Abstract Liver transplantation is the definitive treatment for end-stage liver disease, yet T-cell mediated rejection (TCMR) remains a major challenge. This study aims to identify key genes associated with TCMR and their potential biological processes and mechanisms. The [44]GSE145780 dataset was subjected to differential expression analysis, weighted gene co-expression network analysis (WGCNA), and machine learning algorithms to pinpoint key genes associated with TCMR. Gene Set Enrichment Analysis (GSEA), immune infiltration analysis, and regulatory networks were constructed to ascertain the biological relevance of these genes. Expression validation was performed using single-cell RNA-seq (scRNA-seq) data and liver biopsy tissues from patients. We identified 5 key genes (ITGB2, FCER1G, IL-18, GBP1, and CD53) that are associated with immunological functions, such as chemotactic activity, antigen processing, and T cell differentiation. GSEA highlighted enrichment in chemokine signaling and antigen presentation pathways. A lncRNA-miRNA-mRNA network was delineated, and drug target prediction yielded 26 potential drugs. Evaluation of expression levels in non-rejection (NR) and TCMR groups exhibited significant disparities in T cells and myeloid cells. Tissue analyses from patients corroborated the upregulation of GBP1, IL-18, CD53, and FCER1G in TCMR cases. Through comprehensive analysis, this research has identified 4 genes intimately connected with TCMR following liver transplantation, shedding light on the underlying immune activation pathways and suggesting putative targets for therapeutic intervention. Supplementary Information The online version contains supplementary material available at 10.1038/s41598-024-74874-8. Keywords: T-cell mediated rejection, Liver transplant rejection, Single-cell RNA sequencing, Enrichment analysis, Immune analysis Subject terms: Transplant immunology, Allotransplantation, Bioinformatics Introduction In 1963, Dr. Starzl pioneered human liver transplantation, marking a significant milestone in medical history^[45]1. While advancements in surgical techniques and perioperative care were made, early postoperative rejection resulted in one-year survival rates of only about 30% before the 1980s^[46]2. The introduction of immunosuppressive drugs, particularly cyclosporin A, revolutionized the field by significantly improving patient outcomes^[47]3. Subsequent drugs like tacrolimus and mycophenolate further enhanced survival rates. However, 15-35% of liver transplant recipients still face T-cell-mediated rejection (TCMR) within two years post-transplant, highlighting the need for further molecular research to improve early diagnosis and treatment^[48]4,[49]5. Recent advancements in next-generation sequencing, machine learning, and bioinformatics tools have dramatically enhanced our ability to analyze complex post-transplant data. These technologies allow for a detailed examination of immune cell diversity and gene expression changes associated with transplant rejection. Integrating omics research with clinical findings provides a deeper understanding of liver transplant rejection mechanisms and facilitates the development of personalized treatment strategies^[50]6. High-throughput sequencing technologies have enabled extensive studies on gene and protein expression alterations in both human and animal models, shedding light on the immune processes involved in rejection^[51]7–[52]10. Research has identified specific transcriptional markers in peripheral blood as predictive indicators of rejection. For instance, proteomic analysis has highlighted Heme Oxygenase-1 (HO-1) as a potential biomarker for predicting acute rejection after liver transplantation^[53]11. Additionally, RNA sequencing (RNA-seq) has revealed significant increases in CXCL8 in liver tissues of pediatric patients with subclinical rejection post-transplantation. Clinical studies have shown that serum CXCL8 levels can diagnose subclinical rejection with high accuracy^[54]12. In conclusion, identifying novel biomarkers for liver transplant rejection and elucidating their molecular mechanisms is crucial for early diagnosis and effective treatment. This study analyzes liver biopsy transcriptome data from post-liver transplant patients using publicly available databases. By integrating bulk and single-cell RNA sequencing (scRNA-seq) with machine learning algorithms, we identified five key genes associated with TCMR. We further explored the pathways and biological processes enriched by these genes, providing insights into the