Abstract Background Resistance to existing therapies is a major cause of treatment failure in patients with refractory and relapsed B-cell non-Hodgkin’s lymphoma (r/r B-NHL). Therapy-induced senescence (TIS) is one of the most important mechanisms of drug resistance. Methods This study used single-cell RNA sequencing to analyze doxorubicin-induced senescent B-NHL cells. C-C chemokine receptor 7 (CCR7) expression in patients with aggressive B-NHL was assessed using immunohistochemistry and flow cytometry. Lentiviral transfection was used to target CCR7 expression in Raji and SU-DHL-2 cells. Protein localization was visualized through immunofluorescence, while western blotting and co-immunoprecipitation were used to analyze protein expression and interactions. Cell proliferation was measured with the Cell Counting Kit-8 assay, and senescent cells were detected using senescence-associated β-galactosidase staining. The stemness of cells was evaluated through colony and sphere formation assays. Transwell assays assessed cell migration and invasion. Finally, inhibitors GS143 and Y27632 were used to examine the effect of IKBα and ARHGAP/RhoA inhibition on B-NHL-TIS. Results Here we identified a distinct group of TIS, composed of memory B-cell population characterized by strong positive expression of CCR7, which was significantly elevated in TIS population compared with normal proliferating and autonomously senescent lymphoma cell populations. Additionally, CCR7 expression was significantly upregulated in patients with r/r B-NHL, and was an independent prognostic factor in B-NHL, with high CCR7 expression being strongly associated with poor prognosis. In vitro results indicated that CCL21 induced migration and invasion of B-NHL cells via CCR7, while blocking CCR7 reduced doxorubicin-induced migration and invasion of these cells. Furthermore, B-NHL-TIS regulated by CCR7 and exhibited enhanced phenotypic and functional stemness features, including the upregulation of stemness markers, increased colony-forming, invasive and migratory capabilities. Mechanistically, blocking CCR7 reversed the stemness characteristics of senescent B-NHL cells by inhibiting the activation of ARHGAP18/IKBα signaling. Conclusions Together, TIS promotes the stemness of B-NHL cells via CCR7/ARHGAP18/IKBα signaling activation and targeting CCR7/ARHGAP18 might overcome the chemoresistance of senescent B-NHL cells by inhibiting stemness acquisition and maintenance. Keywords: B cell, Cytokine, Stem cell, Chemotherapy, Hematologic Malignancies __________________________________________________________________ WHAT IS ALREADY KNOWN ON THIS TOPIC * Therapy-induced senescence (TIS) contributes to drug resistance in B-cell non-Hodgkin’s lymphoma (B-NHL), with C-C chemokine receptor 7 (CCR7) implicated in cancer progression. WHAT THIS STUDY ADDS * This study demonstrates that CCR7 enhances the stemness of TIS B-NHL cells through the ARHGAP18/IKBα pathway, which contributes to chemoresistance. HOW THIS STUDY MIGHT AFFECT RESEARCH, PRACTICE OR POLICY * Targeting the CCR7/ARHGAP18 signaling pathway may offer a new approach to overcoming chemoresistance in relapsed B-NHL by reducing stemness in senescent cells. Introduction B-cell non-Hodgkin’s lymphoma (B-NHL) is the most common type of lymphoid malignancy in adults and is highly heterogeneous in clinical manifestation and prognosis. Although rituximab-based immunochemotherapy has been applied for several decades in B-NHL and the remission rate is dramatically improved, treatment failure with disease recurrence remains a major problem.[53]^1 2 Thus, the molecular mechanisms of B-NHL recurrence and drug resistance need further elucidation. Cellular senescence has been reported as a potent tumor-suppressive mechanism, mediated by the cell cycle arrest program and the senescence-associated secretory phenotype (SASP), which induces immuno-surveillance of pre-malignant cells.[54]^3 4 However, senescent tumor cells can re-enter the cell cycle and resume proliferation when external stressors are removed or environmental conditions change.[55]^5 6 Studies show that conventional chemotherapy often induces senescence in tumor cells, and this therapy-induced senescence (TIS) correlates strongly with poor clinical outcomes.[56]7,[57]9 TIS is responsible for tumor relapse and distant metastasis in post-treatment stage, by conferring the resistance and fueling the repopulation of remaining cancerous cells.[58]^10 Moreover, a study by Milanovic et al in Nature reveals that TIS promotes stem-cell-related properties of malignant cells via the activation of Wnt signaling.[59]^11 Exploring the mechanisms driving senescent tumor cells to acquire therapeutic resistance and stemness is crucial for understanding clinical resistance and recurrence in patients and for improving treatment strategies. C-C chemokine receptor 7 (CCR7) is the first identified lymphocyte-specific G protein-coupled receptor, and it has only two specific ligands: C-C chemokine ligand 21 (CCL21) and CCL19.[60]^12 The core function of the CCL19/CCL21/CCR7 axis is to induce dendritic cells carrying CCR7 to migrate directionally to draining lymph nodes, a process closely related to the occurrence and development of lymphoma.[61]^13 CCR7 is also highly expressed in various tumor cells, including B-NHL cells, and its expression is associated with a high risk of tumor metastasis through the induction of angiogenesis, lymph angiogenesis, and epithelial-mesenchymal transition (EMT).[62]13,[63]15 Currently, research on the involvement of chemokine receptors in the regulation of senescence mainly focuses on the well-studied receptors CXCR2 and CXCR4, with fewer studies on other receptors. CXCR2 is transactivated by p53 and significantly induces p38-mediated cellular senescence, exhibiting strong protumor activity.[64]^16 Both CXCR2 and CXCR4 have been reported to promote renal tubular cell senescence and renal fibrosis through β-catenin, a key molecule in Wnt signaling.[65]^17 18 Canonical Wnt and Notch pathways are significantly enriched in TIS and play central roles in stem-cell renewal in hematological malignancies.