Abstract Objective Despite advances in understanding systemic immune responses to Mycobacterium tuberculosis (Mtb), the localized immune dynamics within infected lymph nodes, particularly cell-type-specific transcriptional reprogramming and intercellular crosstalk, remain poorly defined, impeding targeted therapy development. By means of single-cell transcriptomics, the objective was to dissect the immune microenvironment and map intercellular crosstalk in Mtb-infected cervical lymph nodes, with the purpose of uncovering the mechanisms of localized immunity and immunopathology. Methods Paired Mtb-positive cervical swollen lymph nodes (SLN) and adjacent Mtb-negative normal-appearing lymph nodes (NLN) from five cervical LNTB patients were analyzed using single-cell RNA sequencing (scRNA-seq). Computational network modeling (CellChat) and flow cytometry validation were employed to map immune cell heterogeneity and cell–cell communication. Results ScRNA-seq identified ten T cell subsets, ten B cell subsets, and six myeloid subsets, revealing conserved frequencies but substantial transcriptional reprogramming in SLN. The SLN exhibited extensive upregulation of pro-inflammatory pathways across T, B, and myeloid cells, accompanied by minimal alterations in subset frequencies. IL1B + macrophages in SLN showed an enrichment of genes associated with oxidative phosphorylation, antigen presentation, and inflammasome-related genes. The SLN demonstrated an increased cell–cell communication driven by the crosstalk between macrophage and CD8 + T/NKT cells. Validation through flow cytometry confirmed comparable proportions of immune subsets between the SLN and the NLN, which was consistent with the findings of scRNA-seq. Conclusions This study delineates a spatially coordinated immune strategy in cervical LNTB. In this context, Mtb infection induces transcriptional and metabolic reprogramming rather than subset redistribution. The strengthened interaction between macrophages and T cells emphasizes that cellular immunity serves as the main impetus for bacterial containment. Nevertheless, there are trade-offs between inflammation and tissue integrity. These insights provide a framework for developing therapies that target intercellular networks to achieve a balance between immunity and pathology in LNTB. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-025-00763-y. Keywords: Cervical Lymph Node Tuberculosis, Single-cell RNA sequencing, Transcriptional reprogramming, Cell–Cell communication Introduction Globally, it is estimated that one-third of people are infected with Mycobacterium tuberculosis (Mtb), the agent responsible for tuberculosis (TB). Generally, the lungs are the most common site of infection and disease, but extrapulmonary tuberculosis (EPTB) accounted for 16% of notified TB cases in 2020, as reported by the World Health Organization (WHO) Global Tuberculosis Report [[42]1]. In clinical practice, EPTB at atypical sites is frequently diagnosed incidentally[[43]2]. The challenges in diagnosis arise from nonspecific clinical manifestations and the frequent absence of systemic symptoms, which often lead to delayed recognition and the initiation of treatment [[44]3, [45]4]. Notably, EPTB demonstrates particular predilection for immunocompromised populations including patients with diabetes or HIV infection as well as vulnerable age groups [[46]5]. Of the EPTB, cervical lymph node TB (LNTB) is one of frequent forms [[47]6]. However, they paradoxically also harbor Mtb replication and elimination [[48]7–[49]11]. Despite advances in understanding systemic immune responses to Mtb, the localized immune dynamics within infected lymph nodes (LNs), particularly the interplay between cellular subsets, remain poorly defined. This knowledge gap impedes the development of targeted therapies aimed at enhancing bacterial clearance while mitigating immunopathology. Conventional studies of LNTB have largely focused on bulk transcriptional profiling or immunohistochemical characterization, revealing upregulated inflammatory pathways and altered lymphocyte frequencies in infected nodes [[50]12–[51]15]. For instance, Elevated type 1 and 17 cytokines and reduced IL-1β at infection sites suggest a skewed immune milieu favoring bacterial persistence [[52]16]. However, these approaches lack resolution to dissect cell-type-specific responses or intercellular crosstalk, which is a critical driver of immune coordination. Single-cell RNA sequencing (scRNA-seq) has revolutionized immunology by enabling deconvolution of heterogeneous tissues into functionally distinct subsets [[53]17]. However, to date, no study has systematically mapped cell–cell communication networks in human LNTB. Cell–cell communication mediated by ligand-receptor interactions orchestrates immune cells recruitment, activation, and effector functions [[54]18]. In TB, dendritic cells (DCs) and macrophages deliver Mtb antigens to LNs, priming T cell responses [[55]11], while the cytokine cross-talk between myeloid and lymphoid cells determines the outcomes of infection. For example, IFN-γ secreted by CD4 + T cells enhances the bactericidal activity of macrophage, while excessive IL-1β signaling may exacerbate tissue