molecular mechanisms of TCMR and potential therapeutic targets in liver transplantation. Results Research workflow We identified LTR-DEGs by intersecting DEGs between the TCMR and NR groups with TCMR-related genes elucidated in the WGCNA. Following that, GO and KEGG analyses were conducted to explore the functions and pathways of these LTR-DEGs. A PPI network and machine learning algorithms were subsequently employed to pinpoint 5 key genes: ITGB2, FCER1G, IL-18, GBP1, and CD53. Single-gene GSEA analysis further revealed their biological function. Additionally, immune infiltration analysis was conducted to assess fluctuations in immune cells during TCMR and to examine the associations between these key genes and immune cells. Furthermore, we also constructed a lncRNA-miRNA-mRNA network pertinent to key genes and predicted targeted drugs. The scRNA-seq data validated the differential expression of these key genes in distinct cell types. Lastly, we confirmed the expression levels of these genes in liver biopsy samples from post-transplantation patients (Fig. [55]1A). Fig. 1. [56]Fig. 1 [57]Open in a new tab Identification of differentially expressed genes (DEGs) and key module genes. (A) The workflow of the study. (B–C) Volcano plot and heatmap of DEGs; Orange represents upregulated genes, gray represents genes with no significant difference, and green represents downregulated genes. (D) The sample clustering diagram shows an outlier sample; red represents T-cell-mediated rejection (TCMR) samples and white represents no rejection (NR) samples. (E) Re-cluster after removing outlier samples. (F) Analysis of network topology for various soft-threshold powers. (G) Clustering dendrogram of DEGs, genes are divided into different modules. (H) Heatmap of module-trait correlations. Each gene depicts the correlation coefficients and p-values. Genes are colored according to correlation intensity: red for positive and blue for negative, as per the color legend. Identification of DEGs and key module genes A total of 180 DEGs were obtained between the TCMR and NR groups, including 171 upregulated genes and 9 downregulated genes (Fig. [58]1B–C, Supplementary Table [59]1). To further identify genes associated with TCMR, WGCNA was conducted. Sample clustering results showed the presence of an outlier sample ([60]GSM4332835), which was removed and the samples were re-clustered (Fig. [61]1D–E). When the soft threshold was set to meet a scale-free distribution (Fig. [62]1F), a total of 10 modules were obtained through the dynamic tree cutting algorithm (Fig. [63]1G). Among them, the MEblue module (cor = 0.79, p = 5e-36) showed the highest correlation with TCMR (Fig. 1[64]1H). Therefore, this module was considered the key module, and the 5,498 genes within this module were defined as key module genes for subsequent analysis. Acquisition and functional enrichment of LTR-DEGs Based on the intersection of DEGs and key module genes, 119 L-DEGs were identified (Fig. [65]2A). Enrichment analysis revealed that these LTR-DEGs are involved in 525 GO terms and 38 KEGG pathways. The GO analysis showed that the biological processes (BP) predominantly relate to cytokine-mediated signaling pathways, granulocyte chemotaxis, myeloid cell migration, and antigen processing and presentation. The cellular components (CC) are mainly associated with the secretory granule membrane, MHC protein complex, and MHC class II protein complex. The molecular functions (MF) are chiefly related to chemokine activity, chemokine receptor binding, MHC protein complex binding, and G protein-coupled receptor binding (Fig. [66]2B, Supplementary Fig. [67]1A, Supplementary Table [68]2). KEGG enrichment analysis included pathways such as antigen processing and presentation, chemokine signaling pathways, and cytokine-cytokine receptor interaction (Fig. [69]2C, Supplementary Fig. [70]1B, Supplementary Table [71]3). This suggests that these LTR-DEGs are primarily involved in activating and regulating immune cells, as well as processing and presenting antigens, thereby initiating specific immune responses. Fig. 2. [72]Fig. 2 [73]Open in a new tab Definition and functional analysis of DEGs associated with liver transplant rejection reactions (LTR-DEGs). (A) Venn diagram illustrates LTR-DEGs by overlapping DEGs and key module genes. (B) Lollipop diagram of LTR-DEGs’ Gene Ontology (GO) enrichment analysis. BP: biological process. CC: cellular components. MF: molecular functions. (C) Lollipop diagram of LTR-DEGs’ Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, displaying the 20 