
Single‑cell RNA sequencing analysis of intrahepatic cholangiocarcinoma reveals SPP1 facilitates disease progression via interaction with CD4+ T cells
- Authors:
- Published online on: June 10, 2025 https://doi.org/10.3892/ol.2025.15135
- Article Number: 390
-
Copyright: © Lin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Intrahepatic cholangiocarcinoma (ICC), the second most common primary hepatobiliary cancer, is among the most aggressive malignancies, with a 5-year survival rate of ~5% (1). Largely attributable to the absence of pathognomonic symptoms during early tumorigenesis and the suboptimal performance of conventional surveillance strategies in high-risk populations, patients who are diagnosed with liver cancer frequently present at advanced stages of the disease, which typically makes curative liver resection unfeasible (2). Additionally, patients diagnosed with ICC often experience minimal benefits from chemotherapy treatment, a phenomenon driven by intrinsic chemoresistance mechanisms and the tumor's desmoplastic stroma that impedes drug penetration (3,4). Therapeutic approaches, including targeted therapy or immunotherapy, have shown promise and are currently being assessed in clinical trials (5). Therefore, further exploration of effective treatments is essential to extend the survival of patients with ICC.
The tumor microenvironment (TME) is characterized by complex interactions between different cell types, particularly tumor cells and immune cells, such as tumor-associated macrophages and cancer-associated fibroblasts (6). The interactions between various cell types within the TME are associated with tumor progression and are also a promising therapeutic target (6). The TME is crucial for use in assessing the prognosis of patients with ICC and the malignancy of tumors, with stromal density, immune cell spatial distribution and angiogenic patterning providing actionable insights into metastatic potential and therapeutic resistance (7). Single-cell RNA sequencing (scRNA-seq) has provided valuable insights into the nature of the TME by assessing the gene expression levels and immune cell types present in this environmen (7). However, while a number of studies have reported scRNA-seq data of ICC cells (1,8), there is still a need for further studies on patients with ICC to understand the complexities of the TME and how it influences this disease.
The human secreted phosphoprotein (SPP1) gene, also known as osteopontin, encodes a protein with a function similar to cytokines. The highly acidic nature of this protein serves a crucial role in regulating the turnover of the extracellular matrix (ECM). This regulatory capacity is further modulated through post-translational modifications including N-linked/O-linked glycosylation and transglutamination-induced polymerization. These biochemical alterations exhibit context-dependent functionality: While glycosylation may obscure specific interaction domains, transglutamination conversely enhances SPP1's structural stability and ligand-binding specificity. Such plasticity allows spp1 to differentially execute its cytokine-like functions across various cellular microenvironments and tissue matrices (9–11). A recent study reported that SPP1 expression could determine the different subtypes of the TME. Single-cell transcriptomic analyses in ICC reveal two molecularly distinct TME subtypes: The S100P+SPP1-subtype (iCCAphl) and the S100P-SPP1+ subtype (iCCApps). These subtypes correlate with divergent clinical outcomes, where the S100P+SPP1-subtype is associated with improved survival compared to SPP1+ counterparts (12). Furthermore, SPP1 orchestrates tumor microenvironment reprogramming involving SPP1-mediated crosstalk between tumor cells and stromal components, driving immunosuppression, metabolic reprogramming and ECM reorganization to promote tumor invasion and immune evasion (13). CD44 is a widely expressed cell surface glycoprotein that serves as a receptor for hyaluronic acid and other ECM components. It is highly expressed in various cancer cells and is involved in cell adhesion, migration and proliferation. Its expression pattern is often associated with tumor aggressiveness and poor prognosis (14). SPP1 acts as a ligand for CD44 (15) and it has been previously reported that SPP1 interacts with CD44 to modulate cell signaling and the activation of neoplastic cells, resulting in tumor metastasis and progression (16). Additionally, the interactions between SPP1 and CD44 promote stem cell features in the glioma perivascular niche (17) and SPP1 suppresses T-cell activation (18). Therefore, SPP1 may have multiple key functions in tumor progression and the TME. However, the mechanisms of action by which SPP1 modulates the TME phenotype of ICC have remained largely elusive.
The present study provided a comprehensive analysis of the complexity of the TME and transcriptomic features of ICC by analyzing the scRNA-seq of clinical surgical samples and online datasets. The cell types and interactions between lymphocytes and other immune cells were identified. Further analysis demonstrated that the cell types, gene expression levels and interactions of immune cells were markedly different in patients with early-stage ICC compared with patients with late-stage ICC. Furthermore, the present study established a CD4+ T cell/SPP1-CD44 axis, which may serve as a direct link to regulate the development of ICC.
Materials and methods
Data preparation
In the present study, clinical datasets were utilized to evaluate the scRNA-seq results obtained from The National Center for Biotechnology Information Gene Expression Omnibus (GEO; https://www.ncbi.nlm.nih.gov/geo/; GSE138709) (1). Samples in this dataset (GSE138709) had been collected from 5 patients with ICC, including 3 male patients and 2 female patients. The original databases did not include the age for all patients; therefore, the present study is unable to provide an overall age range or median age for the patient cohort.
Sequencing and raw data processing
All sequencing reads were aligned to the human genome (reference no. GRCh38) using Cell Ranger software (version 3.0.2; 10× Genomics). Subsequently, the refined feature barcode matrix was utilized as the basis for data analysis.
scRNA data analysis
The Seurat package (version 5.0.2; http://satijalab.org/seurat/articles/install.html) was used for processing and analyzing the scRNA-seq data in R language (version 4.3.1, 2023-06-16). The individual libraries were transformed into Seurat objects. The data were filtered to focus on genes expressed across ≥10 cells and each cell exhibited gene expression levels within the range of 500–20,000 genes, with mitochondrial transcripts accounting for <15% of the total. The cutoffs were set after performing a quality control evaluation of each library. Subsequently, the data underwent logarithmic transformation for normalization and variable genes were identified through an in-depth mean and variance analysis. The genes were scaled and principal components were derived. To determine the most informative principal components, an elbow plot was used and the leading components were applied for dimensionality reduction using the Uniform Manifold Approximation and Projection (UMAP) dimensionality algorithm (19).
The main cell populations present in both the stromal tissue and immune compartments were identified by analyzing the average expression levels of particular gene markers in each cluster: i) Malignant, epithelial cell adhesion molecule; ii) B cells, B cell scaffold protein with ankyrin repeats 1 and CD79A; iii) CD4 cells, CD3D and CD4; iv) CD8 cells, CD3E and CD8A; v) macrophages, CD14 and apolipoprotein E; vi) myeloid cells, SP100A9 and lysozyme (LYZ); vii) dendritic cells (DCs), CD1C and C-type lectin domain containing 9A (CLEC9A); viii) natural killer (NK) cells, neural cell adhesion molecule 1 and NK cell granule protein 7; ix) hepatocytes, apolipoprotein C1 and fatty acid binding protein 1; and x) fibroblasts, decorin and actin α2, smooth muscle. Initially, the dataset was filtered to include only these specific cells. There were only 568 cholangiocytes in the original literature, resulting in cholangiocyte-specific markers showing limited representation in our results (GSE138709) (1). Following the initial filtering, analysis was performed to identify subpopulations of T cells and myeloid cells, as previously described (19).
