Abstract Background CXCR7 (ACKR3) has been well-supported as a promoter of growth and metastasis in hepatocellular carcinoma (HCC). Both CXCR7 and Hippo signaling play roles in organ development. We aimed to verify the involvement of Hippo-YAP signaling in CXCR7-regulated HCC proliferation, migration, and invasion. Methods HCCLM3 cells were transfected with si-CXCR7, pcDNA-CXCR7, or related control RNA/empty vector. Cell proliferation was assessed using the Cell Counting Kit-8 (CCK-8), and mRNA and protein levels were measured via quantitative real-time PCR (qPCR) and Western blotting. Colony formation assays were conducted to evaluate proliferation capacity, and Transwell assays were used to assess invasion and migration. Transcriptome data from the TCGA-LIHC dataset were analyzed to investigate the potential effects of CXCR7 in HCC. Results si-CXCR7 inhibited cell proliferation in HCCLM3 cells, while pcDNA-CXCR7 promoted it. Migration and invasion were suppressed by si-CXCR7 but enhanced by pcDNA-CXCR7. Patients with higher CXCR7 expression in the TCGA-LIHC dataset had lower overall survival rates and increased gene transcription. The CXCR7-high expressing samples were characterized by the activation of several pathways, including PI3K-AKT signaling, calcium signaling, and the Hippo signaling pathway. si-CXCR7 reduced the relative protein levels of Gαq/11 and GαS while increasing phosphorylated LATS and phosphorylated YAP. Opposite trends in these proteins were observed with pcDNA-CXCR7. Finally, the inhibitory effects of si-CXCR7 on cell proliferation, migration, and invasion were reversed by the YAP inhibitor verteporfin. Conclusion We suggest that CXCR7 promotes the growth and metastasis of HCC cells, at least in part, by inactivating the Hippo-YAP signaling pathway. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-025-02324-6. Keywords: ACKR3, CXCL12, Liver cancer, Metastases, RDC1, TAZ Highlights * CXCR7 regulates HCC progression possibly through the inactivation of the Hippo-YAP signaling pathway. * YAP inhibition reverses the oncogenic effects of CXCR7, including cell proliferation, migration, and invasion. * Transcriptome analysis from the TCGA-LIHC dataset links high CXCR7 expression to PI3K-AKT signaling, the calcium signaling, MAPK, as well as Hippo signaling pathway. Introduction Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide [[32]1], accounting for over 80% of all liver cancers and ranking among the top causes of cancer-related deaths in many countries [[33]2]. The high mortality rate and extremely low 5-year overall survival rate of HCC are primarily due to the challenges in early detection [[34]3]. Therefore, exploring the complex regulation of metastasis in the tumor microenvironment (TME) is critical for developing effective therapeutic strategies [[35]4], particularly for HCC. CXC chemokine receptor (CXCR) signaling, mediated by CXCL12 (SDF1), CXCR4, and CXCR7, plays a pivotal role in organ development and normal liver regeneration [[36]5–[37]7], and is widely dysregulated in various cancers [[38]8]. In HCC, the co-expression of CXCL12 and CXCR4/CXCR7 is strongly associated with patient survival rates [[39]9]. CXCR7, also known as ACKR3 or RDC1, exhibits a much higher affinity for CXCL12 than CXCR4, underscoring its pivotal role in CXCL12-CXCR4/CXCR7 chemokine signaling within the TME [[40]10]. CXCR7 functions as a CXCL12 scavenger receptor, predominantly expressed in liver endothelial cells, with its expression further upregulated in HCC [[41]11]. It regulates cell migration, particularly in endothelial cells, by establishing local CXCL12 gradients [[42]12]. Through β-arrestin-dependent or Gαi-dependent mechanisms, CXCR7 activates signaling pathways such as ERK/Akt, MAPK, and JAK/STAT. These activated pathways subsequently can promote processes like epithelial-to-mesenchymal transition (EMT), angiogenesis, migration, and tumor cell invasion [[43]13, [44]14]. Notably, the downregulation of CXCR7 has been shown to inhibit the proliferation, invasion, and migration of