Skip to main content
Erschienen in: BMC Cancer 1/2023

Open Access 01.12.2023 | Research

Integrated single-cell and bulk RNA sequencing analysis identifies a cancer-associated fibroblast-related gene signature for predicting survival and therapy in gastric cancer

verfasst von: Zhiyang Zhou, Sixuan Guo, Shuhui Lai, Tao Wang, Yao Du, Junping Deng, Shun Zhang, Ge Gao, Jiangnan Zhang

Erschienen in: BMC Cancer | Ausgabe 1/2023

Abstract

As the dominant component of the tumor microenvironment, cancer-associated fibroblasts (CAFs), play a vital role in tumor progression. An increasing number of studies have confirmed that CAFs are involved in almost every aspect of tumors including tumorigenesis, metabolism, invasion, metastasis and drug resistance, and CAFs provide an attractive therapeutic target. This study aimed to explore the feature genes of CAFs for potential therapeutic targets and reliable prediction of prognosis in patients with gastric cancer (GC). Bioinformatic analysis was utilized to identify the feature genes of CAFs in GC by performing an integrated analysis of single-cell and transcriptome RNA sequencing using R software. Based on these feature genes, a CAF-related gene signature was constructed for prognostic prediction by LASSO. Simultaneously, survival analysis and nomogram were performed to validate the prognostic predictive value of this gene signature, and qRT–PCR and immunohistochemical staining verified the expression of the feature genes of CAFs. In addition, small molecular drugs for gene therapy of CAF-related gene signatures in GC patients were identified using the connectivity map (CMAP) database. A combination of nine CAF-related genes was constructed to characterize the prognosis of GC, and the prognostic potential and differential expression of the gene signature were initially validated. Additionally, three small molecular drugs were deduced to have anticancer properties on GC progression. By integrating single-cell and bulk RNA sequencing analyses, a novel gene signature of CAFs was constructed. The results provide a positive impact on future research and clinical studies involving CAFs for GC.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12885-022-10332-w.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CAFs
cancer-associated fibroblasts
GC
gastric cancer
TME
tumor microenvironment
ECM
extracellular matrix
scRNA-seq
single-cell RNA sequencing
RNA-seq
transcriptome RNA sequencing
TCGA
The Cancer Genome Atlas
GEO
Gene Expression Omnibus
OS
overall survival
PCA
principal component analysis
LASSO
least absolute shrinkage selection operator
DEGs
differentially expressed genes
HLA
human leukocyte antigen

Background

The latest statistics for GLOBOCAN 2020 showed that gastric cancer (GC) was the fifth most frequently diagnosed cancer, and the incidence is especially high in Eastern Asian countries, and it is extremely harmful with mortality rate that ranks the fourth in cancer-related death after lung cancer, colorectal cancer, and liver cancer [1, 2]. Following the successful application of targeted therapy and immunotherapy in clinical practice, we present a novel strategy in advanced GC. However, the therapeutic efficacy did not achieve the desired improvement, which emphasizes the need to focus not only on the tumor cells themselves but also on the significance of the surrounding environment, the tumor microenvironment (TME), which comprises all nontumor cells and their noncellular components, such as the extracellular matrix (ECM) and soluble molecules [3]. The crosstalk between tumor cells and the TME directly influences tumor cell growth and cancer progression. Among the different nontumor cells, cancer-associated fibroblasts (CAFs) deserve special attention.
Activated fibroblasts, also defined as CAFs, have long been considered to coevolve with tumor cells as the dominant component of the tumor stroma [4]. CAFs secrete a variety of cytokines, growth factors and chemokines that form fertile soil for the growth of tumor cells; for example, CAFs secrete interleukin-6 (IL-6) or interleukin-11 (IL-11), resulting in tumor progression and the development of chemotherapeutic resistance [57]. In turn, tumor cells secrete numerous factors such as transforming growth factor-β (TGF-β), epidermal growth factor (EGF) and C-X-C motif chemokine ligand 12 (CXCL12), which can activate and educate CAFs [8]. Accumulating studies have confirmed that CAFs are involved in almost every aspect of tumors, including tumorigenesis, metabolism, invasion, metastasis and drug resistance, and CAFs provide an attractive therapeutic target [911].
Notably, researchers are presently unable to achieve breakthroughs in developing viable therapies for CAFs owing to the highly dynamic heterogeneity of CAFs. Indeed, CAFs have diverse potential cellular origins, including resident fibroblasts, mesenchymal stem cells, adipocytes, epithelial cells, mesothelial cells and endothelial cells, and form various subpopulations in different tumor types [8, 10, 12]. Additionally, CAF heterogeneity could possibly be the result of a common precursor in cells at various stages of differentiation that have adopted distinct states based on signaling cues both inside and outside the TME. Currently, α-smooth muscle actin (αSMA), fibroblast-specific protein 1 (FSP1), fibroblast activation protein (FAP), platelet-derived growth factor receptor-α (PDGFRα), PDGFRβ, discoidin domain-containing receptor 2 (DDR2), insulin-like growth factor-binding protein 7 (IGFBP7), caveolin‐1 (CAV1), CD90 (Thy1), tenascin‐C (TNC), periostin (POSTN), podoplanin (PDPN), decorin (DCN), desmin, vimentin and integrin β1 are considered activated CAF markers, and no single specific biomarker can categorize the whole CAF population or distinguish CAFs from all other cell types [8, 10, 13, 14]. As a result, identifying CAFs is extremely difficult and poses a huge challenge for targeted treatment of CAFs.
Additionally, the exploration of the prognostic value of CAFs is also an important reference for individualized treatment, and numerous studies have attempted to validate CAFs as potential pathological indicators of tumor prognosis. In this regard, αSMA serves as a hallmark of prognostic factors. Immunohistochemical (IHC) staining analysis of hepatocellular carcinoma (HCC) patients shows a significantly shorter disease-free survival rate in patients with tumors overexpressing α-SMA [15, 16], and the same negative correlation was shown in colorectal cancer (CRC) and breast cancer [17, 18]. Furthermore, the differential expression signatures of specific genes in CAFs can be used as prognostic tools. In CRC research, Alexandre et al. revealed that high expression levels of the 4-gene signature identify patients with poor prognosis in the CAF cluster [19]. Zou et al. also reported a 12-gene signature of CAFs and its high expression was significantly correlated with pathological and increased clinical events of tumor progression of HCC [20]. However, these CAF-related signatures do not overlap, which presents the same nonspecificity concern in the application of CAFs. Therefore, further clarification of the relationship between CAFs and prognosis and its value in predicting survival will also accelerate the transition from basic CAF research to clinical application. Therefore, the exploration of new biomarkers of CAFs will be of significance.
In this study, we aimed to identify the gene signature of CAFs in GC by performing an integrated analysis of single-cell RNA sequencing (scRNA-seq) and transcriptome RNA sequencing (RNA-seq) with data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). Based on CAF-related genes, we constructed a risk score for prognostic prediction by LASSO, and the analysis revealed that the risk score can be an independent prognostic factor. Then, we established a nomogram model to perform quantitative scores derived from the risk score and other clinicopathological features. In addition, we aimed to identify promising small molecular drugs for gene therapy of CAF-related gene signatures in GC patients.