damage. Despite its pivotal role, the spatial and functional organization of these interactions in Mtb-infected LNs remains poorly understood. Do infected nodes exhibit unique signaling hubs that differentiate them from adjacent normal nodes? How do specific immune subsets, such as macrophages and CD8 + T cells, collaborate to contain Mtb? Addressing these questions requires a systems-level analysis of intercellular communication. Here, we combined scRNA-seq with computational network modeling to comprehensively map the immune landscape and cell–cell communication dynamics in paired swollen lymph nodes (SLN) and normal-appearing lymph nodes (NLN) from cervical LNTB patients. We identified 30 subsets of immune cell and revealed extensive transcriptional reprogramming in SLN. Significantly, CellChat analysis uncovered an enhanced global interaction strength in SLN, which was driven by the crosstalk between macrophage and CD8 + T cell/NKT cells through the IFN-II and IL-1 pathways. These findings not only delineate the cellular basis of localized immune responses in LNTB but also provide a model for targeting intercellular networks to improve TB control. Methods and materials Isolation of cervical LNs for scRNA-seq Five patients diagnosed with cervical LNTB were recruited based on clinical manifestations and microbiological confirmation of Mtb infection through either culture or Xpert MTB/RIF assay (Cepheid, USA). Exclusion criteria included HIV co-infection and prior exposure to anti-TB therapy, ensuring no confounding effects from immunocompromised status or drug interventions. The detailed demographic and clinical characteristics of the cohort are presented in Supplementary Table 1. During surgical resection, paired samples were collected from each patient: (i) one cervical SLN exhibiting pathological features of LNTB, and (ii) one or two adjacent NLN serving as internal controls. NLN were defined as nodes meeting the following criteria: (1) normal anatomical size (bean-shaped, < 1 cm in diameter), (2) absence of gross pathological features (necrosis, suppuration or granuloma formation), and (3) microbiological confirmation of Mtb negativity via Xpert MTB/RIF assay. This sampling strategy resulted in a total cohort of 5 SLN and 8 NLN specimens for downstream analyses. Immediately post-excision, tissues were transferred to sterile petri dishes and gently washed with ice-cold phosphate-buffered saline (PBS) (Solarbio, Beijing, China) to remove peripheral blood contamination. Preparation of single-cell suspensions for scRNA-seq LN samples were cut into approximately 1 mm^3 pieces and immersed in RPMI-1640 medium (Thermo Fisher Scientific, Massachusetts, USA) Supplemented with 10% fetal bovine serum (FBS) (ScienCell, San Diego, California, USA). A portion of tissue fragments was reserved for the detection of Mtb through the Xpert MTB/RIF assay (Cepheid, USA), while the remaining tissue was Subjected to enzymatic digestion in a 37℃ water bath for 30 min. The resultant cell Suspension was filtered through a 40-μm nylon cell strainer and centrifuged at 300 g for 5 min at 4℃. Following the removal of the supernatant removal, the pellet was resuspended in erythrocyte lysis buffer (Solarbio, Beijing, China) and incubated on ice for 5 min to Eliminate residual red blood cells. After being washed twice with PBS, the pellet was resuspended in PBS containing 2% FBS for the preparation of a single-cell RNA library. Single-cell RNA library construction and sequencing According to the manufacturer's protocol, single-cell capturing and downstream Library constructions were carried out using a 10× Chromium Single Cell 3' Reagent Kit (V2 Chemistry). Briefly each cell suspension was loaded onto a Chromium single-cell chip, partitioning oil, reverse transcription (RT) reagents, and a collection of gel beads. The captured cells were lysed, and the RNA released was barcoded through RT in individual Gel Bead-In-Emulsions (GEMs). RT was performed using a C1000 TouchTM Thermal Cycler (Bio-Rad, California, USA). Complementary DNA (cDNA) was generated and amplified by PCR. The quality of amplified cDNA was assessed using an Agilent 4200 (performed by BGI Technology, Shenzhen, China). Single-cell libraries were then constructed and normalized based on the manufacturer's standard parameters. The libraries were sequenced using DNBSEQ T1 sequencer with a paired-end 100-bp (PE100) reading strategy (performed by BGI technology, Shenzhen, China). ScRNA-seq data processing and cell annotation The raw gene expression matrix was generated from each sample using Cell Ranger (v6.1.2) [[56]19]. Pipeline is coupled with human reference genome version GRCh38. Filtered matrices were analyzed with the Seurat R package (v 3.2.0) [[57]20]. Quality control was performed based on the following criteria: 1) Cell filtering: Cells with fewer than 200 detected genes or > 90% of the maximum gene count per sample were excluded. 2) Mitochondrial read filtering: Cells in the top 15% of mitochondrial read ratios were removed. 