CXCR7 overexpression promotes EMT and upregulates the expression of stem marker DCLK1
CXCR7 is highly expressed in many cancers and has predominantly pro-metastatic roles in cancer[26]. EMT is the initial step of metastasis which is characterized by decreasing of E-cadherin in epithelial cells as well as increasing of vimentin in mesenchymal cells. To explore whether CXCR7 contribute to EMT and stemness, we performed RNA-sequencing in CXCR7-overexpressing HCT116 cells and control cells. Among the differentially expressed genes, vimentin and ZEB1 were significantly increased and the intestinal stem cell marker DCLK1 was also markedly enhanced when CXCR7 was overexpressed (Fig. 1A). The most significantly upregulated genes were listed in Supplementary Table 2. To further confirm the association of CXCR7 on the regulation of EMT and DCLK1, colorectal cells of HCT116, HT29 and SW620 cells were infected by lentivirus expressing CXCR7 (LV-CXCR7) and siRNA targeting CXCR7 (CXCR7-KD). The results showed that Vimentin were prominently upregulated with concurrent downregulation of E-cadherin in CXCR7-overexpressing cells compared with control (Fig. 1B, C). Transwell assay indicated the enhanced invasive capacity in CXCR7-overexpressing CRC cells than that of control cells (Supplementary Fig. 1). In contrast, CXCR7 knockdown produced opposite effects (Fig.1C), indicating CXCR7 overexpression triggered EMT in CRC. In parallel, as a stem marker, DCLK1 has similar changes in line with EMT progression, suggesting overexpression of CXCR7 enhanced multipotency with the progression of CRC.
To further determine the clinical relevance of CXCR7 expression with EMT and cell stemness, we analyzed the correlation of expression of CXCR7 with vimentin and DCLK1 in human CRC tissues by Gene Expression Profiling Interactive Analysis (GEPIA) (http://GEPIA.cancer-pku.cn/index.html) using TCGA datasets. Notably, we found a robust and statistically significant association between vimentin, DCLK1 and CXCR7 (R = 0.58 and 0.49 respectively, p = 0) (Fig.1D). Furthermore, we evaluated the expression of vimentin and DCLK1 as prognostic gene signature by using CRC datasets, we found that high vimentin and DCLK1 mRNA expression significantly correlated with a worse prognosis (Fig. 1E). Collectively, these findings suggest CXCR7 overexpression promotes EMT and upregulates the expression of stem marker DCLK1, which is associated with poor clinical outcome.
CXCR7 overexpression contributes to EMT by repressing miR-124-3p and miR-188-5p
In order to investigate the mechanism that CXCR7 signal activation contributes to CRC progression and EMT, miRNA sequencing was performed in above HCT116 cells (HCT116Control and HCT116LV-CXCR7), and the significantly upregulated and downregulated miRNAs are listed in Supplementary Table 3. Among the differentially expressed miRNAs, miR-124-3p and miR-188-5p were significantly downregulated in HCT116LV-CXCR7 comparing with HCT116Control, indicating that these miRNAs were downregulated by CXCR7 activation (Fig. 2A). To verify the results, we determine the expression of these miRNAs in CRC cells in response to activation of the CXCL12/CXCR7 axis. As a result, RT-QPCR analysis showed that miR-124-3p and miR-188-5p were significantly downregulated upon activation of the CXCL12/CXCR7 axis. In contrast, knockdown of CXCR7 markedly elevated these miRNAs in HCT116 and HT29 cells (Fig. 2B). CXCL12 is the common ligand for activation of CXCR4 and CXCR7. As the atypical chemokine receptor, CXCR7 could mediate biased signal activation independent of activation of heterotrimeric G proteins. Importantly, to elucidate whether activation of CXCR7 repressed the expression of miRNAs through biased signaling, we used AMD3100, the specific inhibitor of CXCR4, to exclude the effect of activation of CXCL12/CXCR4. The results showed that CXCL12/CXCR7 produced a similar effect on the downregulation of miR-124-3p and miR-188-5p with or without the treatment of AMD3100. Specifically, the biased signal of CXCL12/CXCR7 can profoundly suppressed miR-124-3p and miR-188-5p. (Fig. 2B).