Materials and methods

Data acquisition

We downloaded the expression matrix of 414 GC and 36 normal gastric samples, and 387 GC samples contained overall survival (OS) data. The clinical information included age, gender, pathologic stage, grade and fraction genome altered, which were procured from the UCSC Cancer Genomics Browser. As a validation set, the GSE62254 dataset including 300 GC samples was downloaded, simultaneously containing OS information generated via the GPL570 platform. In addition, we downloaded the single-cell transcriptome expression profiles of 158,641 cells in 40 samples (29 GC samples and 11 normal samples) from GSE183904 via the GEO database.

Estimation of immune infiltration

The Microenvironment Cell Populations-counter (MCP-counter) package has been applied to study the cellular composition of the microenvironment [21], which uses the gene expression matrix to produce the scores of immunocytes and stromal cells [22]. Therefore, the mRNA data were translated into nontumor cell infiltration levels within the TME using the MCP-counter package of R software.

Processing of single-cell RNA-seq data

We generated a “Seurat” object based on the transcriptome sequencing data of 158,641 cells using the “Seurat” package [23]. The top 2000 genes with highly variable features accounting for cell-to-cell differences were identified by variance analysis and subjected to data scaling and centering. These variable genes were further used for principal component analysis (PCA) with linear dimensionality reduction. The top 35 principal components (PCs) were applied for graph-based clustering (res = 0.4) to identify distinct groups of cells. The cell clusters were visualized based on the “UMAP” method of dimensionality reduction. Clusters were annotated through the “SingleR” package based on the reference gene list of 713 samples from the “HumanPrimaryCellAtlasData” function [24].

Risk assessment model construction and evaluation

In the creation of innovative clinical prediction models, the least absolute shrinkage selection operator (LASSO) regression model is typically utilized [25]. Based on the gene signature generated by LASSO, we calculated the risk score for each patient by applying the following formula:
$$Risk\;score\;=\;\sum_{i=1}^n\beta i\ast i$$
\(\beta i\) refers to the coefficients of each gene; \(i\) represents the expression value of the gene; and \(n\) is the number of genes selected.

Clinical value of the risk assessment model

The samples were divided into high-risk and low-risk groups by the threshold of median score, and the high- and low-risk groups were further analyzed for differential expression with human leukocyte antigens (HLA) and immune checkpoints.
A nomogram was constructed to calculate an individual’s probability of OS by using the package “rms” of R software. In the nomogram, the samples were scored according to the risk assessment model and clinical indicators. The final sum of the scores was expected to be the corresponding 1-, 3-, and 5-year survival probability. The calibration curve was drawn by comparing the predicted probability of the nomogram with the Kaplan–Meier estimate of the observed survival probability.

Gene set enrichment analysis (GSEA)

To further research the potential mechanism between diverse risk groups (median value), we performed GSEA [26]. GSEA was performed to find enriched terms that were predicted to have a correlation with the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway in C2 (“c2.cp.kegg.v7.4.symbols”) [27]. P < 0.01 and FDR (false discovery rate) q < 0.05 were considered to indicate statistical significance.

Identification of potential small molecule drugs

The connectivity map (CMAP) database (http://​www.​broadinstitute.​org) was used to predict potential drugs that may reverse or induce the biological states of GC based on the differentially expressed genes (DEGs). The DEGs were submitted to the CMAP database to search for small molecular drugs that could be used for GC treatment. The enrichment scores ranged from –1 to 1. A negative score suggested that the drug could be beneficial for GC treatment.

Validation of the gene signature expression

The human gastric mucosa epithelial cell line (GES-1) and six GC cell lines (AGS, HGC-27, MKN-45, SGC-7901, MGC-803 and BGC-823) were cultured in Dulbecco’s modified Eagle’s medium (DMEM) or Roswell Park Memorial Institute (RPMI) 1640 medium with 10% fetal bovine serum following the recommended conditions of cell culture. Total RNA was extracted by using TransZol Up, and cDNA was synthesized and mixed with primers (Supplementary Table S1), and placed on the machine following the manufacturer’s protocols. The relative expression of the gene signature mRNA was analyzed by the 2−ΔΔCt method with glyceraldehyde 3-phosphate dehydrogenase (GAPDH) as the internal reference gene.
Twenty pairs of GC tissues and matched adjacent normal tissues were proceeded to validate the expression of the gene signature mRNA in the same operational and statistical manner as described above.
The protein expression levels of the gene signature were compared between normal and malignant tissues with The Human Protein Atlas (HPA: https://​www.​proteinatlas.​org/​).

Statistical analysis

All statistical analyses were performed by using R 4.0.2. The “limma” package was used to analyze the DEGs between tumor and normal samples. The “Survival” package was used to assess the association of each gene with survival. The Survival predictive accuracy of the risk assessment model was assessed based on a time-dependent ROC curve analysis, and survival rates were calculated using the Kaplan–Meier method. The significance of differences between survival curves was determined using the log-rank test. Student's t-test was used to determine the statistical significance of the differences. P values were two-tailed.

Results

Identification of the feature genes for modeling

To explore the TME in GC patients, we conducted nontumor cell infiltration analysis using the MCP-counter package and examined the connection between cell abundance and OS in TCGA and GSE62254 datasets. We noticed that the higher the abundance of fibroblasts was, the poorer the survival of patients (Fig. 1A-B). Subsequently, we performed scRNA-seq profiling to identify CAF-related marker genes. The top 10 genes that were significantly differentially expressed in the cell samples were discovered using variance analysis (Figure S1). The genes that were strongly related in each component were screened using the principal component analysis (PCA) approach. Figures S2-3 show a heatmap and dot plot of the top 30 significantly correlated gene.
The first 35 PCs represented the main deviations of the cells (Figure. S4). According to the “UMAP” algorithm and “SingleR”, 158,641 cells were aggregated into 25 clusters and 8 cell types, respectively (Fig. 1C-E). As the cutoff criteria was P value = 1.0E-5, a total of 10,640 DEGs between GC tissue of 9822 fibroblasts and normal tissue of 3138 fibroblasts were identified.
In the TCGA dataset, 10,091 DEGs between GC samples and normal samples were obtained by differential analysis, including 1093 downregulated genes and 8998 upregulated genes (Fig. 1F). We regarded the DEGs that had a consistent downregulated/upregulated trend with the single-cell results as the stable fibroblast-associated DEGs. Additionally, by survival analysis, 6389 prognosis-related genes were obtained. From the intersection of the stable CAF-related DEGs and the prognosis-related genes, a total of 280 feature genes overlapped for further modeling (supplementary Table S2).