3) Doublet removal: Potential doublets were identified and eliminated using Doublet Detection (v3.0; 10.5281/zenodo.2678041). Cell cycle phase was assessed via the CellCycleScoring function in Seurat. After quality filtering, expression matrices were normalized, and highly variable genes (2,000 genes) were selected for principal component analysis (15 PCs). Dimensionality reduction and clustering were performed using Uniform Manifold approximation and projection (UMAP) and the Louvain algorithm (resolution = 0.01). Differentially expressed genes (DEGs) for each cluster were determined using the FindAllMarkers function with the following parameters: Wilcoxon rank-sum test with Bonferroni-adjusted P < 0.05; Minimum log2 fold change (|log2FC|) > 0.25; Genes detected in ≥ 25% of cells within the cluster and showing a ≥ 0.2-fold difference compared to other clusters. To resolve finer cellular heterogeneity, major clusters were re-clustered at a higher resolution (resolution = 0.5). Subpopulation identities were annotated based on canonical marker genes. Flow cytometry analysis SLN and NLN were Surgically resected from patients diagnosed with cervical LNTB. Single-cell Suspensions were prepared by mechanically dissociating tissues using a syringe plunger followed by filtration through a 70 μm cell filter. Residual erythrocytes were lysed using ACK Lysing Buffer (Thermo Fisher, CA, USA). For Flow Cytometry, Fc receptors were blocked using CD16/CD32 monoclonal antibody (BD, 553,141). Cells were stained with antibodies against surface proteins CD45-APC-Cy7 (BD, 368,516), CD19-APC(BD, 555,415), CD14-Bv421(BD, 325,628), CD3-PE-Cy7(BD, 557,851), CD4-Percp (BD, 652,823), CD8-FITC(BD, 555,643). Surface staining was performed in 1× FACS Buffer for 30 min at 4 °C in the dark. Cells were washed once with PBS (with 2% FBS) and resuspend in 200 μl FACS Buffer. Stained cells were immediately analyzed on BD FACSymphony™A3 (BD Biosciences, CA, USA) and Flow-Jo software (v10.0, Tree Star, Inc.) was used for post-acquisition analysis. Cell population frequencies were quantified using FlowJo’s gating strategies. Gating strategies are shown in Supplementary Fig. 1A. Statistical differences between SLN and NLN were evaluated using the paired Wilcoxon signed-rank test. P-values were adjusted for the false discovery rate (FDR), and a significance threshold of P < 0.05 was applied. DEGs between SLN and NLN DEGs from each cell type between SLN and NLN samples were identified using the "FindMarkers" function in Seurat with default parameters. A Bonferroni-adjusted P < 0.05 and |log2FC|> 0.25 was used to define significant DEGs. Pathway enrichment analysis To determine the significantly enriched pathways, the DEGs were mapped to the Kyoto Encyclopedia of Genes and Genomes (KEGG) database for pathway enrichment analysis using the Dr.Tom network platform of BGI ([58]http://report.bgi.com). The calculated P-values were adjusted for FDR, with FDR < 0.05, as a threshold. KEGG pathways satisfying this condition were defined as significantly enriched pathways. Gene set enrichment analysis (GSEA) To further explore the functional implications of transcriptional differences in cell types between the SLN and NLN groups, GSEA was performed. Initially, DEGs analysis was conducted between the SLN and NLN groups, with a threshold of absolute |log2FC|> 0.1 and Bonferroni-adjusted P < 0.05. GSEA was implemented using the R package “clusterProfiler” to identify Gene Ontology (GO) Biological Process terms that were enriched in either the SLN or NLN groups. The significance of enrichment was assessed with 1,000 permutations, and gene sets meeting FDR < 0.25 and P < 0.05 were considered statistically significant. Visualization of the enriched pathways was performed using dot plots and ridge line plots to highlight normalized enrichment scores (NES) and leading-edge genes. Cell − cell communication analysis ScRNA-seq data from all LN samples were analyzed to investigate intercellular communications. This analysis was conducted using the R CellChat package version 2.1.0 [[59]21, [60]22]. The single-cell RNA-seq data were processed as follows: Seurat objects containing normalized gene expression matrices and cluster annotations for SLN and NLN samples were converted into CellChat objects. Ligand-receptor interactions were annotated using the CellChatDB database [[61]22]. Communication probabilities between cell clusters were calculated by integrating the gene expression levels of ligands and receptors. Interactions were filtered if either the ligand- or receptor-expressing cluster contained fewer than 10% cells, ensuring robustness against low-count noise. The SLN and NLN CellChat objects were merged to enable a direct comparison of interaction networks. The total numbers of interactions and interaction strength (summed communication probabilities) were compared between SLN and NLN. For each cell cluster, differential incoming (receiver) and outgoing (sender) interaction strengths were calculated and visualized in two-dimensional space using Wilcoxon rank-sum tests (FDR < 0.05). Pathway-specific information flow was defined as the sum of communication probabilities across all ligand-receptor pairs within a pathway. Upregulated or downregulated ligand-receptor pairs in SLN were extracted and mapped to their cellular sources and targets. Statistical analysis All statistical analyses