Abstract Esophageal squamous cell carcinoma (ESCC) is associated with a highly immunosuppressive tumor microenvironment (TME), driven in part by cancer-associated fibroblasts (CAFs) that promote immune evasion through the secretion of CXCL12. CXCL12 interacts with the CXCR4 receptor on immune cells, disrupting CD8^+ T cell migration and anti-tumor function. To address this, we developed an innovative siRNA-based therapeutic approach targeting CXCL12 in CAFs. Using lipid nanocarriers (LNCs) as delivery vehicles, we engineered LNCs@si-CXCL12 nanoparticles to specifically silence CXCL12 expression in CAFs. In vitro studies demonstrated that LNCs@si-CXCL12 restored CD8^+ T cell migration and inhibited ESCC cell proliferation and migration. In vivo experiments in a spontaneous ESCC mouse model showed that CXCL12 silencing through nanoparticle delivery significantly reduced tumor growth, enhanced CD8^+ T cell-mediated tumoricidal activity, and improved overall survival. These findings highlight the potential of siRNA-loaded nanoparticles targeting CXCL12 as a novel therapeutic strategy to reprogram the immunosuppressive TME and enhance immune responses in ESCC. This approach provides a promising avenue for improving treatment outcomes and overcoming immune evasion in ESCC. Graphical abstract [54]graphic file with name 12951_2025_3476_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-025-03476-x. Keywords: Esophageal squamous cell carcinoma, Tumor microenvironment, Cancer-associated fibroblasts, CXCL12, SiRNA nanoparticles Introduction Esophageal squamous cell carcinoma (ESCC) is one of the most common malignant tumors in the digestive system, involving multiple cell types and intricate molecular pathways in its pathogenesis. Research has revealed the diverse impacts of genetic factors, environmental factors, and lifestyle on ESCC development. For instance, mutations in genes such as TP53, CDKN2A, PIK3CA, and MYC lead to cell apoptosis and proliferation imbalances, thereby promoting tumor formation [[55]1–[56]4]. Additionally, environmental factors like smoking and alcohol consumption, along with unhealthy dietary habits and chronic gastroesophageal reflux, are significant contributors to the increased incidence of ESCC [[57]5–[58]7]. Cancer-associated fibroblasts (CAFs) play a crucial role in ESCC development. They activate the STAT3 signaling pathway by secreting cytokines like IL-6, promoting tumor growth and infiltration. They also enhance the migration and invasiveness of tumor cells by secreting CXCL12, which binds to CXCR4 [[59]8–[60]10]. Furthermore, CXCL12 can activate signaling pathways, including PI3K/AKT and MAPK, further promoting tumor growth and infiltration [[61]11]. Tumor fibrosis plays a key role in the occurrence and development of cancer, while the application of nanocarriers in gene delivery provides new ideas and methods to improve the success rate of tumor treatment. Tumor fibrosis refers to the loss of normal structure in the connective tissue surrounding tumor cells in the tumor microenvironment (TME), resulting in the formation of excessive collagen and other matrix components. This process is usually caused by the combined actions of cancer cells, CAFs, immune cells, and other components of the TME. Fibrosis increases the mechanical strength of the tumor, providing a microenvironment that supports tumor cell growth, migration, and metastasis. Fibroblasts in the tumor stroma secrete growth factors that stimulate tumor cell proliferation. Additionally, tumor fibrosis often leads to limited drug uptake and distribution, affecting the efficacy of systemic therapies. The density of the tumor stroma may also shield against radiotherapy or chemotherapy, thus reducing treatment efficacy. Furthermore, the fibrotic TME alters the infiltration pattern of immune cells, impairing effective anti-tumor immune responses and allowing the tumor to escape immune surveillance. Recent studies have suggested that the extent of tumor fibrosis can serve as a prognostic factor, helping to predict disease progression and treatment response in patients [[62]12–[63]14]. Nanocarriers, which are micron- or nanometer-sized platforms used for drug or gene delivery, can effectively improve therapeutic efficiency. In cancer therapy, especially in gene delivery, nanocarriers enhance the delivery efficiency of genes by selectively targeting tumor cells via surface-modified ligands, while reducing toxicity to normal cells [[64]15]. Some gene delivery systems use nanocarriers to improve the solubility of water-insoluble gene drugs, thereby enhancing their in vivo stability and bioavailability. The design of nanocarriers enables controlled release of drugs or genes, extending their action time in the TME and improving therapeutic efficacy. Additionally, nanocarriers help overcome the biological barriers of the cell membrane, improving the efficiency of gene entry into cells, which is critical for most gene therapy approaches [[65]16–[66]18]. The TME plays a crucial role in cancer progression and metastasis by influencing various cellular processes such as proliferation, invasion, and immune evasion [[67]19]. Chemokines, such as CXCL12 (also known as stromal cell-derived factor 1, SDF-1), are key components of the TME and regulate interactions between cancer cells and stromal cells. It is well-known that CXCL12 promotes tumor growth, angiogenesis, and metastasis by binding to its receptor CXCR4, which is often overexpressed in various cancers. Therapeutic strategies targeting the CXCL12/CXCR4 signaling pathway have become a promising approach in cancer treatment [[68]20, [69]21]. Using small interfering RNA (siRNA) to silence CXCL12 expression is one such approach, similar to shRNA (short hairpin RNA). siRNA technology can specifically and effectively silence genes by targeting mRNA degradation, thereby reducing the expression of the corresponding proteins. In the context of the TME, silencing CXCL12 with siRNA can disrupt the CXCL12/CXCR4 axis, reducing the signaling through this pathway. This disruption can have a profound impact on tumor progression, as the CXCL12/CXCR4 signaling pathway is involved in processes such as tumor cell migration, invasion, and metastasis. By silencing CXCL12 expression in CAFs and other stromal cells, the interaction between cancer cells and the microenvironment can be altered, potentially reducing tumor invasiveness and metastatic potential [[70]22, [71]23]. Studies have shown that siRNA-mediated CXCL12 silencing targeting the CXCL12/CXCR4 pathway can inhibit tumor growth and metastasis in preclinical models [[72]24, [73]25]. Additionally, this method can enhance the efficacy of other anti-cancer therapies by making tumor cells more sensitive to treatment. However, further studies are needed to comprehensively understand the impact of CXCL12 silencing on the TME and optimize the use of siRNA therapy in cancer treatment. Nanotechnology targeting CAFs provides an innovative therapeutic strategy, precisely delivering therapeutic drugs to key TME cell types [[74]26]. For example, a tailored PLGA nanoparticle formulation can simultaneously deliver chemotherapy drugs and siRNA targeting HGF, effectively transforming activated CAFs into a quiescent state, thus significantly inhibiting tumor proliferation, migration, and invasion and improving tumor permeability [[75]27]. Additionally, Bingyuan Fei and colleagues investigated a novel nanomedicine that modulates CAFs, normalizes tumor vasculature, and sequentially sensitizes photodynamic therapy (PDT), effectively enhancing drug accumulation in the tumor, alleviating hypoxia in the TME, and ultimately boosting the immune response induced by PDT [[76]28]. Therefore, based on the above background, this research project will comprehensively analyze the interactions between CAFs and ESCC cells using transcriptome and single-cell RNA-seq technologies. The project aims to design nano-spheres targeting CAFs to deliver si-CXCL12, silencing CXCL12 gene expression to modulate the TME and enhance the anti-tumor response of CD8^+ T cells. Through this highly precise nano-delivery system, we seek to uncover new mechanisms by which CAFs regulate tumor growth and immune evasion in ESCC and provide potential new strategies for clinical therapy. Materials and methods Clinical sample collection This study adhered to the ethical principles of the Helsinki Declaration and was approved by our hospital's Clinical Ethics Committee (No. 2022-197-01). Written informed consent was obtained from all registered patients. Ten ESCC patients without prior drug treatment or other underlying diseases were selected at our institution for the surgical acquisition of cancer tissue and adjacent normal tissue [[77]29, [78]30]. The ESCC cancer tissue and corresponding adjacent normal tissue were fixed in formalin and embedded in paraffin for histopathological diagnosis. All tissue sections were evaluated by experienced pathologists and patients'TNM staging was classified according to the seventh edition TNM staging standard published by the AJCC [[79]31]. The clinical and pathological characteristics of ESCC patients are presented in Table S1. Regular follow-ups were conducted for all patients to obtain their clinical outcomes. Bulk RNA-seq analysis Eighty-two ESCC bulk RNA-seq data were downloaded from The Cancer Genome Atlas (TCGA) database ([80]https://portal.gdc.cancer.gov/). The tumor cell composition in the tissue was analyzed using the R language packages “CIBERSORT” (immunocyte proportions), “MCPcounter” (cell abundance scores), and “xCell” (cell enrichment scores). Samples were grouped into high and low Fibroblasts based on the median calculation results for subsequent gene differential analysis, taking the intersection [[81]32]. 