A 9-gene risk assessment model for predicting OS

We obtained a gene signature containing 9 genes by reducing the dimensionality of these 280 feature genes with the LASSO Cox regression model (Fig. 2A-B). Then, Cox analysis was performed on the 9 genes to construct a risk assessment model. The coefficient of each gene was obtained (supplementary Table S3). Survival analysis based on the median risk score showed that survival time was significantly shorter in the high-risk group than in the low-risk group (Fig. 2C). Simultaneously, we studied the differences in the abundance of the two groups. We found that patients in the high-risk group also had a higher abundance of fibroblasts, which was consistent with our previous results. Then, ROC curves were drawn to verify the risk assessment model, and the AUC values of 1-, 3- and 5-year survival were 0.670, 0.661 and 0.729, respectively (Fig. 2E). In addition, we further validated the model with the GSE62254 dataset (Fig. 2D, F).

Risk score as a good differentiator and an independent prognostic factor

The differential expression of human leukocyte antigens (HLA) and immune checkpoints in the high- and low-risk groups was analyzed in the TCGA dataset (Fig. 3A-B) and the GSE62254 dataset (Fig. 3C-D). we could find significant differences in the relevant targets between the high- and low-risk groups.
Univariate and multivariate Cox regression analyses were performed in the TCGA dataset, and the risk score was significantly associated with OS in univariate Cox regression analysis (HR = 2.135, 95% CI = 1.506–3.026, P < 0.001; Fig. 3E). Likewise, multivariate analysis showed that the risk score was an independent prognostic indicator in GC (HR = 2.218, 95% CI = 1.559–3.156, P < 0.001; Fig. 3F).

Construction of the nomogram and calibration curves

We constructed the nomogram by combining the risk score with other clinicopathological risk factors. The nomogram showed that our risk score was the most important factor among the various clinical parameters (Fig. 4A). In addition, calibration curves revealed that the predicted and actual survival rates were well matched at 1-, 3-, and 5-years (Fig. 4B-E).

The risk assessment model had a favorable prognostic prediction in patients with different clinical characteristics

The risk score was used to predict the OS of the patients according to age, gender, pathologic stage, grade and altered fraction genome. We could see that the survival time of the high-risk group was obviously shorter than that of the low-risk group in each group (Fig. 5A-J). Therefore, we could use the risk assessment model to predict the OS of patients in clinical practice, providing a strong reference for doctors to adjust the treatment promptly.

GSEA and small molecular drug screening

We performed differential expression analysis between the high- and low-risk groups according to the median risk score. A total of 5803 DEGs, including 439 downregulated DEGs and 5364 upregulated DEGs, were finally identified using the screening criteria: P value < 1E-5 (Fig. 6B). We used the GSEA method to analyze a whole-genome dataset of GC samples between the different risk groups to further understand the molecular mechanism. GSEA analysis using c2 as a reference gene set revealed that biological processes of ECM receptor interaction, focal adhesion, gap junction, cell adhesion molecules cams and cytokine receptor interaction were significantly related to the high-risk group (Fig. 6A).
All the DEGs related to risk score were divided into upregulated and downregulated groups, which were uploaded to the CMAP database. Three small molecular drugs with anticancer properties on GC progression were identified (enrichment score < 0 and p < 0.0001): genistein, adiphenine and biomycin (Supplementary Table S4). The chemical structures of these drugs are shown (Fig. 6C).

Validating the Expression of nine Genes

We used quantitative reverse transcription PCR (qRT–PCR) assays to compare the expression of the gene signature in GC cell lines and normal cell line. Different differential expression can be found in different cell lines, and the results are detailed in Fig. 7. The mRNA levels of GLT8D1, NRP1, PPP1R26, SERPINE1, TMSB15A and ZFYVE27 showed high expression overall, and AARSD1 was highly expressed only in HGC-27, MGC-803 and BGC-823 cells, yet GPX3 and OLFM3 were significantly lower in GC cell lines. Again, as shown in Fig. 8, there is a significant difference in the relative expression levels of these genes in tumor tissue and matched adjacent normal tissues. In addition, immunohistochemical staining was performed to confirm the protein expression of the gene signature using the HPA online site. We compared the differential expression of seven target genes in normal and malignant tissues, except for TMSB15A and OLFM3. The degree of staining for GPX3 and PPP1R26 was stronger in normal tissues than in cancer tissues; on the other hand, ZFYVE27 showed the opposite trend (Fig. 9).