in this study were performed using R (v4.3.3) or GraphPad Prism (version 10.0). The statistical methods and the associated threshold for each analysis were detailed in the preceding method sections. Results Characterization of the major cell types in cervical LNs To investigate the local immune response to Mtb infection in cervical LNTB patients, five paired SLN and NLN samples were collected and analyzed by scRNA-seq (Fig. [62]1A). Subsequent to quality control, 38,968 cells, with a median of 1,311 genes per cell, were obtained from SLN samples and 45,735 cells, with a median of 1,203 genes per cell, were obtained from NLN samples (Supplementary Table 2). Unsupervised clustering performed using the Seurat algorithm classified all the analyzed cells into three major categories: T cells (CD3D + CD3E +), B cells (CD79A + CD79B +), and myeloid cells (CD68 + LYZ +) (Fig. [63]1B and C). Fig. 1. [64]Fig. 1 [65]Open in a new tab Characterization of total cell in cervical LNs from LNTB patients. A Schematic representation of the experimental workflow: Paired SLN and NLN samples were collected from 5 LNTB patients, followed by single-cell RNA sequencing (10 × Genomics) and integrative bioinformatic analysis. B UMAP visualization of 84,703 high-quality cells color-coded by annotated cell types. Major immune populations include T cells (red), B cells (green) and myeloid cells (blue). C Feature plots displaying expression gradients of lineage-defining markers (CD3D and CD3E for T cells, CD79A and CD79B for B cells, CD68 and LYZ for myeloid cells). Gray-to-blue gradient indicates normalized expression levels. D Boxplots comparing the proportions of B, T and myeloid cells between the SLN and NLN samples. The x-axis represents the cell type, and the y-axis shows the proportion of each cell type. Data are presented as mean ± standard deviation, * adjust P < 0.05 considered statistically significant, “ns” indicates non-significant differences Within the total cell population, T cells and B cells were the major constituents, while myeloid cells accounted for only a small proportion. Comparative analysis showed no significant differences in the relative frequencies of these three cell types between the SLN and NLN samples (Fig. [66]1D). To confirm these findings, we independently analyzed SLN and NLN samples from three additional LNTB patients using flow cytometry. Consistent with scRNA-seq results, no significant differences were observed in the proportions of T cells, B cells, or myeloid cells between the SLN and NLN groups (Supplementary Fig. 1A and B). Identification of 10 T cell subsets in cervical LNs To explore the T cell Subsets in cervical LNs from LNTB patients, we re-clustered all the T cell cluster. As a result, we identified 11 clusters based on the distinct expression of associated markers (Fig. [67]2A). While most clusters expressed canonical T cell markers CD3D and CD3E, Cluster 10 was identified as NK cells based on cytotoxic genes (GNLY, TRDC, KLRB1) and anti-inflammatory SPINK2 [[68]23] (Fig. [69]2B, C, Supplementary Figure S2A-B). Cluster 9 represented NKT cells with dual cytotoxicity (GZMA/B, NKG7). A distinct exhausted CD4 + T cell subset (Cluster 8), marked by immune checkpoint molecules (CTLA4, PDCD1) and lymphoid chemokine CXCL13 (Fig. [70]2B, C), indicative of chronic antigen exposure in Mtb infection. Two T sub-clusters exhibiting high expression of CD8A and CD8B were respectively categorized as naïve CD8 + T cells (CCR7 + SELL +) and effector CD8 + T cells (GZMA + GZMK + NKG7 +). Interestingly, six subsets (Clusters 1–5, 7) displayed ambiguous CD4 and CD8 expression profiles yet were classified into two types of naïve T cells (CCR7 + SELL +): XIST + naïve T cells and RPL21 + naïve T cells; three central memory T cells (S100A4 + GPR183 +): S100A4 + Tcm, NR4A1 + Tcm, and GADD45G + Tcm; and regulatory T cells (Tregs) (FOXP3 + IL2RA +) (Fig. [71]2B, C, Supplementary Figure S2A-B). These findings underscore the functional diversification of T cells in response to Mtb, emphasizing transcriptional reprogramming rather than changes in subset frequencies. Fig. 2. [72]Fig. 2 [73]Open in a new tab Characterization of T cell subsets between the SLN and NLN samples. A UMAP plot displaying T cell clusters identified across five paired SLN and NLN samples. The left panel represents all cell types. The right panel focuses on the distribution of samples, with each dot representing an individual cell and colors indicating different samples. B Dot plot of canonical marker gene expression (columns) across clusters (rows). Dot size reflects the percentage of expressing cells; color intensity denotes mean normalized expression (log2 scale). C Violin plots highlighting differential expression of canonically cell marker genes across clusters. The x-axis represents the T cell subsets, and the y-axis shows the normalized expression levels. D The GSEA analysis highlighting biological processes enriched in SLN samples compared to NLN samples. The x-axis represents the running enrichment score, and the y-axis indicates the rank in the ordered dataset. E Boxplots comparing the proportions of various T cell subsets between the SLN and NLN samples. The x-axis represents