10 × genomics Fresh tumor tissues and adjacent non-tumor tissues from three ESCC patients were collected for single-cell RNA sequencing (scRNA-seq). The samples were washed with 0.04% PBS-BSA, minced with a surgical blade, and digested with papain enzyme (P4762, Sigma, Germany) at 37 °C for 30 min. Following digestion, the cell pellets were resuspended in a DNase-inhibitor solution. The suspended single cells were filtered through a pre-wetted 40 μm cell strainer with HBSS, and then centrifuged at 300×g for 5 min at 4 °C. Subsequently, the cell pellets were resuspended in 10 ml of 30% Percoll in PBS, centrifuged at 700×g for 10 min. After removing the surface debris, the cells were resuspended in FACS buffer (PBS, 1% BSA) and stained with Zombie NIR Fixable Viability Kit (423105, BioLegend, USA) for viability assessment. FACS sorting was performed using BD FACS Aria II cell sorter, and the sorted cells were collected in a capture medium (PBS containing 0.04% BSA). Cell counting was done using a CountessTMII automated cell counter with trypan blue staining. Cells with higher than 80% viability were loaded onto the 10 × Genomics Chromium chip following the manufacturer’s instructions [[82]33]. Single-cell cDNA library preparation and quality control analysis of scRNA-seq data sets Gel bead-in-emulsion (GEM) was generated by mixing single-cell suspension, gel beads, and oil through the 10 × Genomics chromium controller. After droplet formation, the sample was added to PCR tubes and subjected to reverse transcription using the T100 Thermal Cycler (Bio-Rad): incubation at 53 °C for 45 min, followed by 85 °C for 5 min, and then held at 4 °C. The cDNA was synthesized, amplified, and evaluated for quality using the Agilent Bioanalyzer 2100. For library construction, P5 primers, Read 2 (reads 2 sequencing primer sites), Sample Index, and P7 primers were added. The resulting library underwent quality control before sequencing using Illumina HiSeq4000 PE125. Further analysis of scRNA-seq data was performed using 10 × Cell Ranger (version 2.2.0). Illumina sequencing machine base calling files (BCLs) were converted to generate FASTQ files, followed by alignment, filtering, barcode, and UMI counting. The small mouse reference transcriptome (mm10) was aligned using STAR (Spliced Transcripts Alignment to a Reference). Cell Ranger conducted primary quality control (QC) on the FASTQ files to produce high-quality data [[83]33]. Single-cell transcriptomic analysis The “Seurat” package in R software was utilized to analyze single-cell data, following quality control standards of nFeature_RNA > 50 & percent.mt (5. To reduce the dimensionality of the scRNA-Seq data set, principal component analysis (PCA) based on the top 1500 highly variable genes was performed. The Elbowplot function provided by the Seurat package was used to select the top 20 principal components (PCs) for downstream analysis. Clustering identification was conducted using the FindClusters function by Seurat, with a resolution set to the default value (res = 1). Subsequently, the t-SNE algorithm was applied to reduce nonlinear dimensionality of the scRNA-seq sequencing data. Seurat package was used to filter marker genes for various cell subgroups. Lastly, cell annotation was performed based on known cell lineage-specific marker genes, and cell communication analysis was carried out using the “CellChat” package [[84]34, [85]35]. Isolation and cultivation of CAFs and NFs ESCC-related CAFs and paired normal fibroblasts (NFs) were isolated from fresh tumor tissue and adjacent non-tumor tissue (at least 5 cm away from the tumor edge) of three ESCC patients. Tissues were cut into small pieces (1–2 mm^3), washed with PBS, digested with 1 mg/mL type II collagenase (1,148,090, Sigma-Aldrich, USA) at 37 °C in a 5% CO[2] environment for 2 h, followed by filtration and centrifugation to collect cell pellets. The cell pellets were resuspended in RPMI 1640 medium (12,633,012, ThermoFisher, USA) containing 10% FBS (10,099,141, ThermoFisher, USA), plated in 25 cm^2 culture flasks, and after 30 min, non-adherent cells (primarily tumor cells, as fibroblast adhesion time (< 30 min) is much shorter than tumor cells adhesion time (typically over 1 h)) were removed. Fibroblasts were identified after 2–3 passages using immunofluorescence for fibronectin and Cytokeratin to distinguish primary NF/CAF isolates (Figure S1A). Subsequently, CAF marker α-SMA and FAPα were identified using immunofluorescence (Figure S1B), resulting in uniform NFs or CAFs cells for further analysis [[86]36]. Isolation of peripheral blood mononuclear cells (PBMC) from human peripheral blood Blood samples were collected from 10 healthy voluntary blood donors, all of whom provided written informed consent and complied with the ethical principles of the Helsinki Declaration. The study was approved by the Clinical Ethics Committee of our institution (No. 2022-197-01). The PBMCs were isolated using a PBMC isolation kit (P8610, Beijing Solebao Technology Co., Ltd., China). Fresh anticoagulated whole blood supplemented with 20 U/mL heparin (H8060, Beijing Solebao Technology Co., Ltd., China) was diluted with an equal volume of PBS. The diluted blood was layered onto the separation solution in a centrifuge tube, ensuring a clear interface between the two liquids. The tube was centrifuged at room temperature at 1000×g for 30 min, resulting in visible layering. The white membrane layer containing cells was carefully collected and transferred to a clean 15 mL centrifuge tube. The cells were washed with 10 mL PBS, centrifuged at 250×g for 10 min, the supernatant was discarded, and the cells were resuspended in 5 mL PBS and centrifuged at 250×g for another 10 min. The supernatant was removed, and the cells were resuspended for further use. CD8 T cells were isolated from PBMC using CD8 MicroBeads (130-045-201, Miltenyi Biotec, Germany). The purity of CD8 T cells was confirmed to be 95% using anti-CD3 cell antibody (981,004, Biolegend, USA), with the Jurkat cell line (iCell, h117) serving as a control. Jurkat is a well-known cell line with a high frequency of CD88^+ T cells, and anti-CD8 cell antibody (344,722, Biolegend, USA) with a flow cytometer (Figure S2) [[87]37]. All experiments were carried out with CD8 T cells at a concentration of 1.5 × 10^6 cells/mL in X-VIVO 15 medium (04-418Q, LONZA, Switzerland) containing 10% FBS. The cells were stimulated with 100 IU/mL IL-2 (130-097-743, Miltenyi Biotec, Germany) and 10 μL/mL CD3/CD28 T cell activator (11131D, Thermo Fisher Scientific, USA) for 48 h [[88]38]. Cell culture and lentiviral transduction The human ESCC cell line TE-1 was purchased from Shanghai iCell (iCell-h211, Shanghai, China), and Het-1A was obtained from ATCC (CRL-2692). The cells were cultured in RPMI 1640 medium (11,875,119, Thermo Fisher, USA) supplemented with 10% fetal bovine serum and 1% penicillin–streptomycin (15,140,148, Gibco, CA, USA), and maintained in a humidified atmosphere with 5% CO2 at 37°C. Cell passaging was performed when the cell growth density reached approximately 80%. For the construction of lentiviral plasmids, the overexpression plasmid pHAGE-CMV-MCS-IRES-ZsGreen, helper plasmids pSPAX2 and pMD2.G, shRNA plasmid pSuper-retro-puro, and helper plasmids gag/pol, VSVG were co-transfected into 293 T cells (CL-0005, Wuhan PuNuosi Life Technology Co., Ltd., Hubei, China) using Lipo2000 (11,668,500, Invitrogen, USA). The cell culture supernatant was collected after 48 h, filtered through a 0.45 μm filter, and the virus was harvested by centrifugation. The concentrated virus was collected after 72 h, and the titers were determined by mixing the two batches of virus. For stable cell line construction, cells in the logarithmic growth phase were digested with trypsin, resuspended to a concentration of 5 × 10^4 cells/mL, and seeded into 6-well plates at 2 mL per well. After overnight incubation at 37 °C, cells were infected with lentivirus at a concentration of 100 μL/mL. After 48 h, the efficiency of GFP expression was observed under a fluorescence microscope. Stable cell lines were established by changing the culture medium to a complete medium containing 2 μg/mL puromycin (A1113803, Gibco, USA) 72 h post-infection and continued for 5 days. The expression levels of target genes in different groups of cells were then detected by Reverse Transcription Quantitative Polymerase Chain Reaction (RT-qPCR). CAF cell grouping: Control (no treatment group), sh-NC (sham lentivirus control group), sh-CXCL12-1 (sh-CXCL12 knockdown lentivirus group 1), sh-CXCL12-2 (sh-CXCL12 knockdown lentivirus group 2); CD8 T cell grouping: sh-NC (sham lentivirus control group), TE-1 + NFs (co-culture of TE-1 and NFs group), TE-1 + CAFs (co-culture of TE-1 and CAFs group), TE-1 + CAFs-sh-NC (co-culture of TE-1 and CAFs-sh-NC group), TE-1 + CAFs-sh-CXCL12 (co-culture of TE-1 and CAFs-sh-CXCL12 group); CAFs-sh-NC (CD8^+T cell and CAFs-sh-NC co-culture group), CAFs-sh-CXCL12 (CD8^+T cell and CAFs-sh-CXCL12 co-culture group). The lentivirus was purchased from Shanghai Hanheng Biotechnology Co., Ltd. (Shanghai, China), and this company completed the primer sequences and plasmid construction. All experimental procedures were performed according to the manufacturer’s instructions and repeated three times. The shRNA sequences can be found in Table S2 [[89]39]. Co-culture system of CD8^+T cells and CAFs Isolated CD8^+T cells were cultured and expanded under CD3/CD28/IL2 stimulation, while CAFs were transduced with sh-CXCL12 lentivirus. Twelve hours before co-culture with CD8 T cells, CAFs were seeded in a culture plate at a density ten times that of CD8^+T cells. After 48 h of co-culture, cells were fixed for immunofluorescence staining, and CD8^+T cells were collected for flow cytometry analysis [[90]40]. Co-culture system of CAFs and TE-1 cells To evaluate the impact of NFs and CAFs on TE-1 cells, a Transwell co-culture system was utilized. NFs or CAFs were co-cultured with TE-1 cells using 0.4 µm Transwell chambers (3412, Corning, USA). The cells were adjusted to a density of 1 × 10^5/mL, with TE-1 cells seeded in the upper chamber and 1.5 mL of CAFs or NFs in the lower chamber. Co-cultures were maintained for 48 h, following which TE-1 cells were extracted for further analysis [[91]41]. RT-qPCR Total RNA from tissues or cells was extracted using TRIZOL (15,596,018, Invitrogen, USA) following the manufacturer’s instructions, and RNA concentration was determined. For mRNA analysis, cDNA was synthesized using the reverse transcription kit (4,368,814, Applied Biosystems, USA) per the manufacturer’s protocol, with the cDNA diluted to 50 ng/μL for subsequent quantitative PCR. Each reaction consisted of 2 μL of cDNA in a total volume of 25 μL, and amplification was done using the ViiA 7 Real-Time PCR System (Applied Biosystems, USA). Primers used in this study were synthesized by the Dalian Takara Company (Table S3). Relative gene expression levels were calculated using the comparative cycle threshold method (2^−∆∆CT method) with β-actin as the internal control [[92]42], where ∆∆Ct = ∆Ct [test]–∆Ct [control], ∆Ct = Ct [target]–Ct [internal], and relative gene expression level = 2^−∆∆Ct. Each experiment was repeated three times. Enzyme-linked immunosorbent assay (ELISA) The concentrations of relevant factors in cell culture supernatants were determined using human CXCL12 ELISA kit (JL19499, Shanghai Jianglai Biological, China), human CXCL11 ELISA kit (ab289695, Abcam, UK), human CXCL9 ELISA kit (JL14160, Shanghai Jianglai Biological, China), human CXCL10 ELISA kit (JL11028, Shanghai Jianglai Biological, China), and human CCL4L2 ELISA kit (LS-C521156, LifeSpan Biosciences, USA) following the respective