Abstract Ischemia–reperfusion injury (IRI) is one of the leading causes of mortality and disability worldwide. Owing to its complex pathogenesis, there is still a lack of effective therapeutic targets in clinical practice, and exploring the mechanism and targets of IRI is still a major clinical challenge. This study aimed to explore the genetic alterations in leukocytes in peripheral blood after ischemia–reperfusion, aiming to discover new biomarkers and potential therapeutic targets. KAS-Seq (Kethoxal-assisted single-strand DNA sequencing) was used to obtain gene expression profiles of circulating leukocytes in a porcine ischemia–reperfusion model at 24, 48, and 72 h post-ischemia‒reperfusion. This method integrated genes that exhibited regular changes over time. In this study, we thoroughly analyzed the dynamic changes in gene expression post-IRI, revealing significant enrichment in key signaling pathways that regulate immune responses and T-cell activation over time. Our identification of the interleukin-7 receptor (IL7R) was particularly striking, as it plays a crucial molecular role in IRI. Additionally, using database mining technology, we confirmed the close relationship between IL7R and IRI, explored the interaction between interferon-γ (IFNG) and IL7R in T-cell activation, and clarified their joint influence on ischemia–reperfusion injury. Using KAS-Seq analysis of leukocytes from peripheral blood, we successfully delineated the temporal patterns of gene expression and changes in signal transduction pathways in porcine models of ischemia–reperfusion. Subsequent in-depth analysis identified IL7R as a potential novel therapeutic target for IRI. The pivotal role of this gene in modulating immune responses provides innovative avenues for the development of IRI treatments. Keywords: KAS-Seq, Ischemia–reperfusion injury, Leukocytes, IL7R, Target Subject terms: Genetics, Gene expression, Immunogenetics Introduction Ischemia is a condition in which the body’s blood supply is restricted, leading to a lack of oxygen and other nutrients necessary for cell metabolism^[46]1,[47]2. Prolonged ischemia can cause irreversible damage to many organs, such as the heart, brain, kidneys, liver, lungs, and intestines^[48]3. Reperfusion is an immediate intervention to restore blood supply, but the damage caused by the sudden restoration of blood flow to an organ with long-term ischemia, known as reperfusion injury, can be more severe and potentially fatal^[49]4. Numerous previous studies have indicated that the pathogenesis of IRI is associated with changes related to neuroinflammation, neutrophil regulation, and other mechanisms^[50]5–[51]8. However, owing to the complexity of these underlying mechanisms, there is currently no fully effective treatment for IRI. The limitations of current clinical treatments for IRI have prompted the search for more effective treatments. Therefore, further exploration into new mechanisms and targets of IRI is still necessary. Previous studies have clearly indicated that inflammation plays a central role in IRI, mainly through the activation of key immune cells such as neutrophils and macrophages. During the activation of these cells, a variety of proteases and proinflammatory cytokines are released. These molecules not only exacerbate the local inflammatory response but also may further contribute to tissue damage.^[52]9–[53]11. In addition, the release of these inflammatory cytokines can affect apoptosis, pyroptosis, and autophagy^[54]12,[55]13. Although previous studies have focused on specific cell types or mechanisms, such as neutrophil activation and macrophage polarization, they have often overlooked the overall impact of white blood cell populations on IRI and changes in global gene expression. Therefore, there is an urgent need to conduct a comprehensive analysis of differential gene expression in leukocytes. This approach can not only enhance our understanding of the immune response to IRI but also uncover potential new therapeutic targets, thereby providing more comprehensive and precise strategies for clinical treatment. Second-generation sequencing technology has played a significant role in exploring the molecular mechanisms underlying IRI. One innovative approach is KAS-Seq (kethoxal-assisted single-strand DNA sequencing)^[56]14, which offers a straightforward, effective, and sensitive method for studying transcriptional regulation and enhancer activity through single-strand DNA sequencing. Instantaneous ssDNA during transcription provides a more direct in situ reading of transcriptional activity than does the RNA itself^[57]14–[58]20. Thus, the application of KAS-Seq in elucidating the molecular mechanism underlying IRI has significant implications for advancing disease research. In this study, KAS-Seq technology was employed to analyze ssDNA in peripheral blood leukocytes of porcine IRI model to explore key dynamic gene changes and potential mechanisms following IRI, and reveal potential therapeutic targets. Specifically, we collected leukocyte samples at 0, 24, 48, and 72 h post-IRI and used KAS-Seq to map the dynamic changes in ssDNA. In-depth analysis revealed that IL7R is a potential key target in IRI, suggesting a role for IL7R in T-cell activation pathways. Our goal is to pinpoint the core pathways and targets involved in the immune response to IRI, thereby offering new insights and inform potential therapeutic