HCC cells [[45]15, [46]16]. Hippo signaling is a critical regulator of tissue homeostasis, organ development, regeneration, and cancer progression, with context-dependent effects [[47]17]. This evolutionarily conserved pathway mainly involves serine/threonine-protein kinases 4/3 (MST1/2), large tumor suppressor kinases (LATS) 1/2, yes-associated protein (YAP), and its paralog WW domain-containing transcription regulator protein 1 (WWTR1, also known as transcriptional co-activator with PDZ-binding motif [TAZ]) [[48]18]. In HCC, Hippo signaling restricts tumor progression by phosphorylating YAP/TAZ to destabilize them, thereby inhibiting the transcription of cancer-promoting genes [[49]19]. Specifically, activating Hippo signaling inhibits the migration and invasion of HCC cells, as demonstrated by the effects of Pien Tze Huang, Tubuloside B, and miR-100 [[50]16, [51]20, [52]21]. The healthy liver has a strong regenerative capacity [[53]22], and both CXCR7 and Hippo signaling are involved in organ development and HCC progression. Interestingly, a novel feedback loop between Hippo signaling and CXCR7 has been identified in gastric cancer by Wang et al. CXCR7 can dephosphorylate YAP via the Gαq/11-ROCK-LATS axis, enhancing YAP’s nuclear localization, which in turn upregulates CXCR7 transcription, thereby promoting gastric cancer progression [[54]23]. However, whether CXCR7-mediated chemokine signaling interacts with Hippo signaling in HCC remains unknown. This study aims to verify the involvement of Hippo-YAP signaling in CXCR7-regulated HCC cell proliferation, migration, and invasion. Materials and methods Cell line information and cell transfection The hepatic carcinoma cell line HCCLM3, obtained from Shanghai FuHeng Biotechnology Co., Ltd. (Catalog #FH0096), was utilized in this study. This cell line exhibits high metastatic potential and adherent growth characteristics. Cultivation was carried out in DMEM culture medium with high glucose (Catalog #C3113-0500, VivaCell, Shanghai, China), supplemented with 10% fetal bovine serum (Catalog #04-001-1ACS, VivaCell) and 1% penicillin–streptomycin (Catalog #BL505A, Biosharp, Hefei, China). Based on the gene sequence of CXCR7 ([55]NM_020311.3), an overexpression sequence and a small interfering RNA (siRNA) targeting CXCR7 (5′-CGCUCUCCUUCAUUUACAUUU-3′) were designed and synthesized by Shanghai Genechem Co., Ltd. The coding sequence (CDS) of CXCR7 was cloned into the pcDNA3.1 vector (pCAMBIA1301, Catalog #HZB800277, HZbscience, Hangzhou, China) to generate the recombinant overexpression vector pcDNA-CXCR7. As corresponding controls, si-NC (sense: 5′-ACGUGACACGUUCGGAGAAUU-3′ and antisense: 5′-UUCUCCGAACGUGUCACGU-3′) and an empty vector were used. HCCLM3 cells were resuspended and adjusted to a concentration of 8 × 10^5 cells/well before being seeded into a 6-well plate. For transfection, the overexpression plasmid or si-CXCR7 was mixed with PEI reagent (Catalog #23966-2, YEASEN, Shanghai, China) in Opti-MEM™ medium (Invitrogen, USA) and pre-incubated at room temperature for 20 min to form DNA-PEI complexes. The transfection complexes were then added to the cells in serum-free medium. After a 6-h incubation, the medium was replaced with complete growth medium, and the cells were further incubated at 37 °C for 24 h. Transfection efficiency was subsequently assessed. CCK-8 activity assay and IC50 determination of verteporfin Cells in the logarithmic growth phase were seeded into a 96-well plate at a density of 3000 cells/well, with each group replicated three times, and incubated at 37 °C. At predetermined time points (0, 24, 48, 72, and 96 h), CCK-8 solution (Catalog #C0037, Beyotime, Shanghai, China) was added, and the plates were returned to the incubator for an additional hour of incubation. Wells containing only the cell culture medium and CCK-8 solution, without cells, served as blank controls. Absorbance was measured at 450 nm, and the data were recorded. Verteporfin (VP; Catalog #HY-B0146, ECM, USA) was dissolved in DMSO and further diluted with PBS. HCCLM3 