the T cell subsets, and the y-axis shows the proportion of each subset. Data are presented as mean ± standard deviation, * adjust P < 0.05 considered statistically significant, “ns” indicates non-significant differences. F Boxplots showing the number of up-regulated and down-regulated DEGs in each T cell subset between the SLN and NLN samples. G KEGG pathway analysis with DEGs for each T cell subset. Adjust P < 0.05 is considered to be statistical significance T cell subsets exhibit functional activation in cervical SLN To interrogate functional differences in T cells between SLN and NLN, we conducted GSEA on DEGs. The DEGs in SLN were significantly enriched in biological process related to T cell activation and differentiation (Fig. [74]2D), suggesting enhanced functional activity within the SLN. Notably, the analysis of T cell subset proportions revealed a markedly low abundance of CD4 + T cells, a finding corroborated by independent flow cytometry validation (Supplementary Figs. 1A and B). Although the frequencies of major T cell subsets were comparable between SLN and NLN (Fig. [75]2E), within each T cell subset, distinct transcriptional differences were observed. In the SLN group, the number of upregulated genes across subsets ranged from 38 to 119, In contrast, the number of downregulated genes was Substantially fewer, with only 10–20 genes showing reduced expression (Fig. [76]2F and Supplementary Tables S3-S12). Notably, KEGG pathway analysis revealed that the upregulated DEGs were associated with apoptosis, TNF/IL-17 signaling, and MAPK activation (Fig. [77]2G), indicating a strong inflammatory environment in the SLN. Collectively, these findings suggest that Mtb infection induces transcriptional reprogramming in T cells, prioritizing effector functions and inflammatory signaling over changes in subset frequency, thereby orchestrating localized immune responses in LNTB. Ten B cell subsets identified by scRNA-seq in cervical LNs The total B cells were re-clustered into 12 distinct clusters based on the expression of associated markers, including ten B cell subsets (CD79A +/CD79B +) alongside monocytic cells (cluster 11: LYZ +/CST3 +) and cycling T cells (cluster 10: CD3D +/CD3E +/MKI67 +) (Fig. [78]3A-C and Supplementary Figure S3A-B). Naïve B cell subsets (clusters 1/3/4/7/8) were characterized by a high level of co-expression of MS4A1 and IGHD and were further stratified into five functional subsets based on the specific enrichment of marker genes: IGHD + naïve (IGHD/TCL1A/IGHM/PLPP5), CD69 + naïve (CD69/ZNF331/CD83/NR4A1), PSME2 + naïve (PSME2/NME1/NME2), ISG15 + naïve (ISG15/IFI6/IFIT3/IFIT1), and NEAT1 + naïve (NEAT1). Memory B cells (clusters 0/2, MS4A1 +/CD27 +) diverged into AIM2 + memory B subset (AIM2/TNFRSF13B) and HSPA1A + memory B subset (HSPA1A). While germinal center (GC) B cells (clusters 5/6, MS4A1 +/CD27 +/CD38 +) comprised RGS13 + GC B subset (CCDC144A/RGS13/MEF2B/LMO2) and HMGB2 + GC B subset (HMGB2/STMN1/HIST1H4C/TUBA1B/HMGN2). The unique plasma B subset (cluster 9, MZB1 +/CD38 +/IGHG4 +) was specifically enriched with antibodies including such as IGHG4, IGHG1, IGHG3, IGLC1, and IGHA1. Fig. 3. [79]Fig. 3 [80]Open in a new tab Characterization of B cell subsets between the SLN and NLN samples. A UMAP plot displaying B cell subsets identified across five paired SLN and NLN samples. The left panel represents all cell types. The right panel focuses on the distribution of samples, with each dot representing an individual cell and colors indicating different samples. B Dot plot of canonical marker gene expression (columns) across clusters (rows). Dot size reflects the percentage of expressing cells; color intensity denotes mean normalized expression (log2 scale). C Violin plots highlighting differential expression of canonically cell marker genes across clusters. The x-axis represents the cell subsets, and the y-axis shows the normalized expression levels. D The GSEA analysis highlighting biological processes enriched in SLN samples compared to NLN samples. The x-axis represents the running enrichment score, and the y-axis indicates the rank in the ordered dataset. E Boxplots comparing the proportions of various B cell subsets between the SLN and NLN samples. The x-axis represents the B cell subsets, and the y-axis shows the proportion of each subset. Data are presented as mean ± standard deviation, * adjust P < 0.05 considered statistically significant, “ns” indicates non-significant differences (F) Boxplots showing the number of up-regulated and down-regulated DEGs in each B cell subset between the SLN and NLN samples. G KEGG pathway analysis with DEGs for major B cell subsets. Adjust P < 0.05 is considered to be statistical significance Functional divergence of B cell subsets in cervical LNs GSEA of DEGs in total B cells between SLN and NLN revealed significant enrichment of pathways related to adaptive immunity, lymphocyte activation regulation, and T cell priming in SLN (Fig. [81]3D), underscoring enhanced functional engagement of B cells during Mtb infection. As shown in Fig. [82]3E, four major B cell subsets were identified: AIM2 + memory B cells, IGHD + naïve B cells, HSPA1A + memory B cells, and CD69 + naïve B