To gain an insight into the molecular mechanism of these miRNAs on CRC progression and EMT, two mRNA target-predicting algorithms (MiRDB and Targetscan) were utilized to identify the potential downstream targets of miR-124-3p and miR-188-5p. Vimentin and DCLK1 were predicted to be the potential target gene of miR-124-3p and miR-188-5p respectively. To verify this, HCT116, HT29 and SW620 cells were transfected with miR-124-3p and miR-188-5p mimics and inhibitors respectively, and the results showed that the expression of vimentin and DCLK1 was significantly suppressed by miR-124-3p and miR-188-5p mimics, while substantially enhanced by miR-124-3p and miR-188-5p inhibitors (Fig. 2C, 2D). Of note, we also observe the regulation of DCLK1 by miR-124-3p, possibly due to indirect impact on gene expression imposed by multiple targets of miRNAs. Furthermore, luciferase reporter assay was performed to confirm the direct binding of miR-124-3p and miR-188-5p with Vimentin 3′-UTR and DCLK1 3′-UTR. The predicted binding sites of the miRNAs with wild type and mutant 3′-UTR luciferase reporter constructs are shown in Fig. 2E. In line with these results, the binding was abolished by mutation of the binding site of these miRNAs on Vimentin 3′-UTR and DCLK1 3′-UTR, suggesting that these miRNAs could directly bind to Vimentin 3′-UTR and DCLK1 3′-UTR and regulate its expression at the post-transcriptional level (Fig.2F). Taken together, these results establish that CXCR7 biased signal activation contributes to CRC progression and EMT by repressing miR-124-3p and miR-188-5p which targeting Vimentin and DCLK1.
YAP1 manipulates CXCR7 biased activation-induced EMT by repressing miR-124-3p and miR-188-5p
EMT is a highly dynamic and reversible process, conferring cancer cells with the plasticity for distant dissemination and stemness properties. YAP1/TEAD transcriptional activation has emerged as the core regulator in EMT by cooperation with ZEB1 and AP-1[27]. In this case, we hypothesize that YAP1 promotes EMT and stemness via the regulation of miRNAs in CRC cells.
As shown in Fig. 3A, YAP1 silencing strongly enhanced the expression of miR-124-3p and miR-188-5p. Of relevance, the mRNA levels of stemness marker DCLK1 and mesenchymal marker vimentin were remarkably reduced with YAP1 depletion (Fig. 3B). Consistently, knockdown of YAP1 with two different siRNAs led to a pronounced decrease of DCLK1 and vimentin proteins in CRC cells (Fig. 3C).
As a transcriptional coactivator, YAP1 translocated into nucleus to exert the regulatory effects with TEAD that has DNA-binding domains. To further interrogate whether nuclear YAP1 orchestrates the expression of vimentin and DCLK1 by regulating miRNAs, we transfected CRC cells with a construct expressing the constitutive active form of YAP1 (YAP5SA), resistant to LATS-mediated phosphorylation, which directly leads to YAP1 nuclear translocation. Intriguingly, enforced expression of YAP5SA rescued, to a large extent, the marked downregulation of DCLK1 and Vimentin caused by YAP1 knockdown (Fig. 3D). More importantly, enforced expression of YAP5SA resulted in a drastic reduction of miR-124-3p and miR-188-5p and it could also attenuate the prominent elevation of these miRNAs in YAP1 knockdown cells (Fig. 3E). The results suggest that YAP5SA could rescue the suppression of EMT marker caused by silencing of YAP1 through repression of miR-124-3p and miR-188-5p. Altogether, these data reveal that nuclear YAP1 is critically involved in promoting EMT and stemness by repressing miR-124-3p and miR-188-5p in CRC cells.
CXCR7/β-arr1-mediated biased signal induces YAP1 nuclear translocation in CRC cells
Since nuclear YAP1 plays a crucial role in regulation of EMT by repression of miR-124-3p and miR-188-5p, next, we explored whether CXCL12/CXCR7 biased activation promoted YAP1 nuclear translocation. Notably, CXCL12 induced the reduction of YAP1 in the cytoplasm paralleled with YAP1 nuclear accumulation, which was potentiated by overexpression of CXCR7 (Fig. 4A). The CXCL12-induced YAP1 nuclear accumulation was also confirmed by immunofluorescence analysis in HCT116 and HT29 cells, showing the time-dependent nuclear translocation of YAP1 and reached plateau after 60 min stimulation by CXCL12 (Fig. 4B, 4C and Supplementary Fig. 2).