cells were seeded into a 96-well plate at a density of 5,000 cells/well, with each condition replicated in triplicate. Cells were then treated with varying concentrations of Verteporfin (0, 2, 4, 6, 10, 14 μM) and incubated at 37 °C for 72 h. The cytotoxicity of Verteporfin was evaluated using the CCK-8 assay, measuring absorbance at 450 nm to determine the IC50 value. A Verteporfin concentration of 16 μM (median for 16 and 18 μM) was selected for subsequent experiments according to its calculated IC50 (about 17.90 μM). Bioinformatics analysis of CXCR7 Gene expression data specific to CXCR7 in LIHC cases, along with corresponding clinical data, were retrieved from the TCGA database (The Cancer Genome Atlas, [56]https://portal.gdc.cancer.gov). Patients were divided into high and low CXCR7 expression cohorts using the median expression level as a cutoff. Survival times and statuses were used to construct Kaplan–Meier survival curves, with a log-rank test assessing the statistical significance of survival differences between the two groups. Differential expression analysis was conducted using the Wilcoxon rank-sum test, applying criteria of |log2 Fold Change|> 1 and a p-value < 0.05 to identify significantly altered genes. The volcano plot of the differentially expressed genes (DEGs), including key DEGs, was visualized using the R package “ggplot2.” Enrichment analysis of Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and Gene Set Enrichment Analysis (GSEA) were conducted using the R packages clusterProfiler and org.Hs.eg.db. Genes highly correlated with CXCR7 were identified using GEPIA ([57]http://gepia.cancer-pku.cn/), while immune infiltration was assessed using Timer 2.0 ([58]http://timer.cistrome.org/). Quantitative real-time PCR (qRT-PCR) Total RNA was extracted from cells using Trizol Reagent (Catalog #15596026, Invitrogen) and purified by phenol–chloroform extraction, following the manufacturer's instructions. The obtained RNA was converted into cDNA using a reverse transcription system (Catalog #R333-01, Vazyme, Nanjing, China). Quantitative PCR analysis of CXCR7 was performed on a Bio-Rad CFX96 system using ChamQ Universal SYBR qPCR Master Mix (Catalog #R711-03, Vazyme) with the following primer sequences: GAPDH forward (F): 5′-GTTCGTCATGGGTGTGAACC-3′, reverse (R): 5′-CATCCACAGTCTTCTGGGTG-3′; CXCR7 F: 5′-TGTCCTCACCATCCCAGTCT-3′, R: 5′-CTGCCGAAGAGGTTGATGGA-3′. The 2^−ΔΔCt method was used to assess the relative expression levels of CXCR7 compared to the control group, with GAPDH as the internal reference. Western blotting Antibodies used in this study include: CXCR7 (1:3000, 60 kDa, Catalog #60216-1-Ig, Proteintech, Shanghai, China), Gα q/11 (1:500, 41 kDa, Catalog #sc-365906, Santa Cruz, USA), Gα s (1:500, 49 kDa, Catalog #sc-135914, Santa Cruz), phosphorylated (p)-LATS (1:1000, 140 kDa, Catalog #8654S, CST, USA), LATS (1:1000, 140 kDa, Catalog #17049-1-AP, Proteintech), p-YAP (1:1000, 72 kDa, Catalog #13008T, CST), YAP (1:1000, 72 kDa, Catalog #abs134112, absin), and β-actin (1:150,000, 42 kDa, Catalog #T0022, Affinity, USA). Protein concentration was determined (Catalog #P0010, Beyotime), and the samples were separated on a 12% SDS-PAGE gel, followed by semidry transfer. Membranes (0.22 µm) were blocked with a solution containing 5% non-fat milk and 0.1% Tween. Membranes were then incubated with primary antibodies overnight at 4 °C. The following day, membranes were exposed to the corresponding secondary antibody at room temperature for one hour. Immunoblots were visualized using an enhanced chemiluminescence system (Catalog #G2014, Servicebio, Wuhan, China), and images were captured on GenoSens1800 autoradiography film (Shanghai Qinxiang Scientific Instrument Co., Ltd., China). β-actin was used for normalization. Colony formation assay Cells were resuspended and adjusted to a concentration of 1 × 10^4 cells/mL. Cells were then seeded at a density of 300 cells per well in a 6-well plate, with 2 mL of fresh culture medium added. Plates were incubated at 37 °C in a 5% CO[2] atmosphere. The