Discussion

Emerging clinical applications of targeted therapy and immunotherapy underscore the importance of the TME, the complex regulatory network of which poses great challenges to therapeutic efficacy. In light of the dominant sector in the TME and its functional heterogeneity, CAFs have gradually become an intense area of research. On the one hand, it is important for further exploration of tumor mechanisms in CAFs to develop more novel therapeutic targets, but the prediction of prognosis is also a vital part of clinical decisions. However, the specific markers and origin of CAFs remain controversial. In this study, by integrated single-cell and RNA sequencing analysis, a novel signature in GC was developed to identify feature genes of CAFs.
CAFs, as the absolute dominant component of the tumor stroma, secrete various components that participate in constituting and remodeling the ECM. We observed that the higher the abundance of fibroblasts was, the poorer the survival of patients with GC. The reason for this may be that the dense ECM forms a physical barrier that promotes tumor progression and prevents drug penetration [28]. As with the results of our analysis, fibroblast content can be utilized to predict prognosis, which has been validated in numerous tumors [2933]. In particular, there is a highly aggressive subtype of GC with a very poor prognosis –scirrhous gastric cancer (SGC), which is characterized by rapid infiltration and proliferation of tumor cells with extensive stromal fibrosis [34]. In this fibrotic TME of SGC, researchers explored the biological behavior by constructing SGC cell lines and mouse models [35], gradually depicting the crosstalk between tumor cells and CAFs [34, 36].
CAFs have been demonstrated to promote migration and EMT in GC by activating the JAK2/STAT3 signaling pathway through the secretion of IL-6 [5], as well as activation of the ERK1/2-SP1-ZEB2 pathway via the secretion of IL-33 [37]. Other factors induced by CAFs, such as IL-11, IL-22, IL-17a, FGF9, TGFβ1, lumican, LOXL2, SDR1 and CXCL12, are also involved in the migration and invasion of GC [3840]. Likewise, CAF-derived galectin-1 and HGF can promote angiogenesis, supporting the progression of GC [41, 42]. Acquired drug resistance severely affects patient treatment prognosis. Numerous studies have shown that CAFs play an important role in mediating drug resistance [43]. CAFs can regulate drug resistance via the secretion of the IL-11-mediated gp120/JAK/STAT3/Bcl2 pathway [7], and activate the PI3K/AKT signaling pathway by generating IL-8, which causes NF-B activation and cisplatin resistance [44]. In addition, Yang et al. found that CAFs can promote chemoresistance by mediating VEGF/NRP2 signaling via CXCL12 secretion [45]. Emerging evidence has demonstrated that CAFs can also affect tumor progression and drug resistance by forming extracellular vesicles (EVs). Studies have shown that CD9-positive exosomes generated from CAFs can be taken up by SGC cells, which promote cancer cell migration and invasion by activating the MMP2 signaling pathway [46]. Similarly, exosomal circ_0088300 derived from CAFs promotes GC malignancy by activating miR-1305/JAK/STAT1 [47], and annexin A6 in CAF-EVs induces drug resistance via activation of β1 integrin-FAK-YAP [48]. Nonetheless, CAF-derived exosomal miRNA-34 and miRNA-139 could inhibit the progression of GC [49, 50]. Collectively, the mystery of the diverse biological functions of CAFs is gradually being unraveled, for which we will also further explore the value of their clinical application.
In this study, we performed scRNA-seq profiling to reveal the fibroblast subset and identify CAF-related marker genes. A total of 280 feature genes were obtained with the intersection of the stable CAF-related DEGs and the prognosis-related genes. By LASSO Cox regression, we successfully constructed and validated a novel 9-gene CAF-related signature to predict the prognosis of GC, and the signature was confirmed as an independent predictor of OS by univariate and multivariate Cox regression analyses. Most of these CAF-related genes are associated with tumorigenesis and cancer progression, including GLT8D1 [51], GPX3 [52], NRP1 [53], PPP1R26 [54], SERPINE1 [55], and TMSB15A [56]. Of these, we focused on SERPINE1, one of the genes upregulated in this gene signature, which encodes plasminogen activator inhibitor-1 (PAI-1). Studies confirm that its overexpression is involved in the progression and unfavorable outcomes in various cancers [55]. Sakamoto et al. proved that PAI-1 from CAFs stimulated esophageal squamous cell carcinoma (ESCC) cell migration and invasion through contact with LRP1 via phosphorylation of Akt and Erk1/2 [57]. Furthermore, CAFs induced M2 polarization in macrophages by secreting CXCL12, which in turn induced PAI-1 secretion and enhanced the malignant behavior of HCC [58]. As a result, the gene signature we constructed can serve as a target reference for CAFs in tumor research. However, the detailed mechanisms in GC warrant further investigation.
Subsequently, we tried to discover promising small molecular drugs for gene therapy of CAF-related gene signatures in GC patients. Traditional Chinese herbal extracts have been demonstrated to be effective in slowing the progression of GC. For example triptonide, a small molecule (MW358) extracted from Tripterygium wilfordii Hook F, efficiently inhibits development and metastasis by blocking the oncogenic Notch1 and NF-B signaling pathways [59]. Wang et al. discovered that several natural products inhibit CAF activity in a series of investigations. By rectifying aberrant microRNA expression, astragaloside IV and treponil restricted the malignancy-promoting capacity of CAFs [60, 61]. In contract, Paeoniflorin suppressed the malignancy of CAFs by decreasing its IL-6 secretion [62]. In this study, we screened three small molecular drugs for the treatment of CAFs. The one with the most significant p value is the one we are interested in, Genistein is a phytoestrogen and a naturally occurring chemical constituent found primarily in legumes. It has anticancer properties, and studies have shown that by targeting distinct biological processes, it can suppress the growth of various cancer cells [63]. In regard to GC research, genistein inhibits tumor cell proliferation by suppressing cancer stem cell-like properties and inducing G2/M arrest [64, 65], as well as improving chemotherapy sensitivity by inhibiting ERK1/2 activity [64]. Nevertheless, the practical application of these potentially therapeutic small molecule compounds requires further exploration and validation.

Conclusions

We identified a novel CAF-related gene signature for GC by integrating single-cell and bulk RNA sequencing analysis, and these differentially expressed genes might become valuable prognostic and therapeutic targets. We also validated them by multiple approaches and successfully screened genistein, adiphenine and viomycin as potential therapeutic drugs for the treatment of GC patients. However, further studies are still needed to validate the specific mechanisms and effectiveness of these differential genes and therapeutic drugs in GC.