cells, which together accounted for nearly 80% of the total B cell population. Analogous to the observations in T cells, no significant differences were observed in the frequency of these B cell subsets between the SLN and the NLN (Fig. [83]3E). Transcriptomic analysis identified a greater number of upregulated DEGs in the SLN when compared to the NLN, while only a relatively small number of genes showed significant downregulation (Fig. [84]3F and Supplementary Tables S13–S21). Notably, GC B cell subsets exhibited minimal transcriptional differences between the two groups (Fig. [85]3F and Supplementary Tables S13–S21). KEGG pathway analysis further linked the upregulated DEGs to apoptosis, IL-17/TNF signaling, and MAPK activation (Fig. [86]3G), suggesting a pro-inflammatory B cell phenotype in SLN. These findings suggest that B cells may contribute to the amplification of local inflammatory responses during Mtb infection through the modulation of cytokine signaling and survival pathways, despite the maintenance of stable subset frequencies. Five myeloid cell subsets identified by scRNA-seq in cervical LNs The entire population of myeloid cells were re-clustered into seven distinct clusters based on the cell-specific marker genes, including three macrophage subsets (CD68 +), two plasmacytoid DC (pDC) subsets (CLEC4C + LILRA4 +), and two mesenchymal stem cell (MSC) subsets (DCN + COL3A1 +) (Fig. [87]4A-C and Supplementary Figure S4A, B). The macrophage subsets were classified as IL1B +, C1QC +, and S100B + subset, all of which displayed high expression of the M1-polarization marker CD86, indicating a pro-inflammatory phenotype within this cell population. Notably, the IL1B + macrophages expressed a high level of the pro-inflammatory cytokine IL1B and associated mediators (CCL3L1, S100A8,S100A9), suggesting specialized roles in inflammatory cascades. Both pDC subsets highly expressed cytotoxic and immune-regulatory genes (GZMB, JCHAIN, DUSP5, GPR183, TCL1A, and PTGDS) (Fig. [88]4B). Fig. 4. [89]Fig. 4 [90]Open in a new tab Characterization of myeloid cell subsets between the SLN and NLN samples. A UMAP plot displaying myeloid cell subsets identified across five paired SLN and NLN samples. The left panel represents all cell types. The right panel focuses on the distribution of samples, with each dot representing an individual cell and colors indicating different samples. B Dot plot of canonical marker gene expression (columns) across clusters (rows). Dot size reflects the percentage of expressing cells; color intensity denotes mean normalized expression (log2 scale). C Violin plots highlighting differential expression of canonically cell marker genes across clusters. The x-axis represents the cell subsets, and the y-axis shows the normalized expression levels. D The GSEA analysis highlighting biological processes enriched in the SLN samples compared to NLN samples. The x-axis represents the running enrichment score, and the y-axis indicates the rank in the ordered dataset. E Boxplots comparing the proportions of various myeloid cell subsets between SLN and NLN samples. The x-axis represents the myeloid cell subsets, and the y-axis shows the proportion of each subset. Data are presented as mean ± standard deviation, * adjust P < 0.05 considered statistically significant, “ns” indicates non-significant differences. F Boxplots showing the number of up-regulated and down-regulated DEGs in each myeloid cell subset between the SLN and the NLN samples. G KEGG pathway analysis with DEGs for IL1β + macrophages and MSC1. Adjust P < 0.05 is considered to be statistical significance Assessment of myeloid cell subset differences between SLN and NLN samples To explore the functional heterogeneity among myeloid cells in SLN as compared to NLN, GSEA was conducted on DEGs obtained from myeloid populations. This analysis revealed that DEGs in SLN were significantly enriched in pathways related to embryonic organ morphogenesis, G protein-coupled receptor signaling, and inflammatory responses (Fig. [91]4D). Cellular composition analysis demonstrated comparable frequencies of myeloid subsets between SLN and NLN (Fig. [92]4E), with IL1B + macrophages and GZMB + pDCs collectively dominating. Subsequent transcriptional profiling uncovered subset-specific gene expression changes. In particular, IL1B + macrophages in SLN exhibited 313 DEGs, including 249 upregulated and 64 downregulated genes (Fig. [93]4F and Supplementary Table S22-23). The upregulated genes were strongly associated with pro-inflammatory signaling pathways, including IL-17, TNF, NOD-like receptor, and NF-κB signaling (Fig. [94]4G). Strikingly, MSC1 (a distinct myeloid subset) displayed 500 DEGs, with 428 genes significantly upregulated. These genes were enriched in pathways related to oxidative phosphorylation, TNF signaling, and antigen presentation machinery (Fig. [95]4G). In contrast, other myeloid subsets showed minimal transcriptional divergence between the SLN and NLN, suggesting a general functional stability across LN conditions. These findings highlight the activation of specific myeloid subsets, particularly IL1B + macrophages and MSC1, in driving inflammatory and metabolic reprogramming in LNTB. Enhanced