colony size was periodically monitored, and the medium was refreshed regularly to maintain optimal growth conditions. After approximately two weeks, cells were fixed with tissue fixative (Catalog #P0099, Beyotime) for 10 min to preserve colony morphology and then stained with a 0.2% crystal violet solution (Catalog #[59]C02121, Beyotime) for 15 min to visualize the cell clusters. Images of the colonies were captured for analysis. Transwell assay Cells were resuspended and seeded into a 96-well plate at a density of 5 × 10^4 cells onto the upper chamber of a Transwell insert, while the lower chamber was filled with culture medium supplemented with 10% serum. The Transwell plate was then incubated for 24 h. For the invasion assay, the membrane was precoated with Matrigel. Briefly, Matrigel (Catalog #111005, GENOM Bio, Hangzhou, China) was diluted with serum-free medium to a final concentration of 1 mg/mL. The diluted Matrigel was added to the center of the chamber and incubated at 37 °C for 4 h, followed by incubation at 4 °C overnight to allow for proper gelation. Migratory or invasive cells were fixed and stained using a 0.1% crystal violet solution for 10 min. Images were captured under identical parameters. Statistical analysis Protein gray scale analysis was performed using ImageJ software (1.48v, NIH, USA). Results are expressed as mean ± SD. The significance between two groups was evaluated using an unpaired two-tailed Student's t-test, while significance among multiple groups was assessed using one-way analysis of variance followed by Tukey's post hoc test. A p-value < 0.05 was considered significant. Results CXCR7 functions as an oncogene in HCC To explore the role and molecular mechanisms of CXCR7 in the biological characteristics of HCC, our study utilized transfection techniques to introduce both an overexpression plasmid (pcDNA-CXCR7) and siRNA targeting CXCR7 (si-CXCR7). Evidence from RT-qPCR and Western blotting confirmed the effectiveness of these transfections; si-CXCR7 successfully induced a marked decrease in CXCR7 mRNA and protein expression (Fig. [60]1a, p < 0.01), while pcDNA-CXCR7 transfection led to a substantial increase in both (Fig. [61]1b, p < 0.01). Results from the CCK-8 assay demonstrated that HCCLM3 cells overexpressing CXCR7 showed enhanced viability over time compared to the control, whereas suppression of CXCR7 resulted in a reduction in cellular viability (Fig. [62]1c, p < 0.01). During a two-week colony formation assay, we observed that the number of colonies formed in the si-CXCR7 group was notably lower than in the control group, whereas the pcDNA-CXCR7 group showed a significant increase in colony formation (Fig. [63]1d, e, p < 0.05). Moreover, Transwell assays revealed that inhibition of CXCR7 resulted in a significant decrease in the number of migrating and invading cells, while overexpression of CXCR7 yielded the opposite outcomes (Fig. [64]1f–i). These findings suggest that high expression of CXCR7 in HCC is associated with enhanced cell activity, proliferation, migration, and invasion, indicating that CXCR7 plays a role in promoting cancer in HCC. Fig. 1. [65]Fig. 1 [66]Open in a new tab Chemokine C-X-C Motif Receptor 7 (CXCR7) promotes proliferation, migration, and invasion in hepatocellular carcinoma (HCC) cells. HCCLM3 cells were transfected with non-targeting control siRNA (si-NC), CXCR7 siRNA (si-CXCR7), pcDNA3.1 (Vector), or pcDNA3.1-CXCR7 (pcDNA-CXCR7). a RT-qPCR analysis of relative CXCR7 mRNA levels. b Western blot analysis of relative CXCR7 protein levels. c Cell viability was assessed using the CCK-8 assay. d, e Colony formation assays were conducted to assess cell proliferation. Transwell assays were used to evaluate cell migration f, g and invasion h, i. For A, B, E, and G: *p < 0.05, **p < 0.01; for C: **p < 0.01 versus si-NC, ##p < 0.01 versus Vector CXCR7 correlates with the gene signature of the Hippo pathway in HCC The activation of the Hippo signaling pathway and its correlation with CXCR7 were evaluated through bioinformatics