Acknowledgements

We thank the peer reviewers for their input, which improved this manuscript. And we thank all researchers and staff who uploaded and maintained data in TCGA and GEO databases for their great efforts. Meanwhile, we would like to express our gratitude to the team of director Li of Gastrointestinal Surgery department of the First Affiliated Hospital of Nanchang University for the source of the cells.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors report no conflicts of interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209–49.CrossRef Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209–49.CrossRef
2.
Zurück zum Zitat Sekiguchi M, Oda I, Matsuda T, Saito Y. Epidemiological trends and future perspectives of Gastric Cancer in Eastern Asia. Digestion. 2022;103:22–8.CrossRef Sekiguchi M, Oda I, Matsuda T, Saito Y. Epidemiological trends and future perspectives of Gastric Cancer in Eastern Asia. Digestion. 2022;103:22–8.CrossRef
3.
Zurück zum Zitat Xiao Y, Yu D. Tumor microenvironment as a therapeutic target in cancer. Pharmacol Ther. 2021;221: 107753.CrossRef Xiao Y, Yu D. Tumor microenvironment as a therapeutic target in cancer. Pharmacol Ther. 2021;221: 107753.CrossRef
4.
Zurück zum Zitat Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016;16:582–98.CrossRef Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016;16:582–98.CrossRef
5.
Zurück zum Zitat Wu X, Tao P, Zhou Q, et al. IL-6 secreted by cancer-associated fibroblasts promotes epithelial-mesenchymal transition and metastasis of gastric cancer via JAK2/STAT3 signaling pathway. Oncotarget. 2017;8:20741–50.CrossRef Wu X, Tao P, Zhou Q, et al. IL-6 secreted by cancer-associated fibroblasts promotes epithelial-mesenchymal transition and metastasis of gastric cancer via JAK2/STAT3 signaling pathway. Oncotarget. 2017;8:20741–50.CrossRef
6.
Zurück zum Zitat Ham IH, Oh HJ, Jin H, et al. Targeting interleukin-6 as a strategy to overcome stroma-induced resistance to chemotherapy in gastric cancer. Mol Cancer. 2019;18:68.CrossRef Ham IH, Oh HJ, Jin H, et al. Targeting interleukin-6 as a strategy to overcome stroma-induced resistance to chemotherapy in gastric cancer. Mol Cancer. 2019;18:68.CrossRef
7.
Zurück zum Zitat Ma J, Song X, Xu X, Mou Y. Cancer-Associated Fibroblasts Promote the Chemo-resistance in Gastric Cancer through Secreting IL-11 Targeting JAK/STAT3/Bcl2 Pathway. Cancer Res Treat. 2019;51:194–210.CrossRef Ma J, Song X, Xu X, Mou Y. Cancer-Associated Fibroblasts Promote the Chemo-resistance in Gastric Cancer through Secreting IL-11 Targeting JAK/STAT3/Bcl2 Pathway. Cancer Res Treat. 2019;51:194–210.CrossRef
8.
Zurück zum Zitat Tao L, Huang G, Song H, Chen Y, Chen L. Cancer associated fibroblasts: an essential role in the tumor microenvironment. Oncol Lett. 2017;14:2611–20.CrossRef Tao L, Huang G, Song H, Chen Y, Chen L. Cancer associated fibroblasts: an essential role in the tumor microenvironment. Oncol Lett. 2017;14:2611–20.CrossRef
9.
Zurück zum Zitat Bu L, Baba H, Yoshida N, et al. Biological heterogeneity and versatility of cancer-associated fibroblasts in the tumor microenvironment. Oncogene. 2019;38:4887–901.CrossRef Bu L, Baba H, Yoshida N, et al. Biological heterogeneity and versatility of cancer-associated fibroblasts in the tumor microenvironment. Oncogene. 2019;38:4887–901.CrossRef
10.
Zurück zum Zitat Biffi G, Tuveson DA. Diversity and biology of Cancer-associated fibroblasts. Physiol Rev. 2021;101:147–76.CrossRef Biffi G, Tuveson DA. Diversity and biology of Cancer-associated fibroblasts. Physiol Rev. 2021;101:147–76.CrossRef
11.
Zurück zum Zitat Sung JY, Cheong JH. New immunometabolic strategy based on cell type-specific metabolic reprogramming in the Tumor immune microenvironment. Cells. 2022;11(5):768.CrossRef Sung JY, Cheong JH. New immunometabolic strategy based on cell type-specific metabolic reprogramming in the Tumor immune microenvironment. Cells. 2022;11(5):768.CrossRef
12.
Zurück zum Zitat Chen Y, McAndrews KM, Kalluri R. Clinical and therapeutic relevance of cancer-associated fibroblasts. Nat Rev Clin Oncol. 2021;18:792–804.CrossRef Chen Y, McAndrews KM, Kalluri R. Clinical and therapeutic relevance of cancer-associated fibroblasts. Nat Rev Clin Oncol. 2021;18:792–804.CrossRef
13.
Zurück zum Zitat Nurmik M, Ullmann P, Rodriguez F, Haan S, Letellier E. In search of definitions: Cancer-associated fibroblasts and their markers. Int J Cancer. 2020;146:895–905.CrossRef Nurmik M, Ullmann P, Rodriguez F, Haan S, Letellier E. In search of definitions: Cancer-associated fibroblasts and their markers. Int J Cancer. 2020;146:895–905.CrossRef
14.
Zurück zum Zitat Joshi RS, Kanugula SS, Sudhir S, Pereira MP, Jain S, Aghi MK. The role of Cancer-associated fibroblasts in Tumor progression. Cancers (Basel). 2021;13(6):1399.CrossRef Joshi RS, Kanugula SS, Sudhir S, Pereira MP, Jain S, Aghi MK. The role of Cancer-associated fibroblasts in Tumor progression. Cancers (Basel). 2021;13(6):1399.CrossRef
15.
Zurück zum Zitat Lau EY, Lo J, Cheng BY, et al. Cancer-Associated Fibroblasts Regulate Tumor-Initiating Cell Plasticity in Hepatocellular Carcinoma through c-Met/FRA1/HEY1 Signaling. Cell Rep. 2016;15:1175–89.CrossRef Lau EY, Lo J, Cheng BY, et al. Cancer-Associated Fibroblasts Regulate Tumor-Initiating Cell Plasticity in Hepatocellular Carcinoma through c-Met/FRA1/HEY1 Signaling. Cell Rep. 2016;15:1175–89.CrossRef
16.
Zurück zum Zitat Fang M, Yuan J, Chen M, et al. The heterogenic tumor microenvironment of hepatocellular carcinoma and prognostic analysis based on tumor neo-vessels, macrophages and α-SMA. Oncol Lett. 2018;15:4805–12. Fang M, Yuan J, Chen M, et al. The heterogenic tumor microenvironment of hepatocellular carcinoma and prognostic analysis based on tumor neo-vessels, macrophages and α-SMA. Oncol Lett. 2018;15:4805–12.
17.
Zurück zum Zitat Tsujino T, Seshimo I, Yamamoto H, et al. Stromal myofibroblasts predict disease recurrence for colorectal cancer. Clin Cancer Res. 2007;13:2082–90.CrossRef Tsujino T, Seshimo I, Yamamoto H, et al. Stromal myofibroblasts predict disease recurrence for colorectal cancer. Clin Cancer Res. 2007;13:2082–90.CrossRef
18.