cell–cell communications in SLN samples To systematically compare cell–cell communications dynamics between the SLN and the NLN, a computational network analysis was carried out using CellChat. The SLN exhibited a higher number of interactions (SLN: 9,614 vs. NLN: 8,275) and a stronger interaction strength intensity (SLN = 285.919 vs. NLN = 214.418), indicating a systemic enhancement of cellular crosstalk in the infected nodes (Fig. [96]5A). Visualization via a heatmap confirmed more extensive connectivity across all cell types in the SLN. Among them, C1QC + macrophages, MSC1, and MSC2 emerged as the predominant receivers of incoming signals, while MSC1 and HMGB2 + GC B cells showed heightened outgoing signaling activity (Fig. [97]5B). Notably, naïve CD8 + T cells, MSC2, and NKT cells ranked as the top three cell types with elevated incoming interaction strength in SLN, whereas MSC1 and S100B + macrophages dominated outgoing interactions. Critically, macrophages in SLN demonstrated intensified crosstalk with naïve CD8 + T cells and NKT cells, suggesting their central role in coordinating adaptive immunity. Fig. 5. [98]Fig. 5 [99]Open in a new tab Characterization of cell–cell communications among all cell types between the SLN and NLN samples. A The number of interactions and interaction strength among all cell types between the SLN and NLN. B Heatmaps show differential number of interactions (left) and differential interaction strength (right) among all cell types in the overall signaling patterns between the SLN and NLN. The color bars on the top show the total incoming signal for each column, while the bars on the right show the total outgoing signal for each row. Red and blue color indicates increases and decreases, respectively in signal in the SLN compared to the NLN. C Identification and visualization of conserved and specific signaling pathways. Orange color indicate pathways that were expressed only or higher in the SLN; Green color indicate those that were expressed only or higher the NLN. D Heatmaps of the overall signaling patterns of the SLN (left) and NLN (right). Circle plots show the INF-II (E) and IL-1 (F) signaling networks among all cell types between the SLN and NLN Comparative pathway analysis identified 18 SLN-specific pathways (VISFATIN, TGFb, PTN, CD46, CD80, PERIOSTIN, NOTCH, GAS, TWEAK, XCR, OSM, KIT, HSPG, DESMOSOME, EPHB, PROS, RA, and BMP) and 4 NLN-enriched pathways (SEMA3, ICOS, CD160, and SELL) (Fig. [100]5C). Shared pathways showed SLN-specific hyperactivation, including MK, CCL, and CD40 signaling (Fig. [101]5D). Cell-type-restricted pathways were also identified: PD-L1 signaling localized to monocytes and exhausted CD4 + T cells in both groups, while NCAM signaling linked MSC1 and NKT cells (Fig. [102]5D). Focusing on antimicrobial responses, IFN-II signaling, which is critical for the control of Mtb), originated from CD8 + effector T cells, NKT cells, and T cell subsets in the SLN. This signaling targeted macrophages (C1QC +/IL1B +) and plasma B cells (IGHG4 +) (Fig. [103]5E). In contrast, the NLN limited IFN-II signaling to NKT → macrophage interactions. Similarly, IL-1 signaling, a key innate immune driver, was amplified in the SLN. IL1B + and C1QC + macrophages transmitted signals to both NKT cells and MSC1, whereas the NLN restricted IL-1 reception to NKT cells (Fig. [104]5F). These findings establish macrophages as the central orchestrators of inflammatory crosstalk in the SLN, bridging the innate and adaptive immunity through spatially coordinated signaling. Discussion Despite its significant clinical implications, EPTB receives far less research attention than pulmonary TB due to its minimal infectiousness. Following Mtb infection, LNs are the first path of immune cell movement and are the most often infected area in EPTB [[105]24, [106]25]. Understanding their immune microenvironment—specifically whether these sites foster protective immunity or drive pathology—is fundamental to disease control. Conventional bulk RNA-seq approaches fail to resolve cellular heterogeneity, whereas scRNA-seq enables comprehensive characterization of distinct cell subsets and cell states [[107]17]. Notably, within infected individuals, only select LNs develop pathological changes while adjacent LNs appear histologically normal, presumably reflecting differential Mtb containment or clearance. By comparing the immune responses in abnormal lymph nodes to those in normal-appearing lymph nodes from individuals with LNTB, a deeper insight into the pathogenesis of LNTB can be gained. Our single-cell transcriptomic analysis of paired SLN and NLN obtained from cervical LNTB patients provides deep insights into the immune microenvironment of LNTB. Although previous studies have described broad inflammatory signatures in Mtb-infected LNs [[108]12, [109]16], our work advances the field by systematically mapping cell-type-specific transcriptional reprogramming and intercellular communication networks. The enhanced global cell–cell communication in SLN, which is characterized by macrophage-centric signaling hubs and amplified IFN-II/IL-1 pathways suggests a spatially coordinated immune strategy to counter Mtb, albeit with potential