analysis. CXCR7 expression was significantly higher in HCC tissues compared to adjacent normal tissues; however, their mean expression levels were relatively similar (Fig. [67]2a). Across different disease stages, CXCR7 levels showed no significant variation (Fig. [68]2b). The top 70 genes predicted to be similar to CXCR7 demonstrated relatively low Pearson correlation coefficients (Fig. [69]2c). Furthermore, immune infiltration did not differ significantly between the top 10 samples with the highest and lowest CXCR7 expression (Fig. [70]2d). These findings suggest that CXCR7 signaling is broadly dysregulated in HCC. The activation of the Calcium signaling pathway, Chemokine signaling pathway, Hippo signaling pathway, MAPK signaling pathway, and PI3K-Akt signaling pathway was confirmed through GSEA ontology analysis (Fig. [71]2e). Additionally, a heatmap illustrating the expression of genes involved in the Hippo signaling pathway is presented in Fig. [72]2f. Patients with high CXCR7 expression exhibited significantly worse prognoses compared to those with low expression (p = 0.016), as shown in Fig. [73]3a. The volcano plot revealed a total of 7152 upregulated DEGs, such as MUC5AC, KJRREL2, IGFN1, and MUCL3, alongside 1,112 downregulated genes (Fig. [74]3b). The number of upregulated genes was more than 6.4 times higher than the number of downregulated genes, suggesting a dominant effect of CXCR7 on gene upregulation. GO enrichment analysis of these DEGs showed that Biological Processes (BP) were primarily enriched in axon development, cell junctions, and external encapsulating structure organization. Cellular Component (CC) terms were enriched in collagen-containing extracellular matrix, cell-substrate junctions, and synaptic membranes (Fig. [75]3c). Molecular Function (MF) terms were enriched in channel activity, passive transmembrane transporter activity, and ion channel activity, highlighting the multifaceted impact of CXCR7 on cellular functions (Fig. [76]3c). Moreover, KEGG pathway enrichment analysis demonstrated significant enrichment in the PI3K-Akt signaling pathway, calcium signaling pathway, and MAPK signaling pathway, implicating the involvement of these critical signaling cascades in CXCR7-mediated effects (Fig. [77]3d). Interestingly, in our current research, we also found that the Hippo signaling pathway was significantly enriched. Fig. 2. [78]Fig. 2 [79]Open in a new tab Bioinformatics analysis of CXCR7 in HCC and its correlation with the Hippo signaling pathway. HCC samples were obtained from the TCGA-LIHC dataset. a Relative RNA expression of CXCR7 in HCC tissues compared to adjacent normal tissues. b Relative RNA expression of CXCR7 across different stages of HCC. c Predicted genes most closely related to CXCR7. d Immune infiltration in the top 10 samples with the highest and lowest CXCR7 expression. e GSEA ontology analysis of the pathways of interest. f Heatmap of Hippo signaling pathway genes arranged according to the relative RNA levels of CXCR7 for each sample *p < 0.05 by Pearson correlation for panels c & f, and by Two-way ANOVA for panel d. The “*” marked in red indicates upregulation, and marked in blue indicates downregulation in the ACKR3 high group Fig. 3. [80]Fig. 3 [81]Open in a new tab Bioinformatics analysis based on CXCR7 expression in patients with LIHC. Patients were divided into high-expression (N = 184) and low-expression (N = 184) groups based on the median expression level of the CXCR7 gene. a Kaplan–Meier survival curves were generated to compare the survival rates between the two groups. b A volcano plot was used to depict differential gene expression. GO enrichment analysis c and KEGG enrichment analysis d were performed on the differentially expressed genes CXCR7 regulates Hippo-YAP signaling in HCC cells Several key components of the Hippo signaling pathway, including Gαq/11, Gαs, LATS, and YAP were evaluated with Western blotting. The result showed that suppression of CXCR7 levels led to a reduction in cellular Gαq/11 and Gαs protein levels, which in