Zurück zum Zitat Surowiak P, Murawa D, Materna V, et al. Occurence of stromal myofibroblasts in the invasive ductal breast cancer tissue is an unfavourable prognostic factor. Anticancer Res. 2007;27:2917–24. Surowiak P, Murawa D, Materna V, et al. Occurence of stromal myofibroblasts in the invasive ductal breast cancer tissue is an unfavourable prognostic factor. Anticancer Res. 2007;27:2917–24.
19.
Zurück zum Zitat Calon A, Lonardo E, Berenguer-Llergo A, et al. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat Genet. 2015;47:320–9.CrossRef Calon A, Lonardo E, Berenguer-Llergo A, et al. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat Genet. 2015;47:320–9.CrossRef
20.
Zurück zum Zitat Zou B, Liu X, Gong Y, et al. A novel 12-marker panel of cancer-associated fibroblasts involved in progression of hepatocellular carcinoma. Cancer Manag Res. 2018;10:5303–11.CrossRef Zou B, Liu X, Gong Y, et al. A novel 12-marker panel of cancer-associated fibroblasts involved in progression of hepatocellular carcinoma. Cancer Manag Res. 2018;10:5303–11.CrossRef
21.
Zurück zum Zitat Petitprez F, Vano YA, Becht E, et al. Transcriptomic analysis of the tumor microenvironment to guide prognosis and immunotherapies. Cancer Immunol Immunother. 2018;67:981–8.CrossRef Petitprez F, Vano YA, Becht E, et al. Transcriptomic analysis of the tumor microenvironment to guide prognosis and immunotherapies. Cancer Immunol Immunother. 2018;67:981–8.CrossRef
22.
Zurück zum Zitat Becht E, Giraldo NA, Lacroix L, et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol. 2016;17:218.CrossRef Becht E, Giraldo NA, Lacroix L, et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol. 2016;17:218.CrossRef
23.
Zurück zum Zitat Stuart T, Butler A, Hoffman P, et al. Comprehensive integration of single-cell data. Cell. 2019;177:1888-1902.e1821.CrossRef Stuart T, Butler A, Hoffman P, et al. Comprehensive integration of single-cell data. Cell. 2019;177:1888-1902.e1821.CrossRef
24.
Zurück zum Zitat Aran D, Looney AP, Liu L, et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat Immunol. 2019;20:163–72.CrossRef Aran D, Looney AP, Liu L, et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat Immunol. 2019;20:163–72.CrossRef
25.
Zurück zum Zitat Ueno D, Kawabe H, Yamasaki S, Demura T, Kato K. Feature selection for RNA cleavage efficiency at specific sites using the LASSO regression model in Arabidopsis thaliana. BMC Bioinformatics. 2021;22:380.CrossRef Ueno D, Kawabe H, Yamasaki S, Demura T, Kato K. Feature selection for RNA cleavage efficiency at specific sites using the LASSO regression model in Arabidopsis thaliana. BMC Bioinformatics. 2021;22:380.CrossRef
26.
Zurück zum Zitat Zito A, Lualdi M, Granata P, et al. Gene set enrichment analysis of interaction networks weighted by node centrality. Front Genet. 2021;12: 577623.CrossRef Zito A, Lualdi M, Granata P, et al. Gene set enrichment analysis of interaction networks weighted by node centrality. Front Genet. 2021;12: 577623.CrossRef
27.
Zurück zum Zitat Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28:27–30.CrossRef Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28:27–30.CrossRef
28.
Zurück zum Zitat Sung JY, Cheong JH. Prognosis-related gene signature is enriched in cancer-associated fibroblasts in the stem-like subtype of gastric cancer. Clin Transl Med. 2022;12: e930.CrossRef Sung JY, Cheong JH. Prognosis-related gene signature is enriched in cancer-associated fibroblasts in the stem-like subtype of gastric cancer. Clin Transl Med. 2022;12: e930.CrossRef
29.
Zurück zum Zitat Liu X, Yao L, Qu J, et al. Cancer-associated fibroblast infiltration in gastric cancer: the discrepancy in subtypes pathways and immunosuppression. J Transl Med. 2021;19:325.CrossRef Liu X, Yao L, Qu J, et al. Cancer-associated fibroblast infiltration in gastric cancer: the discrepancy in subtypes pathways and immunosuppression. J Transl Med. 2021;19:325.CrossRef
30.
Zurück zum Zitat Sandberg TP, Stuart M, Oosting J, Tollenaar R, Sier CFM, Mesker WE. Increased expression of cancer-associated fibroblast markers at the invasive front and its association with tumor-stroma ratio in colorectal cancer. BMC Cancer. 2019;19:284.CrossRef Sandberg TP, Stuart M, Oosting J, Tollenaar R, Sier CFM, Mesker WE. Increased expression of cancer-associated fibroblast markers at the invasive front and its association with tumor-stroma ratio in colorectal cancer. BMC Cancer. 2019;19:284.CrossRef
31.
Zurück zum Zitat Hosein AN, Brekken RA, Maitra A. Pancreatic cancer stroma: an update on therapeutic targeting strategies. Nat Rev Gastroenterol Hepatol. 2020;17:487–505.CrossRef Hosein AN, Brekken RA, Maitra A. Pancreatic cancer stroma: an update on therapeutic targeting strategies. Nat Rev Gastroenterol Hepatol. 2020;17:487–505.CrossRef
32.
Zurück zum Zitat Zhi K, Shen X, Zhang H, Bi J. Cancer-associated fibroblasts are positively correlated with metastatic potential of human gastric cancers. J Exp Clin Cancer Res. 2010;29:66.CrossRef Zhi K, Shen X, Zhang H, Bi J. Cancer-associated fibroblasts are positively correlated with metastatic potential of human gastric cancers. J Exp Clin Cancer Res. 2010;29:66.CrossRef
33.
Zurück zum Zitat Zhang J, Li S, Zhao Y, et al. Cancer-associated fibroblasts promote the migration and invasion of gastric cancer cells via activating IL-17a/JAK2/STAT3 signaling. Ann Transl Med. 2020;8:877.CrossRef Zhang J, Li S, Zhao Y, et al. Cancer-associated fibroblasts promote the migration and invasion of gastric cancer cells via activating IL-17a/JAK2/STAT3 signaling. Ann Transl Med. 2020;8:877.CrossRef
34.
Zurück zum Zitat Yashiro M, Hirakawa K. Cancer-stromal interactions in scirrhous gastric carcinoma. Cancer Microenviron. 2010;3:127–35.CrossRef Yashiro M, Hirakawa K. Cancer-stromal interactions in scirrhous gastric carcinoma. Cancer Microenviron. 2010;3:127–35.CrossRef
35.
Zurück zum Zitat Yashiro M, Matsuoka T, Ohira M. The significance of scirrhous gastric cancer cell lines: the molecular characterization using cell lines and mouse models. Hum Cell. 2018;31:271–81.CrossRef Yashiro M, Matsuoka T, Ohira M. The significance of scirrhous gastric cancer cell lines: the molecular characterization using cell lines and mouse models. Hum Cell. 2018;31:271–81.CrossRef