trade-offs between bacterial containment and immunopathology. The macrophages in SLN communication networks aligns with their dual roles as Mtb reservoirs and immune sentinels [[110]10, [111]11]. We observed that IL1B + macrophages, which exhibited marked upregulation of inflammation-related genes, engaged extensively with NKT cells via IL-1 and TNF signaling. This interaction likely amplifies IFN-γ production from cytotoxic lymphocytes, a mechanism critical for macrophage activation and intracellular pathogen control [[112]26–[113]30]. However, excessive IL-1β signaling may result in tissue damage [[114]31, [115]32]. Strikingly, while SLN macrophages displayed transcriptional activation, their frequency remained unchanged compared to NLN. This paradox mirrors findings in pulmonary TB granulomas, where macrophage functional polarization, rather than abundance, determines infection outcomes. Our data thus reinforce the paradigm that Mtb subverts myeloid cell plasticity to establish chronic infection niches. The expanded IFN-II signaling network in SLN—with CD8 + T/NKT cells as key IFN-γ sources—contrasts sharply with the attenuated responses observed in peripheral blood of TB patients [[116]33–[117]36]. This compartmentalization suggests that SLN serve as localized hubs for anti-Mtb effector functions, potentially compensating for systemic immune exhaustion. IFN-γ is known to enhance macrophage autophagy and antigen presentation, while chronic IFN-γ exposure may also induce T cell exhaustion. These findings highlight the need for therapies that balance localized immunity with systemic tolerance. Despite constituting nearly 50% of LN immune cells, B cell subsets showed minimal frequency changes between SLN and NLN. However, transcriptional profiling revealed significant upregulation of ISG15 and IFIT1 in naïve B cells. These genes are associated with antiviral responses. Conventionally, B cells are regarded as having a secondary role in the defense against TB, Nevertheless, our data suggest they may participate in Mtb control through non-canonical mechanisms, such as cytokine secretion or antigen-independent T cell priming. The absence of the Mtb-specific antibody detection in our study limits definitive conclusions, but high levels of IGHG1/IGHG4 in plasma cells hints at localized humoral responses. Future studies integrating BCR sequencing and spatial proteomics could clarify their role. The current study provides valuable insights into the immune microenvironment of cervical LNTB but has several limitations that warrant further discussion. The absence of healthy controls limits the ability to distinguish between baseline immunological variation and Mtb-specific changes. While the study employed rigorous bioinformatic pipelines, potential batch effects arising from technical variability were not explicitly addressed. Batch effects could confound transcriptional comparisons between the SLN and NLN groups, particularly given the small cohort size. Future studies should incorporate batch correction methods to ensure robust cross-sample comparisons. The cohort (n = 5 patients) may underrepresent the diversity of cervical LNTB immunopathology, especially given the heterogeneity of Mtb strains and host genetic backgrounds. Additionally, all samples were sourced from a single geographic region, which may limit the global applicability of findings. CellChat inferred enhanced cell crosstalk in SLN, these predictions rely on ligand-receptor databases and expression probabilities. Functional assays are needed to validate these interactions and establish causality. Last, the absence of paired peripheral blood mononuclear cell (PBMC) data precludes comparisons between local LN immunity and systemic immune responses. Such comparisons could elucidate compartmentalized immune exhaustion or activation patterns in LNTB. In conclusion, our single-cell transcriptomic study uncovers the complex immunological architecture of Mtb-infected cervical LN, providing the first comprehensive atlas of immune cell heterogeneity in cervical LNTB. We identified 10 T cell, 10 B cell, and 6 myeloid cell subsets, revealing remarkable functional specialization within each lineage. While SLN and NLN exhibited comparable immune subset frequencies, SLN demonstrated widespread transcriptional reprogramming across cell types, characterized by upregulation of pro-inflammatory pathways and metabolic remodeling. This chronic inflammatory milieu, coupled with enhanced intercellular communication networks centered on macrophage-T cell interactions, likely drives both anti-mycobacterial responses and pathological LN enlargement. By mapping the ligand-receptor axes governing immune crosstalk, this work paves the way for developing precision immunotherapies targeting cellular crosstalk in LNTB, while offering a framework to dissect compartmentalized immunity in other EPTB manifestations. Supplementary Information [118]Supplementary Material 1.^ (3.1MB, pdf) [119]Supplementary Material 2.^ (10.1KB, xlsx) [120]Supplementary Material 3.^ (10.2KB, xlsx) [121]Supplementary Material 4.^ (92KB, xlsx) [122]Supplementary Material 5.^ (83KB, xlsx) [123]Supplementary Material 6.^ (72.6KB, xlsx) Acknowledgements