kit instructions [[93]43]. Western blot Cell pellets were lysed in 200 μL of Lysis Buffer in 1.5 mL Eppendorf tubes. After homogenization, the tubes were placed on ice for 10 min, centrifuged (3000×g, 4 ℃, 10 min), and the supernatant was transferred to pre-chilled Eppendorf tubes. The protein concentration was measured using the BCA assay, and the samples were denatured in Loading Buffer (99 ℃, 10 min) and stored at − 20 ℃. Proteins were separated based on molecular weight, with 20 μL of each sample loaded per well. PVDF membranes were activated with methanol and proteins were transferred using a BioRad electrophoresis system with adjusted voltage or current based on protein size. The membranes were blocked with 5% BSA at room temperature for 1 h and then incubated with respective primary antibodies against CXCL12 (ab155090, 1:5000, Abcam, UK), CXCR4 (ab181020, 1:1000, Abcam, UK), GAPDH (ab8245, 1:1000, Abcam, UK) at 4 ℃ overnight. The membranes were washed with TBST (containing 0.1% Tween 20) and incubated with secondary antibodies at room temperature for 1 h, followed by additional washes. The chemiluminescent substrate was applied, images were captured using a gel imaging system, and protein bands were quantified using AlphaView SA software (Version: 3.4.0) [[94]44]. CCK-8 experiment The proliferation of cells was assessed using a CCK-8 assay kit (40203ES60, Yeasen, Shanghai, China). Log-phase cells were taken and adjusted to a concentration of 5 × 10^4 cells/mL with a complete culture medium. The cells were seeded into a 96-well culture plate, with 100 μL of cell culture medium added to each well. The plate was then incubated in a cell culture incubator for 0 h, 24 h, 48 h, and 72 h. After discarding the supernatant, the fresh culture medium was added, followed by 10 μL of CCK-8 solution to each well. The plate was then further incubated at 37 °C for 2 h. The absorbance (A) was measured using a Multiskan FC microplate reader (51,119,080, Thermo Fisher Scientific, USA) at a wavelength of 450 nm. The proliferation rate (%) was calculated as [(A [control group]–A [experimental group])/A [control group]] × 100% [[95]45, [96]46]. Three replicate wells were set up for each group, and the experiment was repeated three times. EDU staining Cell proliferation was detected using an EDU assay kit (C0075S, purchased from Biyuntian, Shanghai, China). Cells were treated with EdU(10 μM)-containing medium at different conditions, then incubated at 37 °C for 2 h. The culture medium was removed, and the cells were fixed with 1 mL of 4% paraformaldehyde at room temperature for 15 min. After washing with PBS three times for 3 min each, the cells were permeabilized with 1 mL of 0.3% Triton X-100 in PBS at room temperature for 15 min. The cells were then stained with Hoechst 33,342 for 10 min, mounted, observed and photographed under a microscope (IX73, OLYMPUS, Japan). Five random fields were captured for each sample for counting, and the rate of EDU-positive cells was calculated [[97]47]. The experiment was repeated three times. CD8^+ T cell cytotoxicity and exhaustion detection CD8 T cells were treated differently, cultured for 4 days, and then collected. The cells were resuspended in 2% FBS in PBS and labeled with PE/Cy7-conjugated Anti-CD8 (ab217344, 1:500, Abcam, UK), Anti-Granzyme B (ab255598, 1:200, Abcam, UK), Anti-perforin (ab47226, 1:1000, Abcam, UK), Anti-PD-1 (ab300425, 1:1000, Abcam, UK), and Anti-IFN-gamma (ab224197, 1:1000, Abcam, UK) at 4 °C for 30 min. The cells were washed twice with 2% FBS in PBS and analyzed using a BD LSRFortessa flow cytometer (BD Bioscience, New Jersey, USA). Data analysis and quantification of experimental results were performed using BD FACSDiva software [[98]48–[99]51]. Flow cytometry analysis of cell apoptosis Apoptosis of cells was assessed using the Annexin V-FITC/PI double staining method. Cells were seeded at a density of 2 × 10^5 cells/well in 6-well plates. The cells were collected in a 15 mL centrifuge tube, centrifuged at 800 g, and the supernatant was discarded. The cell pellet was washed twice with PBS and resuspended in 500 μL binding buffer according to the instructions of the BD Bioscience cell apoptosis assay kit (556,547, BD Bioscience, USA). Subsequently, 5 μL of FITC and 5 μL of PI were added to the suspension, mixed well, and incubated for 15 min. Cell apoptosis was detected using a BD FACSCalibur flow cytometer [[100]52]. Annexin V-FITC positive cells were considered apoptotic cells. The experiment was repeated three times. Transwell assay for invasion and migration For invasion and migration assays, 8 μm Transwell inserts (3422, Corning, USA) in 24-well plates were used. For invasion assay, matrix gel (no need for migration assay) was added to each insert and incubated at 37 °C for 2 h before the experiment. ESCC cells were digested, washed with PBS, resuspended in serum-free culture medium, and adjusted to a cell density of 3 × 10^5 cells/mL. Three inserts were prepared for each group, with 200 μL of cell suspension added. The lower chamber was filled with 700 μL of complete culture medium, and the inserts were incubated at 37 °C with 5% CO[2] for 48 h. After fixation with methanol for 30 min, the inserts were stained with 0.05% crystal violet for 5 min, and photographed under a microscope (IX73, OLYMPUS, Japan) after removing internal cells with a cotton swab. Migration assay followed the same steps as invasion assay but without adding matrix gel. Images were processed and quantified using ImageJ software [[101]29]. The experiment was repeated three times. Immunofluorescence staining Cells were seeded in confocal cell dishes. After 24 h, the cells were washed twice with PBS, or tissue slices were fixed in 4% PFA for 15 min, washed three times with 0.3% PBST for 5 min each time. The cells were then blocked with 5% goat serum (diluted in 0.3% PBST) at room temperature for 1 h. Subsequently, the cells were incubated with diluted primary antibodies including rabbit anti- α-SMA (1:50, 14,395-1-AP, Proteintech, USA), mouse anti-FAP (1:50, sc-65398, Santa Cruz Biotechnology, USA), rabbit anti-CD8 (ab237709, 1:50, Abcam, UK), and mouse anti-GZMB (MA1-80734, 1:100, Invitrogen, USA) at room temperature for 1 h. The cells were washed three times with 0.3% PBST for 10 min each time. Then, the cells were incubated with secondary antibodies conjugated with red fluorescent protein (Cy3) including goat anti-rabbit secondary antibody (ab6939, 1:500, Abcam, UK), or with secondary antibodies conjugated with green fluorescent protein (FITC) including goat anti-mouse antibody (ab6785, 1:500, Abcam, UK), or with secondary antibodies conjugated with green fluorescent protein (FITC) including goat anti-rabbit secondary antibody (ab6717, 1:500, Abcam, UK) at room temperature for 1 h. After washing three times with 0.3% PBST for 10 min each time, the coverslips were air-dried, and the cells were observed and photographed under a microscope (IX73, OLYMPUS, Japan) [[102]44, [103]53]. 3D spheroid invasion assay: in this assay 3D MCTS developed for TE-1 cells were embedded in basement membrane extract (R&D Systems, MN, USA) to provide extracellular matrix around the tumor spheroids. The embedded spheroids were treated with ARV, VEM, and their combination, each at a concentration of 1 μM. The treatment protocol followed was similar to the 3D spheroid cell viability assay, during which tumor growth, invasion percentage, and invasive pseudopod length were analyzed. Images of the MCTS were captured every other day using the Evos imaging system (Thermo Fisher Scientific, MA, USA) [[104]54]. Serum stability test Nanoparticles were incubated with fresh human serum at a concentration of 0.5 nmol/mL for 24 h. Protein binding in human serum was measured using Sephadex G-50 size exclusion chromatography (GE Healthcare Illustra, UK). 25 µL of serum samples were tested at different time points. Subcutaneous transplantation and metastasis models in humanized CD34^+ mouse models Sixty CD34^+ humanized mice, huHSC-(M-NSG) (NM-NSG-017), were purchased from Shanghai Model Organisms Center, Inc. (Shanghai, China) and housed in SPF-grade animal facilities with a humidity of 60–65% and temperature of 22–25 °C. After acclimating for one week, the mice were observed for their health status before the experiment. In the subcutaneous transplantation experiment, CD34^+ humanized mice, huHSC-(M-NSG), were injected subcutaneously with TE-1 cells (5 mice per group). TE-1 cells (4 × 10^6 cells/mouse) were mixed with CAFs (2 × 10^5 cells/mouse), suspended in a mixture of PBS and Matrigel (1:1), and then injected subcutaneously into the right axilla of the mice. Tumor length (L), width (W), and volume (V) were measured every other day starting from day 7. Tumor volume was calculated using the formula V = (L × W^2) × 0.5, and a tumor growth curve was plotted. After 35 days, the mice were euthanized with 150 mg/kg isoflurane (Sigma-Aldrich, USA), and tumors were excised, weighed, and photographed. The subcutaneous transplantation experiment groups included: huHSC-(M-NSG) mice injected with TE-1 + NFs (co-injection of TE-1 and NFs cells), TE-1 + CAFs (co-injection of TE-1 and CAFs cells), TE-1 + CAFs-sh-NC (co-injection of TE-1 and CAFs-sh-NC cells), and TE-1 + CAFs-sh-CXCL12 (co-injection of TE-1 and CAFs-sh-CXCL12 cells) [[105]44]. In the lung metastasis model experiment (6 mice per group), TE-1 and CAFs mixed cells were co-cultured for 48 h, TE-1 stable cells were digested with trypsin to obtain single-cell suspension, and 10^6 TE-1 cells (in 100 μL PBS) were injected into the mice via tail vein. After 5 weeks, mice were euthanized with 150 mg/kg isoflurane, lungs were isolated, and lung metastases were observed through H&E staining and histological examination. This experiment protocol and animal use were approved by the Institutional Animal Care and Use Committee (No. GZRCH-AE-20211012). The lung metastasis model experiment groups included: huHSC-(M-NSG) mice injected with TE-1 + NFs (TE-1 cells co-cultured with NFs before injection), TE-1 + CAFs (TE-1 cells co-cultured with CAFs before injection), TE-1 + CAFs-sh-NC (TE-1 cells co-cultured with CAFs-sh-NC before injection), and TE-1 + CAFs-sh-CXCL12 (TE-1 cells co-cultured with CAFs-sh-CXCL12 before injection) [[106]55]. Preparation and characterization of lipid nanocarriers (LNCs)@si-CXCL12 Materials and reagents required for the preparation and characterization of LNCs@si-CXCL12 include 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-amino-2000 (DSPE-PEG2000) and DSPE-PEG2000-maleimide (DSPE-PEG-Mal) purchased from Avanti Polar Lipids; stearic acid (SA) and polyethyleneimine (PEI 1800) purchased from Aladin; 1-[3-(dimethylamino)propyl]−3-ethylcarbodiimide hydrochloride (EDC) and N-Hydroxysuccinimide (NHS) from J&K Scientific. The si-CXCL12 (5’-CCAGAGCCAACGTCAAGCATCTGAA-3’) was synthesized by GenePharma. Firstly, SA was dissolved in DMSO, EDC and NHS were added, and the mixture was stirred at room temperature for 2 h to activate the carboxyl groups on SA. Subsequently, PEI 1800 was added, and the N/P ratio was set to 15:7 [[107]56] and the stirring continued for 24 h. The residual SA was extracted using ethyl acetate, then further purification of the obtained PSA through dialysis and freeze-drying. By performing the Michael addition reaction, Anti-FAPα (PA5-99458, Invitrogen, USA) and DSPE-PEG2000-Mal were coupled in a chloroform/methanol solution, and the final product was obtained by dialysis and stored at – 20 °C. DSPE-PEG, DSPE-PEG-Anti-FAPα, and PSA were dissolved in methanol at a mass ratio 8:2:1 to form a liposomal membrane, which was hydrated with PBS and sonicated to form micelles. Finally, si-CXCL12 was mixed with the micelles at a weight ratio of 1:5, and the mixture was incubated at room temperature for 15 min to form LNCs@si-CXCL12 complex [[108]57]. Dynamic light scattering (DLS) and Zeta potential analysis were used to determine the particle size and surface charge of the self-assembled complex, and the morphology of the complex was observed under transmission electron microscopy (TEM). Stability test of LNCs@si-CXCL12 The physical stability of freshly prepared LNCs@si-CXCL12 was monitored at different time points, including particle size, zeta potential, and encapsulation efficiency (%), for a period of 3 months. During this period, LNCs@si-CXCL12 were stored at 4 °C [[109]58]. Establishment of spontaneous ESCC mouse model Beijing Vital River Laboratory Animal Technology Co., Ltd. (strain code: 219) provided female C57BL/6 mice aged 6 to 7 weeks. The mice were housed in specific pathogen-free (SPF) animal facilities with controlled room temperature and humidity at 40–60%, following a 12 h light–dark cycle. To induce spontaneous ESCC, mice were fed with drinking water containing 100 μg/mL 4-nitroquinoline 1-oxide (4-NQO, HY-33354, MCE, USA) continuously for 16 weeks. Subsequently, mice were provided with sterile pure water to promote ESCC formation. Throughout the experiment, the mice's activity, water intake behavior, body weight, and survival status were observed and recorded every two days. At weeks 16, 30, and 32 of feeding, the entire esophageal tissue of the mice was stained with H&E to detect the induced ESCC process. Additionally, total RNA was extracted from the esophageal tissue of mice fed for 28 weeks to detect the expression of CXCL12 and CXCR4. Ki67 and α-SMA staining were also performed on the esophageal tissue. At week 30 of the experiment, 4-NQO-induced ESCC mice were randomly divided into two groups and intraperitoneally injected with 200μL PBS or 10mg/kg LNCs@si-CXCL12 every 2 days for two weeks. After the treatment, the esophageal tissues of the mice were collected, photographed using a dissecting microscope, and the number and diameter of tumor lesions were recorded [[110]59]. H&E staining H&E staining was performed using the hematoxylin and eosin staining kit (C0105, Beyotime, China). Briefly, lung tissues were fixed in 10% neutral buffered formalin at 4 °C for 24 h, followed by dehydration, embedding in paraffin, sectioning, deparaffinization in xylene, gradient alcohol hydration, and distilled water rinsing. The tissues were then stained with hematoxylin for 5–10 min; excess stain was removed with deionized water for approximately 10 min, followed by eosin staining for 30 to 2 min. Subsequently, the tissues were dehydrated in gradient alcohol, cleared in xylene [[111]29, [112]30], and mounted using neutral resin or other mounting medium. Slides were observed and photographed under an inverted microscope (IX73, OLYMPUS, Japan). Immunohistochemistry staining Tissue blocks were fixed in 4% paraformaldehyde phosphate buffer solution for 12 h. Following routine deparaffinization in xylene and gradient alcohol hydration, the tissues were boiled in 0.01M citrate buffer for 15–20 min, cooled to room temperature, PBS washed, and incubated in freshly prepared 3% H[2]O[2] methanol for 10 min at room temperature. Subsequently, the tissues were incubated with 10% goat serum blocking solution (16210072, Gibco, USA) for 20 min at room temperature. Primary antibodies against CXCL12 (17402-1-AP, 1:100, Proteintech, USA), CXCR4 (ab181020, 1:100, Abcam, UK), and α-SMA (ab5694, 1:200, Abcam, UK) were added and incubated for 1 h at room temperature, followed by PBS wash. Secondary antibodies, goat anti-rabbit IgG (ab6721, 1:500, Abcam, UK) or goat anti-mouse IgG (ab6789, 1:500, Abcam, UK), were then added and incubated for 1 h at room temperature. After PBS wash, the samples were incubated with SP (streptavidin-peroxidase) for 30 min at 37 °C, followed by DAB color development for 5–10 min, stop reaction with water for 10 min, counterstained with hematoxylin for 2 min, differentiated in hydrochloric acid ethanol, water rinsed for 10 min, dehydrated, cleared, and mounted. The stained slides were observed and photographed under a microscope (IX73, OLYMPUS, Japan), processed and quantified using ImageJ. Staining intensity and extent of positive staining were scored, and the scores were summed for comparison [[113]30]. Three pathologists independently performed slide scoring, and agreement between observers was achieved. Staining intensity was graded as follows: 0 (negative), 1 (weakly positive), 2 (moderately positive), and 3 (strongly positive). The percentage of positive cells was also divided into four categories: 0–25% positive cells scored as 1; 26–50% as 2; 51–75% as 3; and more than 75% as 4. A total score of ≤ 1 defined negative and a score > 1 defined positive. Statistical analysis The research data was analyzed using SPSS software (version 21.0, IBM, USA). Continuous data were presented as mean ± standard deviation. Normality and homogeneity of variance were initially assessed. If the data met the criteria of normal distribution and homogeneity of variance, non-paired t tests were applied for intergroup comparisons, while one-way analysis of variance or repeated measures analysis of variance were used for comparisons among multiple groups. Pearson correlation analysis was utilized to examine the relationship between two variables. A significance level of P < 0.05 was considered statistically significant. Results Bioinformatics analysis reveals close association of chemokines (CXCL12, CXCL1, CXCL8, CCL20) with ESCC-CAFs The TME is mainly composed of immune cells, CAFs, endothelial cells, and extracellular matrix, playing crucial roles in promoting tumor cell proliferation, inhibiting tumor cell apoptosis, and inducing immune evasion [[114]60, [115]61]. CAFs communicate bidirectionally with tumor cells and other cells in the TME, mediating cell-derived factors, chemokines, growth factors, and exosomes. CAFs enhance tumor proliferation and induce immune escape in cancer cells, making them important anticancer targets [[116]62, [117]62, [118]63]. We downloaded ESCC bulk RNA-seq data from the TCGA database in this study. Using the cell abundance scores calculated by MCPcounter and cell enrichment scores by xCell, we categorized samples into the high Fibroblasts group and the low Fibroblasts group based on the median Fibroblasts score. Differential analysis revealed 535 upregulated genes and 338 downregulated genes by MCPcounter; 1060 upregulated genes and 262 downregulated genes by xCell (Fig. [119]1A–D). Growing evidence suggests that CAFs secreted chemokines influence tumor initiation and progression [[120]64]. We collected 49 human Chemokines [[121]65] and identified one upregulated gene (CXCL12) and three downregulated genes (CXCL1, CXCL8, CCL20) by intersecting Chemokines with MCPcounter and xCell results (Fig. [122]1E–F). Additionally, we have presented the expression of CXCL12, CXCL1, CXCL8, and CCL20 in MCPcounter and xCell analysis. The data indicated that CXCL12 expression in the Fibroblasts score High group was significantly higher than in the Fibroblasts score Low group, while CXCL1, CXCL8, and CCL20 expression levels were significantly lower in the Fibroblasts score High group. These results suggest a significant correlation between the increase in Fibroblasts score and the expression levels of these inflammation-related genes, implying that fibroblasts may play an important role in the regulation of these genes (Fig. [123]1G–H). Fig. 1. [124]Fig. 1 [125]Open in a new tab Analysis of ESCC-CAFs-related genes using bioinformatics tools. Note: A Heatmap of the top 100 differentially expressed genes related to Fibroblasts cell count in TCGA-ESCC analyzed by MCPcounter (Low: 41, High: 40); B Volcano plot of differentially expressed genes related to Fibroblasts cell count in TCGA-ESCC analyzed by MCPcounter (Red: high expression, Blue: low expression, Black: no difference); C Heatmap of the top 100 differentially expressed genes related to Fibroblasts in TCGA-ESCC analyzed by xCell (Low: 41, High: 40); D Volcano plot of differentially expressed genes related to Fibroblasts in TCGA-ESCC analyzed by xCell (Red: high expression, Blue: low expression, Black: no difference); E–F Venn diagrams showing the intersection of upregulated and downregulated genes obtained from Chemokines, MCPcounter, and xCell analyses; (G-H) Box plots of CXCL12, CXCL1, CXCL8, CCL20 expression in MCPcounter and xCell analyses (Fibroblasts score Low: n = 41, Fibroblasts score High: n = 40, * indicates p < 0.05, ** indicates p < 0.01, *** indicates p < 0.001) Based on the above results, we conducted bioinformatics analysis on ESCC bulk RNA-seq data from the TCGA database and identified four chemokines (CXCL12, CXCL1, CXCL8, CCL20) closely associated with ESCC-CAFs. Quality control, filtering, and PCA of scRNA-seq data from brain tissues of ESCC patients To further investigate the role of CAFs in the development of ESCC, we obtained a scRNA-seq dataset of ESCC patient esophageal tissues through high-throughput sequencing. Using the Seurat package for data integration, we first examined the number of genes detected in each cell (nFeature_RNA), the total mRNA molecules detected (nCount_RNA), and the percentage of mitochondrial genes (percent. mt) in the scRNA-seq data. The results showed that most cells had nFeature_RNA < 7500, nCount_RNA < 10^5, and percent.mt less than 25%. In the ESCC2 sample, percent.mt ranged from 20 to 75%, and nFeature_RNA and nCount_RNA were lower compared to other samples (Figure S3A). We filtered out low-quality cells using the criteria of nFeature_RNA > 50 and percent.mt < 10, resulting in an expression matrix containing 22,685 genes and 23,676 cells. Correlation