turn resulted in an elevation of LATS and YAP phosphorylation (Fig. [82]4a, b, p < 0.05). Conversely, the overexpression of CXCR7 caused an increase in intracellular Gαq/11 and Gαs protein levels, leading to a decrease in the phosphorylation levels of LATS and YAP (Fig. [83]4a, b, p < 0.05). This evidence suggests that CXCR7 is involved in regulating the Hippo-YAP signaling pathway, influencing cellular outcomes through alterations in protein expression and phosphorylation states. Fig. 4. [84]Fig. 4 [85]Open in a new tab CXCR7 overexpression inhibits Hippo-YAP signaling. HCCLM3 cells were transfected with si-NC, si-CXCR7, Vector, or pcDNA-CXCR7. a, b Western blot analysis was performed to measure the protein levels of Gαq/11, Gαs, p-LATS, LATS, p-YAP, and YAP, followed by densitometric analysis to quantify the immunoblot images. *p < 0.05, **p < 0.01 CXCR7 enhanced proliferation, migration and invasion by regulating Hippo-YAP signaling in HCC cells Verteporfin, a photosensitizer and YAP inhibitor, was utilized to elucidate the interactions and functional significance of CXCR7 modulation on the Hippo-YAP pathway. As shown in Fig. [86]5 (p < 0.05), following treatment with Verteporfin, HCCLM3 cells exhibited a marked enhancement in cell viability, accompanied by increased proliferation, migration, and invasion capabilities. Notably, in cells transfected with si-CXCR7 and subsequently treated with Verteporfin, the previously inhibited cell activity, proliferation, migration, and invasion were significantly reversed. These findings suggest a critical role for CXCR7 in modulating the effects of Verteporfin on cancer cell behavior, implicating its potential as a therapeutic target for interventions aimed at controlling tumor progression. Fig. 5. [87]Fig. 5 [88]Open in a new tab CXCR7 promotes proliferation, migration, and invasion by regulating Hippo-YAP signaling. HCCLM3 cells were transfected with si-NC, si-CXCR7, Vector, or pcDNA-CXCR7, followed by treatment with the YAP inhibitor Verteporfin (VP). a Cell viability was assessed using the CCK-8 assay. b, c Colony formation assays were conducted to assess cell proliferation. Transwell assays were used to evaluate cell migration (d, e) and invasion (f, g). For a: **p < 0.01 versus si-NC + PBS, ^##p < 0.01 versus si-CXCR7 + PBS; for c, e, and g: *p < 0.05, **p < 0.01 Discussion Angiogenesis is a universally significant therapeutic target that facilitates tumor growth, progression, metastasis, and chemoresistance [[89]24]. CXCR7, which is highly expressed in liver endothelial cells [[90]11], can be further upregulated under hypoxic conditions [[91]25], a key stimulus for inducing angiogenesis, normal liver regeneration, and HCC proliferation [[92]26, [93]27]. The promotive role of HCC derived CXCR7 on metastases, angiogenesis and tumor growth has been explored in earlier studies [[94]28, [95]29]. Several signaling pathways have been implicated in CXCR7-associated angiogenesis and tumor progression, including MEK/ERK, NF-κB, p38 MAPK and PI3K/AKT signaling pathways [[96]30–[97]33]. Additionally, CXCR7 contributes to HCC progression by functioning as a chemokine regulator, shaping the tumor microenvironment. Elevated CXCR7 expression promotes the recruitment of M2 macrophages, which facilitates immune escape in HCC [[98]34, [99]35]. In line with these findings, our study also demonstrated that silencing CXCR7 inhibited proliferation, colony formation, migration, and invasion of HCCLM3 cells, whereas increasing CXCR7 expression produced the opposite effects. Bioinformatics analysis was subsequently performed to gain a comprehensive understanding of CXCR7 in HCC using the TCGA-LIHC dataset. Our analysis revealed that CXCR7 expression was significantly dysregulated in HCC. The number of upregulated DEGs in samples with high CXCR7 expression was more than 6.4 times greater than in samples with low CXCR7 expression. This result suggests that CXCR7 has a broad impact on gene transcription in HCC, potentially activating or enhancing a wide range of signaling pathways. GO