strategies for its clinical management. Materials and methods Animal model In this study, we selected three pigs weighing from 20 to 25 kg and approximately 4 months old as experimental subjects for intravenous induced anesthesia. Under strict aseptic procedures, we intubated the pigs and maintained gas anesthesia. Subsequently, we made an accurate skin incision at the femoral artery and controlled the total blood loss to approximately 400 ml through an arterial catheter to simulate ischemia in the body. Then, we carefully inserted a pre-inflated balloon catheter into the artery, blocking blood flow for 30 min. Throughout the entire balloon intervention process, we continuously monitored the animals’ vital signs, including heart rates and blood pressures. As the balloon was slowly deflated and blood flow in the femoral artery was restored, it marked the beginning of the reperfusion phase. We collected blood samples at key time points of 0, 24, 48, and 72 h after blood withdrawal and reperfusion. In light of the animals’ blood loss and fluctuations in blood pressure, we promptly supplemented physiological saline or lactated Ringer’s solution to maintain stable blood volume and blood pressure. Postoperatively, we conducted a meticulous examination of the surgical site to ensure that there were no complications such as bleeding or hematoma. Ultimately, these animals were safely transferred to the recovery area, where they were closely monitored to evaluate their recovery progress. Study participants In this investigation, we utilized KAS-Seq sequencing technology to map the genomic landscape of pigs during various stages of ischemia–reperfusion. The study included a total of 12 samples, with 3 designated as control samples at 0 h and the remaining 9 representing the 24-, 48-, and 72-h postischemia reperfusion intervals. The KAS-Seq libraries for all samples were sequenced using the Illumina NovaSeq 6000 platform. Data processing involved comparative analyses at each time point—0, 24, 48, and 72 h—to construct a comprehensive KAS-Seq atlas. Through temporal analysis, we sought to pinpoint the key mechanisms driving IRI over time. By examining these temporal shifts, we identified potential therapeutic targets. Furthermore, we conducted target analysis utilizing relevant single-cell sequencing omics data, corroborating that the identified key targets are indeed closely linked to transcriptional activity and are both genuine and efficacious, as revealed by KAS-Seq comparisons. Leukocyte gDNA extraction and quantity assessment Five-milliliter porcine peripheral blood samples were collected in BD Vacutainer®EDTA tubes at 0, 24, 48, and 72 h post-ischemia reperfusion, and leukocytes were isolated and extracted (Becton, Dickinson and Company, product No. 367525). All blood samples were sent to the laboratory within 24 h. The plasma was separated by centrifugation at 1350×g for 12 min. The plasma was then transferred to a 2 ml centrifuge tube (AXYGEN, MCT-200-C) and centrifuged again at 1350×g for 12 min. The upper plasma was carefully removed and retained, while the bottom precipitate containing leukocytes was transferred to a new 2 ml centrifuge tube and immediately cryopreserved in a gradient. Leukocyte DNA was extracted from plasma via the Quick-DNA™ Miniprep Plus Kit (ZYMO, D4069), and DNA concentrations were measured via a Qubit 3.0 fluorometer (Thermo Fisher Scientific, [59]Q33216). The extracted DNA samples were then stored at − 80 °C for future use. Prior to constructing the KAS-Seq library, we performed nucleic acid electrophoresis to determine the size of the DNA fragments, ensuring their suitability for library construction. KAS-Seq library construction and sequencing First, the ground tissues were cultured in complete medium containing 5 mM N3-ketoaldehydes at 37 °C and 5% CO[2] for 10 min. After culture, tissue samples were collected, and genomic DNA (gDNA) was extracted from the cells via the PureLink Genomic DNA Mini-kit (Thermo, K182002). One microgram of extracted gDNA was dissolved in 95 µL of DNA elution buffer. Subsequently, 5 µL of 20 mM DBCO-PEG4-biotin (DMSO solution, Sigma, 760749) and 25 mM K3BO3 were added, and the mixture was incubated at 37 °C with gentle shaking for 1.5 h. Next, 5 µL of RNase A (Thermo, 12091039) was added to the reaction system, and the mixture was incubated at 37 °C for 5 min. Biotinized DNA was purified via a DNA Clean & Concentrator-5 kit (Zymo, D4013). The purified gDNA was dissolved in 100 µL of water, and the DNA fragments were broken to a size of 150–350 bp via a Bioruptor Pico ultrasound apparatus for 30 cycles in the mode of 30 s pulses/30 s intervals. Five percent of the DNA fragments were retained as control samples, and the remaining 95% of the DNA was incubated with 10 µL of prewashed Dynabeads MyOne Streptavidin C1 (Thermo, 65001) at room temperature for 15 min to enrich the biotin-labeled DNA. After incubation, the beads were washed and heated in 15 µL of H2O at 95 °C for 10 min to elute the DNA. Finally, the DNA library was constructed via an Accel-NGS Methyl-Seq DNA library kit (Swift, 30024), and the library was sequenced on an Illumina NovaSeq 6000 sequencing platform in the double-ended 150-bp mode. The goal was to obtain approximately 30 million reads for each library^[60]15. Mapping and identification of enriched regions via KAS-Seq A comprehensive quality control analysis of the obtained raw sequencing