For each patient, the subpopulations of T cells and myeloid cells were examined by isolating the corresponding subset based on patient identity and processing the raw data according to the aforementioned methodology. Patient data with <100 cells in either the myeloid or T-cell categories for the individual patient analysis were excluded (19). The marker identification function within the Seurat package was used to identify the signature genes associated with each cluster (19).
The visualizations, including UMAPs, heat maps, violin plots and box plots, displayed gene expression levels that were normalized using the Scran method (version 1.10.2; http://bioconductor.org/packages/release/bioc/vignettes/scran/inst/doc/scran.html) (20). The calculated expression values were derived from the raw expression data.
Correction of batch effect
To analyze the myeloid and T-cell populations that exhibited clustering specific to individual patients, the Harmony algorithm (version 1.2.3; http://github.com/immunogenomics/harmony) was used to mitigate the influence of patient-related variations, also known as batch effects (19). A cross-verification of the analysis outcomes for the clusters and their associated markers with those from a single-library cluster was conducted to ensure that the Harmony algorithm focused solely on technical variations, without accounting for biological factors (21).
InferCNV analysis
The inferCNV software (Bioconductor; version 3.10; http://bioconductor.riken.jp/packages/3.10/bioc/html/infercnv.html) was used to estimate the initial copy number variants (CNVs) (22). CNVs for each region were calculated based on cellular expression levels, using a cutoff value of 0.1 to refine the data. All cells, with the exception of epithelial cells, were categorized as healthy cells. Subsequently, a denoising procedure was used to generate refined CNV profiles. The diffusion pseudotime analysis was performed using Monocle3 software (Bioconductor; version 3.10; http://bioconductor.jp/packages/3.10/bioc/html/monocle.html) on an anndata object that had been processed with Harmony correction and the results were converted into a Seurat-compatible format (23).
Subpopulation delineation
The established clusters of immune cell subpopulations, specifically myeloid and T cells, were labeled based on the average expression levels of the subsequent genetic indicators: i) Macrophages, CD14 and apolipoprotein E; ii) myeloid cells, SP100A9 and LYZ; iii) DCs, CD1C and CLEC9A; iv) CD8 cells, CD8A and CD8B; v) CD4 cells, forkhead box (FOXP3, FOXP6 and CD4; and vi) proliferative cells, marker of proliferation Ki-67.
Gene set enrichment analysis (GSEA)
The fgsea package (version 1.26.0; http://bioconductor.org/fgsea.html) was employed to perform a comprehensive analysis of gene set enrichment based on the outcomes derived from analysis of differential gene expression patterns, following the approach as previously described (24). The msigdbr package was used to retrieve the hallmark gene sets from the extensive Molecular Signatures Database collection (version 7.5.1; http://cran.r-project.org/msigdbr) (19,25).
Single-cell regulatory network inference and clustering (SCENIC) pathway evaluation
SCENIC pathway analysis was performed using the motifs database associated with RcisTarget (version 1.20.0; http://www.bioconductor.org/RcisTarget.html) in conjunction with the GRNboost algorithm (SCENIC; version 1.3.1) (26,27). The RcisTarget package was utilized to identify binding sequences for transcription factors within the provided gene list (26). The AUCell package (version 1.22.0; http://bioconductor.org/AUCell.html) was utilized to assess the activity levels of each regulon group across individual cells (26).
Data analysis online
Survival of patients with ICC was assessed utilizing the Cancer Genome Atlas dataset through the Gene Expression Profiling Interactive Analysis (GEPIA2) database (http://gepia2.cancer-pku.cn/). The KM-plotter website (https://kmplot.com/analysis/) is the most sophisticated online survival analysis tool, performing all calculations in real time. It contains the gene expression information and clinical case data of 364 patients with liver cancer. The best cutoff value of gene expression was automatically set. The details of this dataset were mentioned by Menyhárt (28). For the Kaplan-Meier survival curves, the follow-up time of patients was ≤5 years, and the log-rank test was performed to observe the statistical difference.
The Metabolic gEne RApid Visualizer (MERAV) website (http://merav.wi.mit.edu/) is designed to analyze human gene expression across a large variety of arrays (4,454 arrays). It contains the gene expression of 7 normal liver tissues and 14 liver primary cancer tissues. The differential expression analysis of SPP1 was conducted directly online.
Experimental model
A total of 2 patients with ICC were thoroughly evaluated to confirm their eligibility (aged >18 years, had no contraindications for surgery or infectious diseases) based on established medical criteria and provided written informed consent prior to participation, ensuring the patients were fully aware of the study's purpose, procedures and potential risks. At last, a 52-year-old female patient and a 63-year-old male patient with ICC from The University of Hong Kong Shenzhen Hospital were enrolled. Detailed information of the patients is presented in Table SI.
Reverse transcription-quantitative PCR (RT-qPCR)
Total RNA was isolated from paraffin-embedded tumor tissues through a dewaxing process, followed by the utilization of TransZol reagent (TransGen Biotech Co., Ltd.), according to the manufacturer's protocol. A total of 1 µg RNA was used for cDNA synthesis using the TransScript First-Strand cDNA Synthesis Kit (TransGen Biotech Co., Ltd.). The thermocycling procedure used for template amplification was set as deformation (94°C, 5 sec), annealing (60°C, 15 sec) and extension stage (72°C, 10 sec) for 40 cycles. RT-qPCR reactions were performed in triplicate with the TransStart Tip Green qPCR SuperMix (TransGen Biotech Co., Ltd.) containing SYBR Green dye and detected by the CFX96 real-time PCR system (Bio-Rad Laboratories, Inc.). Primers for PCR were as follows: Human SPP1 forward, 5′-CGAGGTGATAGTGTGGTTTATGG-3′ and reverse, 5′-GCACCATTCAACTCCTCGCTTTC-3′; human beta-actin forward, 5′-CACCATTGGCAATGAGCGGTTC-3′ and reverse, 5′-AGGTCTTTGCGGATGTCCACGT-3′. The relative quantification method was used for quantifying mRNA level of the target gene, beta-actin was the reference gene and used for normalization, and then the normalized values were compared to obtain the fold change. The comparative Cq value (2−ΔΔCq method) was used to calculate relative quantitative data (29).