most significantly different pathways^[74]13–[75]15. Screening and expression analysis of key genes A PPI network constructed from the 119 L-DEGs consisted of 114 nodes and 735 edges (Fig. [76]3A). Ten candidate genes (CYBB, ITGB2, FCER1G, IL-18, CXCL11, GBP1, PLEK, CD53, LAPTM5, and C3AR1) were ultimately selected for further analysis from hub gene networks derived through four algorithms (Fig. [77]3B, Supplementary Fig. [78]2). LASSO regression analysis then selected 5 LASSO feature genes (ITGB2, FCER1G, IL-18, GBP1, and CD53) (Fig. [79]3C–D). The SVM model had the lowest error rate with six genes (GBP1, FCER1G, ITGB2, CYBB, IL-18, and CD53) (Fig. [80]3E). Overlapping LASSO and SVM-RFE feature genes, five key genes were identified (ITGB2, FCER1G, IL-18, GBP1, and CD53) (Fig. [81]3F). Expression analysis showed significantly higher expression of these genes in the TCMR group than in the NR group (Supplementary Fig. [82]3). The ROC curves showed that the AUC values of the five genes were greater than 0.9, indicating that the five genes had good diagnostic performance. It also indicated the good performance of both SVM-RFE and LASSO methods (Supplementary Fig. [83]4). These genes, as novel biomarkers and therapeutic targets, provide deeper insight into TCMR mechanisms and may guide future diagnostic and therapeutic strategies. Fig. 3. [84]Fig. 3 [85]Open in a new tab Construction of the Protein-Protein Interaction (PPI) network and key gene screening. (A) The nodes indicate proteins, and the letters represent gene symbols. (B) A Venn diagram illustrates candidate genes by overlapping the hub genes of the 4 algorithms. (C, D) The results of least absolute shrinkage and selection operator (LASSO) COX regression analysis. The dotted line on the left indicates the position with the smallest cross-validation error. At this position (Lambda.min), one identifies the corresponding log (Lambda) value on the horizontal axis. The upper horizontal axis displays the number of feature genes to find the optimal log (Lambda) value, identifying the relevant genes and their coefficients, and explaining the proportion of residuals in the model. (E) When the gene count is six, the error rate is at its lowest. (F) Venn diagram illustrates key genes by overlapping the results of two machine algorithms. Single-gene GSEA analysis Single-gene GSEA analysis identified that key genes are mainly enriched in antigen processing and presentation, cell cycle, T cell receptor pathway, chemokine signaling pathway, NK cell-mediated cytotoxicity, Toll-like receptor signaling pathway, etc. (Supplementary Fig. [86]5, Supplementary Tables [87]4–[88]8). These findings indicate complex functions of key genes in processes like antigen recognition, activation of immune cells, and regulation of immune responses. Immune-related analysis of key genes Bar graphs displayed the proportions of 28 immune cells in each sample (Fig. [89]4A). Significant differences in 27 immune cells were observed between the TCMR and NR groups, with a greater degree of differential immune cell infiltration in the TCMR group (Fig. [90]4B). Correlation analysis showed that CD53 had the strongest positive correlation with Th1 cells. There was a significant positive correlation between key genes and all differential immune cells (Fig. [91]4C, Supplementary Tables [92]9–[93]10). Fig. 4. [94]Fig. 4 [95]Open in a new tab Immune cell profiling in TCMR and NR groups. (A) Bar graph of immune scores for 28 immune cell types between TCMR and NR groups. (B) Comparative scoring of 28 immune cell types in two groups of samples. (C) Correlation between key genes and immune cells, the x-axis represents immune cells, and the y-axis represents biomarkers. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, ns: p > 0.05. Drug prediction for key genes and construction of lncRNA-miRNA-mRNA network We have identified 26 therapeutic drugs targeting three central genes by the DGIdb database (Fig. [96]5A). The network included 19 drugs targeting ITGB2 (ERLIZUMAB, LIFITEGRAST, cyclophosphamide, EFALIZUMAB, etc.), 7 drugs targeting IL-18 (IBOCTADEKIN, THYROXINE, etc.), and one drug targeting FCER1G (ASPIRIN). To identify the lncRNA-miRNA-mRNA network of key genes in TCMR, several online databases (mirTarbase and starBase) were used. The network comprised 4 mRNAs (ITGB2, FCER1G, IL-18, and GBP1), 8 miRNAs (hsa-miR-26b-5p, hsa-miR-1225-3p, hsa-miR-335-5p, etc.), and 43 lncRNAs ([97]AC008040.1, HAGLR, [98]AL158206.1, FGD5-AS1, [99]AC069281.2, etc.) (Fig. [100]5B, Supplementary Tables [101]11–[102]12). Fig. 