analysis of sequencing depth showed a correlation coefficient of r = − 0.1 between nCount_RNA and percent. mt and r = 0.84 between nCount_RNA and nFeature_RNA in the filtered data, indicating good data quality for further analysis (Figure S3B). Subsequently, we performed gene expression variance analysis on the filtered cells, selecting the top 2000 highly variable genes based on variance for downstream analysis (Figure S3C). Before conducting PCA dimensionality reduction, standard preprocessing was performed. The RunPCA function performed PCA on the top 2000 highly variable genes, revealing no apparent batch effects between samples (Figure S3D). We displayed the major component genes of the first 6 PCs (Figure S4A) and generated a heatmap of the first 6 PCs using the DimHeatmap function (Figure S4B). Visualizing the significance of the PCs using the JackStrawPlot function showed that important PCs, typically with small p-values (above the dashed line), could effectively capture the information in the highly variable genes selected earlier (Figure S3E). Additionally, by combining the JackStrawPlot analysis with the ElbowPlot function, we selected the top 9 PCs with a PC p value < 0.05 for subsequent t-SNE analysis (Figure S3F). Through quality control, filtering, and PCA of scRNA-seq data from brain tissues of ESCC patients, we obtained high-quality filtered cell data and selected the top 9 PCs for further analysis. Cell clustering and analysis of cell communication unveiled the significant involvement of the CXCL12-CXCR4 signaling axis in cell interactions We first analyzed the distribution of cells across different clusters in each sample (Figure S6A), which revealed distinct patterns between Normal and ESCC samples. Next, we performed t-SNE clustering analysis based on the top 9 PCs, resulting in the classification of all cells into 21 clusters (Figure S5A). We then identified the marker genes for each cluster and visualized the top 10 most highly expressed cluster-specific marker genes (Figure S5B). Finally, we compared the cluster distribution between Normal and ESCC samples, showing that Normal cells were mainly enriched in Clusters 0, 3, 4, 6, 8, 9, 10, 11, 12, 13, 14, 15, 16, and 20, while ESCC cells were predominantly found in other clusters (Figure S5C). Using the Bioconductor/R package “SingleR” combined with the CellMarker database, we annotated the marker genes of different cell types in ESCC to the 21 cell clusters (Table S2), resulting in the annotation of 12 cell types (Mast cells, CD8^+ T cells, T cells, B cells, Fibroblasts, Macrophages, Monocyte, Stem cells, Endothelial cells, Monocyte, CAFs, Plasma cells). The quantities of different cell types between Normal and ESCC were significantly different, with Cluster#18 annotated as CAFs (Figure S5D). Comprehensive analysis based on samples, clusters, and cell types revealed the distribution of CXCL12, CXCL1, CXCL8, and CCL20 expression. Results showed that CXCL12 was more prominently expressed in Fibroblasts (Cluster 4, 14), CAFs (Cluster 18), and Endothelial cells (Cluster 13); CXCL1 was mainly expressed in CAFs (Cluster 18) and Stem cells (Cluster 10); CXCL8 exhibited high expression in Macrophages (Cluster 8), Monocyte (Cluster 9), Stem cells (Cluster 10, 16), and Mast cells (Cluster 20); while CCL20 did not show significant distribution or expression in any cells (Figure S5E; Figure S6B-E). Both CXCL12 and CXCL1 showed high expression in CAFs. Subsequently, using the"CellChat"package, cell communication analysis was conducted, where gene expression data of cells were inputted, and interactions between ligands, receptors, and their auxiliary factors were simulated to explore the interactions between CXCL12 and CXCL1 in CAFs and immune cells and receptors in the TME. Initially, the CXCL signaling pathway-mediated cell interactions showed that CAFs exhibited the strongest interactions with CD8^+ T cells, T cells, and B cells (Figure S5F; Figure S6F-G). Further calculations on all ligand-receptor-mediated cell interactions and visualization of significant ligand-receptor pairs revealed a significant interaction between CAFs and CD8^+ T cells, CAFs and T cells, as well as CAFs and B cells mediated by the CXCL12/CXCR4 ligand-receptor pair (Figure S5G). Additionally, the receptor CXCR4 was prominently distributed and expressed in CD8^+ T cells (Cluster 1, 6), T cells (Cluster 2, 11, 12), and B cells (Cluster 3, 5, 7) (Figure S5H; Figure S5E; Figure S6H), consistent with the cell communication results. Furthermore, analysis of RNA-seq data from GTEx normal esophagus samples merged with ESCC RNA-seq data revealed that the expression levels of CXCL12 and CXCR4 in the tumor group were significantly higher than in the normal group (Figure S5I). In summary, we discovered a strong interaction between the CXCL12-CXCR4 cell signaling axis in CAFs and CD8^+ T cells, T cells, and B cells in ESCC through single-cell clustering and cell communication analysis. Overexpression of CXCL12 in human ESCC-CAFs Previous studies have shown that in pancreatic ductal adenocarcinoma, activated pancreatic stellate cells reduce the migration of CD8^+ T cells towards the tumor stroma through the CXCL12/CXCR4 axis, thereby preventing CD8^+ T cells from approaching cancer cells. The secretion of CXCL12 by pancreatic stellate cells can suppress effective anti-tumor immune responses [[126]66]; in ovarian cancer, the CXCL12-CXCR4 axis has also been confirmed to have an immunosuppressive effect [[127]67]. Furthermore, CXCL12 and its receptor CXCR4 are highly expressed in ESCC [[128]68]. Based on these findings, we will further investigate how CAFs regulate CD8^+ T cells via the CXCL12/CXCR4 axis to influence the development of ESCC. Firstly, we examined the expression of CXCL12 in human ESCC-CAFs, and the results from H&E staining and immunohistochemistry showed that CXCL12 is significantly expressed in the stroma of ESCC, while there is slight expression in the stroma of normal tissues (Fig. [129]2A). Immunofluorescence staining revealed that α-SMA, a marker protein for CAFs [[130]69], was co-expressed with CXCL12 in a significantly increased number of stromal cells in ESCC tissues. The markedly enhanced co-localization of α-SMA and CXCL12 in the ESCC stroma indicates high CXCL12 expression in CAFs within the TME (Fig. [131]2B). Fig. 2. [132]Fig. 2 [133]Open in a new tab Levels of CXCL12 expression in ESCC-CAFs. Note: A Hematoxylin and eosin staining and immunohistochemical testing were conducted to assess the pathological changes of ESCC clinical tissues and the expression levels of CXCL12 (50 μm and 25 μm magnification, Normal: n = 10, Tumor: n = 10, * indicates p < 0.05 compared to the Normal group); B Immunofluorescence staining was used to detect the expression of α-SMA and CXCL12 in ESCC clinical tissues (50 μm, n = 10); C RT-qPCR was performed to detect the expression of α-SMA and FAPα in NFs and CAFs cells; D RT-qPCR was carried out to measure the levels of CXCL12 expression in NFs and CAFs cells (* indicates p < 0.05 compared to the NFs group); E ELISA was utilized to determine the levels of CXCL12 expression in NFs and CAFs cells (* indicates p < 0.05 compared to the NFs group). Each cell experiment was repeated three times To further explore the characteristics of CAFs and their interactions with tumor cells, we isolated CAFs and paired NFs from tumor tissue and adjacent non-tumor tissue, and conducted identification (Figure S1). RT-qPCR analysis of α-SMA and FAPα showed that the expression of α-SMA and FAPα is significantly increased in CAFs compared to NFs (Fig. [134]2C). RT-qPCR and ELISA results demonstrated that compared to the NFs group, the expression of CXCL12 is significantly increased in the CAFs group (Fig. [135]2D–E). These results indicate that CXCL12 is highly expressed in human ESCC-CAFs. CAFs secrete CXCL12 to inhibit the tumor-killing activity of CD8^+ T cells Previous studies have shown that CD8^+ T cells are a critical component of anti-tumor immunity, and the success of immunotherapy relies on the activation of effective cytotoxic T-cell responses [[136]38]. Therefore, we further investigated whether CAFs affect the infiltration of CD8^+ T cells by secreting CXCL12. Firstly, we constructed two CXCL12 silencing sequences and detected the silencing efficiency in CAFs cells isolated from ESCC tumors using RT-qPCR. The results showed that compared to the sh-NC group, the expression levels of CXCL12 were significantly decreased in the sh-CXCL12-1 and sh-CXCL12-2 groups (Fig. [137]3A). Since CXCL12 is a secretory protein, ELISA analysis of the supernatant of CAFs cells revealed that the CXCL12 concentration was also significantly reduced in the sh-CXCL12-1 and sh-CXCL12-2 groups compared to the sh-NC group (Fig. [138]3B), with sh-CXCL12-1 showing higher efficiency. Therefore, the sh-CXCL12-1 (sh-CXCL12) silencing sequence was used for subsequent experiments. Fig. 3. [139]Fig. 3 [140]Open in a new tab Impact of CXCL12 secreted by CAFs on the function of CD8^+ T cells. Note: A RT-qPCR was conducted to evaluate the silencing efficiency of CXCL12 in CAFs cells; B ELISA was utilized to measure the levels of CXCL12 in the supernatant of CAFs cells; C Schematic representation of co-culturing; D RT-qPCR was performed to detect the levels of CXCR4 in the co-culture system; E Western blot analysis was used to assess the expression changes of CXCL12 and CXCR4 in the co-culture system; F Immunofluorescence was performed to observe the chemoattractant capability of CAFs from various groups for CD8^+ T cells; G Flow cytometry was used to detect the changes in the expression of PD-1 and IFN-γ in CD8^+ T cells in the co-culture system, and quantitative results were obtained; H Flow cytometry was employed to measure the changes in the expression of GrzB and Perforin in CD8^+ T cells in the co-culture system; I Flow cytometry was used to quantify changes in PD-1 and IFN-γ expression in CD86^+ macrophages in the co-culture system; J Flow cytometry was also used to evaluate GrzB and Perforin levels in CD86^+ macrophages; K PD-1 and IFN-γ expression was assessed in CD56^+ NK cells; and L GrzB and Perforin expression was analyzed in CD56 + NK cells. *p < 0.05, ***p < 0.001; all cell experiments were replicated three times Next, we co-cultured CD8^+ T cells with CXCL12-silenced CAFs (GFP labeled) (Fig. [141]3C) and detected the expression of CXCL12 and its receptor CXCR4 in the co-culture system. The results showed that compared to the CAFs-sh-NC group, the expression of CXCL12 and CXCR4 was significantly reduced in the CAFs-sh-CXCL12 group, while their expression levels were significantly elevated in the oe-CXCL12 group compared to the oe-NC group (Fig. [142]3D–E). As CXCR4 is the