Immunohistochemistry (IHC) staining
The tissues collected were fixed in 10% neutral formalin at room temperature for 24 h. Subsequently, the tissue underwent gradient alcohol dehydration, was cleared with xylene and was embedded in paraffin. Subsequently, 4 µm-thick slices were prepared for IHC staining. In briefly, paraffin sections of excised tumor tissue were dewaxed with xylene and processed using a gradient of ethanol concentrations. Tissue slices were washed with PBS before staining. Endogenous peroxidase activity was quenched with 3% H2O2 for 30 min. Subsequently, the tissues were blocked with 3% BSA in PBS for 30 min at room temperature and incubated overnight at 4°C with antibody against human SPP1 (1:100; cat. no. ER1802-16; Hangzhou HuaAn Biotechnology Co., Ltd.). Subsequently, slices were incubated with HRP-conjugated secondary antibody (1:1,000 dilution; cat. no. 7074S; Cell Signaling Technology, Inc.) for 1 h at room temperature after washing with PBS three times. The immune complex was visualized using DAB (Vector Laboratories, Inc.; Maravai LifeSciences) as a chromogen. The reaction was terminated by washing with distilled water, and the sections were counterstained with 0.5% hematoxylin staining solution (cat. no. C0107; Beyotime Institute of Biotechnology) for 5 min at room temperature and mounted with neutral resin. Images were captured using a BX53 microscope (Olympus Corp.).
Statistical analysis
Statistical analysis and figure generation were performed using R (version 4.3.1; http://cran.rstudio.com/bin/windows/base/old/4.3.1/). Statistical differences in data between paired groups were analyzed using Student's t-tests and the outcomes were presented as the mean ± standard deviation.
Results
Transcriptomic analysis of individual cells in ICC
To investigate the TME characteristics of ICC, the data of tumor tissues and adjacent tissues collected from 5 patients with ICC were downloaded from the GEO database. The details of the patients with ICC are listed in Table SII. Prior to transcriptomic analysis, histopathological validation had been performed using hematoxylin-eosin staining to confirm the malignant features of tumor regions and normal architecture of adjacent tissues. IHC staining of cytokeratin 19, a cholangiocyte-specific marker, further verified the origin of the tumor cells (Fig. S1A and B). The scRNA-seq data encompassed 4 tumor tissue samples and 3 samples of adjacent normal tissue (1). Finally, after quality control, an scRNA-seq dataset consisting of a total of 20,412 cells from the database was produced.
T cells and macrophages exhibit differences in the TME of ICC tumor tissue
Using graph-based clustering (30) to analyze the cells from the aforementioned patients with ICC, 10 main cell types were identified, including tumor cells, hepatocytes, B cells, CD4+ T cells, CD8+ T cells, macrophages, DCs, fibroblast, monocytes and NK cells (Fig. 1A and B) (31). All the cell types were characterized using known markers (Figs. 1C and S1C). A UMAP plot was used to assess the distribution of these cells in tumor tissues (Fig. 1D) and adjacent tissues (Fig. 1F). The distribution of different cell types in the tumor and adjacent tissues from ICC samples indicated that macrophages were significantly increased in tumor tissues, whereas T cells were markedly reduced (Fig. 1E, G and H). The numbers of different cell types in every patient are displayed in Tabl SIII and Table SIV.
Increased ratio of regulatory T cells to central memory T cells in the TME of ICC tumor tissues
To determine the influence of ICC tumor cells on T cells, the presence of infiltrating T lymphocytes (TILs) in ICC tumor tissues was compared with the surrounding non-tumor tissues. A total of 9,654 T cells were shown and 7 subsets of TILs were identified in ICC tissues (Fig. 2A). The expression levels of certain marker genes (Figs. 2B and S2A) and the percentage of each cell type subset in each patient with ICC were analyzed (Fig. 2C). The percentage of subsets of TILs in tumor tissues and the surrounding non-tumor tissues of each patient were assessed (Fig. 2D and E). A UMAP plot was used to measure the distribution of T-cell subsets in tumor tissues (Fig. 2F) and adjacent tissues (Fig. 2G). These data demonstrate that central memory T cells [CD4-interleukin 7 receptor (IL7R) T cells] were decreased, while exhausted T cells [CD4-C-X-C motif chemokine ligand 13 (CXCL13) T cells] and regulatory T cells (CD4-FOXP3 T cells) were increased in the tumor tissue of X23 compared with adjacent tissues (Fig. 2H). Furthermore, the ratio of regulatory T cells to central memory T cells was higher in the TME of ICC tumor tissues compared with adjacent tissues (Fig. 2I). The expression level of CD44 in T cells in tumor tissues was higher compared with that in adjacent tissues (Fig. S2B).
A volcano plot demonstrated that 875 genes were upregulated and 354 genes were downregulated in T cells in tumor tissues compared with adjacent tissues (Fig. 2J). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis demonstrated that differential gene expression mainly occurred in ‘response to interleukin-7, ‘signal transduction by p53’, ‘positive thymic T cell selection’ and ‘DNA damage response’ (Fig. 2K). The expression levels of the top 10 highest differentially expressed genes were measured and their association with the prognosis of patients with ICC was analyzed. It was shown that the basic leucine zipper ATF-like transcription factor gene exhibited increased expression levels in T cells in ICC tumor tissues compared with adjacent tissues and exerted a notable influence on the prognosis of patients with ICC (Fig. S2C and D). This finding was also reported by a previous study (32).
The diffusion pseudotime analysis provided a window for studying cellular dynamic processes, facilitating the prediction of tumor cell development and lineage diversification. The developmental trajectories of the subsets of T cells were analyzed (Figs. 2L and S2E). Pseudotime analysis showed that central memory T cells and regulatory T cells in adjacent tissues had similar gene clusters but had different gene clusters in tumor tissues (Fig. S2E). In addition, most regulatory T cells in tumor tissues were distributed at, or close to, the pseudotime branching point 1 (Fig. S2E), which indicated that these cells were undergoing cell programmed changes such as cell fate differentiation. Thus, both cell proliferation and reprogramming could potentially account for the increased ratio of regulatory T cells to central memory cells in tumor tissues.
Association between upregulation of SPP1 in ICC tumor cells and poor prognosis
To detect the transcriptomic features of ICC tumor cells, scRNA-seq data including tumor cells, hepatocytes and cholangiocytes were analyzed (Fig. 3A). The expression level of each cell group marker gene was assessed (Fig. 3B). CNVs, which comprise amplifications and deletions, could notably accelerate the adaptive evolution and development of cancer (33). The results of the present study demonstrated that ICC tumor cells contained several amplification and deletion variants in the entire chromosome. Specifically, the amplification variants in chromosomes 1, 7, 8, 11 and 21 and the deletion variants in chromosomes 3, 6, 16, 17 and 22 (Fig. 3C). KEGG pathway analysis results demonstrated that ICC tumor cells had different gene expression levels in the ‘PI3K-Akt signaling pathway’, ‘focal adhesion’ cascade, ‘ECM receptor-interaction’ cascade, ‘cell cycle pathway’ and ‘p53 signaling pathway’ (Fig. 3D). Furthermore, a volcano plot was used to investigate the differential gene expression of ICC tumor cells. Finally, 176 genes were identified as upregulated and 228 genes and as downregulated in ICC tumor cells compared with adjacent cells (Fig. 3E).