5. [103]Fig. 5 [104]Open in a new tab Networks of key genes-drug interaction and lncRNA-miRNA-mRNA. (A) Drug-target network diagram for key genes, with green rectangles representing drugs and red shapes representing key genes. (B) lncRNA-miRNA-mRNA network diagram, where red triangles represent mRNAs, green circles represent miRNAs, and blue rectangles represent lncRNAs. Red lines in the diagram indicate interactions between miRNAs and mRNAs, while grey lines indicate interactions between lncRNAs and miRNAs. Expression analysis of key genes in different cell clusters After quality control, scRNA-seq data were available for subsequent analysis (Supplementary Fig. [105]6A). Cell screening and standard data processing yielded 2000 highly variable genes (Fig. [106]6A). Subsequently, we further analyzed the top 20 PCs of the inflection point through principal component analysis (PCA) (Supplementary Fig. [107]6B). To assess the effectiveness of the clustering results, cell flow Sankey diagrams at different resolutions were drawn (Supplementary Fig. [108]6C). Unsupervised clustering divided these cells into 11 cell clusters, visualized using t-SNE (Fig. [109]6B). Based on the expression patterns of marker genes in each cell cluster, these cell clusters were divided into 8 cell subtypes (T cells, B cells, myeloid cells, etc.) (Fig. [110]6C–D). The clustering results for the NR and TCMR groups were presented separately (Fig. [111]6E–F), with the highest proportion of T cells in both the TCMR and NR groups (Fig. [112]6G). The proportions of endothelial cells, hepatocytes, and neutrophils were significantly higher in the NR group than in the TCMR group. Expression validation results showed key genes were primarily expressed in T cells, myeloid cells, and NK cells (Fig. [113]7A–E). Significant differences in the expression of key genes between T cells and myeloid cells were observed between NR and TCMR groups. Fig. 6. [114]Fig. 6 [115]Open in a new tab Cell clustering analysis of single-cell RNA sequence (scRNA-seq) data. (A) Red dots represent high-variability genes, and black dots represent invariant genes; the greater the height on the y-axis, the larger the variance and difference of the genes. The names of the top 10 high-variability genes are also displayed. (B) t-distributed stochastic neighborhood embedding (t-SNE) plot colored by different cell clusters. (C) Bubble chart of classic marker genes for each cell group. (D) t-SNE plot of cell clustering annotation results. (E) Cell clustering annotation results (NR group). (F) Cell clustering annotation results (TCMR group). (G) Proportion of each cell group among all cells. Fig. 7. [116]Fig. 7 [117]Open in a new tab Expression differences of key genes across various cell groups. (A) Expression level differences of CD53 across different cell groups. (B) Expression level differences of FCER1G across different cell groups. (C) Expression level differences of GBP1 across different cell groups. (D) Expression level differences of IL-18 across different cell groups. (E) Expression level differences of ITGB2 across different cell groups. Key genes expression validation Immunohistochemical staining confirmed the expression levels of five key genes in TCMR patients and controls. Results showed FCER1G, IL-18, GBP1, and CD53 were upregulated in TCMR liver tissues compared to controls, while ITGB2 expression did not differ significantly (Fig. [118]8). These findings indicate FCER1G, IL-18, GBP1, and CD53 have potential as predictive indicators of immune rejection post-liver transplantation and may play significant roles in immune activation during rejection. Fig. 8. [119]Fig. 8 [120]Open in a new tab Immunohistochemical staining of key genes in liver biopsy tissue. Discussion Liver transplantation is a proven treatment for end-stage liver disease. However, TCMR substantially risks graft failure and elevates patient mortality, highlighting the imperative for investigating its molecular determinants^[121]16. Such inquiries are fundamental to enhancing early detection and therapeutic interventions. While the original study utilized the dataset to develop a diagnostic system for rejection, this study delves deeper into the molecular mechanisms of TCMR and identifies potential drug targets, thus providing a valuable complement to the original research^[122]17. Recent studies have increasingly integrated machine learning algorithms with bioinformatics methods to create decision models that aid in identifying potential diagnostic or prognostic biomarkers and therapeutic targets, as well