[66]^11 Notably, CCR7 has been reported to promote cancer cell stemness by activating the Notch[67]^19 and JAK/STAT[68]^20 pathways. However, the role of CCR7 in tumor TIS, particularly in altering the stemcell-related properties of lymphoma senescence, remains unclear. In this work, we perform single-cell RNA sequencing (ScRNA-seq) on chemotherapeutic drug doxorubicin (DOX) induced B-NHL-TIS model and identify a lymphoma TIS cell subset which consists of a type of memory B-cell population characterized by the CCR7+. We report here that chemotherapy-induced cellular senescence enhances the stemness of aggressive B-NHL by activating CCR7 signaling, promoting resistance and relapse. Methods and materials Human cell lines The human B-NHL cell lines Raji, Namalwa, SU-DHL-4, OCI-LY3, SU-DHL-2 and acute leukemia cell lines HL-60, MOLM-13 were obtained from the China Center for Type Culture Collection. Cells were maintained in Roswell Park Memorial Institute-1640 (Hyclone, Logan, Utah, USA; SH30809.01) with the addition of 10% fetal bovine serum (Lonsera, Uruguay; S711-001S). Cell cultures were maintained and incubated at 37°C in humidified air with 5% CO[2]. Patients From March 2021 to October 2023, a total of 112 patients with aggressive B-NHL, including 56 with diffuse large B-cell lymphoma (DLBCL), 3 with Burkitt lymphoma, and 53 with other types of aggressive B-NHL, as well as 6 patients with reactive lymph node hyperplasia (RLN) and 29 healthy controls, were enrolled in this study at the Second Affiliated Hospital of Anhui Medical University. Healthy individuals and RLN served as controls. The clinical characteristics of patients with B-NHL and controls are presented in [69]online supplemental table S1. Initial evaluation, staging, and response assessment of all patients with B-NHL were according to The Lugano International Conference on Malignant Lymphoma. All healthy volunteers included in this study demonstrated normal liver and kidney function, were free from autoimmune diseases, and had no history of using immunosuppressive drugs. TIS model constructing and ScRNA-seq Human B-NHL cell lines Raji (2×10^5 cells/mL) were treated with 20 nM, 40 nM and 80 nM DOX (Beyotime, Shanghai, China, SC0159) by repeated pressure in vitro and was induced to senescence as previously reported.[70]^21 The optimal concentration for induction was determined through staining with senescence-associated β-galactosidase kit (SA-β-Gal, Beyotime, Shanghai, China, C0602). Subsequently, single-cell suspensions (2×10^5 cells/mL) were loaded onto microfluidic devices and ScRNA-seq libraries were constructed according to the Singleron GEXSCOPETM protocol by GEXSCOPETM Single-Cell RNA Library Kit (Singleron Biotechnologies, Nanjing, China, 4180011). Libraries were diluted to 4 nM, pooled, and sequenced on an Illumina HiSeq X using 150 bp paired-end reads. Lentiviral transfection Raji and SU-DHL-2 cells were seeded at 1×10^5 cells per well in a 24-well plate. Human shCCR7 and CCR7 overexpression lentiviral vector plasmids (eRFP-puro, Gene ID 1236 and [71]NM_001838.4, constructed by Hanbio Tech, Shanghai, China) were added at an optimal multiplicity of infection of 50 for shCCR7 and 100 for CCR7 overexpression, based on pilot experiments. Lipofectamine 3,000 (Thermo Fisher Scientific, Waltham, USA, L3000075) was prepared at 5 µg/mL in Opti-MEM medium (Thermo Fisher Scientific, Waltham, USA, 31985062). Then, it was mixed with the virus and incubated at room temperature for 20 min. The mixture was added to the cells and incubated for 24 hours. After 24 hours, the medium was replaced with a complete medium and the cells were cultured for 72 hours. Cells were observed for fluorescence under a fluorescence microscope (Zeiss, Germany). Puromycin (Thermo Fisher Scientific, Waltham, USA, A1113803) was added at a concentration of 1.5 µg/mL to select successfully transfected cells. The surviving cells were subsequently expanded and cryopreserved for further use. Inhibitors treatment GS143 (IKBα inhibitor, MedChemExpress, New Jersey, USA, HY-110261) and Y27632 (RhoA/Rock inhibitor, MedChemExpress, New Jersey, USA, HY-10071) lyophilized powders were dissolved in dimethyl sulfoxide (Sangon Biotech, Shanghai, China, ST038) to prepare 10 mM stock solutions. The final working concentrations of the inhibitors GS143 and Y27632 were 0, 0.1, 1, 10, and 100 µM, and they were incubated with Raji cells (2×10⁶ cells/mL) for 1 hour. After incubation, the inhibitor-containing medium was replaced with DOX for induction as described previously. Statistical analysis Statistical analyses were conducted using SPSS V.19.0 software (IBM, Chicago, Illinois, USA) and GraphPad Prism V.8.0.2 (GraphPad Software, La Jolla, California, USA). For quantitative data following a normal distribution, the t-test was used for comparisons between two groups, while one-way analysis of variance (ANOVA) was applied for comparisons among multiple independent samples. For quantitative data that did not meet normal distribution criteria, the Mann-Whitney U test was used for comparing two independent samples, and non-parametric ANOVA was employed for comparisons among multiple samples. Categorical variables were presented as counts and percentages (n (%)), with comparisons made using either the χ^2 test or Fisher’s exact test. Overall survival (OS) was defined as the duration from the date of diagnosis to either death or the last follow-up, and progression-free survival (PFS) was defined as the time from the start of treatment to disease progression. Groups with high and low CCR7 expression were formed based on a predetermined cut-off value, and differences between these groups were assessed using the log-rank test. Survival analysis was performed using the Kaplan-Meier method, with between-group differences in survival curves evaluated via the log-rank test. Univariate and multivariate Cox proportional hazards regression models were used to evaluate the influence of various factors on OS. A p value of <0.05 was considered statistically significant. Results Identification of senescent cell subpopulations in the chemotherapy-induced senescent B-NHL cells model Various concentrations of DOX (20, 40, and 80 nM) were used to construct the TIS model of Raji cells in vitro. It was found that 40 nM DOX had significant advantages in constructing the TIS model of Raji cells, hence, it was used for subsequent experiments ([72]figure 1A,B). Subsequently, a total of 