The SPP1 gene was among the most upregulated genes in ICC tumor cells (Fig. 3F). To further assess the association of SPP1 with ICC tumors, analysis of data from the GEPIA2 disease database was performed, which demonstrated a significant upregulation of SPP1 levels in ICC tumors compared with normal tissue and in stage IV of ICC (Fig. S3A and B). Furthermore, the sequencing results were further verified using biological experiments. SPP1 mRNA levels were significantly increased in tumor tissues compared with adjacent tissues from both patients with ICC (Fig. S3C). The SPP1 protein level was also demonstrated to be upregulated in tumors compared with adjacent tissues using IHC staining (Fig. S3D). The results demonstrate that SPP1 expression is elevated in ICC tumor tissues. Thus, the role of SPP1 in ICC immune microenvironment modulation was evaluated. SPP1 was first identified in osteosarcoma cells to serve a role of mediating osteoblast adhesion (34). The data from normal liver, primary hepatocellular carcinoma (HCC) and primary ICC microarrays showed a significant increase in SPP1 expression levels in liver carcinoma, particularly in ICC, compared with normal liver tissues (Fig. 3G). Furthermore, the expression level of SPP1 had a significant association with adverse prognosis in patients with ICC (Fig. 3H). In addition, S100A2 and stratifin (SFN) were also associated with the prognosis of patients with ICC (Fig. S3E-G) (35,36).
SPP1 expression and the ratio of regulatory T cells to central memory T cells are increased in late-stage ICC
Tumor stage is an important clinical diagnostic index for tumor prognosis. To identify an association between the expression levels of the SPP1 gene and the ICC tumor stage, the expression level of SPP1 in tumor cells at different ICC stages was analyzed. ICC stages T1-2 were defined as early-stage and ICC stages T3-4 were defined as late-stage. Tumor cells from patient X25 were excluded from the datasets due to the absence of tumor tissue data in the public database. Therefore, tumor cells from patients X20, X23 and X24 were used as the early-stage group and tumor cells from patient X18 as late-stage (Fig. 4A). The volcano plot indicated that there were 69 upregulated genes and 101 downregulated genes in late-stage tissues compared with early-stage tissues (Fig. S4A).
The differentially expressed genes and their association with the prognosis of patients with ICC were further investigated. These results demonstrated that the expression level of SPP1 was higher in late-stage ICC compared with earlier stages (Fig. 4B). Furthermore, aldo-keto reductase family 1 member C2 and S100P exhibited increased expression levels, while IL-32 and vitronectin exhibited decreased expression levels in the late-stage ICC tumors compared with early-stage ICC tumors, and all of the aforementioned genes were significantly associated with the prognosis of patients with ICC (Fig. S4B-F). KEGG pathway analysis showed that the early- and late-stage groups exhibited different expression levels of genes associated with ‘cellular senescence’, the ‘p53 signaling pathway’ and ‘ferroptosis’ (Fig. S4G).
Furthermore, a UMAP plot was utilized to analyze the subset of CD4+ T cells present in the late and early stages of ICC tumor tissues (Fig. 4C and D). An increase in the ratio of regulatory T cells to central memory T cells in the tumor tissues of late-stage ICC was demonstrated (Fig. 4E). Pseudotime analysis showed distinct gene expression clusters of various T-cell types that were different between the early and late stages of ICC (Figs. 4F and S4H). Compared with early-stage tissues, both central memory T cells and regulatory T cells in late-stage tissues were located at the pseudotime branching points 2 and 3, which suggested that these two subtypes of CD4+ T cells underwent cell reprogramming and differentiation during ICC tumor progression.
ICC tumor cells interact with T cells and macrophages to facilitate tumor progression through SPP1/CD44 interactions
To identify the interactions between tumor and immune cells, a list of 1,800 reported interactions were compiled from previously published literature. This list included ligand-receptor pairs from several families, such as cytokines, chemokines, TNFs and receptor tyrosine kinases, along with interactions between the ECM and integrins (37). A bubble plot was employed to predict the ligand-receptor interactions that contributed to the signaling between the tumor cells and immune cell clusters in ICC (Fig. 5A). The details of SPP1-CD44, macrophage migration inhibitory factor-CD74 and CD44, CXCL12-C-X-C motif chemokine receptor type 4 and CCL3-C-C motif chemokine receptor type 1 interactions in the microenvironment of ICC were compiled in a chord diagram (Fig. 5B-E).
Discussion
ICC is an aggressive form of cancer characterized by a scarcity of effective treatment modalities and poor prognosis (1). Immunological therapy is an emerging treatment strategy for this disease that has the potential to improve treatment efficacy and prolong the survival of patients with ICC. However, the complex immune microenvironment of ICC limits the effectiveness of immunotherapy for patients. scRNA-seq is a powerful and effective method that can be used to understand the detailed immune microenvironment of different types of cancer.
T cell-based immunotherapy is a promising strategy for the systemic treatment of cancer. CD8+ T lymphocytes, particularly CD8+ cytotoxic T cells, can directly eliminate cancer cells via specific major histocompatibility complex class I interactions. The function of CD4+ T cells in immunotherapy has been previously reported in both mouse models and clinical studies involving patients, which shows that various subsets of CD4+ T cells mediate the effects of antitumor immune responses (38). However, there is still limited research on the roles of CD4+ T cells in the oncogenesis and progression of ICC, which hinders the development of effective immunotherapy strategies against ICC tumors.
In the present study, characteristics of the TME of ICC tumor tissues were identified and features of the immune microenvironment in both early- and late-stage ICC were further analyzed. The scRNA-seq data demonstrated that the number of CD4+ T cells was increased in ICC tumor tissues. When activated by antigen-presenting cells, uncommitted CD4+ T cells can differentiate into a range of effector T-cell subsets, as well as memory T cells (39,40). The results demonstrated that the proportion of central memory T cells (CD4-IL7R T cells), regulatory T cells (CD4-FOXP3 T cells) and exhausted T cells (CD4-CXCL13 T cells) were higher in ICC tumor tissues compared with those in adjacent normal tissues. Central memory T cells are potent antitumor immune cells, distinguished by their prolonged in vivo survival and their robust capacity for self-renewal (41). However, regulatory T cells and exhausted T cells exerted a pro-tumor effect (42). The number of CD4+ T cells was higher in the tumor tissues of late-stage ICC (T3 stage) compared with those in early-stage ICC (T2 stage). Furthermore, the proportion of regulatory T cells to central memory T cells was elevated both in ICC tumor tissues compared with adjacent liver tissues and in late-stage ICC tumor tissues (T3 stage) compared with early-stage tissues (T2 stage). These results demonstrated that the ICC tumor TME is in a hyper-immunosuppressive state, particularly regarding CD4+ T cells. In addition, the ratio of CD4+ T-cell subsets to other subsets, rather than the absolute subset cell number, is critical for the growth and advancement of ICC tumors. Therefore, the development of techniques and methods that can reduce the proportion of inhibitory CD4+ cells (regulatory T cells and exhausted CD4+ T cells) could effectively alleviate or restrict the growth of ICC tumors.