receptor for CXCL12, CXCL12 silencing leads to the downregulation of CXCR4 due to the absence of ligand binding, consistent with previous reports [[143]22, [144]70]. Immunofluorescence results demonstrated that the number of bound CD8^+ T cells in the CAFs-sh-CXCL12 group was significantly decreased compared to the CAFs-sh-NC group, and significantly increased in the oe-CXCL12 group compared to controls (Fig. [145]3F). We hypothesized that CAFs may recruit CD8^+ T cells through secreting CXCL12 to induce T cell exhaustion, thereby promoting immunosuppression. In tumor tissues, functional exhaustion of CD8^+ T cells is characterized by decreased IFN-γ and high expression of programmed cell death protein-1 (PD-1) [[146]71, [147]72]. Flow cytometry analysis of PD-1 and IFN-γ levels in CD8^+ T cells revealed that the expression of PD-1 was significantly reduced, while IFN-γ levels were significantly increased in the CAFs-sh-CXCL12 group compared to the CAFs-sh-NC group and IFN-γ levels were upregulated in CD8^+ T cells from the CAFs-sh-CXCL12 group, whereas the opposite trend was observed in the oe-CXCL12 group (Fig. [148]3G), further validating our hypothesis. To further evaluate the impact of this phenomenon on the tumor-killing function of CD8^+ T cells, we analyzed the expression of key cytotoxic markers, Granzyme B (GrzB), and Perforin in CD8^+ T cells co-cultured with CAFs [[149]73]. The results showed that compared to the CAFs-sh-NC group, the expression of Perforin and GrzB was significantly increased in the CAFs-sh-CXCL12 group. The number of CD8^+ T cells bound to CAFs also significantly increased in the oe-CXCL12 group versus the oe-NC group, indicating a substantial enhancement in the tumor-killing function of CD8^+ T cells (Fig. [150]3H). Similar results were observed in CD86^+ macrophages and CD56^+ NK cells (Fig. [151]3I–L). In conclusion, we found that in ESCC, CAFs secreting CXCL12 inhibit the tumor-killing effect of CD8^+ T cells, CD86^+ macrophages, and CD56^+ NK cells, while silencing CXCL12 enhances their tumor-killing potential. Validation of the promotion of ESCC growth and metastasis by CAFs through CXCL12 secretion To further investigate the regulatory role of CAFs on ESCC, CAFs were co-cultured with TE-1 cells. ELISA results of TE-1 cells showed a significant increase in CXCL12 expression in the TE-1 + CAFs group compared to the TE-1 + NFs group. Additionally, there was a significant decrease in the expression of CXCL12 in the TE-1 + CAFs-sh-CXCL12 group compared to the TE-1 + CAFs-sh-NC group (Figure S7A). Subsequently, validation was conducted through CCK-8, EdU staining, flow cytometry, and Transwell assays. The results revealed that compared to the TE-1 + NFs group, the TE-1 + CAFs group demonstrated significantly increased cell viability, proliferation, migration, and invasion, with reduced apoptosis. In contrast, the TE-1 + CAFs-sh-CXCL12 group exhibited significantly decreased cell viability, proliferation, migration, and invasion, with increased apoptosis compared to the TE-1 + CAFs-sh-NC group (Figure S7B-F). These in vitro experiments indicate that CAFs play a key role in promoting the growth and metastasis of ESCC cells through CXCL12 secretion. To further confirm the effect of CAF-secreted CXCL12 on tumor growth and metastasis in vivo, TE-1 cells were mixed with NFs or CAFs and injected into the axilla of CD34^+ humanized mice. The experimental results showed a significant increase in tumor volume and weight in the TE-1 + CAFs group compared to the TE-1 + NFs group. Furthermore, there was a significant reduction in tumor volume and weight in the TE-1 + CAFs-sh-CXCL12 group compared to the TE-1 + CAFs-sh-NC group (Fig. [152]4A–C). Immunofluorescence (IF) analysis demonstrated a significant decrease in intratumoral CD8 T cells and activated CD8 T cells (GZMB +) in the TE-1 + CAFs group compared to the TE-1 + NFs group. Conversely, the TE-1 + CAFs-sh-CXCL12 group showed a significant increase in intratumoral CD8 T cells and activated CD8 T cells (GZMB +) compared to the TE-1 + CAFs-sh-NC group (Fig. [153]4D). Results from the lung metastasis model revealed a significant increase in lung metastatic foci in the TE-1 + CAFs group compared to the TE-1 + NFs group. Conversely, there was a significant reduction in lung metastatic foci in the TE-1 + CAFs-sh-CXCL12 group compared to the TE-1 + CAFs-sh-NC group (Fig. [154]4E–F). These in vivo experiments demonstrate that CAF-secreted CXCL12 significantly promotes the growth and lung metastasis of ESCC cells. Fig. 4. [155]Fig. 4 [156]Open in a new tab Effects of CAF-secreted CXCL12 on the growth and metastasis of ESCC. Note: A Subcutaneous transplantation tumor changes in humanized mice with CD34^+ cells (n = 6); B Growth curve of subcutaneous transplantation tumors in humanized mice with CD34^+ cells (n = 5); C Variation in the weight of subcutaneous transplantation tumors in humanized mice with CD34^+ cells (n = 6); D Immunofluorescence analysis of CD8 T cell and activation quantity within the transplantation tumors of humanized mice with CD34^+ cells (red: CD8, green: GZMB, yellow arrow: GZMB inside CD8 T cells, scale bar: 25 μm, n = 6); E–F Metastatic foci in the lungs of humanized mice with CD34^+ cells and corresponding H&E staining images (yellow circle: metastatic foci, scale bar: 50 μm; n = 6). (* indicates p < 0.05 compared to the TE-1 + NFs group, # indicates p < 0.05 compared to the TE-1 + CAFs-sh-NC group) Construction of nano-spheres targeting CAFs for si-CXCL12 delivery Based on in vitro and in vivo experimental results, we observed that in ESCC, CAFs not only inhibit the tumor-killing function of CD8^+ T cells by secreting CXCL12 but also promote the proliferation and metastasis of ESCC cells. In light of this, we propose developing a nano-particle strategy targeting CAFs, where these nano-particles can deliver siRNA (si-CXCL12) to specifically target and knock down the expression of CXCL12 in CAFs. Through this approach, we aim to weaken the inhibitory effect of CAFs on CD8^+ T cell function, thereby enhancing their tumor-killing activity, while inhibiting the proliferation and metastasis of ESCC cells, providing a new therapeutic strategy for ESCC treatment. The preparation process involves constructing the core structure using materials such as SA and polyethyleneimine (PEI), activating carboxyl groups and coupling with PEI through EDC and NHS chemical reactions. Subsequently, the antibody Anti-FAPα is conjugated with DSPE-PEG2000-Mal through a Michael addition reaction to form the targeting molecule. This structure is consistent with the characteristics of polymeric lipid complexes, which enable drug loading within the core or on the surface [[157]74, [158]75]. Finally, these components are mixed in specific proportions, processed with ultrasound to form nano-particles, and complexed with CXCL12 siRNA to form LNCs@si-CXCL12 nano-particles (Fig. [159]5A). The DLS measurement shows the average diameter of LNCs@si-CXCL12 as 80.5 ± 2.8 nm (Fig. [160]5B), with a Zeta potential of 23.29 mV (Fig. [161]5C). TEM reveals that the nano-spheres have a spherical morphology with uniform size distribution (Fig. [162]5D). Including DSPE-PEG2000 enhances the colloidal stability of liposomes, providing them with long-circulating stealth properties. In PBS containing 10% FBS, the size of LNCs@si-CXCL12 remains unchanged at 37 °C for 72 h (Fig. [163]5E), indicating high stability under normal physiological conditions. Additionally, the release rate of LNCs@si-CXCL12 reaches 58% at 24 h (Fig. [164]5F). Gel electrophoresis further confirmed the systemic stability of the nanoparticles (Fig. [165]5G). In 3D multicellular tumor spheroid (MCTS) co-culture assays, LNCs@si-CXCL12 treatment significantly reduced the size of spheroids derived from TE-1 cells, whereas spheroids treated with blank LNCs continued to grow over five days. Moreover, LNCs-treated spheroids exhibited smooth, regular surfaces with dense cores and minimal peripheral apoptosis, while LNCs@si-CXCL12-treated spheroids had irregular surfaces and abundant apoptotic cells at the periphery, indicating potent cytotoxicity (Fig. [166]5H). Spectral data confirmed the successful synthesis of PSA using SA and PEI 1800 (Fig. [167]5I). Long-term stability assessments over three months showed no significant changes in particle size, polydispersity index, or zeta potential (Fig. [168]5J). The encapsulation efficiency remained above 95% at the end of the observation period (Fig. [169]5K). The LNCs@si-CXCL12 nanoparticles also showed no significant changes in particle size or zeta potential during storage (Fig. [170]5L). Western blot analysis verified the successful covalent conjugation of Anti-FAPα to DSPE-PEG2000-Mal, as indicated by a clear shift in protein band size (Fig. [171]5M). Serum stability assays demonstrated that LNCs@si-CXCL12 maintained structural integrity under physiological conditions (Fig. [172]5N). Fig. 5. [173]Fig. 5 [174]Open in a new tab Preparation and characterization of LNCs@si-CXCL12. Note: A Schematic illustration of LNCs@si-CXCL12; B DLS measurement of the size distribution of LNCs@si-CXCL12; (C) Zeta potentials of LNCs and LNCs@si-CXCL12; D TEM images of LNCs and LNCs@si-CXCL12, bar = 100nm; E Stability of LNCs@si-CXCL12 at 37 ℃ in PBS containing 10% FBS; (F) Release efficiency of LNCs@si-CXCL12 within 24 h; G Gel electrophoresis verified the systemic stability of conjugated nanoparticles; H 3D tumor spheroid co-culture assay demonstrated the anti-tumor and anti-fibrotic potential of LNCs@si-CXCL12; I Spectral analysis confirmed PSA synthesis using SA and PEI 1800; J Particle size and potential statistics, K encapsulation efficiency detection; L LNCs@si-CXCL12 Particle size and potential statistics; M Western blot validated Anti-FAPα-DSPE-PEG2000-Mal conjugation; N Serum stability assay of nanoparticles. All experiments were repeated three times By successfully implementing the above experiments, we confirm that the nano-spheres LNCs@si-CXCL12 targeting CAFs exhibit good stability and uniformity under physiological conditions and show favorable targeting effects on tumor tissues. CAFs exhibit excellent targeted uptake of LNCs@si-CXCL12 with low cytotoxicity To further observe the targeted uptake of CAFs by LNCs@si-CXCL12, we co-cultured these nano-complexes with CAFs and evaluated the expression level of CXCL12 by RT-qPCR. The results showed that the expression of CXCL12 in CAFs decreased by approximately 65% after treatment, and this inhibitory effect persisted for at least 14 days (Fig. [175]6A), indicating that LNCs@si-CXCL12 effectively inhibits the transcription of CXCL12 in CAFs. Fig. 6. [176]Fig. 6 [177]Open in a new tab Targeted uptake and cytotoxicity of LNCs@si-CXCL12 