terms enriched include extracellular matrix organization, collagen-containing extracellular matrix, ion channel activity, gated channel activity, and extracellular matrix structural constituents. These enriched terms could support the roles of CXCR7 in cell migration, particularly through pathways related to extracellular matrix and cell adhesion [[100]8]. The most significantly enriched KEGG pathways are PI3K-AKT, calcium signaling, and MAPK signaling, which have been reported to be regulated by CXCR7 to promote HCC progression, invasion, and migration [[101]30, [102]31]. The Hippo signaling pathway was also significantly enriched by KEGG, which the activation was confirmed by GSEA ontology. Both Hippo signaling and CXCL12-CXCR7/CXCR4 are important in organ development, and CXCR7 can form a positive feedback loop with Hippo signaling (via YAP expression) to promote gastric cancer progression [[103]23]. The enrichment of the Hippo signaling pathway suggests that such a feedback loop may also be involved in tumor progression in HCC. The Hippo signaling pathway has been recognized as a critical regulator of organ size [[104]36]. When Hippo signaling is activated, the downstream effectors YAP/TAZ are phosphorylated (rendered inactive) and restricted to the cytoplasm; upon Hippo pathway inactivation, YAP/TAZ are dephosphorylated (activated) and transported into the nucleus, where they enhance the expression of target genes to increase cell proliferation and survival, enhance stemness, and promote tumor progression. Numerous studies have highlighted the key role of the Hippo signaling pathway in HCC [[105]18, [106]37]. Activation of Hippo signaling suppresses HCC progression by affecting multiple aspects, including proliferation and fate reprogramming, tumor microenvironment, and metabolic reprogramming [[107]18]. Similar to CXCR7, activation of YAP/TAZ in peritumoral tissues has been shown to exert suppressive effects on HCC progression [[108]38], which is contrary to what happens in HCC cells. Consistent with findings characterized by Wang et al. in gastric cancer [[109]23], we further demonstrated that Hippo signaling is regulated by CXCR7 in HCC cells. Specifically, Gαq/11 and Gαs protein levels were reduced, while p-LATS/LATS and p-YAP/YAP levels were increased with si-CXCR7 treatment; conversely, pcDNA-CXCR7 had the opposite effects on these proteins. Co-treatment with the YAP inhibitor verteporfin reversed the suppressive effects of si-CXCR7 on HCC cells, including cell viability, colony formation, migration, and invasion. Our results support the conclusion that Hippo signaling can be inhibited by CXCR7 in HCC cells, promoting proliferation, colony formation, migration, and invasion, as concluded in Fig. [110]6. Fig. 6. Fig. 6 [111]Open in a new tab Interaction of CXCR7 and Hippo signaling to promote HCC progression While our findings provide evidence for the interplay between CXCR7 and Hippo signaling in HCC, there are limitations. The in vitro tests were only carried out with HCCLM3 cells, it is not known whether the transfection can mimic physiological expression levels, and in vivo consequences of CXCR7 remain unexplored. CXCR7 has been reported to influence a wide range of signaling pathways, requiring further investigation to delineate its direct and indirect roles. Most importantly, both CXCR7 and Hippo signaling could have different implications in HCC and adjacent normal tissues, so their expression and interactions across cell types should be determined. Future studies should focus on validating these findings in vivo and exploring therapeutic strategies targeting the CXCR7-Hippo axis. Conclusion Collectively, our findings indicate that two groups of morphogens, CXCL12-CXCR4/CXCR7 and Hippo signaling, may interact to promote HCC progression. CXCR7 inhibits Hippo signaling by activating LATS and YAP, which promotes the proliferation, colony formation, migration, and invasion of HCCLM3 cells. Supplementary Information [112]Additional file 1 (JPG 2376 KB)^ (3.8MB, jpg) Acknowledgements