Beyond the immunosuppressive TME landscape, the genomic profiling performed in the present study revealed recurrent chromosomal amplification (Chr1, 7, 8, 11, 21) and deletion (Chr3, 6, 16, 17, 22) variants in ICC tumor cells. The detection of specific amplification and deletion variants in ICC tumor cells provides valuable insights into the genetic landscape of this malignancy. Amplifications on chromosomes 1, 7, 8, 11 and 21 may indicate regions of oncogene enrichment, while deletions on chromosomes 3, 6, 16, 17 and 22 could suggest loss of tumor suppressor genes. For instance, amplifications in chromosome 1q (encompassing MDM2) and chromosome 8q (containing MYC) are frequently associated with enhanced cell proliferation, apoptosis resistance and metabolic reprogramming in solid tumors (43,44). Conversely, deletions in chromosome 17p (spanning TP53) reduced the tumor suppressor activity, genomic instability and chemoresistance patterns observed in biliary tract malignancies (45,46). Notably, the chromosome 11q amplification encompasses FGF family genes, which are implicated in promoting tumor cell proliferation and survival in ICC (47). Future studies should explore whether these aberrations correlate with clinicopathological features (metastasis, survival) or therapeutic responses to targeted agents.
The SPP1 gene is located at human chromosome 4 and encodes a protein similar to cytokines, which modulates the turnover of the ECM. Previous studies reported that SPP1 serves as both a predictive biomarker and a potential therapeutic target for various types of cancer, including head and neck squamous cell carcinoma and lung adenocarcinoma (48,49). SPP1 gene expression and its downstream effects are regulated by certain upstream regulators, such as cell adhesion to the ECM, the movement of leukocytes, organization of the ECM and signaling pathways that are dependent on integrins. In addition, SPP1 serves a vital role in the process of liver regeneration following a partial hepatectomy (50). SPP1 expression was upregulated in HCC. A previous study reported an association between elevated serum SPP1 levels and several adverse outcomes, including worse overall survival and disease-free survival, advanced HCC stage, larger tumor size and the presence of vascular invasion following surgical resection in patients with HCC (50). Furthermore, it has been reported that SPP1 is a potential serum biomarker for HCC (50). However, it remains largely elusive how SPP1 contributes to the development of ICC. In the present study, a significant upregulation of SPP1 expression levels was observed in ICC tumors, particularly in late-stage ICC, using the GEPIA2 database. Furthermore, RT-qPCR and IHC results demonstrated that SPP1 expression was markedly elevated in tumor tissues compared with adjacent non-tumor tissues. These findings provide evidence that elevated SPP1 expression levels are associated with a worse prognosis and progression of ICC. Therefore, SPP1 may potentially be used as a new tumor marker for ICC.
A previous study reported that SPP1 is elevated during inflammation and is associated with the infiltration and differentiation of immune cells (51). In a mouse model of obesity induced by a high-fat diet, knocking out SPP1 and neutralizing SPP1 ameliorated inflammation in adipose tissue and enhanced insulin sensitivity (50). Mechanistically, SPP1 interacted with various immune cells (e.g., macrophages, T cells and cancer-associated fibroblast cells) (48,52,53) and molecules (e.g., CD44, ITGB1) (54,55) to create an immunosuppressive microenvironment. For instance, SPP1 in breast cancer acts as an autocrine and paracrine factor, promoting proliferation and recruiting and polarizing macrophages into a pro-tumorigenic state. Its inhibition reduces recurrence and enhances the efficacy of immunotherapy, underscoring its crucial role in the immune microenvironment (56). Furthermore, SPP1 promotes cancer cell migration and proliferation by interacting with growth factor receptors (EGFR, PDGFR, VEGFR) and signaling pathways (PI3K/AKT pathway, MAPK/ERK pathway, JAK/STAT pathway) that are involved in cell motility and survival (57,58). In late-stage ICC, increased expression of SPP1 has been associated with the enhancement of cancer cell invasion and metastasis, which are hallmarks of aggressive disease progression. SPP1 achieves this by modulating the activity of signaling molecules such as the PI3K-Akt signaling cascade, focal adhesion cascade, ECM-receptor interaction cascade, cell cycle pathway and p53 signaling cascade, which are known to regulate cell migration, proliferation and survival. SPP1 expression contributes to the maturation and migration of DCs by interaction with CD44 (59). An association has been previously reported between high SPP1 expression levels in adipose tissue and macrophage recruitment (50). Cytoplasmic SPP1 promoted the migration of macrophages by interacting with the CD44-ezrin-radixin-moesin complex (50). The knockout of SPP1 delayed liver regeneration in a mouse model by reducing hepatic macrophage and neutrophil infiltration, along with insufficient activation of STAT3 signaling and IL6 in Kupffer cells (60,61). Following the activation of T cells, SPP1 serves a notable role in promoting the differentiation of Th1 and Th17 cells (18).
SPP1 overexpression in tumor cells could regulate TME features through surrounding immune cells in a receptor-ligand pattern. The SPP1-CD44 interaction serves a critical role in tumor progression and metastasis in various types of cancers. For example, the SPP1-CD44 interaction promotes tumor progression and has been recognized as mediating the interplay between macrophages and HCC cells (62). Furthermore, the SPP1-CD44 axis promotes cancer stemness and metastasis in pancreatic tumors (54). The SPP1-CD44 axis has also been reported to mediate crosstalk between macrophages and cancer cells in gliomas, highlighting its potential as a promising therapeutic target for the treatment of gliomas (63). Additionally, the SPP1-CD44 interaction has been identified as a promising target for combined immunotherapy, offering a novel perspective for clinical approaches targeting ICC through modulation of effector T-cell infiltration (64).
In conclusion, the present study revealed that elevated SPP1 expression in ICC tumor cells correlates with ICC progression and poor prognosis. The findings demonstrated a dual immunomodulatory role of SPP1 within the TME: First, CD44 expression was significantly upregulated across all T-cell subsets, particularly in tumor-infiltrating CD4+ T cells, which showed substantial enrichment in ICC tissues. Notably, late-stage ICC exhibited an increased ratio of immunosuppressive regulatory T cells (Tregs) to antitumor central memory T cells within the CD4+ population, indicative of a progressively immunosuppressive TME. Second, ligand-receptor analysis of scRNA-seq data identified critical interactions between SPP1-expressing tumor cells and CD44-bearing CD4+ T cells, suggesting the SPP1-CD44 axis serves as a key mediator of tumor-immune crosstalk. Collectively, these findings position the SPP1-CD44 interaction as both a prognostic biomarker and a promising therapeutic target. Future studies should validate its potential for combination immunotherapy strategies aimed at reprogramming the immunosuppressive TME while directly targeting tumor-stromal communication pathways.
There are certain limitations to the present study and experimental design. Firstly, for the patient data being collected from online databases and included in the analysis, it only partially reflects the general clinical case pattern. Secondly, due to the small sample size, there is a lack of reliable correlation analysis between clinical manifestations and pathological features of tumors. Therefore, only indirect assessments can be made with the help of online pathological databases. Thirdly, the variability in treatment protocols among patients poses a challenge to the reliability and convincing nature of the survival outcomes reported. Despite best efforts to analyze the data thoroughly, the heterogeneity in treatment approaches underscores a notable limitation that cannot be overlooked. Due to the constraints of the current dataset, the present study was unable to provide survival outcomes that are unaffected by these inconsistencies. Therefore, these limitations will be addressed in future work by increasing the sample size to enhance the reliability and generalizability of the findings. By doing so, potential confounding factors can be accounted for to provide more robust survival outcome data.