by CAFs. A qRT-PCR detected CXCL12 transcript levels on days 1 and 14 following co-culture with LNCs@si-CXCL1; B and C Illustrating the uptake of LNCs@si-CXCL12 by CAFs; D Uptake by 293 T cells was minimal (scale bar = 25 μm); E–G CCK-8 assay was performed to observe the impact of LNCs@si-CXCL12 concentrations ranging from 0 to 100 mg/mL on the viability of CAFs, TE-1, and Het-1A cells. *** indicates p < 0.001 compared to the PBS/LNCs group. Cell experiments were repeated three times Given that LNCs@si-CXCL12 includes antibodies targeting the surface molecule FAPα of CAFs for targeting purposes, we used LNCs without adding FAPα antibodies as a control group. Both types of LNCs were labeled with Cy5.5 and co-cultured with CAFs and TE-1 cells. The results showed a significant increase in the uptake of FAPα-linked LNCs@si-CXCL12 in CAFs (Fig. [178]6B), while the uptake efficiency of LNCs and LNCs@si-CXCL12 was similar in TE-1 cells and 293 T cells (Fig. [179]6C–D), indicating a significant targeting effect of LNCs@si-CXCL12 on CAFs [[180]76, [181]77]. Furthermore, we assessed the cytotoxicity of LNCs@si-CXCL12. Using the CCK-8 assay, we tested the impact of LNCs@si-CXCL12 at concentrations ranging from 0 to 100 mg/mL on CAFs, TE-1 cells, and human esophageal epithelial cells Het-1A. The results showed that after 6 h of treatment, the viability of all three cell types remained above 80%. However, CAFs exhibited slightly lower cell viability compared to TE-1 and Het-1A cells, possibly due to the higher endocytic capacity of CAFs towards LNCs@si-CXCL12 (Fig. [182]6E–G). In summary, LNCs@si-CXCL12 demonstrates excellent targeting capability towards CAFs, effectively inhibiting CXCL12 expression without significantly impacting cell viability. LNCs@si-CXCL12 inhibits tumor growth in spontaneous ESCC mice and enhances immune response To further investigate whether LNCs@si-CXCL12 could attenuate the inhibitory effect of CAFs on CD8^+ T cell function, thereby enhancing their tumor-killing activity and inhibiting ESCC growth, we established a spontaneous ESCC mouse model by feeding with 4-NQO (Fig. [183]7A). LNCs@si-CXCL12 maintained stable knockdown of CXCL12 mRNA in tumor tissues up to day 14 post-treatment (Fig. [184]7B). Western blotting revealed increased expression of fibrosis-associated proteins in the ESCC group (Fig. [185]7C). At the 16th week of modeling, H&E staining results showed a relatively orderly structure of esophageal epithelial tissues without apparent abnormalities; by the 30th week, the esophageal epithelial structure became disordered, with cellular atypia, indicating the formation of intraepithelial neoplasia. However, after 2 weeks of treatment with LNCs@si-CXCL12, tissue slices in the 32nd week showed decreased neoplastic cells, effectively inhibiting tumor progression (Fig. [186]7D). Fig. 7. [187]Fig. 7 [188]Open in a new tab Therapeutic effects of LNCs@si-CXCL12 on spontaneous ESCC mice. Note: A Schematic diagram of spontaneous ESCC mouse model induced by feeding with 4-NQO; B qPCR analysis of CXCL12 mRNA expression in CAFs; C Western blot analysis of fibrosis-related proteins in mouse tumor tissues; D H&E-stained images of esophageal tissues from mice at 16, 30, and 32 weeks during modeling period (scale bar =  100 μm); E Statistical analysis of body weight changes in each group of mice; and Survival curve of mice in each group (n = 10); F Representative images of esophageal tissues from each group of mice and statistical analysis of tumor numbers; G–H Tumor volume and WBC count in each group; (I) Expression of Ki67 in tumor tissues of each group of mice (scale bar =  100 μm, magnified scale bar =  25 μm); J Immunofluorescence showing co-localization of αSMA and CXCL12 in tumor tissues (scale bar =  25 μm); K Flow cytometry analysis of IFNγ and GzmB expression in CD8^+ T cells within tumor tissues of each group of mice. ** indicates p < 0.01 compared to the PBS group, *** indicates p < 0.001 Regarding monitoring mouse weight, the treatment with LNCs@si-CXCL12 did not negatively impact mouse weight. Survival curve analysis revealed that compared to the LNCs group, LNCs@si-CXCL12 significantly prolonged the survival time of ESCC mice (p = 0.037) (Fig. [189]7E). Further analysis of esophageal tissues indicated a significant reduction in tumor numbers after treatment with LNCs@si-CXCL12 (Fig. [190]7F), along with a significant decrease in the number of Ki67-positive cells within the tumors (Fig. [191]7G). Antitumor scoring criteria confirmed the therapeutic effect of LNCs@si-CXCL12 in vivo [[192]54, [193]78]. Furthermore, immunofluorescence analysis showed that LNCs@si-CXCL12 treatment significantly reduced the number of cells co-expressing α-SMA and CXCL12 within tumor tissues, indicating targeted silencing of CXCL12 in CAFs (F[194]ig. [195]7I). Tumor weight and WBC count were also markedly decreased (Fig. [196]7G–H). Corroborated by immunohistochemistry (Figure S8A). This further confirms the inhibition of tumor development in spontaneous ESCC mice by LNCs@si-CXCL12. Furthermore, by detecting the expression of the effector molecules IFNγ and GzmB produced by CD8^+ T cells in tumor tissues, we found that LNCs@si-CXCL12 treatment significantly increased the expression of these cytokines (Fig. [197]7J, K), indicating that the treatment stimulated the immune system's response against the tumor. Moreover, LNCs@si-CXCL12 treatment markedly decreased the expression of CXCL12 and CXCR4 in tumor tissues, while VEGF levels remained unaffected (Figure S8B). Treatment with LNCs@si-CXCL12 did not cause morphological damage to vital organs such as the heart, liver, spleen, lungs, and kidneys in mice (Figure S9A), and the main biochemical indicators in mouse serum, including ALT, AST, BUN, and CR, remained within normal ranges after treatment (Figure S9B–E), indicating the safety of LNCs@si-CXCL12 treatment. To explore the underlying molecular mechanisms, transcriptome analysis was performed on tumor tissues before and after treatment. Volcano plot analysis (Fig. [198]8A) revealed that CXCL12 was significantly downregulated, while immune activation-related genes such as Ccl7 and Igfbp2 were significantly upregulated, suggesting enhanced immune responses in the TME. Moreover, genes involved in stromal remodeling and metastasis, including Pdgfrl and Fgfr2, were downregulated. A heatmap (Fig. [199]8B) further illustrated the distinct gene expression profiles between the control and treatment groups, with upregulation of immune-related genes (Ccl7, Igfbp2) and downregulation of tumor progression genes (CXCL12, Fgb, Pax5), as well as fibrosis-related genes (Pdgfrl, Serpina3k). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed significant enrichment of pathways including chemokine signaling, complement and coagulation cascades, and cytokine–cytokine receptor interactions. Notably, differential expression of CCL7, CCL8, CCL12, CXCL12, and ACKR4 suggested enhanced immune infiltration. Changes in FGA, FGB, FGG, and SERPINA1A implicated a role in inhibiting tumor progression via coagulation regulation. Other pathways affected included PPAR signaling, insulin secretion, calcium signaling, and cytoskeletal regulation (e.g., TNNC1, TNNT3, MYH4) (Fig. [200]8C). Gene Ontology (GO) functional analysis revealed that LNCs@si-CXCL12 promoted immune infiltration via chemokine signaling and immunoglobulin-mediated immune response, while inhibiting adaptive ECM remodeling. Molecular function enrichment showed significant representation of chemokine activity, CCR receptor binding, and extracellular matrix structural constituents (Fig. [201]8D). Fig. 8. [202]Fig. 8 [203]Open in a new tab Transcriptomic analysis. Note: A Volcano plot displaying differentially expressed genes (DEGs) in tumor tissues after LNCs@si-CXCL12 treatment; B Heatmap showing hierarchical clustering of DEGs; C KEGG pathway enrichment analysis of DEGs; D GO functional enrichment analysis of DEGs Additionally, protein–protein interaction (PPI) network analysis and modular clustering using Cytoscape’s MCODE tool identified six significant functional modules (Figure S10A–F), mainly involved in TME regulation, immune response, metabolic modulation, and ECM remodeling, further validating the mechanistic role of LNCs@si-CXCL12 in tumor suppression (Fig. [204]9). Fig. 9. [205]Fig. 9 [206]Open in a new tab Schematic representation of the molecular mechanism by which CAFs upregulate CXCR4 expression via CXCL12 secretion to promote ESCC growth and metastasis In conclusion, LNCs@si-CXCL12 inhibits ESCC tumor growth and activates the tumor-killing function of CD8^+ T cells, demonstrating potential therapeutic value in suppressing ESCC development and enhancing tumor immune responses. Discussion This study's findings reveal a positive correlation between CXCL12 and the quantity of CAFs in ESCC, while other chemokines such as CXCL1, CXCL8, and CCL20 demonstrate a negative correlation with the quantity and activity of CD8^+ T cells in ESCC. This discovery emphasizes chemokines’ diverse and complex roles in tumor development. Building upon this foundation, our research utilized scRNA-seqg technology to further elucidate the close interplay between the CXCL12-CXCR4 axis in CAFs and CD8^+ T cells, highlighting the pivotal role of this signaling axis in regulating the immune response in ESCC. Previous studies have shown that CXCL12 promotes the polarization and activation of M1-type macrophages, enhancing their cytotoxicity against pathogens and tumor cells, and regulates their pro-inflammatory responses and iNOS expression via CXCR4 binding (PMID: 34864921; 34847760). In addition, CXCL12 enhances NK cell activation and cytotoxicity, partly by modulating surface markers such as CD56 and NKp46 to promote tumor cell recognition (PMID: 34079127; 36781697). These results, together with our own, suggest that CXCL12 can reprogram the TME and exert complex immunomodulatory effects. Additionally, a targeted nanosphere, LNCs@si-CXCL12, was developed to efficiently deliver siRNA, selectively downregulating CXCL12 expression in CAFs, thereby diminishing the inhibitory effect of CAFs on CD8^+ T cell function and restoring or enhancing CD8^+ T cells'immune clearance capability against ESCC. Through both in vitro and in vivo experiments, this nanosphere has demonstrated significant inhibition of tumor cell proliferation and metastasis while enhancing the tumor-killing function of CD8^+ T cells, indicating its potential clinical utility. Notably, we employed single-cell RNA sequencing (scRNA-seq) to investigate the CXCL12–CXCR4 axis and revealed its tight interaction between CAFs and CD8⁺ T cells, underscoring the pivotal role of this signaling pathway in orchestrating the immune response within