5,330 cells were subjected to ScRNA-seq analysis, comprising 3,261 cells from the control group and 2,069 cells from the DOX-induced TIS group. Using t-distributed stochastic neighbor embedding (t-SNE), the combined samples (comprising TIS and control groups) were categorized into eight primary B-cell subpopulations ([73]figure 1C). Clusters 1, 4, 5, and 7 were defined as plasma cell-like (MZB1, IGKC, JCHAIN), cluster 2 as memory B cells (CD27, CD19, PAX5, CD86), and clusters 3, 6, and 8 as undistinguishable B cells (MS4A1, CD27, PAX5, IGHM, MZB1, IGKC, JCHAIN) ([74]online supplemental figure S1A–K). Samples from the control group predominantly clustered in clusters 1, 4, 5, 6, 7, and 8, while those from the DOX-induced group primarily clustered in clusters 2, 3, and 8 ([75]figure 1D,E). Substantial variances in gene expression profiles were observed between the two groups, suggesting that chemotherapy-induced markedly modified the gene expression profile in B-NHL cells. Subsequent Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed notable enrichment of cell cycle and DNA replication pathways in clusters 1, 3, 4, 5, 7, and 8 (non-senescent cell clusters), whereas proteins involved in cell cycle and DNA replication in clusters 2 and 6 (senescent cell clusters) showed significant downregulation ([76]figure 1F–H and [77]online supplemental figure S2A–P). It suggested that cluster 2 depicted a DOX-induced senescent cell population in the chemotherapy-induced senescent B-NHL cells model (TIS group), whereas cluster 6 represented an autonomous senescent cell population (control group). Meanwhile, chemotherapy-induced senescent B-NHL cells exhibited significant enrichment of Nuclear Factor kappa B(NF-κB) pathway, whereas NF-κB signaling was significantly downregulated in non-senescent cells (cluster 1) ([78]online supplemental figure S2B,J). Gene expression profile analysis using ScRNA-seq revealed the expression of chemokine factor family including CCR7, NF-κB family including NFKBIA, and lymphocyte survival protective factor including BCL2A1, were significantly higher in cluster 2 of senescent B-NHL cells compared with other non-senescent clusters or autonomous senescent clusters ([79]figure 1I). The violin plot results showed the distribution of the top nine marker genes of the senescent cluster 2 in various cell subpopulations. Notably, CCR7 exhibited the most significant expression difference between the senescent cluster and the non-senescent clusters ([80]figure 1J). These results suggested that activation of CCR7 and NF-κB pathway might play a crucial role in the formation and function of senescent B-NHL cells. Figure 1. Identification of senescent cell population in a DOX-induced B-NHL-TIS model. (A) Chemical staining showing SA-β-Gal positive senescent cells after induction of Raji cells with 20, 40 and 80 nM DOX. (B) The proportion of SA-β-Gal positive senescent cells in Raji cells induced by 20, 40 and 80 nM DOX. (C) ScRNA-seq identified major senescent cell subpopulations in the senescence model. t-SNE dimensionality reduction analysis showing the clustering of cells in the combined sample (colored by cell group). (D) t-SNE dimensionality reduction analysis joint sample cell clustering situation (colored by sample). (E) The proportion of each cell cluster in two groups of samples. (F) Violin plots of cell cycle protein enrichment for cluster 1–8. (G) Violin plots of DNA replication protein enrichment for cluster 1–8. (H) Normalized Enrichment Scores (NES) of the cell cycle and DNA replication pathways are presented for clusters 1–8. NES values indicate the degree of enrichment for these pathways within each cluster, with positive values representing significant upregulation. Pathways with NES values above 1.5 are considered highly enriched, while those below −1.5 indicate downregulation. (I) Heatmap displaying the most significantly expressed marker genes in each cell population from the combined samples. (J) Violin plots show the distribution of significantly different genes in each cell population. Validation of marker gene expression in senescent cell populations in the B-NHL-TIS in vitro model. **p<0.01, ***p<0.001, ****p<0.0001. B-NHL, B-cell non-Hodgkin’s lymphoma; DOX, doxorubicin; SA-β-Gal, senescence-associated β-galactosidase; TIS, therapy-induced senescence; t-SNE, t-distributed stochastic neighbor embedding. [81]Figure 1 [82]Open in a new tab Stemness characteristics analysis of chemotherapy-induced senescent B-NHL cells In pseudotime trajectory analysis, we found that clusters 1, 4, 5, and 7, which concentrated on the left side of the trajectory, were identified as terminally differentiated plasma cell types. Cluster 2, which concentrated on the right side of the trajectory, was identified as early-stage differentiating cells, with differentiation direction from right to left ([83]figure 2A). These results indicated the chemotherapy-induced senescent B-NHL cell population was distributed in the earliest stage of cell differentiation, suggesting that senescent B-NHL cells might possess the potential capability of stemness. We then performed cluster analysis of the significantly differentially expressed genes and senescence-related genes. CCR7 was downregulated along the pseudotime trajectory, reflecting dynamic changes in various gene modules ([84]figure 2B,C). Next, the stemness characteristics of chemotherapy-induced senescent cells were investigated in this study. The expression levels of stemness markers CD34, CD44, CD150, and LGR5 were significantly higher in the senescent cell group compared with the control group ([85]figure 2D). Moreover, the TIS group demonstrated a significantly enhanced ability for stemness colony formation compared with the control group on day 3 ([86]figure 2E), and exhibited strong positive expression of CCR7, as well as co-expression with the stemness marker CD44 and the senescence marker H3K9me3, as observed using fluorescence confocal microscopy ([87]figure 2F). This indicates that chemotherapy-induced cellular senescence enhances the phenotypic and functional stemness features of B-NHL cells. Figure 2. Analysis of the phenotypic and functional stemness features in B-NHL-TIS cell populations. (A) Cell trajectory plot colored according to cell clusters. (B) Changes