as elucidating the molecular mechanisms of disease pathogenesis. Several studies on kidney transplant TCMR have employed machine learning algorithms in the analysis of transcript expression data to explore diagnostic biomarkers and potential immunotherapeutic targets for TCMR^[123]18,[124]19. However, similar research has not been conducted for liver transplant TCMR. This study builds on these approaches by combining Single-cell RNA-seq analysis to identify genes closely associated with TCMR in liver transplantation and further clarifies the differential expression of key genes across various cell populations, exploring potential immune activation pathways in the context of liver transplant TCMR. In this study, we utilized SVM-RFE and LASSO for feature selection, as both methods are highly effective in handling high-dimensional datasets and identifying key genes in complex biological processes. SVM-RFE combines the nonlinear classification capabilities of the SVM model with a recursive feature elimination strategy, enabling the automated selection of the most performance-contributive feature subset, particularly excelling in handling high-dimensional data and nonlinear relationships. LASSO, through L1 regularization, achieves sparse feature selection, which not only reduces model complexity but also enhances model interpretability, making it especially suitable for high-dimensional data scenarios. Both methods do not require assumptions about the specific functional form between features and the target variable, with LASSO performing particularly well in the presence of multicollinearity^[125]20,[126]21. Therefore, in the context of optimizing model performance, automating feature selection, and processing high-dimensional data, choosing SVM-RFE and LASSO as feature selection methods offers distinct advantages. We also recognize that future research could further explore other feature selection techniques to more comprehensively evaluate their performance and optimize our analytical processes. In the initial phase of our research, we identified DEGs between the TCMR and NR groups from the [127]GSE145780 dataset and combined a WGCNA network to select LTR-DEGs. GO and KEGG enrichment analyses connected these LTR-DEGs with immune functions such as cytokine activation and antigen processing. Following this, we constructed a PPI network and used 4 algorithms to further identify the candidate genes. Machine learning further validated the reliability of our findings, pinpointing five key genes (ITGB2, FCER1G, IL-18, GBP1, and CD53) linked to TCMR. Further interrogation using scRNA-seq confirmed that the five key genes were upregulated in both T cells and myeloid cells within the TCMR group. ITGB2/CD18 encodes an integrin beta chain protein, which is specifically expressed by leukocytes and can form corresponding β2 integrin heterodimers with four different known alpha chains, participating in cell adhesion and cell surface-mediated signal transduction^[128]22. Mac-1 (CD11b/CD18) and LFA-1 (CD11a/CD18), as members of the β2 integrin family, play critical roles in mediating leukocyte adhesion to target structures and other immune cells^[129]23. Research found that their interaction with ICAM-1 facilitates adhesion, crucial for lymphocyte regulation and inflammation control during early liver transplant rejection^[130]24; this finding is consistent with our research results. Additionally, in mucosal biopsies from lung transplant recipients, ITGB2 has been identified as a key gene associated with lung transplant rejection^[131]25. And in a rabbit model of heterotopic heart transplantation, antibody therapy targeting ITGB2 has shown significant efficacy in preventing transplant rejection^[132]26. Leukocyte surface antigen CD53, a member of the tetraspanin family, is a transmembrane signaling and pro-inflammatory protein widely expressed in various types of immune cells^[133]27,[134]28. Literature reports that CD53 can induce homotypic cell adhesion by activating β2 integrin LFA-1 in NK, T, and B cells, promoting adhesion and migration of immune cells^[135]29,[136]30. Additionally, studies have shown that CD53 stimulation enhances T cell proliferation. It drives the transition of naive T cells to effector/memory phenotypes. These effects have been observed in vitro in human T cells and in genetically modified mice. Further investigation revealed CD53’s role in regulating migration rate and stability of CD45RO on T cell surfaces. This regulation impacts TCR signal transduction. The absence of CD53 results in a loss of CD45RO