In conclusion, the present study provides valuable insights into the role of SPP1 in ICC, highlighting its potential as a therapeutic target and prognostic marker. The findings suggest that targeting the SPP1-CD44 axis may offer a promising strategy for ICC treatment by modulating the tumor immune microenvironment. Future research should build on these results to further explore the mechanisms underlying SPP1's functions and to develop effective therapeutic approaches for patients with ICC.
Supplementary Material
Supporting Data
Supporting Data
Acknowledgements
Not applicable.
Funding
The present work was supported by The Shenzhen Science and Technology Innovation Commission Fundamental Research Key Projects (grant nos. JCYJ20210324120200001, JCYJ20210324101805014 and JCYJ20240813113038049) and the Sanming Project of Medicine in Shenzhen (grant no. SZSM202211017).
Availability of data and materials
The data generated in the present study may be requested from the corresponding author.
Authors' contributions
XL, HP, KL, WC, XS, CN, HL and DY contributed to the study conception and design. DY drafted the manuscript and supervised the study. HL conceptualized the study and drafted the manuscript. XL wrote the original draft. HP analyzed the data and produced figures and tables. KL was involved in visualization. WC provided professional suggestions. XS performed data curation. CN edited the manuscript. XL and HP confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.
Ethics approval and consent to participate
All of the patients involved provided written informed consent and the present study was approved by the Medical Research Ethics Committee of The University of Hong Kong Shenzhen Hospital [approval no. Lun (2021)122].
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Zhang M, Yang H, Wan L, Wang Z, Wang H, Ge C, Liu Y, Hao Y, Zhang D, Shi G, et al: Single-cell transcriptomic architecture and intercellular crosstalk of human intrahepatic cholangiocarcinoma. J Hepatol. 73:1118–1130. 2020. View Article : Google Scholar : PubMed/NCBI | |
Loeuillard E, Yang J, Buckarma E, Wang J, Liu Y, Conboy C, Pavelko KD, Li Y, O'Brien D, Wang C, et al: Targeting tumor-associated macrophages and granulocytic myeloid-derived suppressor cells augments PD-1 blockade in cholangiocarcinoma. J Clin Invest. 130:5380–5396. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yuan H, Lin Z, Liu Y, Jiang Y, Liu K, Tu M, Yao N, Qu C and Hong J: Intrahepatic cholangiocarcinoma induced M2-polarized tumor-associated macrophages facilitate tumor growth and invasiveness. Cancer Cell Int. 20:5862020. View Article : Google Scholar : PubMed/NCBI | |
Zheng C, Zheng L, Yoo JK, Guo H, Zhang Y, Guo X, Kang B, Hu R, Huang JY, Zhang Q, et al: Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell. 169:1342–1356.e16. 2017. View Article : Google Scholar : PubMed/NCBI | |
Kim RD, Chung V, Alese OB, El-Rayes BF, Li D, Al-Toubah TE, Schell MJ, Zhou JM, Mahipal A, Kim BH and Kim DW: A Phase 2 multi-institutional study of nivolumab for patients with advanced refractory biliary tract cancer. JAMA Oncol. 6:888–894. 2020. View Article : Google Scholar : PubMed/NCBI | |
Song G, Shi Y, Zhang M, Goswami S, Afridi S, Meng L, Ma J, Chen Y, Lin Y, Zhang J, et al: Global immune characterization of HBV/HCV-related hepatocellular carcinoma identifies macrophage and T-cell subsets associated with disease progression. Cell Discov. 6:902020. View Article : Google Scholar : PubMed/NCBI | |
D'Avola D, Villacorta-Martin C, Martins-Filho SN, Craig A, Labgaa I, von Felden J, Kimaada A, Bonaccorso A, Tabrizian P, Hartmann BM, et al: High-density single cell mRNA sequencing to characterize circulating tumor cells in hepatocellular carcinoma. Sci Rep. 8:115702018. View Article : Google Scholar : PubMed/NCBI | |
Chai X, Wang J, Li H, Gao C, Li S, Wei C, Huang J, Tian Y, Yuan J, Lu J, et al: Intratumor microbiome features reveal antitumor potentials of intrahepatic cholangiocarcinoma. Gut Microbes. 15:21562552023. View Article : Google Scholar : PubMed/NCBI | |
Jono S, Peinado C and Giachelli CM: Phosphorylation of osteopontin is required for inhibition of vascular smooth muscle cell calcification. J Biol Chem. 275:20197–1203. 2000. View Article : Google Scholar : PubMed/NCBI | |
Christensen B, Kazanecki CC, Petersen TE, Rittling SR, Denhardt DT and Sørensen ES: Cell type-specific post-translational modifications of mouse osteopontin are associated with different adhesive properties. J Biol Chem. 282:19463–19472. 2007. View Article : Google Scholar : PubMed/NCBI | |
Oyama M, Kariya Y, Kariya Y, Matsumoto K, Kanno M, Yamaguchi Y and Hashimoto Y: Biological role of site-specific O-glycosylation in cell adhesion activity and phosphorylation of osteopontin. Biochem J. 475:1583–1595. 2018. View Article : Google Scholar : PubMed/NCBI | |
Song G, Shi Y, Meng L, Ma J, Huang S, Zhang J, Wu Y, Li J, Lin Y, Yang S, et al: Single-cell transcriptomic analysis suggests two molecularly subtypes of intrahepatic cholangiocarcinoma. Nat Commun. 13:16422022. View Article : Google Scholar : PubMed/NCBI | |
Ma L, Wang L, Khatib SA, Chang CW, Heinrich S, Dominguez DA, Forgues M, Candia J, Hernandez MO, Kelly M, et al: Single-cell atlas of tumor cell evolution in response to therapy in hepatocellular carcinoma and intrahepatic cholangiocarcinoma. J Hepatol. 75:1397–1408. 2021. View Article : Google Scholar : PubMed/NCBI | |
Paulis YW, Huijbers EJ, van der Schaft DW, Soetekouw PM, Pauwels P, Tjan-Heijnen VC and Griffioen AW: CD44 enhances tumor aggressiveness by promoting tumor cell plasticity. Oncotarget. 6:19634–19646. 2015. View Article : Google Scholar : PubMed/NCBI | |
Katagiri YU, Sleeman J, Fujii H, Herrlich P, Hotta H, Tanaka K, Chikuma S, Yagita H, Okumura K, Murakami M, et al: CD44 variants but not CD44s cooperate with beta1-containing integrins to permit cells to bind to osteopontin independently of arginine-glycine-aspartic acid, thereby stimulating cell motility and chemotaxis. Cancer Res. 59:219–226. 1999.PubMed/NCBI | |