in expression modules of differentially expressed genes during the pseudo time process. (C) Expression changes of differentially expressed genes from the start to the end of the pseudo time process. Expression profile of stemness markers in the TIS model. (D) Expression levels of stemness markers CD34/CD44/CD150/LGR5 in the control group and TIS group detected using qRT-PCR. (E) Clonogenic capacity of cells in the control group and TIS group. (F) Co-expression of CCR7 and CD44/H3K9me3 in cells detected using immunofluorescence in the control group and TIS group. ****p<0.0001. B-NHL, B-cell non-Hodgkin’s lymphoma; CCR7, C-C chemokine receptor 7; DOX, doxorubicin; mRNA, messenger RNA; qRT-PCR, quantitative Reverse Transcription Polymerase Chain Reaction; TIS, therapy-induced senescence. [88]Figure 2 [89]Open in a new tab Abnormally high expression of CCR7 is related to poor prognosis in patients with B-NHL To investigate the expression levels of CCR7, CCL21, and CCL19, Immunohistochemistry (IHC) analysis was performed on lymph node tissue from patients with B-NHL and RLN. The results showed that CCL21 and CCR7 exhibited strong positive expression in the lymph nodes of patients with B-NHL, while CCL19 expression was negative. Additionally, CCR7 expression was significantly higher in patients with B-NHL compared with those with RLN, while CCL21 showed significant expression in both groups ([90]figure 3A). IHC analysis was also performed on bone marrow biopsy samples from patients with newly diagnosed (ND) B-NHL with or without clinically confirmed bone marrow involvement. The results demonstrated that the levels of CCL21 and CCR7 were higher in the bone marrow involvement group compared with the non-involvement group. Furthermore, EMT-related markers, vimentin and N-cadherin, were elevated, while E-cadherin was reduced in the bone marrow involvement group compared with the non-involvement group ([91]figure 3B). These findings suggest that abnormally high expression of CCL21 and CCR7 is associated with malignant bone marrow invasion in patients with ND B-NHL. Figure 3. The abnormally high expression of CCR7 is associated with bone marrow invasion and poor prognosis of patients with B-NHL. (A) The expression of CCL21/CCL19/CCR7 in serial sections of lymph node tissue from patients with reactive lymph node hyperplasia, DLBCL, and Burkitt lymphoma was assessed by Immunohistochemistry, scale bar, 20 µm (×40). (B) The expression of CCL21/CCR7 and epithelial-mesenchymal transition-related indicators vimentin, N-cadherin, E-cadherin in bone marrow biopsy samples of patients with B-NHL with and without bone marrow invasion was detected by Immunohistochemistry, scale bar, 50 µm (×20). The expression of CCR7 (C) and CCL21 (D) in peripheral blood of healthy controls (HC, n=9) and patients with B-NHL with newly diagnosed (ND, n=15), completely remission (CR, n=10) and relapsed/refractory (r/r, n=23) was analyzed by qRT-PCR. (E) The percentage of CCR7+cells on the peripheral blood B lymphocyte membrane surface in HC (n=29), ND (n=32), CR (n=39) and r/r (n=41) groups was detected by flow cytometry. (F) The percentage of CCR7+cells on the membrane surface of abnormal B lymphocytes and T lymphocytes in the peripheral blood of patients with B-NHL (n=97) was detected by flow cytometry. (G) The receiver operating characteristic curve for CCR7 expression in patients with B-NHL (n=112). (H) Kaplan-Meier survival curve of OS according to CCR7 expression in B lymphocytes of patients with B-NHL (n=112). (I) Kaplan-Meier survival curve of PFS according to CCR7 expression in B lymphocytes of patients with B-NHL (n=82). *p<0.05, ***p<0.001, ****p<0.0001. CCR7, C-C chemokine receptor 7; B-NHL, B-cell non-Hodgkin’s lymphoma; CCL21, C-C chemokine ligand 21; CCL19, C-C chemokine ligand 19; BMI, bone marrow involvement; qRT-PCR, quantitative Reverse Transcription Polymerase Chain Reaction; AUC, area under the curve; OS, overall survival; PFS, progression-free survival. [92]Figure 3 [93]Open in a new tab To further analyze the differences in CCR7 expression among patients with B-NHL at different disease stages, quantitative Reverse Transcription Polymerase Chain Reaction (qRT-PCR) and flow cytometry were used to detect CCR7 levels in peripheral blood from patients with ND, complete remission (CR), relapsed or refractory (r/r) B-NHL, and healthy individuals. The levels of CCL21/CCR7 in the ND and r/r groups were significantly higher than those in healthy individuals and the CR group. Simultaneously, the expression of CCR7 in the r/r group was markedly elevated compared with the ND group ([94]figure 3C,D). Flow Cytometry (FCM) results revealed that the expression of CCR7 on B lymphocytes in the peripheral blood of patients with B-NHL was significantly higher than that in healthy controls. Additionally, r/r patients also exhibited significantly higher CCR7 levels compared with ND patients ([95]figure 3E). Further analysis of CCR7 expression on the membrane surface of different cell subpopulations in patients with B-NHL revealed that CCR7 was scarcely expressed in the monocytes and granulocytes, but was significantly expressed in lymphocytes. Notably, it was significantly expressed in the B lymphocyte subpopulation, showing markedly higher levels than in T lymphocyte subsets ([96]figure 3F). To further evaluate the association between CCR7 expression and survival in patients with B-NHL, we followed the survival status of 112 patients with B-NHL. A receiver operating characteristic curve was generated based on the percentage of CD197+B lymphocytes in patients with B-NHL. The area under the curve was 0.8876 (95% (CI): 0.8269 to 0.9483, p<0.0001) and the cut-off value is 88.24% ([97]figure 3G). The relationship between CCR7 expression in peripheral blood and survival of patients with B-NHL was further evaluated. Patients with B-NHL were divided into a high CCR7 expression group and a low expression group according to the cut-off value. OS (median survival time 6 vs 33 months, p<0.001) and PFS (median survival time 4 months vs 6 months, p<0.001) of patients with B-NHL in the CCR7 high expression group were significantly shorter than those in the low expression group by Kaplan-Meier survival analysis (p<0.001) ([98]figure 3H,I), suggesting that patients with