36.
Zurück zum Zitat Miki Y, Yashiro M, Moyano-Galceran L, Sugimoto A, Ohira M, Lehti K. Crosstalk Between Cancer Associated Fibroblasts and Cancer Cells in Scirrhous Type Gastric Cancer. Front Oncol. 2020;10: 568557.CrossRef Miki Y, Yashiro M, Moyano-Galceran L, Sugimoto A, Ohira M, Lehti K. Crosstalk Between Cancer Associated Fibroblasts and Cancer Cells in Scirrhous Type Gastric Cancer. Front Oncol. 2020;10: 568557.CrossRef
37.
Zurück zum Zitat Zhou Q, Wu X, Wang X, et al. The reciprocal interaction between tumor cells and activated fibroblasts mediated by TNF-α/IL-33/ST2L signaling promotes gastric cancer metastasis. Oncogene. 2020;39:1414–28.CrossRef Zhou Q, Wu X, Wang X, et al. The reciprocal interaction between tumor cells and activated fibroblasts mediated by TNF-α/IL-33/ST2L signaling promotes gastric cancer metastasis. Oncogene. 2020;39:1414–28.CrossRef
38.
Zurück zum Zitat Ham IH, Lee D, Hur H. Role of Cancer-associated fibroblast in Gastric Cancer progression and resistance to treatments. J Oncol. 2019;2019:6270784.CrossRef Ham IH, Lee D, Hur H. Role of Cancer-associated fibroblast in Gastric Cancer progression and resistance to treatments. J Oncol. 2019;2019:6270784.CrossRef
39.
Zurück zum Zitat Izumi D, Ishimoto T, Miyake K, et al. CXCL12/CXCR4 activation by cancer-associated fibroblasts promotes integrin β1 clustering and invasiveness in gastric cancer. Int J Cancer. 2016;138:1207–19.CrossRef Izumi D, Ishimoto T, Miyake K, et al. CXCL12/CXCR4 activation by cancer-associated fibroblasts promotes integrin β1 clustering and invasiveness in gastric cancer. Int J Cancer. 2016;138:1207–19.CrossRef
40.
Zurück zum Zitat Qin Y, Wang F, Ni H, et al. Cancer-associated fibroblasts in gastric cancer affect malignant progression via the CXCL12-CXCR4 axis. J Cancer. 2021;12:3011–23.CrossRef Qin Y, Wang F, Ni H, et al. Cancer-associated fibroblasts in gastric cancer affect malignant progression via the CXCL12-CXCR4 axis. J Cancer. 2021;12:3011–23.CrossRef
41.
Zurück zum Zitat Tang D, Gao J, Wang S, et al. Cancer-associated fibroblasts promote angiogenesis in gastric cancer through galectin-1 expression. Tumour Biol. 2016;37:1889–99.CrossRef Tang D, Gao J, Wang S, et al. Cancer-associated fibroblasts promote angiogenesis in gastric cancer through galectin-1 expression. Tumour Biol. 2016;37:1889–99.CrossRef
42.
Zurück zum Zitat Ding X, Xi W, Ji J, et al. HGF derived from cancer-associated fibroblasts promotes vascularization in gastric cancer via PI3K/AKT and ERK1/2 signaling. Oncol Rep. 2018;40:1185–95. Ding X, Xi W, Ji J, et al. HGF derived from cancer-associated fibroblasts promotes vascularization in gastric cancer via PI3K/AKT and ERK1/2 signaling. Oncol Rep. 2018;40:1185–95.
43.
Zurück zum Zitat Sun Y, Wang R, Qiao M, Xu Y, Guan W, Wang L. Cancer associated fibroblasts tailored tumor microenvironment of therapy resistance in gastrointestinal cancers. J Cell Physiol. 2018;233:6359–69.CrossRef Sun Y, Wang R, Qiao M, Xu Y, Guan W, Wang L. Cancer associated fibroblasts tailored tumor microenvironment of therapy resistance in gastrointestinal cancers. J Cell Physiol. 2018;233:6359–69.CrossRef
44.
Zurück zum Zitat Zhai J, Shen J, Xie G, et al. Cancer-associated fibroblasts-derived IL-8 mediates resistance to cisplatin in human gastric cancer. Cancer Lett. 2019;454:37–43.CrossRef Zhai J, Shen J, Xie G, et al. Cancer-associated fibroblasts-derived IL-8 mediates resistance to cisplatin in human gastric cancer. Cancer Lett. 2019;454:37–43.CrossRef
45.
Zurück zum Zitat Yang Y, Ma Y, Yan S, et al.: CAF promotes chemoresistance through NRP2 in gastric cancer. Gastric Cancer2021. Yang Y, Ma Y, Yan S, et al.: CAF promotes chemoresistance through NRP2 in gastric cancer. Gastric Cancer2021.
46.
Zurück zum Zitat Miki Y, Yashiro M, Okuno T, et al. CD9-positive exosomes from cancer-associated fibroblasts stimulate the migration ability of scirrhous-type gastric cancer cells. Br J Cancer. 2018;118:867–77.CrossRef Miki Y, Yashiro M, Okuno T, et al. CD9-positive exosomes from cancer-associated fibroblasts stimulate the migration ability of scirrhous-type gastric cancer cells. Br J Cancer. 2018;118:867–77.CrossRef
47.
Zurück zum Zitat Shi H, Huang S, Qin M, et al. Exosomal circ_0088300 Derived From Cancer-Associated Fibroblasts Acts as a miR-1305 Sponge and Promotes Gastric Carcinoma Cell Tumorigenesis. Front Cell Dev Biol. 2021;9: 676319.CrossRef Shi H, Huang S, Qin M, et al. Exosomal circ_0088300 Derived From Cancer-Associated Fibroblasts Acts as a miR-1305 Sponge and Promotes Gastric Carcinoma Cell Tumorigenesis. Front Cell Dev Biol. 2021;9: 676319.CrossRef
48.
Zurück zum Zitat Uchihara T, Miyake K, Yonemura A, et al. Extracellular Vesicles from Cancer-Associated Fibroblasts Containing Annexin A6 Induces FAK-YAP Activation by Stabilizing β1 Integrin. Enhancing Drug Resistance Cancer Res. 2020;80:3222–35. Uchihara T, Miyake K, Yonemura A, et al. Extracellular Vesicles from Cancer-Associated Fibroblasts Containing Annexin A6 Induces FAK-YAP Activation by Stabilizing β1 Integrin. Enhancing Drug Resistance Cancer Res. 2020;80:3222–35.
49.
Zurück zum Zitat Shi L, Wang Z, Geng X, Zhang Y, Xue Z. Exosomal miRNA-34 from cancer-associated fibroblasts inhibits growth and invasion of gastric cancer cells in vitro and in vivo. Aging (Albany NY). 2020;12:8549–64.CrossRef Shi L, Wang Z, Geng X, Zhang Y, Xue Z. Exosomal miRNA-34 from cancer-associated fibroblasts inhibits growth and invasion of gastric cancer cells in vitro and in vivo. Aging (Albany NY). 2020;12:8549–64.CrossRef
50.
Zurück zum Zitat Xu G, Zhang B, Ye J, et al. Exosomal miRNA-139 in cancer-associated fibroblasts inhibits gastric cancer progression by repressing MMP11 expression. Int J Biol Sci. 2019;15:2320–9.CrossRef Xu G, Zhang B, Ye J, et al. Exosomal miRNA-139 in cancer-associated fibroblasts inhibits gastric cancer progression by repressing MMP11 expression. Int J Biol Sci. 2019;15:2320–9.CrossRef
51.
Zurück zum Zitat Hu H, Li Z, Zhou Y, et al. GLT8D1 overexpression as a novel prognostic biomarker in human cutaneous melanoma. Melanoma Res. 2019;29:612–20.CrossRef Hu H, Li Z, Zhou Y, et al. GLT8D1 overexpression as a novel prognostic biomarker in human cutaneous melanoma. Melanoma Res. 2019;29:612–20.CrossRef