expression on T cells. Consequently, it alters the expression of CD45 isoforms and diminishes T cell activation^[137]31. In a transcriptomic analysis of post-kidney transplant recipients, CD53 was identified as a biomarker for acute rejection in kidney transplantation^[138]32. FCER1G encodes the γ subunit of the high-affinity immunoglobulin E (IgE) Fc receptor, widely expressed in various types of immune cells. The FCER1G gene is involved in various biological processes, including neutrophil activation, T cell differentiation, immunoglobulin-mediated immune responses, and Fc receptor-mediated signaling pathways^[139]33–[140]35. Similar to this study, FCER1G has been proven to be an upregulated gene highly representative of acute rejection in heart, liver, and kidney transplants in a transcriptome data analysis of various solid organ transplants^[141]36. GBP1 is a GTPase of the dynamin superfamily, involved in the regulation of membrane, cytoskeleton, and cell cycle progression dynamics^[142]37. Furthermore, GBP1 is key to host cell immunity and antibacterial protection, recognizing infection and inhibiting bacterial proliferation by activating inflammasomes and regulating pyroptosis^[143]38. GBP1 is highly expressed in macrophages, endothelial cells, and epithelial cells, with continued high expression levels in T cells after IFN-γ stimulation induced by interferon-γ (IFN-γ)^[144]39. In several studies of renal transplant rejection, researchers have confirmed through transcriptomic analysis and tissue biopsies that GBP1 may serve as a biological characteristic and predictive model for acute rejection^[145]40–[146]42. Recent research have found that GBP1 binds to lipopolysaccharide (LPS) during bacterial infection, mediating the recruitment and activation of inflammatory caspase-4 through cleavage of GSDMD to induce pyroptosis^[147]43,[148]44. IL-18 is a member of the IL-1 family of pro-inflammatory cytokines, originally identified in the cytoplasm of macrophages as an inactive precursor (pro-IL-18). This precursor is transformed into an active mature form by proteolytic cleavage^[149]45. Recent proteomic studies have revealed IL-18 plays a significant biological role in regulating innate and adaptive immunity^[150]46. It also effectively induces IFN-γ production, exerting various immunoregulatory functions in the presence of different cytokines. These functions include regulating the transformation of T cells to regulatory T cells, modulating Th1 and Th2 responses, and participating in Th17 responses^[151]47–[152]49. It has been documented in existing literature that IL-18 plays a significant role in the rejection reactions of various solid organs. In a rat orthotopic liver transplantation model, blocking the binding of IL-18 to its receptor significantly alleviated graft rejection^[153]50. Elevated levels of IL-18 have also been observed in acute rejection reactions following kidney and heart transplantation^[154]51,[155]52.Historically, the maturation of IL-18 has been ascribed primarily to Caspase-1-mediated cleavage within the NLRP3 inflammasome complex^[156]53,[157]54. However, recent studies indicate that in Caspase-4, as part of the non-canonical pyroptosis pathway, can recognize and cleave the same site on pro-IL-18, leading to its activation. This interaction establishes a Caspase-4–IL-18 axis that links non-canonical pyroptosis to adaptive immunity^[158]55. Our findings lend support to the hypothesis that both GBP1 and IL-18 levels increase substantially in liver tissues following TCMR. This suggests that post-transplantation dysbiosis and the aberrant translocation of lipopolysaccharide (LPS) into the liver through the gut-liver axis might provoke non-canonical pyroptotic pathways in liver macrophages and other cells, mediated by GBP1. This cascade potentially triggers TCMR via cytokine release subsequent to cell death. TCMR is characterized by a complex, multifactorial immune response, with extant experimental research underscoring the pivotal role of various immune cells in this process^[159]56. In our study, immune infiltration analysis of two groups of data revealed a significant increase in the majority of immune cells in TCMR tissues, with scRNA-seq clustering analysis further showing the most significant upregulation in T and myeloid cells. Additionally, correlation analysis of the five selected key genes with 28 types of immune cells showed strong correlations with T cells, B cells, myeloid cells, etc. scRNA-seq revealed that the expression of the five key genes in the TCMR group was significantly