data was performed via FastQC software (version 0.11.5). This step is essential to ensure the accuracy and reliability of the data and to build a solid foundation for subsequent analysis. We then used Bowtie2 software (version 2.2.9) to align these qualitatively screened raw sequencing data precisely with the reference pig genome^[61]21 and filtered with SAMtools (version 1.3.1)^[62]22 to preserve unique, nonrepetitive matches. Pair-end reads were extended and converted into BedGraph format via Bedtool2 (version 2.19.1)^[63]23 and then converted to BigWig format for visualization via bedGraphToBigWig from the Integrated Genomics Viewer. MACS2 (version 2.1.1) was used to identify potential KAS-Seq-enriched regions in each sample^[64]22. Regions of less than 1000 base pairs (bp) that occur in two or more samples are used as a uniform reference for each sample. To improve the reliability of the data, we specifically excluded genomic regions that could generate false signals, as identified by data from the ENCODE project (Encyclopedia of DNA Elements). Next, we successfully identified specific regions rich in KAS-Seq signals by comparing the individual peak detection files for each sample with the combined peak detection files. We used the ChIPseeker package to annotate the KAS-Seq-enriched regions, using the gene closest to each region for annotation. Differential analysis and functional analysis RStudio 3.5.0 (version) of the DESeq2 package (version 3.24.3)^[65]24 was used to identify KAS-Seq difference loci (root filter thresholds: p values < 0.05 and |log twofold change|> 0.5). Genes were selected for GO (Gene Ontology) analysis^[66]25, and BP (biological process), CC (cell component) and MF (molecular function) terms were enriched. Protein‒protein interaction (PPI) network analysis We utilized the interactive Gene/Protein Retrieval Search Tool (STRING) database to search for protein‒protein interaction (PPI) data^[67]26. Interactions with a confidence score greater than 0.7 were selected to ensure the reliability of the network data. The collected data underwent rigorous preprocessing to eliminate redundant and self-interacting entries. Using Cytoscape software^[68]27, we constructed a protein–protein interaction (PPI) network for visualization of the collated data. Each protein is depicted as a node, and its interactions are represented as edges, forming a complex network topology. The first 10 highly connected proteins, referred to as central genes, are considered potentially key components of this network.^[69]28. GEO database Three ischemia-reperfusion injury-related microarray datasets, [70]GSE9634, [71]GSE72646, and [72]GSE23160, were downloaded from the GEO Expression Synthesis database^[73]29. Cell culture and cell IR model construction In this study, we utilized two human leukemia T lymphoid cell lines (MOLT-4 and MOLT-6) to establish an IRI model. These cell lines were obtained from the ATCC cell bank and cultured in DMEM/F12 medium supplemented with 10% fetal bovine serum and 1% penicillin–streptomycin at 37 °C in a humidified atmosphere containing 95% CO2. To simulate the IRI process, cells were initially incubated in a three-gas incubator at 37 °C with a gas composition of 1% O2, 94% N2, and 5% CO2 for a duration of 12 h to mimic ischemic conditions. Subsequently, the cells were transferred to a standard incubator for an additional six hours to replicate the reperfusion phase. Through this approach, we successfully simulated the IRI process in vitro, thereby providing a reliable experimental framework for investigating the responses of leukemia cells under conditions of ischemia and reperfusion. This work contributes to a deeper understanding of the mechanisms underlying IRI and offers an experimental basis for developing novel therapeutic strategies. Results Overview of the research process flow Blood is a type of easy-to-obtain biological sample that provides an ideal method for studying the effects of IRI on the immune system. This study aimed to track immune response changes at various time points post-IRI. We collected peripheral blood samples from four groups of model pigs at 0, 24, 48, and 72 h post-IRI, with the 0-h samples serving as the control. After centrifuging the blood to isolate leukocytes, we constructed KAS-Seq libraries from the leukocyte single-stranded DNA and performed in-depth sequencing analysis via high-throughput techniques. We employed visualization and machine learning algorithms to investigate the underlying mechanisms and potential biomarkers of IRI, with the aim of providing new insights and strategies for clinical diagnosis and treatment (Fig. [74]1A). Fig. 1. [75]Fig. 1 [76]Open in a new tab Schematic overview of the study. (A) Porcine femoral artery ischemia–reperfusion was constructed and blood leukocytes were collected at different times after reperfusion. The leukocytes were subjected to KAS-Seq to dynamically map ssDNA alterations. In-depth data analysis was performed to identify key genes and pathways that may contribute to IRI. KAS-Seq global signal distribution expression profile To explore the dynamic changes in KAS-Seq signals induced by IRI over time, KAS-Seq sequencing was performed on leukocyte ssDNA in pig models at 0, 24, 48, and 72 h after IRI. Our observations show that the number of ischemic peaks in different genomic regions varies significantly over time.In particular, significant