B-NHL with low CCR7 expression have a significant survival advantage over patients with high expression levels. Patients were further stratified into groups based on relevant indicators that may affect survival, including gender, age, white blood cell count, hemoglobin level, platelet count, International Prognostic Index, Eastern Cooperative Oncology Group scores and CCR7 expression. COX multivariate regression was used to analyze the association between the above indicators and survival time, and it found that CCR7 (p=0.032) is a prognostic indicator for patients with B-NHL ([99]online supplemental table S2). High CCR7 expression is strongly associated with poor prognosis in patients with B-NHL, indicating it as a potential prognostic marker. CCR7 promoted the invasion and migration of chemotherapy-induced senescent B-NHL cells This study investigated seven hematological malignancy cell lines, comprising two leukemia cell lines (HL-60 and MOLM-13), two Burkitt lymphoma cell lines (Raji and Namalwa), and three DLBCL cell lines (SU-DHL-2, SU-DHL-4, and OCI-LY3), suggested that CCR7 expression in Burkitt lymphoma and DLBCL was significantly higher than that in the leukemia group ([100]online supplemental figure S3A,B). B-NHL cells were stimulated in vitro with various concentrations of the exogenous ligand CCL21. It was observed that at concentrations of 50 ng/mL and 100 ng/mL, CCL21 significantly enhanced the migration and invasion abilities of Raji and SU-DHL-2 cells, respectively ([101]online supplemental figure S3C,D). We analyzed the impact of CCR7 on the migration and invasion abilities of Raji and SU-DHL-2 cells induced by CCL21 using CAP-100, a novel humanized IgG1-blocking monoclonal antibody directed against the ligand-binding site of hCCR7. The results revealed that CCL21 induces migration and invasion of B-NHL cells through CCR7 ([102]online supplemental figure S3E–G). Lentiviral-mediated short hairpin RNA (shRNA) was employed to interfere with CCR7 expression in Raji cells. It was observed that the virus successfully transfected B-NHL cells ([103]figure 4A), with the sh2 groups having the strongest inhibition rate in Raji cells ([104]figure 4B). The cell proliferation ability was reduced in the shCCR7 group compared with the NC group ([105]figure 4C). Inducing the NC group with 40 nmol DOX resulted in a significant increase in the proportion of cells in G0/G1 phase, while the proportion of cells in the S phase and G2/M phase decreased, suggesting that the cell cycle was arrested in the G0/G1 phase ([106]figure 4D), while the shCCR7 group did not show similar results. Using 40 nmol DOX to induce NC and shCCR7 groups, it enhanced the invasion and migration ability of Raji cells ([107]figure 4E,F). However, compared with the NC group, the invasion and migration ability of cells in the shCCR7 group significantly reduced ([108]figure 4E,F). Similar results were obtained in SU-DHL-2 cells ([109]figure 4G–L). These findings suggest that inhibiting CCR7 expression significantly reduces the invasion and migration of B-NHL cells, which are induced by sublethal doses of the chemotherapeutic DOX. Figure 4. Knockdown of CCR7 suppresses the invasive and migratory capabilities induced by sublethal doses of DOX in B-cell non-Hodgkin’s lymphoma. (A) Fluorescence images of stably transfected CCR7 shRNA in Raji cells, with red fluorescence indicating virus-infected cells. (B) Verify the transfection efficiency of stable CCR7 shRNA in Raji cells using qRT-PCR. (C) CCK-8 detection of the proliferation ability of Raji cells stably transfected with CCR7 shRNA. (D) The effect of CCR7 expression on the cell cycle distribution of stable CCR7 shRNA in Raji cells analyzed by flow cytometry. The effect of CCR7 expression on the migration (E) and invasion (F) ability of sublethal doses of DOX-induced Raji cells stably transfected with CCR7 shRNA detected by Transwell (×20). (G) Fluorescence images of stably transfected CCR7 shRNA in SU-DHL-2 cells, with red fluorescence indicating virus-infected cells. (H) Verify the transfection efficiency of stable CCR7 shRNA in SU-DHL-2 cells using qRT-PCR. (I) CCK-8 detection of the proliferation ability of SU-DHL-2 cells stably transfected with CCR7 shRNA. The effect of CCR7 expression on the migration (J) and invasion (K) ability of sublethal doses of DOX-induced SU-DHL-2 cells stably transfected with CCR7 shRNA detected by Transwell (×20). *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001. CCR7, C-C chemokine receptor 7; DOX, doxorubicin; mRNA, messenger RNA; shRNA, short hairpin RNA; qRT-PCR, quantitative Reverse Transcription Polymerase Chain Reaction; CCK8, Cell Counting Kit-8. [110]Figure 4 [111]Open in a new tab Raji cells were infected with lentiviral vectors and Green Fluorescent Protein (GFP) fluorescence expression was successfully observed 72 hours after viral infection ([112]figure 5A). qRT-PCR validation demonstrated a significant increase in CCR7 expression in the overexpression group compared with the empty vector group ([113]figure 5B). Using DOX to induce Vec and overexpression groups, respectively, the proliferation ability of CCR7 overexpressed Raji cells was enhanced ([114]figure 5C). DOX-induced Raji cells showed a significant increase in the proportion of G1 phase cells compared with untreated cells. This was coupled with a significant reduction in the percentage of cells in the G2 phase observed in both DOX-induced vector and overexpression groups. Notably, the proportion of G1 phase cells in the overexpression induction group was significantly higher than that in the vector induction group ([115]figure 5D). In stably transfected Raji cells, the CCR7 overexpression group showed significantly enhanced migration and invasion abilities after DOX induction, compared with the uninduced group or the induced or uninduced empty plasmid group ([116]figure 5E,F). Stable transfection of CCR7 overexpression plasmids into SU-DHL-2 cells ([117]figure 5G,H) did not show significant differences in cell proliferation ability among the groups ([118]figure 5I). The results for cell cycle, migration, and invasion were similar to those observed in Raji cells ([119]figure 5J,K). High expression of CCR7 enhances the invasive and migratory capabilities induced by sublethal doses of