52.
Zurück zum Zitat Cai M, Sikong Y, Wang Q, Zhu S, Pang F, Cui X. Gpx3 prevents migration and invasion in gastric cancer by targeting NFкB/Wnt5a/JNK signaling. Int J Clin Exp Pathol. 2019;12:1194–203. Cai M, Sikong Y, Wang Q, Zhu S, Pang F, Cui X. Gpx3 prevents migration and invasion in gastric cancer by targeting NFкB/Wnt5a/JNK signaling. Int J Clin Exp Pathol. 2019;12:1194–203.
53.
Zurück zum Zitat Mei B, Chen J, Yang N, Peng Y. The regulatory mechanism and biological significance of the Snail-miR590-VEGFR-NRP1 axis in the angiogenesis, growth and metastasis of gastric cancer. Cell Death Dis. 2020;11:241.CrossRef Mei B, Chen J, Yang N, Peng Y. The regulatory mechanism and biological significance of the Snail-miR590-VEGFR-NRP1 axis in the angiogenesis, growth and metastasis of gastric cancer. Cell Death Dis. 2020;11:241.CrossRef
54.
Zurück zum Zitat Zheng T, Lu M, Wang T, Zhang C, Du X. NRBE3 promotes metastasis of breast cancer by down-regulating E-cadherin expression. Biochim Biophys Acta Mol Cell Res. 1865;1869–1877:2018. Zheng T, Lu M, Wang T, Zhang C, Du X. NRBE3 promotes metastasis of breast cancer by down-regulating E-cadherin expression. Biochim Biophys Acta Mol Cell Res. 1865;1869–1877:2018.
55.
Zurück zum Zitat Kubala MH, DeClerck YA. The plasminogen activator inhibitor-1 paradox in cancer: a mechanistic understanding. Cancer Metastasis Rev. 2019;38:483–92.CrossRef Kubala MH, DeClerck YA. The plasminogen activator inhibitor-1 paradox in cancer: a mechanistic understanding. Cancer Metastasis Rev. 2019;38:483–92.CrossRef
56.
Zurück zum Zitat Darb-Esfahani S, Kronenwett R, von Minckwitz G, et al. Thymosin beta 15A (TMSB15A) is a predictor of chemotherapy response in triple-negative breast cancer. Br J Cancer. 2012;107:1892–900.CrossRef Darb-Esfahani S, Kronenwett R, von Minckwitz G, et al. Thymosin beta 15A (TMSB15A) is a predictor of chemotherapy response in triple-negative breast cancer. Br J Cancer. 2012;107:1892–900.CrossRef
57.
Zurück zum Zitat Sakamoto H, Koma YI, Higashino N, et al. PAI-1 derived from cancer-associated fibroblasts in esophageal squamous cell carcinoma promotes the invasion of cancer cells and the migration of macrophages. Lab Invest. 2021;101:353–68.CrossRef Sakamoto H, Koma YI, Higashino N, et al. PAI-1 derived from cancer-associated fibroblasts in esophageal squamous cell carcinoma promotes the invasion of cancer cells and the migration of macrophages. Lab Invest. 2021;101:353–68.CrossRef
58.
Zurück zum Zitat Chen S, Morine Y, Tokuda K, et al. Cancer-associated fibroblast-induced M2-polarized macrophages promote hepatocellular carcinoma progression via the plasminogen activator inhibitor-1 pathway. Int J Oncol. 2021;59(2):59.CrossRef Chen S, Morine Y, Tokuda K, et al. Cancer-associated fibroblast-induced M2-polarized macrophages promote hepatocellular carcinoma progression via the plasminogen activator inhibitor-1 pathway. Int J Oncol. 2021;59(2):59.CrossRef
59.
Zurück zum Zitat Xiang S, Zhao Z, Zhang T, et al. Triptonide effectively suppresses gastric tumor growth and metastasis through inhibition of the oncogenic Notch1 and NF-κB signaling pathways. Toxicol Appl Pharmacol. 2020;388: 114870.CrossRef Xiang S, Zhao Z, Zhang T, et al. Triptonide effectively suppresses gastric tumor growth and metastasis through inhibition of the oncogenic Notch1 and NF-κB signaling pathways. Toxicol Appl Pharmacol. 2020;388: 114870.CrossRef
60.
Zurück zum Zitat Wang ZF, Ma DG, Zhu Z, et al. Astragaloside IV inhibits pathological functions of gastric cancer-associated fibroblasts. World J Gastroenterol. 2017;23:8512–25.CrossRef Wang ZF, Ma DG, Zhu Z, et al. Astragaloside IV inhibits pathological functions of gastric cancer-associated fibroblasts. World J Gastroenterol. 2017;23:8512–25.CrossRef
61.
Zurück zum Zitat Wang Z, Ma D, Wang C, et al. Triptonide inhibits the pathological functions of gastric cancer-associated fibroblasts. Biomed Pharmacother. 2017;96:757–67.CrossRef Wang Z, Ma D, Wang C, et al. Triptonide inhibits the pathological functions of gastric cancer-associated fibroblasts. Biomed Pharmacother. 2017;96:757–67.CrossRef
62.
Zurück zum Zitat Wang ZF, Ma DG, Wang L, et al. Paeoniflorin inhibits migration- and invasion-promoting capacities of Gastric Cancer Associated Fibroblasts. Chin J Integr Med. 2019;25:837–44.CrossRef Wang ZF, Ma DG, Wang L, et al. Paeoniflorin inhibits migration- and invasion-promoting capacities of Gastric Cancer Associated Fibroblasts. Chin J Integr Med. 2019;25:837–44.CrossRef
63.
Zurück zum Zitat Spagnuolo C, Russo GL, Orhan IE, et al. Genistein and cancer: current status, challenges, and future directions. Adv Nutr. 2015;6:408–19.CrossRef Spagnuolo C, Russo GL, Orhan IE, et al. Genistein and cancer: current status, challenges, and future directions. Adv Nutr. 2015;6:408–19.CrossRef
64.
Zurück zum Zitat Huang W, Wan C, Luo Q, Huang Z, Luo Q. Genistein-inhibited cancer stem cell-like properties and reduced chemoresistance of gastric cancer. Int J Mol Sci. 2014;15:3432–43.CrossRef Huang W, Wan C, Luo Q, Huang Z, Luo Q. Genistein-inhibited cancer stem cell-like properties and reduced chemoresistance of gastric cancer. Int J Mol Sci. 2014;15:3432–43.CrossRef
65.
Zurück zum Zitat Liu YL, Zhang GQ, Yang Y, Zhang CY, Fu RX, Yang YM. Genistein induces G2/M arrest in gastric cancer cells by increasing the tumor suppressor PTEN expression. Nutr Cancer. 2013;65:1034–41.CrossRef Liu YL, Zhang GQ, Yang Y, Zhang CY, Fu RX, Yang YM. Genistein induces G2/M arrest in gastric cancer cells by increasing the tumor suppressor PTEN expression. Nutr Cancer. 2013;65:1034–41.CrossRef
Metadaten
Titel
Integrated single-cell and bulk RNA sequencing analysis identifies a cancer-associated fibroblast-related gene signature for predicting survival and therapy in gastric cancer
verfasst von
Zhiyang Zhou
Sixuan Guo
Shuhui Lai
Tao Wang
Yao Du
Junping Deng
Shun Zhang
Ge Gao
Jiangnan Zhang
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2023
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-022-10332-w

Weitere Artikel der Ausgabe 1/2023

BMC Cancer 1/2023 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.