spikes were observed at 48 h post-IRI, and the KAS-Seq signals began to recover by 72 h post-IRI (Fig. [77]2A).Visual analysis revealed that KAS-Seq differences were predominantly distributed in gene functional regions, such as transcriptional start sites, introns, and distal regions (Fig. [78]2B). Notably, significant disparities in the distribution characteristics of these genomic regions were observed at different time points (Fig. [79]2C). Furthermore, we identified distinct variations among gene functional regions, particularly in transcription start sites and introns, across the groups at each time point (Fig. [80]2D,E). The subsequent principal component analysis demonstrated significant aggregation and differentiation among the groups at 0, 24, 48, and 72 h (Fig. [81]2F). Therefore, KAS-Seq markers can effectively differentiate between these time points post-ischemia–reperfusion, holding considerable significance for distinguishing among these four groups. Fig. 2. [82]Fig. 2 [83]Open in a new tab Global and dynamic ssDNA distribution characteristics of leukocytes from KAS-Seq in ischemia–reperfusion. (A) Genome-wide ssDNA distribution from samples collected 0, 24, 48, and 72 h after ischemia–reperfusion(0 h in green, 24 h in black,48 h in yellow, 72 h in pink); (B) Differences in the distribution of gene function at 0 h, 24 h, 48 h and 72 h(0 h in green, 24 h in black, 48 h in yellow, 72 h in pink); (C) 5hmC signal profile of ischemia–reperfusion at 0, 24, 48, and 72 h. (D,E) Peak number of promoter and intron regions at 0 h,24 h,48 h, and 72 h after ischemia–reperfusion D, is promoter, E, is intron regions. (F) PCA (principal component analysis) distinguished injured white blood cell samples(0 h in green, 24 h in black,48 h in yellow, 72 h in orange) after ischemia–reperfusion 0, 24, 48, and 72 h. GO signaling pathway and functional enrichment analysis We conducted a differential gene expression analysis using significance criteria of p < 0.05 and |log2FoldChange|≥ 0.5. This analysis identified 1651 differentially expressed genes (DEGs), including 690 upregulated and 941 downregulated genes, in the 24-h ischemia–reperfusion samples compared to the healthy controls (Fig. [84]3A, Supplementary Table [85]S1). Compared with those in the 24-h sample, there were 1578 DEGs in the 48-h sample, including 858 upregulated and 720 downregulated genes (Fig. [86]3B, Supplementary Table [87]S2). Similarly, there were 1577 DEGs in the 72-h sample compared with the 48-h sample, including 802 upregulated and 775 downregulated genes (Fig. [88]3C, Supplementary Table [89]S3). We conducted unsupervised hierarchical clustering analysis on the top 100 KAS-seq differential sites, which allowed for preliminary differentiation of samples at ischemia–reperfusion time points of 0, 24, 48, and 72 h (Fig. [90]S1A). Our analysis revealed that the signaling pathways enriched with the DEGs were closely related to damage development. For example, multiple pathways are involved in regulating the homeostasis of calcium ions within 24 h, which is consistent with the findings that calcium homeostasis is closely related to IRI^[91]30. Subsequently, signaling pathways related to the immune response became apparent 48 h post-ischemia–reperfusion, and other pathways related to myocardial tissue development and hypoxia level regulation began to appear 72 h post-ischemia–reperfusion (Fig. [92]S1B–D). Studies have shown that IRI can lead to changes in the immune system and that changes in various inflammatory factors can lead to multiple types of organ damage, including myocardial ischemia^[93]31,[94]32. To identify biomarkers of ischemia–reperfusion progression, we conducted a comparative analysis of different stages of detection, including mild ischemia–reperfusion at 24, 48, and 72 h. Mfuzz was utilized to cluster the identified biomarkers into four discrete clusters according to four-time points of ischemia–reperfusion. Cluster 2 tended to be upregulated within 48 h, whereas Cluster 1, Cluster 3, and Cluster 4 tended to be downregulated within 48 h (Fig. [95]3D) (Supplementary Table [96]S5). Concurrently, we performed pathway enrichment analysis on clusters exhibiting similar time trends. It was observed that some typical pathways were functionally rich (Fig. [97]3E,F). For example, in Cluster 2, the major enriched gene signaling pathways were involved in nervous system regulation and development, the immune response, and leukocyte regulation-related functions. In contrast, in Clusters 1, 3, and 4, the related genes were primarily enriched in pathways related to epithelial cell proliferation, leukocyte functions, T-cell regulation, and other immune responses. Previous studies have reported that IRI is associated with nervous system regulation, immune system regulation, and functions^[98]32,[99]33. Therefore, the results suggest that the gene-related changes identified via KAS-Seq may be significantly related to the molecular mechanisms and clinical symptoms of IRI. Fig. 3. [100]Fig. 3 [101]Open in a new tab GO enrichment analysis and function exploration of biomarkers. (A) Volcano map (0 h vs. 24 h). DEGs significantly changed (p-value < 0.05 & |log2FoldChange|≥ 0.5) are marked in red (up) or green (down). The black dots represent the DEGs with no difference. (B) Volcano map (24 h vs. 48 h). DEGs significantly changed (p-value < 0.05 & |log2FoldChange|≥ 0.5) are marked in red (up) or green (down). The black dots represent the DEGs with no difference. (C) Volcano map (48 h vs. 72 h). DEGs significantly changed (p-value < 0.05 & |log2FoldChange|≥ 0.5) are marked in red (up) or green (down). The black dots represent the DEGs with no difference. (D) The four groups identified by Mfuzz analysis showed regulatory trends in ischemia–reperfusion time progression. (E) Differential genes in Cluster 2 were analyzed for GO enrichment pathway. (F) Differential genes in Cluster1, Cluster3, and Cluster 4 were analyzed for signaling pathway enrichment. Analysis of Cluster 2 and immune-related functions and temporal cell landscape changes in Cluster 2 Since the overall signal level was highest at 48 h post-injury, we focused on biomarkers in Cluster 2 that exhibited regulatory trends similar to those observed during disease progression (Supplementary Table [102]S4). Several immune-related biological functions were significantly enriched in the functional enrichment analysis of ischemia–reperfusion in Cluster 2. Therefore, we identified common immune-related genes. A total of 5276 immune-related genes were retrieved from the database, Cluster 2 intersected with immune-related genes, and 178 differential genes were identified at the intersection (Fig. [103]4A and Supplementary Table [104]S6). We used the STRING database to generate a PPI network of the 178 intersecting genes. As shown in Fig. [105]4B, each node represents a cross-gene-encoded protein, each edge represents the correlation confidence between the two targets, and the thickness of the edge indicates the strength of data support. The P value of PPI enrichment was less than 1.0e-16, indicating significant protein interactions in the PPI network.To further clarify the intrinsic biological differences among the 178 immune-related genes, we used the CIBERSORT algorithm to analyze the composition of immune cell types, focusing on the 178 genes with a major distribution, and found that their proportion was significantly increased, particularly in activated memory CD4 T cells (Fig. [106]4C). Additionally, we intersected genes from Clusters 1, 3, and 4 with immune-related genes to identify 212 differentially expressed genes (Fig. [107]S2A and Supplementary Table [108]S7). We used the STRING database to generate a PPI network of 212 target genes (Fig. [109]S2B). Similarly, we clarified the composition of immune cells with 212 immune-related genes and found that their proportion of memory CD4 T cells decreased significantly (Fig. [110]S2C), which was consistent with the results of Cluster 2 (Fig. [111]4C). These results suggest that IRI may be associated with T-cell activity through the regulation of immune-related genes. Fig. 4. [112]Fig. 4 [113]Open in a new tab Immune infiltration analysis was performed for immune-related genes. (A) Venn diagram shows the intersection between immune-related genes and differentially expressed genes (DEGs) in Cluster2; (B) PPI network analysis of Cluster2 and immune-related gene intersection; (C) Characteristic analysis of 178 intergene-mediated immune cell infiltration landscape. Identification of hub genes and potential targets for IRI We further analyzed the hub genes among the 178 immune-related genes identified in Cluster 2 via the Cytoscape plugin CytoHubba (Fig. [114]4A). According to the maximum cluster centrality (MCC) algorithm, the top 10 hub genes were identified in the immune-related dataset of Cluster 2 (Fig. [115]5A). A total of 10 genes in the dataset were identified as hub genes (IFNG, IL7R, TLR4, IL2RA, CCRL2, CD40, CCR7, CCL20, CD5, and CD38). In the PPI network analysis, we identified two hub genes: interferon-gamma (IFNG) and the interleukin-7 receptor (IL7R). The expression levels of these genes were consistent with the overall temporal trend of the signal, showing upregulation from 0 to 48 h and downregulation from 48 to 72 h post-ischemia–reperfusion (Figs. [116]5B,C and [117]2A). Many studies have reported a close association between IFNG and the occurrence and progression of IRI, confirming our results from KAS-Seq sequencing^[118]31. Therefore, in the subsequent phase of our study, we concentrated on exploring the correlation between IL7R and IRI. Initially, we confirmed the expression of IL7R across three GEO datasets ([119]GSE9634, [120]GSE72646, and [121]GSE23160) (Fig. [122]5D–F). In each dataset, the IL7R expression level was increased in the IRI group, which aligns with our findings (Fig. [123]5D–F). Additionally, reports have indicated a correlation between IFNG and IL7R in the context of other diseases^[124]34–[125]36. Therefore, to explore the role of IFNG and IL7R in IRI, we used the STRING database to study the correlation between IFNG and IL7R target genes in IRI. GO signaling pathway enrichment was utilized to analyze potentially relevant pathways in IRI (Fig. [126]5G). Through pathway analysis of these target genes, we identified a series of typical biological pathways responsible for regulating and activating leukocytes and the immune system, as well as pathways associated with T-cell activation. These findings are consistent with the high expression of T-cell specificity observed in our previous analysis, suggesting a critical role for these pathways in the immune response to IRI (Fig. [127]4C). IRI is reported to be closely related to T-cell activation and immune system regulation^[128]37,[129]38. The results of this study