chemotherapeutics DOX in B-NHL cells. Figure 5. High expression of CCR7 enhances the invasive and migratory capabilities induced by sublethal doses of DOX in B-cell non-Hodgkin’s lymphoma. (A) Fluorescence images of stably transfected human CCR7 lentivirus in Raji cells, with red fluorescence indicating virus-infected cells. (B) Verify the transfection efficiency of stable transfection CCR7 lentivirus in Raji cells using qRT-PCR. (C) CCK-8 detection of the proliferation ability of Raji cells stably transfected with CCR7 lentivirus. (D) The effect of CCR7 expression on the cell cycle distribution of sublethal doses of DOX-induced Raji cells stably transfected with CCR7 lentivirus analyzed by flow cytometry. The effect of CCR7 expression on the migration (E) and invasion (F) ability of sublethal doses of DOX-induced Raji cells stably transfected with CCR7 lentivirus detected by Transwell (×20). (G) Fluorescence images of stably transfected human CCR7 lentivirus in SU-DHL-2 cells, with red fluorescence indicating virus-infected cells. (H) Verify the transfection efficiency of stable transfection CCR7 lentivirus in SU-DHL-2 cells using qRT-PCR. (I) CCK-8 detection of the proliferation ability of SU-DHL-2 cells stably transfected with CCR7 lentivirus. (J) The effect of CCR7 expression on the cell cycle distribution of sublethal doses of DOX-induced SU-DHL-2 cells stably transfected with CCR7 lentivirus analyzed by flow cytometry. The effect of CCR7 expression on the migration (K) and invasion (L) ability of sublethal doses of DOX-induced SU-DHL-2 cells stably transfected with CCR7 lentivirus detected by Transwell (×20). *p<0.05, **p<0.01, ***p<0.01, ****p<0.0001.CCR7, C-C chemokine receptor 7; DOX, doxorubicin; mRNA, messenger RNA; qRT-PCR, quantitative Reverse Transcription Polymerase Chain Reaction; CCK8, Cell Counting Kit-8. [120]Figure 5 [121]Open in a new tab CCR7 enhanced the stemness characteristics of senescent B-NHL cells by activating ARHGAP18/IKBα/p65 pathway activation To explore the effect of silencing CCR7 on TIS formation in B-NHL cells, DOX was used to induce cells in NC-CCR7 and shCCR7 groups. The proportion of SA-β-Gal positive cells in the DOX-induced group was significantly higher than in the untreated group. However, CCR7 knockdown significantly reduced SA-β-Gal-positive cells, indicating that silencing CCR7 expression in Raji cells significantly inhibited DOX-induced cellular senescence ([122]figure 6A). We detected CCR7 expression in the DOX-induced NC/shCCR7 group and found that the CCR7 expression levels corresponded with the proportion of SA-β-gal positive cells ([123]figure 6B). To further evaluate the reasons that knockdown of CCR7 inhibited the DOX-induced cellular senescence of B-NHL cells, we tested the stemness function of DOX-induced B-NHL cells. It was observed that the spheroid formation ability and colony formation ability of DOX-induced Raji cells in the shCCR7 group were significantly lower than those in the NC group, and similar results were obtained in the phenotypic stemness features test ([124]figure 6C–E). Similar results were obtained in SU-DHL-2 cells ([125]figure 6G–K). These findings indicate that silencing CCR7 expression significantly inhibited the stemness characteristics and altered the phenotype of B-NHL cells. On the contrary, upregulated expression of CCR7 promoted the stemness characteristics and phenotype of senescent B-NHL cells ([126]figure 7A–E and L–P). Figure 6. Knockdown of CCR7 inhibits the phenotypic and functional stemness features of senescent B-cell non-Hodgkin’s lymphoma cells by suppressing NF-κB pathway activation. (A) Silencing CCR7 expression in Raji cells significantly inhibits the formation of DOX-induced senescence. (B) CCR7 expression in DOX-induced Raji cells which stably transfected with shCCR7. (C) Silencing CCR7 expression significantly suppresses the stemness phenotype of senescent Raji cells. (D) Silencing CCR7 expression in Raji cells significantly inhibits the abilities of DOX-induced sphere formation. (E) Silencing CCR7 expression in Raji cells significantly inhibits the ability of DOX-induced colony formation. (F) Silencing CCR7 expression significantly inhibits the activation of NF-κB pathway in senescent Raji cells. (G) Silencing CCR7 expression in SU-DHL-2 cells significantly inhibits the formation of DOX-induced senescence. (H) CCR7 expression in DOX-induced SU-DHL-2 cells which stably transfected with shCCR7. (I) Silencing CCR7 expression significantly suppresses the stemness phenotype of senescent SU-DHL-2 cells. (J) Silencing CCR7 expression in SU-DHL-2 cells significantly inhibits the abilities of DOX-induced sphere formation. (K) Silencing CCR7 expression in SU-DHL-2 cells significantly inhibits the ability of DOX-induced colony formation. (L) Silencing CCR7 expression significantly inhibits the activation of NF-κB pathway in senescent SU-DHL-2 cells. *p<0.05, **p<0.01, ***p<0.01, ****p<0.0001. CCR7, C-C chemokine receptor 7; DOX, doxorubicin; mRNA, messenger RNA; SA-β-Gal, senescence-associated β-galactosidase; Nuclear Factor kappa B, NF-κB. [127]Figure 6 [128]Open in a new tab Figure 7. Overexpression of CCR7 enhances the phenotypic and functional stemness features of senescent B-cell non-Hodgkin’s lymphoma cells by activating NF-κB pathway activation. (A) Upregulated CCR7 expression significantly enhances the formation of DOX-induced senescence in Raji cells. (B) CCR7 expression in DOX-induced Raji cells stably transfected with CCR7 lentivirus. (C) Upregulated CCR7 expression significantly increases the stemness phenotype of senescent Raji cells. (D) Upregulated CCR7 expression significantly promotes the abilities of DOX-induced sphere formation in Raji cells. (E) Upregulated CCR7 expression significantly promotes the ability of DOX-induced colony formation in Raji cells. (F) Upregulated CCR7 expression significantly enhances the activation of Wnt/β-catenin and NF-κB pathway in senescent Raji cells. (G) Co-IP assay and immunoblotting with ARHGAP18/CCR7 antibody (or IgG) in Raji cells. (H) The effect of the IKBα inhibitor GS143 treatment on ARHGAP18 activation in DOX-induced Raji cells. (I) The effect of RhoA/ROCK inhibitor Y27632 treatment on IKBα activation in DOX-induced Raji cells. (J) The effect of IKBα inhibitor GS143 treatment