ESCC. This aspect of the study offers new insights into the complex crosstalk between tumor cells and the surrounding microenvironment. In parallel, we developed a CAF-targeting nanoparticle system, LNCs@si-CXCL12, which not only enables efficient siRNA delivery but also effectively downregulates CXCL12 expression in CAFs, thereby reversing the suppression of CD8⁺ T cell activity. Previous studies have demonstrated that reducing CXCL12 levels can enhance T cell function and improve the efficacy of immune checkpoint inhibitors such as anti–PD-1/PD-L1 antibodies [[207]79]. CXCL12, as a chemokine, primarily facilitates the recruitment of immunosuppressive cells including MDSCs and regulatory T cells (Tregs) into the TME. Its downregulation can diminish the infiltration of these suppressive cells, thereby facilitating effector T cell access to tumors and enhancing anti-tumor immunity [[208]80, [209]81]. Comparison with existing literature reveals that CXCL12 exhibits similar regulatory patterns in various tumor types. For instance, high expression of CXCL12 in colorectal cancer correlates positively with patient survival rates and can stimulate cancer cell proliferation and metastasis [[210]82]. Similarly, overexpression of CXCL1 in oral squamous cell carcinoma is also associated with tumor proliferation, invasion, and metastasis [[211]83]. The CXCL12/CXCR4 axis exacerbates disease progression by promoting tumor cell migration and invasion, consistent with our observations of CXCL12's role in ESCC, where it enhances tumor invasiveness and metastatic potential by activating CAFs [[212]84, [213]85]. Various strategies have been proposed in research regarding the application of nanomaterials, especially in targeting CAFs for cancer therapy. For example, some studies have demonstrated the design of nanocarriers for specific targeting of CAFs, releasing chemotherapy drugs or RNA interference molecules to suppress the pro-tumor function of CAFs in the TME [[214]27, [215]28]. These studies validate the effectiveness of nanotechnology in precisely delivering therapeutic agents to complex TMEs. Our strategy using the LNCs@si-CXCL12 nanosphere delivery system targeting the downregulation of CXCL12 expression in CAFs is similar, but our approach emphasizes inhibiting tumor growth through immune microenvironment modulation. While this study provides novel insights into the role of CXCL12 in ESCC and demonstrates the application of nanomaterials in therapy, there are limitations. Firstly, although the results of in vitro and in vivo experiments are promising, the complexity of the TME may lead to uncertainty in clinical applications. Future research should explore the impact of other cell types and molecular mechanisms in the TME. Additionally, whether CXCL12 interferes with conventional chemotherapy or other immunotherapies remains unexplored and will be one of our next research directions. Secondly, this study primarily used mouse models rather than human clinical samples; therefore, the clinical relevance of the research findings needs validation through broader clinical trials. Conducting additional experiments in other ESCC cell lines would also improve the generalizability of our findings. Moreover, future investigations will focus on how LNCs@si-CXCL12 impacts other immune cell populations beyond CD8⁺ T cells. Lastly, given that tumor initiation and progression are multifactorial and multistep processes, future studies should further investigate the roles of other cells and molecular mechanisms in the growth and metastasis of ESCC. The use of multiplex spatial immunofluorescence, which retains spatial resolution at the cellular and subcellular level, will allow us to analyze the distribution and activation states of immune cells in the TME, providing deeper mechanistic insights into immune evasion strategies. Incorporating this technology will be a key focus in our future research. Regarding nanomedicine applications, previous studies have shown that nanoparticles can effectively target CAFs and modulate their pro-tumoral functions [[216]27, [217]28], consistent with our approach. However, unlike strategies that solely inhibit CAFs, we emphasize the importance of reprogramming the immune microenvironment. Our strategy focuses not only on suppressing the tumor-promoting properties of CAFs but also on reinvigorating immune effector cells to enhance tumor clearance. This immunomodulatory perspective provides new opportunities for the development of next-generation targeted immunotherapies. In conclusion, This study reveals a positive correlation between CXCL12 and the number of cancer-associated fibroblasts (CAFs) in esophageal squamous cell carcinoma (ESCC), and that CXCL12 exerts an immunosuppressive effect by inhibiting CD8 + T cell function. This finding is consistent with existing studies, particularly regarding the role of the CXCL12/CXCR4 axis in immune evasion and metastasis in various cancers [[218]82, [219]84, [220]85]. We have explored the interaction between the CXCL12-CXCR4 axis and CAFs in regulating immune responses through single-cell RNA sequencing (scRNA-seq). Targeting CAFs with nanoparticles (LNCs@si-CXCL12) that effectively deliver siRNA and reduce CXCL12 expression has reversed the inhibition of CD8 + T cells, enhancing the tumor-killing effect. Previous studies have demonstrated the application of nanoparticles in targeting CAFs and modulating the immune microenvironment [[221]27, [222]28], and our approach further emphasizes strategies to enhance tumor clearance through immune activation, providing new insights for targeted immunotherapy. Overall, our study highlights the role of CXCL12 in ESCC and demonstrates the potential of targeting CXCL12 to improve immune responses, laying the foundation for future therapeutic strategies (Fig [223]9). Supplementary Information [224]12951_2025_3476_MOESM1_ESM.jpg^ (1.7MB, jpg) Additional file 1. Figure S1. Identification of NF and CAFs. Note: Immunofluorescence was conducted to observe the expression of the fibroblast marker gene fibrillinectin and the endothelial cell marker gene Cytokeratin in NF/CAFs; Immunofluorescence was performed to observe the expression of the CAFs markers α-SMA and FAPα in NF/CAFs [225]12951_2025_3476_MOESM2_ESM.jpg^ (586.1KB, jpg) Additional file 2. Figure S2. Purity Detection of CD8+ T Cells. Note: Flow cytometry detection of the purity of the extracted Jurkat cells; Flow cytometry was used to assess the purity of CD8+ T cells extracted from mice [226]12951_2025_3476_MOESM3_ESM.jpg^ (561.4KB, jpg) Additional file 3. Figure S3. scRNA Sample Analysis and Filtering. Note: Violin plots displaying the gene count, mRNA molecule count, and mitochondrial gene proportion of all cells in each sample; Scatter plots showing the correlation between nCount_RNA and percent.mt, nCount_RNA and nFeature_RNA; Identification of highly variable genes among 23676 genes through variance analysis; Scatter plot of cell clustering in samples analyzed by PCA; p-values for the top 40 PCs determined by PCA; Elbow plot analysis determining the use of the top 9 PCs for t-SNE clustering analysis [227]12951_2025_3476_MOESM4_ESM.jpg^ (3.6MB, jpg) Additional file 4. Figure S4. PCA of scRNA-seq Data. Note: PCA of feature genes in the first 6 PCs and heatmap showing the expression levels of feature genes [228]12951_2025_3476_MOESM5_ESM.jpg^ (3MB, jpg) Additional file 5. Figure S5. Cell Clustering and Cell Communication Analysis. Note: t-SNE clustering analysis categorizing cells into 21 cell clusters; Heatmap of the top 10 marker gene expressions for each cell cluster; t-SNE clustering analysis showing the clustering of Normal and Tumor samples; Annotation of 21 cell clusters into 12 cell types using the"SingleR"package in combination with the Cell Marker database; Distribution of CXCL12, CXCL1, CXCL8, and CXCR4 in cell clusters; Cell interaction network diagram mediated by CXCL signaling pathways; Bubble plot of all ligand receptor-mediated cell interactions, with a red box indicating a significant cell interaction relationship between CXCL12/CXCR4-mediated CAFs and CD8+ T cells, T cells, and B cells; Distribution of CXCR4 in the clustering plot; Expression patterns of CXCL12 and CXCR4 in normal and tumor groups; *** indicates P < 0.001 compared to the normal group [229]12951_2025_3476_MOESM6_ESM.jpg^ (2.4MB, jpg) Additional file6Figure S6. Cell Clustering and Cell Communication Analysis. Note: t-SNE cluster analysis of 6 samples; Distribution of CXCL12, CXCL1, CXCL8 in the cluster map; Distribution of CXCL12, CXCL1, CXCL8 in 21 cell clusters; Cell interaction chord diagram and hierarchical diagrammediated by CXCL signaling pathway; Distribution of CXCR4 in 21 cell clusters [230]12951_2025_3476_MOESM7_ESM.jpg^ (6.2MB, jpg) Additional file 7. Figure S7. Effect of CAFs on the Growth and Metastasis of ESCC by Secreting CXCL12. Note: ELISA to detect the expression levels of CXCL12 in TE-1 cells in each treatment group;CCK-8 assay to evaluate the viability of TE-1 cells in each treatment group; EDU assay to measure the proliferation of TE-1 cells in each treatment group; Flow cytometry to detect the apoptosis of TE-1 cells in each treatment group; Transwell assay to assess the migration and invasion of TE-1 cells in each treatment group. * indicates P < 0.05 compared to TE-1+NFs group, # indicates P < 0.05 compared to TE-1+CAFs-sh-NC group. All cell experiments were performed in triplicate [231]12951_2025_3476_MOESM8_ESM.jpg^ (1.4MB, jpg) Additional file 8. Figure S8. Targeting Effect of LNCs@si-CXCL12 on CAFs. Note: Immunohistochemistry to observe the alteration in the expression of the CAF activation marker αSMA in tumor tissues of mice; Western blot analysis to detect the expression of CXCL12,,VEGF and CXCR4 in tumor tissues of different groups. ** indicates P < 0.01 compared to the PBS group, *** indicates P < 0.001. [232]12951_2025_3476_MOESM9_ESM.jpg^ (2.7MB, jpg) Additional file 9. Figure S9. Biological Safety Evaluation of PAMAM@Cur. Note: H&E stained tissue sections of mouse heart, liver, spleen, lung, and kidney for morphological changes post PAMAM@Cur treatment; Evaluation of biochemical parameters ALT, AST, BUN, and CR in mouse serum post-treatment to assess the biocompatibility and safety of PAMAM@Cur. n=6. [233]12951_2025_3476_MOESM10_ESM.jpg^ (1MB, jpg) Additional file 10. Figure S10. PPI Network Modules. Note: Modular clustering of the PPI network using the Cytoscape MCODE plugin, identifying six prominent functional clusters related to TME regulation, immune responses, metabolic pathways, and extracellular matrix remodeling [234]Additional file 11^ (13.4KB, docx) [235]Additional file 12. ^ (12KB, docx) [236]Additional file 13. ^ (12.6KB, docx) Acknowledgements