suggest that IL7R may be a key target of IRI and may coregulate T cells with IFNG to affect IRI. This insight offers a novel perspective on the intricate immune regulatory mechanisms involved in IRI and could guide the development of effective clinical treatment strategies. Fig. 5. [130]Fig. 5 [131]Open in a new tab Hub genes recognized by the cytoHubba plug-in. (A) TOP10 Hub genes recognized by cytoHubba plugin. According to the MCC method, the gradient of color represents the value of the fraction. (B,C) The changing trend of IFNG and IL7R with ischemia–reperfusion injury time. (D–F) Expressions of IL7R in [132]GSE9634, [133]GSE72646, and [134]GSE23160 (blue represents 0 h, red represents IRI group). (G) Gene target networks of IL7R and IFNG in IRI-associated pathways. (Yellow represents target genes; Orange represents related genes; Blue indicates the functional annotation of the target genes). Expression of IL7R in cell models To validate the aforementioned mechanisms and investigate the species-specific differences between pigs and humans, we analyzed the expression of IL7R in human T lymphocytes (MOLT-4 and MOLT-6) under simulated ischemia–reperfusion injury (IRI) hypoxia-reoxygenation conditions. Flow cytometry was employed to detect differences in IL7R expression. The results demonstrated that in both MOLT-4 and MOLT-6 cells cultured under simulated IRI hypoxia-reoxygenation (HR) conditions, the expression levels of IL7R were significantly higher compared to the isotype control antibody staining group (ISO) and the normal culture condition group (Normal) (Fig. [135]6A–G). These findings further corroborate our previous sequencing results. Fig. 6. [136]Fig. 6 [137]Open in a new tab The expression of IL7R in CD4^+ T cells under IRI mimic hypoxia-reoxygenation condition analyzed by flow cytometry. (A) Flow cytometry analysis of MOLT-4 cells stained with isotype control antibody. (B) Flow cytometry analysis of the IL7R expression in MOLT-4 cells cultured under normal condition. (C) Flow cytometry analysis of the IL7R expression in MOLT-4 cells cultured under the IRI mimic hypoxia-reoxygenation (HR) condition. (D) Quantitative analysis of the IL7R positive MOLT-4 cells under different conditions shown in (A–C). (E) Flow cytometry analysis of MOLT-6 cells stained with isotype control antibody. (F) Flow cytometry analysis of the IL7R expression in MOLT-6 cells cultured under normal condition. (G) Flow cytometry analysis of the IL7R expression in MOLT-6 cells cultured under the IRI mimic hypoxia-reoxygenation (HR) condition. (H) Quantitative analysis of the IL7R positive MOLT-6 cells under different conditions shown in (A–C). Data are expressed as mean ± SD. N = 3. ***p < 0.001, ****p < 0.0001. Discussion IRI is a complex pathological process involving multiple organs and mechanisms. Despite the continuous deepening of our understanding of IRI, there remains a significant gap in effective clinical treatment options, highlighting the need for further research and development of targeted therapies. Future research needs to further explore the specific mechanisms of IRI and search for new therapeutic targets and strategies^[138]2. Although numerous studies have indicated that the pathogenesis of IRI is closely related to immune responses, such as T-cell activation, neutrophil burst, and macrophage polarization^[139]39–[140]41, the role of immune responses in IRI has not been fully elucidated. To further investigate the correlation between immunity and IRI, we used Kas-Seq sequencing technology to analyze the single-stranded DNA of leukocytes in the blood to explore the immune-related molecular mechanisms of immune cells in IRI. In addition, research has demonstrated that the pig model holds significant scientific value in IRI studies due to its high degree of homology with human physiology^[141]42. Therefore, in our study, we established a pig model of IRI and collected blood samples at 0, 24, 48, and 72 h post-ischemia–reperfusion, centrifuged them to separate leukocytes for Kas-Seq sequencing and searched for new therapeutic targets related to immunity. We first analyzed the overall distribution of Kas-Seq signals at different time points after the start of ischemia–reperfusion and found that the signals increased at 0 h, 24 h, and 48 h and began to decrease at 72 h, as shown in Fig. [142]1. We subsequently performed enrichment analysis on the DEGs whose expression changed regularly at the four-time points of ischemia–reperfusion. As shown in Fig. [143]2, the DEGs included upregulated (Cluster 2) and downregulated (Cluster 1, Cluster 3, Cluster 4) genes, most of which are closely related to leukocyte regulation pathways. The literature reports that T-cell activation and immune system regulation are related to the IRI process^[144]43–[145]50, thus confirming the obvious correlation between our data and this disease. To further explore the correlation between IRI and immune system regulation, we screened 178 immune-related genes from the upregulated genes and performed immune infiltration analysis on these genes. This analysis revealed that these 178 genes were significantly enriched in activated memory CD4 T cells. Studies have shown that within a certain period of ischemia–reperfusion, the function of CD4^+ T cells in organs such as the heart and liver changes. For example, in studies of myocardial IRI, CD4^+ T cells participate in the process of myocardial