMap database^[59]7 ([60]Lamb et al., 2006), that is, a potential drug might correspond to various gene expression cases. Genes in each case were ranked via taking differential expression values between drug-untreated and drug-treated cell lines, and 6,100 gene lists associated with drugs were then generated. On the basis of identified key SSSIRGs involved in septic shock and 6,100 drug-related cases, we conducted a non-parametric test to explore the relationship between drugs and septic shock. Information of targeting compounds is available in the mechanism of actions (MoAs)^[61]8 that includes transcriptional responses of various human cell lines to perturbagens, structural formulas, and various targets. On the basis of MoA, compounds which may target to septic shock-related SSSIRGs in this study were extracted. Validation of Chromatin Immunoprecipitation Sequencing and Assay for Targeting Accessible-Chromatin With High-Throughput Sequencing Chromatin Immunoprecipitation sequencing (ChIP-seq) and Assay for Targeting Accessible-Chromatin with high-throughput sequencing (ATAC-seq) data were used to validated the regulation mechanism of the network. ChIP-seq and ATAC-seq data were obtained from Cistrome database (see text footnote 5) and [62]GSE139099,^[63]9 respectively ([64]Zheng et al., 2019; [65]Grubert et al., 2020). The WashU Epigenome Browser and Gviz package were used to visualize the binding peaks ([66]Hahne and Ivanek, 2016; [67]Li et al., 2019). Processing of Single-Cell RNA Sequencing Data Many studies supported that COVID-19 severe forms share clinical and laboratory aspects with various pathologies such as hemophagocytic lymphohistiocytosis, sepsis, or cytokine release syndrome. Fatal COVID-19 patients, as would be expected with septic shock patients, produce inflammatory reaction and cytokine storm ([68]Bost et al., 2021). Therefore, single-cell RNA sequencing (scRNA-seq) data of peripheral blood mononuclear cells (PBMCs) from fatal COVID-19 patients were obtained from [69]GSE150728 to determine the specific cellular location of key SSSIRGs and cell cycle regulation in this study, which can further explain the molecular pathogenesis of septic shock spatiotemporally. The preprocessing of scRNA-seq data of seven COVID patients and six healthy samples was conducted using 10x Genomics Chromium^[70]10 ([71]Chen et al., 2020). After demultiplexing, the sequencing results were divided into two paired-end reads fastq files that were then trimmed to eliminate the template switch oligo (TSO) sequence and polyA tail sequence. In addition, clean reads were aligned with the GRCh38 (Version: 100) genome assembly, which were quantified using the Cell Ranger Software (Version 1.0.0).^[72]11 The quantitative gene expression matrices (the row names of matrices were genes and col names were barcodes) were analyzed with Seurat pipeline (Version: 3.2.2) for further analysis ([73]Butler et al., 2018). Only cells with less than 10% mitochondrial gene mapped and expressing more than 100,000 transcripts were extracted for further analysis. In addition, genes expressed in over three single cells were included in follow-up analyses. After the completion of quality control (QC), all samples were integrated into one Seurat object with the function of “IntegrateData,” which were then scaled and normalized with the function of “ScaleData.” The “vst” method was utilized to determine the top 1,500 variable genes. To reduce model dimensionality, principal component analysis (PCA) was initially conducted, and the top 20 PCs were incorporated as input file for Uniform Manifold Approximation and Projection for Dimension Reduction (UMAP) analysis. UMAP plots illustrating cell subclusters were constructed using the “DimPlot” and “RunUMAP” function. Differential Expression Analysis and Cell Type Annotation Genes with significantly differential expression patterns from the top 1,500 variable genes were defined as DEGs with the “wilcox” method using the “FindAllMarkers” function. To identify the cell type of each unsupervised cluster, DEGs in all subclusters were applied as potential references, which were combined