on the stemness and sphere-forming ability of DOX-induced Raji cells. (K) The effect of RhoA/Rock inhibitor Y27632 treatment on sphere-forming ability of DOX-induced Raji cells. (L) Upregulated CCR7 expression significantly increases the formation of DOX-induced senescence in SU-DHL-2 cells. (M) CCR7 expression in DOX-induced SU-DHL-2 cells which stably transfected with CCR7 lentivirus. (N) Upregulated CCR7 expression significantly increases the stemness phenotype of senescent SU-DHL-2 cells. (O) Upregulated CCR7 expression significantly inhibits the abilities of DOX-induced sphere formation in SU-DHL-2 cells. (P) Upregulated CCR7 expression significantly promotes the ability of DOX-induced colony formation in SU-DHL-2. (Q) Upregulated CCR7 expression significantly enhances the activation of Wnt/β-catenin and NF-κB pathway in senescent SU-DHL-2 cells. **p<0.01, ****p<0.0001. CCR7, C-C chemokine receptor 7; DOX, doxorubicin; mRNA, messenger RNA; SA-β-Gal, senescence-associated β-galactosidase; Co-IP, Co-immunoprecipitation; NF-κB, Nuclear Factor kappa B. [129]Figure 7 [130]Open in a new tab Further analysis of the downstream pathways revealed that CCR7 significantly activated the expression of ARHGAP18 and IKBα while inhibiting p65 expression ([131]figures6F,L [132]7F,Q). Meanwhile, we found that CCR7 could significantly activate the canonical stemness pathway, Wnt/β-catenin, including the activation of Wnt1 and β-catenin ([133]figures6F,L [134]7F,Q). To further investigate the potential interaction between ARHGAP18 and CCR7, we conducted a Co-immunoprecipitation(Co-IP) assay. Both ARHGAP18 and CCR7 were identified in the input group, confirming their presence in the sample. The IgG group within the IP set exhibited no bands, ruling out non-specific binding. Conversely, the anti-CCR7 group showed reduced bands for ARHGAP18 and CCR7. Both proteins were found in the precipitated complex, providing evidence of an interaction between ARHGAP18 and CCR7 ([135]figure 7G). Next, we performed rescue experiments using the IKBα inhibitor GS143[136]^22 and the RhoA/ROCK inhibitor Y27632[137]^23 to further explore the underlying mechanisms. First, GS143 and Y27632 significantly inhibited the expression of IKBα and ARHGAP18, respectively, in a dose-dependent manner. Inhibition of the IKBα pathway did not significantly alter ARHGAP18 expression. However, when ARHGAP18 expression was significantly suppressed, the activated IKBα signaling in TIS was notably inhibited ([138]figure 7H1). Notably, inhibiting either IKBα or ARHGAP18 resulted in varying degrees of suppression of DOX-induced sphere formation in Raji cells ([139]figure 7J,K). These results indicate that CCR7 promotes the stemness characteristics and phenotype of senescent B-NHL cells by activating the ARHGAP18/IKBα/p65 signaling pathway. Discussion Cellular senescence is an essential tumor suppressive mechanism, but TIS plays an important role in drug resistance of tumor cells because it is a reversible cellular behavior.[140]^5 6 Pharmacological targeting of TIS cells could mitigate the risk of cancer recurrence and thereby improve the therapeutic efficacy of cancer chemotherapy,[141]^24 25 but is still limited by the high heterogeneity of senescent cells. Currently, the recognized senescent cell marker to identify senescent cells is the measurement of SA-β-Gal activity,[142]^26 which can reflect the increase of lysosomal quality in senescent cells. But in fact, non-senescent cells such as macrophages stain positive for SA-β-Gal, and tissue sections from preclinical mouse models and patient samples can only be detected using frozen sections, to a certain extent. This limits the future clinical application of SA-β-Gal as a specific marker of senescence.[143]^27 Except for SA-β-gal, senescent cells exhibit elevated expression of cell cycle regulators, including Cdkn2a (p16) and Cdkn1a (p21), which play roles in regulating cell cycle arrest. However, p16/p21 may not be used as a specific or sensitive indicator of cellular senescence because not all cells with elevated p16 levels are necessarily in a senescent state, and some senescent tumor cells often lack the expression of p16/p21.[144]^28 29 Therefore, due to a combination of technical issues and the complexity and high heterogeneity of senescence, no clinically universal senescence markers have yet to be truly promoted.[145]^27 Research is crucial to precisely characterize cellular senescence phenotypes and functions in different tumor types. This will help identify specific features of senescence that can be targeted to either inhibit or enhance, without adversely affecting normal cells. In the present study, we used doxorubicin to construct B-NHL-TIS, and then identified the distinctive chemotherapy-induced senescent cell subpopulation using ScRNA-seq analysis. Compared with the non-senescent and autonomous senescent cell subpopulations, the TIS cell subpopulation not only characterized by typical cell cycle proteins deficiency and cellular senescence signaling proteins enrichment, but also had a high expression of chemokine family and the anti-apoptotic BCL2 family and the NF-κB family. The chemokine receptor CCR7 exhibited significant differential expression in the senescent cell population and the non-senescent cell population. This is the first ScRNA-seq report on CCR7 is uniquely overexpressed in chemotherapy-induced senescent B-NHL subpopulation, which may be crucial for understanding the molecular mechanism of TIS and chemoresistance in B-NHL. CCR7 is an important signaling molecule in tumorigenesis and tumor development.[146]1320 30,[147]35 In our study, the expression levels of CCR7 and CCL21 were notably higher in patients with aggressive B-NHL compared with those with RLN. However, CCL19 exhibited almost no expression in the lymph nodes of patients with most ND DLBCL and Burkitt lymphoma. A recent large-scale multicenter survival study on patients with cancer found that in breast and ovarian cancers, patients with high CCL19 expression had a significantly better prognosis compared with those with low CCL19 expression. Furthermore, inhibition of CCL19 expression led to significant cancer progression and increased apoptotic activity,[148]^36 supporting our findings on CCL19 expression in patients with aggressive B-NHL. Further exploration reveals that CCL21 significantly enhances DOX-induced migration and invasion of B-NHL cells through interaction with CCR7. When CCR7 was blocked by CAP-100 (a novel humanized IgG1 blocking monoclonal antibody),[149]^37 38 the CCL21-induced migration and invasion abilities were significantly inhibited. Additionally, CCR7 expression was significantly elevated in patients with r/r B-NHL compared with ND patients, suggesting its potential role in disease progression. High CCR7 expression is an independent prognostic factor for patients with B-NHL, consistent with findings reported in primary DLBCL.