ischemic injury through the HMGB1-TLR4 signaling pathway^[146]51. In allo-orthotopic liver transplantation, depleting anti-CD4 antibodies can reduce neutrophil and macrophage infiltration and proinflammatory gene expression caused by IRI^[147]52, indicating that CD4^+ T-cell activation can affect IRI. We subsequently screened for hub genes from the 178 genes and found that IL7R and IFNG are key target genes. In mouse kidney IRI, IFNG can regulate the migration of neutrophils together with IL17^[148]31. Consequently, we investigated IL7R as a novel target for IRI and validated its differential expression using external datasets. Previous studies have demonstrated that IL7R, an essential cell surface receptor, promotes T cell survival, proliferation, and differentiation primarily through its interaction with IL-7^[149]53–[150]55. In the context of IRI, the expression of IL7R may influence immune cell function, thereby affecting the recovery and repair processes. Unlike previously reported targets, such as oxidative stress, IL7R addresses a broader spectrum of immune responses, offering a unique approach to mitigate IRI. For instance, NRF2 within the NRF2-ARE pathway functions as a transcription factor that activates antioxidant genes and diminishes oxidative stress^[151]56–[152]58. While effective, its influence is confined to the antioxidant response. Similarly, the inhibition of HDAC3 may alleviate inflammation by modulating the JAK1/STAT3 signaling pathway, but its effects are restricted to specific cell types^[153]59–[154]61. Additionally, ALOX12, a lipoxygenase implicated in myocardial IRI, primarily affects lipid metabolism by catalyzing the production of peroxidation products from polyunsaturated fatty acids, rather than engaging in immune regulation^[155]62,[156]63. In contrast, IL7R exerts a more extensive impact on immune responses by regulating T cell activation and promoting the release of cytokines such as IFN-γ. To further study the correlation between IL7R and IFNG and IRI, we analyzed all target genes related to IL7R and IFNG. Interestingly, we found that the genes associated with IL7R and IFNG are cross-enriched in signaling pathways related to immune regulation, including leukocyte activation, migration, and T-cell activation pathways. Given that previous analyses have revealed that global immune-related genes are highly expressed specifically in T cells, we speculate that following IRI, the upregulation of IL7R may regulate the upregulation of IFNG, thereby influencing T-cell activity. To verify the above mechanisms and investigate species-specific differences between pigs and humans, we established a human T-lymphocyte model and verified that IL7R expression was significantly higher in the model group than in the control group. These findings suggest that IL7R is a potential new target for IRI treatment. Compared to established pathways such as macrophage polarization and neutrophil burst, the mechanism of action of IL7R is similar in some respects but may present new challenges in others. Macrophage polarization plays an important role in IRI, as these cells can differentiate into the pro-inflammatory M1 type or the anti-inflammatory M2 type, thereby affecting the inflammatory response and tissue repair^[157]64,[158]65. IL7R may influence the polarization of macrophages by regulating T cell function, thus affecting the outcome of IRI. Additionally, neutrophil bursts are a key component of the early inflammatory response in IRI, and IL7R may regulate this process by influencing interactions between T cells and neutrophils^[159]66,[160]67. In summary, this study not only provides new insights into clinical treatment strategies for IRI but also identifies potential future therapeutic targets. Future studies should further explore the mechanisms underlying IL7R’s role in IRI and evaluate its clinical potential as a therapeutic target. Clinical implications In summary, our findings clearly indicate that the release of single-stranded DNA (ssDNA) markers by leukocytes in peripheral blood can serve as powerful epigenetic biomarkers for revealing the mechanisms of IRI. The data from our study revealed a close association between IRI and the activation of T cells, particularly CD4^+ T cells. Notably, IL7R, identified as a key target in this study, plays a significant role in regulating T-cell activation in conjunction with the upregulation of IFNG, which has a marked effect on the progression of IRI. These findings not only deepen our understanding of the pathophysiological mechanism of IRI but also open up new ideas and potential directions for future clinical treatment. Targeting IL7R is expected to provide entirely new strategies for the development of therapeutic interventions. For example, anti-IL7R monoclonal antibodies can be used to block IL-7 signaling, thereby reducing inflammatory responses and tissue damage in IRI. In conclusion, the results of this study further expand our understanding of the pathophysiological mechanism of IRI and provide new perspectives and potential therapeutic clues for future clinical treatment. Strengths and limitations In this study, the KAS-Seq technique was employed for the first time to detect single-stranded DNA in peripheral blood leukocytes, aiming to evaluate its role in IRI. This approach offers a novel perspective for investigating the molecular mechanisms underlying IRI. Furthermore, this research