Rao G, Wang H, Li B, Huang L, Xue D, Wang X, Jin H, Wang J, Zhu Y, Lu Y, et al: Reciprocal interactions between tumor-associated macrophages and CD44-positive cancer cells via osteopontin/CD44 promote tumorigenicity in colorectal cancer. Clin Cancer Res. 19:785–797. 2013. View Article : Google Scholar : PubMed/NCBI | |
Pietras A, Katz AM, Ekström EJ, Wee B, Halliday JJ, Pitter KL, Werbeck JL, Amankulor NM, Huse JT and Holland EC: Osteopontin-CD44 signaling in the glioma perivascular niche enhances cancer stem cell phenotypes and promotes aggressive tumor growth. Cell Stem Cell. 14:357–369. 2014. View Article : Google Scholar : PubMed/NCBI | |
Klement JD, Paschall AV, Redd PS, Ibrahim ML, Lu C, Yang D, Celis E, Abrams SI, Ozato K and Liu K: An osteopontin/CD44 immune checkpoint controls CD8+ T cell activation and tumor immune evasion. J Clin Invest. 128:5549–5560. 2018. View Article : Google Scholar : PubMed/NCBI | |
Hornburg M, Desbois M, Lu S, Guan Y, Lo AA, Kaufman S, Elrod A, Lotstein A, DesRochers TM, Munoz-Rodriguez JL, et al: Single-cell dissection of cellular components and interactions shaping the tumor immune phenotypes in ovarian cancer. Cancer Cell. 39:928–944.e6. 2021. View Article : Google Scholar : PubMed/NCBI | |
Lun ATL, McCarthy DJ and Marioni JC: A step-by-step workflow for low-level analysis of single-cell RNA-seq data with bioconductor. F1000Res. 5:21222016. View Article : Google Scholar : PubMed/NCBI | |
Korsunsky I, Millard N, Fan J, Slowikowski K, Zhang F, Wei K, Baglaenko Y, Brenner M, Loh PR and Raychaudhuri S: Fast, sensitive and accurate integration of single-cell data with Harmony. Nat Methods. 16:1289–1296. 2019. View Article : Google Scholar : PubMed/NCBI | |
Hao Q, Li J, Zhang Q, Xu F, Xie B, Lu H, Wu X and Zhou X: Single-cell transcriptomes reveal heterogeneity of high-grade serous ovarian carcinoma. Clin Transl Med. 11:e5002021. View Article : Google Scholar : PubMed/NCBI | |
Cao J, Spielmann M, Qiu X, Huang X, Ibrahim DM, Hill AJ, Zhang F, Mundlos S, Christiansen L, Steemers FJ, et al: The single-cell transcriptional landscape of mammalian organogenesis. Nature. 566:496–502. 2019. View Article : Google Scholar : PubMed/NCBI | |
Sergushichev AA: An algorithm for fast preranked gene set enrichment analysis using cumulative statistic calculation. bioRxiv. 0600122016. | |
Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES and Mesirov JP: Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 102:15545–15550. 2005. View Article : Google Scholar : PubMed/NCBI | |
Aibar S, González-Blas CB, Moerman T, Huynh-Thu VA, Imrichova H, Hulselmans G, Rambow F, Marine JC, Geurts P, Aerts J, et al: SCENIC: Single-cell regulatory network inference and clustering. Nat Methods. 14:1083–1086. 2017. View Article : Google Scholar : PubMed/NCBI | |
Bao MM, Kennedy JM, Dolinger MT, Dunkin D, Lai J and Dubinsky MC: Cytomegalovirus colitis in a patient with severe treatment refractory ulcerative colitis. Crohns Colitis 360. 6:otae0142024. View Article : Google Scholar : PubMed/NCBI | |
Menyhárt O, Nagy Á and Győrffy B: Determining consistent prognostic biomarkers of overall survival and vascular invasion in hepatocellular carcinoma. R Soc Open Sci. 5:1810062018. View Article : Google Scholar : PubMed/NCBI | |
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI | |
Traag VA, Waltman L and van Eck NJ: From louvain to leiden: Guaranteeing well-connected communities. Sci Rep. 9:52332019. View Article : Google Scholar : PubMed/NCBI | |
Zhang Q, He Y, Luo N, Patel SJ, Han Y, Gao R, Modak M, Carotta S, Haslinger C, Kind D, et al: Landscape and dynamics of single immune cells in hepatocellular carcinoma. Cell. 179:829–845.e20. 2019. View Article : Google Scholar : PubMed/NCBI | |
Jia C, Ma Y, Wang M, Liu W, Tang F and Chen J: Evidence of omics, immune infiltration, and pharmacogenomics for BATF in a pan-cancer cohort. Front Mol Biosci. 9:8447212022. View Article : Google Scholar : PubMed/NCBI | |
Lauer S and Gresham D: An evolving view of copy number variants. Curr Genet. 65:1287–1295. 2019. View Article : Google Scholar : PubMed/NCBI | |
Sodek J, Chen J, Nagata T, Kasugai S, Todescan R Jr, Li IW and Kim RH: Regulation of osteopontin expression in osteoblasts. Ann N Y Acad Sci. 760:223–241. 1995. View Article : Google Scholar : PubMed/NCBI | |
Sato Y, Harada K, Sasaki M and Nakanuma Y: Clinicopathological significance of S100 protein expression in cholangiocarcinoma. J Gastroenterol Hepatol. 28:1422–1429. 2013. View Article : Google Scholar : PubMed/NCBI | |
Yang Z, Jin Q, Hu W, Dai L, Xue Z, Man D, Zhou L, Xie H, Wu J and Zheng S: 14-3-3σ downregulation suppresses ICC metastasis via impairing migration, invasion, and anoikis resistance of ICC cells. Cancer Biomark. 19:313–325. 2017. View Article : Google Scholar : PubMed/NCBI | |
Kumar MP, Du J, Lagoudas G, Jiao Y, Sawyer A, Drummond DC, Lauffenburger DA and Raue A: Analysis of single-cell RNA-Seq identifies cell-cell communication associated with tumor characteristics. Cell Rep. 25:1458–1468.e4. 2018. View Article : Google Scholar : PubMed/NCBI | |
Richardson JR, Schöllhorn A, Gouttefangeas C and Schuhmacher J: CD4+ T cells: Multitasking cells in the duty of cancer immunotherapy. Cancers (Basel). 13:5962021. View Article : Google Scholar : PubMed/NCBI | |
Kim HJ and Cantor H: CD4 T-cell subsets and tumor immunity: The helpful and the not-so-helpful. Cancer Immunol Res. 2:91–98. 2014. View Article : Google Scholar : PubMed/NCBI | |
Tay RE, Richardson EK and Toh HC: Revisiting the role of CD4+ T cells in cancer immunotherapy-new insights into old paradigms. Cancer Gene Ther. 28:5–17. 2021. View Article : Google Scholar : PubMed/NCBI | |