increased in T and myeloid cell clusters compared to the NR group. Integrating bulk RNA-seq and scRNA-seq analyses, these key genes appear to be pivotal in immune activation during TCMR, significantly influencing antigen presentation and lymphocyte activation between Antigen-Presenting Cell (APC) and T cells. To deepen the understanding of the specific mechanisms of TCMR, we explored differences in miRNA and lncRNA levels based on mRNA differences and constructed an entire ceRNA network, which may be involved in important biological pathways related to TCMR. Currently, some studies have found that these miRNAs affect the progression of certain immune-related diseases by regulating corresponding mRNAs. This holds important guiding significance for transplant rejection, which also involves aberrant activation and dysfunction of immune cells. Research has found that miR-346 expression is present in synovial cells activated by LPS induction of pattern recognition receptors (PRRs) in rheumatoid arthritis patients. This miRNA negatively regulates the IL-18 response in fibroblast-like synoviocytes by inhibiting the transcription of Bruton’s tyrosine kinase. This mechanism has been validated in THP-1 cells, demonstrating miR-346’s inhibitory effect on IL-18^[160]57. Researchers have found through dual-luciferase reporter analysis that miR-130a negatively regulates IL-18, thereby participating in the regulation of primary immune thrombocytopenia^[161]58. The miR-124-3p, which regulates GBP1, acts as a suppressor in various cancers and also plays a role in cell apoptosis and inflammatory lesions^[162]59. This approach potentially offers a new perspective for researching TCMR mechanisms, discovering novel therapeutic targets, and developing biomarkers. Moreover, drug prediction for the five key genes identified 26 therapeutic drugs corresponding to three key genes, with tacrolimus, mycophenolate mofetil, methylprednisolone, and cyclosporine already widely used in the treatment of liver transplant rejection^[163]56,[164]60,[165]61. Previous research has confirmed that colchicine inhibits T cell proliferation by blocking the expression of the IL-2R gene, damaging the antigen recognition process^[166]62. Lifitegrast, an integrin antagonist, has been proven in dry eye disease research to block the antigen transfer process from APC to T cells^[167]63. These targeted drugs may provide new insights for the prevention and treatment of TCMR after liver transplantation. Finally, immunohistochemistry was used to validate the upregulated expression of key genes in human liver biopsy tissues. The significant upregulation of four key genes: GBP1, IL-18, CD53, FCER1G, in the TCMR group further supports their potential as predictive indicators and therapeutic targets for liver transplant TCMR. This finding also provides a theoretical basis for further exploration of their molecular mechanisms. However, we acknowledge the limitations of our study, primarily due to the scarcity of post-transplant liver biopsy samples, leading to insufficient validation. Our data were derived from a public database, which may not capture all possible TCMR case types or patient subgroups, potentially limiting the generalizability of our conclusions. Additionally, there may be unknown systematic biases or technical variations in the data collection and processing that could affect the accuracy of the results. To address this, we incorporated additional datasets for single-cell analysis and immunohistochemistry to enhance the study’s comprehensiveness and validation, but these datasets may also share similar limitations. Furthermore, the specific functions and potential mechanisms of key genes involved in liver transplant rejection require further investigation. Predictions related to miRNA, lncRNA, and targeted drugs were also based on public databases and have yet to be experimentally validated. In future research, we plan to explore more comprehensive, diverse, and high-quality datasets while implementing corresponding foundational experiments to gain deeper insights into gene expression changes and their biological significance in the TCMR process. Conclusions To sum up, this work provides insights into the molecular mechanisms of TCMR through a series of bioinformatics methods such as differential expression analysis, WGCNA, enrichment analysis, immune infiltration analysis, GSEA enrichment analysis, and single-cell analysis. Additionally, drug analysis targeting key genes helps provide more references for the treatment of post-transplant rejection.