delves into the intricate relationship between IRI and immune regulation, as well as T cell activation. While prior studies have indicated a potential involvement of immune cells in IRI, our investigation further elucidates their roles by analyzing changes in KAS-Seq signaling at specific temporal points—particularly concerning T cell activation and immunomodulatory pathways. These findings underscore IL7R as a promising target for therapeutic intervention in IRI; specifically, the upregulation of IL7R and IFNG may influence T cell activation and significantly impact the progression of IRI. Compared to previous investigations, this study not only provides fresh insights into clinical treatment strategies for IRI but also identifies potential future therapeutic targets. Although our study has achieved initial results in exploring the role of IL7R in IRI, we must acknowledge some limitations in the study. First, due to the small sample size, we may not be able to capture all biological variations comprehensively. This sample size limitation can lead to statistical inefficiencies, which in turn affect the reliability and broad applicability of our findings, and limit our ability to dig deeper into the underlying mechanisms. To overcome these limitations, we plan to take the following steps in future studies: Increase the sample size, which will enhance the statistical power of the study and allow us to more accurately assess the role of IL7R in IRI. By increasing the sample size, we expect to be able to more fully reveal the detailed mechanisms of how IL7R affects immune response and tissue damage. Second, while the pig model shares similarities with humans in various aspects, there may be differences in the immune responses between pigs and humans. These discrepancies could have significant implications for understanding the pathological mechanisms of IRI and developing effective treatment strategies. Furthermore, our current study lacks direct validation using human IRI samples, which may impact the clinical translation and applicability of future research findings. To address this limitation, future research will be directed towards two primary avenues. First, in vitro experiments will be conducted utilizing IL7R-expressing cell lines (such as T cells) or human primary cells. CRISPR-Cas9 technology will be employed to either knock out or overexpress the IL7R gene. Subsequently, these cells will be subjected to ischemia–reperfusion conditions to evaluate cell viability, apoptosis rates, and the secretion of inflammatory factors. Furthermore, cells will be treated with IL-7 agonists or inhibitors to further elucidate their effects on cellular function. These in vitro studies will provide critical insights into the specific mechanisms by which IL7R modulates cellular responses during ischemia–reperfusion injury, thereby laying the groundwork for targeted therapeutic interventions. Second, in vivo studies using animal models will involve the development of IL7R gene knockout or overexpression models. For instance, CRISPR-Cas9 technology will be used to knock out the IL7R gene in IRI mouse models to validate its in vivo role. By confirming the role of IL7R in IRI through these animal models, we will be able to bridge the gap between basic research and clinical application, ultimately facilitating the development of more effective therapeutic strategies for IRI. This approach aims not only to enhance our understanding of the mechanisms by which IL7R contributes to IRI but also to provide a critical experimental foundation for developing targeted therapeutic strategies. There are many challenges to translating IL7R into a human therapeutic target. First, drug development needs to consider the complex role of IL7R in the immune system. IL7R plays a key role in the differentiation, development, and maturation of T and B cells, and is essential for adaptive immunity and maintenance of immune homeostasis. However, abnormal IL7R levels are also associated with immunopathology, for example in autoimmune diseases such as type I diabetes and multiple sclerosis, as well as chronic inflammatory diseases such as rheumatoid arthritis, ankylosing spondylitis, and inflammatory bowel disease^[161]68–[162]72. Therefore, drug development targeting IL7R requires careful evaluation of its potential off-target effects and immune system variabilit. Additionally, current drugs targeting IL7R have shown some efficacy in clinical trials, but further optimization is still needed. Conclusions In this study, we conducted KAS-Seq sequencing analysis of leukocytes in porcine blood following ischemia-reperfusion to investigate the consistent changes in gene expression from 0 to 72 h post-ischemia-reperfusion. Our results suggest that analysis of leukocytes via the KAS-Seq technique can capture not only the dynamics of gene expression at different time points after IRI but also the underlying pathological mechanisms closely related to T-cell activation. Importantly, we identified IL7R as a key molecular target for IRI. The upregulation of IL7R and its coregulation with IFNG during T-cell activation may play crucial roles in IRI. This discovery provides a new perspective and treatment strategy for the future clinical treatment of IRI and is expected to lead to more effective treatment plans for patients. Supplementary Information [163]Supplementary Information.^ (1.4MB, zip) Acknowledgements