Aizarani N, Saviano A, Sagar, Mailly L, Durand S, Herman JS, Pessaux P, Baumert TF and Grün D: A human liver cell atlas reveals heterogeneity and epithelial progenitors. Nature. 572:199–204. 2019. View Article : Google Scholar : PubMed/NCBI | |
Kang K, Wang X, Meng C, He L, Sang X, Zheng Y and Xu H: The application of single-cell sequencing technology in the diagnosis and treatment of hepatocellular carcinoma. Ann Transl Med. 7:7902019. View Article : Google Scholar : PubMed/NCBI | |
Shroff RT and Bachini M: Treatment options for biliary tract cancer: Unmet needs, new targets and opportunities from both physicians' and patients' perspectives. Future Oncol. 20:1435–1450. 2024. View Article : Google Scholar : PubMed/NCBI | |
Vincelette ND, Yu X, Kuykendall AT, Moon J, Su S, Cheng CH, Sammut R, Razabdouski TN, Nguyen HV, Eksioglu EA, et al: Trisomy 8 defines a distinct subtype of myeloproliferative neoplasms driven by the MYC-alarmin axis. Blood Cancer Discov. 5:276–297. 2024. View Article : Google Scholar : PubMed/NCBI | |
Popek-Marciniec S, Styk W, Wojcierowska-Litwin M, Chocholska S, Szudy-Szczyrek A, Samardakiewicz M, Swiderska-Kolacz G, Czerwik-Marcinkowska J and Zmorzynski S: Association of chromosome 17 aneuploidy, TP53 deletion, expression and Its rs1042522 variant with multiple myeloma risk and response to thalidomide/bortezomib treatment. Cancers (Basel). 15:47472023. View Article : Google Scholar : PubMed/NCBI | |
Hallek M and Al-Sawaf O: Chronic lymphocytic leukemia: 2022 Update on diagnostic and therapeutic procedures. Am J Hematol. 96:1679–1705. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wang F, Luo M, Qu H and Cheng Y: BAP1 promotes viability and migration of ECA109 cells through KLF5/CyclinD1/FGF-BP1. FEBS Open Bio. 11:1497–1503. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Du W, Chen Z and Xiang C: Upregulation of PD-L1 by SPP1 mediates macrophage polarization and facilitates immune escape in lung adenocarcinoma. Exp Cell Res. 359:449–457. 2017. View Article : Google Scholar : PubMed/NCBI | |
Cai X, Zhang H and Li T: The role of SPP1 as a prognostic biomarker and therapeutic target in head and neck squamous cell carcinoma. Int J Oral Maxillofac Surg. 51:732–741. 2022. View Article : Google Scholar : PubMed/NCBI | |
Song Z, Chen W, Athavale D, Ge X, Desert R, Das S, Han H and Nieto N: Osteopontin takes center stage in chronic liver disease. Hepatology. 73:1594–1608. 2021. View Article : Google Scholar : PubMed/NCBI | |
Morse C, Tabib T, Sembrat J, Buschur KL, Bittar HT, Valenzi E, Jiang Y, Kass DJ, Gibson K, Chen W, et al: Proliferating SPP1/MERTK-expressing macrophages in idiopathic pulmonary fibrosis. Eur Respir J. 54:18024412019. View Article : Google Scholar : PubMed/NCBI | |
Liu L, Zhang R, Deng J, Dai X, Zhu X, Fu Q, Zhang H, Tong Z, Zhao P, Fang W, et al: Construction of TME and Identification of crosstalk between malignant cells and macrophages by SPP1 in hepatocellular carcinoma. Cancer Immunol Immunother. 71:121–136. 2022. View Article : Google Scholar : PubMed/NCBI | |
Storrs EP, Chati P, Usmani A, Sloan I, Krasnick BA, Babbra R, Harris PK, Sachs CM, Qaium F, Chatterjee D, et al: High-dimensional deconstruction of pancreatic cancer identifies tumor microenvironmental and developmental stemness features that predict survival. NPJ Precis Oncol. 7:1052023. View Article : Google Scholar : PubMed/NCBI | |
Nallasamy P, Nimmakayala RK, Karmakar S, Leon F, Seshacharyulu P, Lakshmanan I, Rachagani S, Mallya K, Zhang C, Ly QP, et al: Pancreatic tumor microenvironment factor promotes cancer stemness via SPP1-CD44 axis. Gastroenterology. 161:1998–2013.e7. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zheng Y, Zhao L, Xiong Z, Huang C, Yong Q, Fang D, Fu Y, Gu S, Chen C, Li J, et al: Ursolic acid targets secreted phosphoprotein 1 to regulate Th17 cells against metabolic dysfunction-associated steatotic liver disease. Clin Mol Hepatol. 30:449–467. 2024. View Article : Google Scholar : PubMed/NCBI | |
Shevde LA and Samant RS: Role of osteopontin in the pathophysiology of cancer. Matrix Biol. 37:131–141. 2014. View Article : Google Scholar : PubMed/NCBI | |
Yan Z, Hu X, Tang B and Deng F: Role of osteopontin in cancer development and treatment. Heliyon. 9:e210552023. View Article : Google Scholar : PubMed/NCBI | |
Zhao H, Chen Q, Alam A, Cui J, Suen KC, Soo AP, Eguchi S, Gu J and Ma D: The role of osteopontin in the progression of solid organ tumour. Cell Death Dis. 9:3562018. View Article : Google Scholar : PubMed/NCBI | |
Kawamura K, Iyonaga K, Ichiyasu H, Nagano J, Suga M and Sasaki Y: Differentiation, maturation, and survival of dendritic cells by osteopontin regulation. Clin Diagn Lab Immunol. 12:206–212. 2005.PubMed/NCBI | |
Cui G, Chen J, He J, Lu C, Wei Y, Wang L, Xu X, Li L, Uede T and Diao H: Osteopontin promotes dendritic cell maturation and function in response to HBV antigens. Drug Des Devel Ther. 9:3003–3016. 2015.PubMed/NCBI | |
Wen Y, Feng D, Wu H, Liu W, Li H, Wang F, Xia Q, Gao WQ and Kong X: Defective initiation of liver regeneration in osteopontin-deficient mice after partial hepatectomy due to insufficient activation of IL-6/Stat3 pathway. Int J Biol Sci. 11:1236–1247. 2015. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Zhang L, Ju X, Wang S and Qie J: Single-cell transcriptomic analysis reveals macrophage-tumor crosstalk in hepatocellular carcinoma. Front Immunol. 13:9553902022. View Article : Google Scholar : PubMed/NCBI | |
He C, Sheng L, Pan D, Jiang S, Ding L, Ma X, Liu Y and Jia D: Single-cell transcriptomic analysis revealed a critical role of SPP1/CD44-mediated crosstalk between macrophages and cancer cells in glioma. Front Cell Dev Biol. 9:7793192021. View Article : Google Scholar : PubMed/NCBI | |
Cheng M, Liang G, Yin Z, Lin X, Sun Q and Liu Y: Immunosuppressive role of SPP1-CD44 in the tumor microenvironment of intrahepatic cholangiocarcinoma assessed by single-cell RNA sequencing. J Cancer Res Clin Oncol. 149:5497–5512. 2023. View Article : Google Scholar : PubMed/NCBI |