[150]^39 We analyzed CCR7 expression across various hematological malignancies and observed significant upregulation at both the transcriptional and post-translational levels in DLBCL and Burkitt lymphoma. However, CCR7 expression was low in two leukemia cell lines, indicating that CCR7 may serve as a differential target in B-cell lymphoma subtypes as opposed to leukemia subtypes. Currently, chemotherapy is the primary treatment for B-NHL. Our findings suggest that CCR7 is not only an independent prognostic factor but also a novel therapeutic target for patients with r/r B-NHL. Targeting CCR7 could enable the development of new treatment strategies combining chemotherapy with agents that exploit senescence-inducing mechanisms, offering significant clinical potential. In the pseudotime analysis, we found that the TIS population was distributed at the earliest stage of the pseudotime trajectory, suggesting that senescent B-NHL cells possess self-renewal capability and multipotent differentiation potential. We further investigated whether chemotherapy-induced senescence could enhance the stemness of B-NHL cells and the role that CCR7 plays in this process. Senescent B-NHL cells exhibited typical stem cell characteristics, including the upregulation of stem cell molecular markers, enhanced tumorsphere and colony formation capabilities, and increased invasion and migration abilities. This is consistent with reports that chemotherapy-induced senescence enhances stem cell characteristics and increases tumor initiation potential,[151]^11 promoting stemness and tumorigenicity.[152]^40 Furthermore, the formation of B-NHL-TIS is strictly regulated by CCR7. Targeted inhibition of CCR7 not only suppresses chemotherapy-induced senescence in lymphoma cells but also significantly inhibits the acquisition of phenotypic and functional stemness features. Mechanistically, CCR7 enhances stemness in oral squamous cell carcinoma and MMTV-PyMT mammary cancer cells by activating the JAK/STAT[153]^20 and Notch[154]^19 pathways. Additionally, Wnt signaling plays a central role in the renewal of stem cells and is essential for the development of cancer stem cells in hematologic malignancies.[155]^11 41 42 CCR7 activation leads to a significant enrichment of the wnt1 and β-catenin, while targeted inhibition of CCR7 restricts the expression of wnt1 and β-catenin, supporting the typical Wnt/β-catenin signaling pathway enrichment in B-NHL-TIS. We provide the first direct evidence of NFKBIA and NF-κB pathway activation in the CCR7+B-NHL-TIS population, while NF-κB is significantly inhibited in non-senescent, proliferating B-NHL cells. Second, by further blocking CCR7 and screening for senescence-related proteins and the NF-κB pathway, we found a significant inhibition of RhoA GTPase-activating protein ARHGAP18 and NFKBIA-encoded protein IKBα expression. They exhibited a synergistic trend in the chemotherapy-induced senescent B-NHL cells stage regulated by CCR7. When the TIS process was reversed, ARHGAP18 and IKBα expression was significantly inhibited, while the expression of NF-κB p65 was the opposite. ARHGAP18 is a GTPase-activating protein for RhoA that controls cell migration, proliferation, and survival,[156]^43 and regulates stress-induced senescence in endothelial cells via p16 and retinoblastoma activation.[157]^44 We previously reported that ARHGAP18 is significantly enriched in B-NHL-TIS.[158]^21 The regulatory relationship between CCR7 and ARHGAP18 is unknown, and our results suggest that activated CCR7 binds to its ligand, it is internalized and activates ARHGAP18, which in turn binds to RhoA to promote its inactivation, then acts on the NF-κB pathway[159]^45 as reported. It is worth noting that the loss of RhoA function reported by Hiroki Sasaki et al leads to increased invasiveness and stemness in gastric cancer,[160]^46 which is consistent with our observation of enhanced ARHGAP and increased stemness in lymphoma cells. NF-κB signaling is chronically activated during senescence to promote the SASP.[161]^47 48 NFKBIA is transcriptionally induced as part of a negative feedback mechanism to regulate NF-κB signaling.[162]^49 When NF-κB is activated, one of the target genes it upregulates is NFKBIA, which produces IKBα binding to the p65 DNA-binding sites to inhibit further NF-κB activation.[163]^50 In senescent cells, the SASP lead to persistent, low-level NF-κB activation and constant expression of IKBα to prevent excessive activation, resulting in both NF-κB and IKBα being elevated simultaneously. The activation of CCR7 can enhance NF-κB signaling, leading to the upregulation of cancer stem cell markers such as Wnt1/β-catenin signaling, as well as CD34 and CD44, thereby promoting the stemness of B-NHL-TIS. Herein, our studies provide evidences for a key role of CCR7 in B-NHL-TIS, contributing to the reprogramming of cellular senescence and promote the senescence-associated stemness of B-NHL cells, which has not been reported yet. The activation of the CCR7/ARHGAP18/IKBα signaling cascade is one of the crucial mechanisms by which TIS lymphoma acquires stemness features. In conclusion, this study demonstrated that TIS promotes the stemness of B-NHL cells via CCR7/ARHGAP18/IKBα signaling activation, which might elucidate a potential mechanism in chemoresistance for B-NHL. Targeting the signaling might overcome the chemoresistance of B-NHL cells by inhibiting stemness acquisition and maintenance. supplementary material online supplemental file 1 [164]jitc-13-1-s001.pdf^ (877KB, pdf) DOI: 10.1136/jitc-2024-009356 online supplemental file 2 [165]jitc-13-1-s002.pdf^ (14.4MB, pdf) DOI: 10.1136/jitc-2024-009356 Acknowledgements