β-arr1, previously known as a cytosolic regulator and scaffold of GPCR signaling, has recently been revealed to translocate to the nucleus mediating receptor endocytosis and signal transduction. Therefore, it is likely that β-arr1 shuttles between the cytoplasm and the nucleus mediating CXCL12/CXCR7 biased signal activation. To assess whether β-arr1 could functionally contribute to YAP1 activity regulation consequently to CXCL12/CXCR7 axis activation, we performed co-immunoprecipitation analysis in whole cell lysates derived from HCT116 cells, and found that endogenous YAP1 physically interacted with β-arr1 (Fig. 4D). Here we ask whether the interaction between β-arr1 and YAP1 facilitate the nuclear translocation of YAP1 with the concurrent nuclear shuttling of β-arr1 or not? Immunofluorescence analysis indicated that although CXCL12 stimulation at early stage (for 90 min) led to substantial YAP1 nuclear translocation, β-arr1 and β-arr2 were predominantly located in cytoplasm and not involved in the YAP1 nuclear co-translocation (Fig. 4E). However, we do observe the nuclear translocation of β-arr1 at later stage (> 6 h) of CXCL12 stimulation (Supplementary Fig. 3). Furthermore, nuclear-cytoplasmic fractions indicated that CXCL12 induced dramatic reduction of YAP1 in the cytoplasm accompanied by the significant increase of nuclear YAP1. Concurrently, AMD3100, as a CXCR4 antagonist, was used to rule out any effects of CXCL12/CXCR4 signaling activation (Fig. 4F). These results indicated that CXCL12/CXCR7 biased signal activation substantially promoted nuclear translocation of YAP1. Noticeably, β-arr1 does not exhibit a simultaneous nuclear translocation at early stage of CXCL12 stimulation in CRC cells (Fig. 4F). Then we ask whether β-arr1 is required for YAP1 nuclear translocation upon CXCL12/CXCR7 biased signal activation? As shown in Fig. 4G and 4H, we demonstrated that knockdown of β-arr1 but not β-arr2 interfered with the nuclear translocation of YAP1 in response to CXCL12 stimulation.
These results revealed that CXCL12/CXCR7 biased signal activation promoted YAP1 nuclear translocation by recruiting β-arr1 in the cytoplasm, which could not be abrogated by AMD3100 pretreatment. Overall, these findings establish that CXCR7 activation by CXCL12 induces YAP nuclear enrichment in CRC cells and prove the critical role of β-arr1 in transducing CXCL12/CXCR7-dependent YAP1 cytoplasmic-nuclear shuttling.
YAP1 inhibits miR-124-3p and miR-188-5p expression by recruiting YY1 to the promoter
Nuclear YAP1 functions as a potent transcriptional cofactor by binding with TEAD1. YAP1/TEAD1 interacts with other transcriptional factors to regulate the expression of target genes[28]. Generally, YAP1/TEAD complex activates the oncogenic downstream genes to trigger carcinogenesis. To further explore the inhibitory effects of nuclear YAP1 on the expression of miR-124-3p and miR-188-5p, TransmiR v2.0 database (http://www.cuilab.cn/transmir) and interface of mirTrans (http://mcube.nju.edu.cn/jwang/lab/soft/mirtrans/) were used to predict the transcriptional factor binding sites at the promoter of miR-124-3p and miR-188-5p. As a result, Yin Yang 1 (YY1) is predicted to be the potentially common transcriptional factor that could suppress the expression of these miRNAs.
We hypothesized that YAP1 functions as a transcriptional repressor by interacting with YY1, transcriptionally repressing the expression of miR-124-3p and miR-188-5p, thereby promoting EMT and metastasis. To prove this, HCT116 and SW620 cells are used to confirm whether YY1 was involved in the regulation of miR-124-3p and miR-188-5p expression. As shown in Fig. 5A, miR-124-3p and miR-188-5p were robustly upregulated by YY1 silencing. Expectedly, as shown in Fig. 5B-5C, knockdown of YY1 led to remarkably downregulation of mesenchymal marker vimentin in SW620 cells, consistently, the expression of stem cell marker DCLK1 was also profoundly impaired with YY1 depletion. These results indicated that YY1 was implicated with the regulation of EMT and stemness. To further assess the specificity of YY1 in promoting stemness and EMT, we transfected CRC cells with a construct expressing YY1 with HA-tag which led to YY1 overexpression. Remarkably, cells with YY1 silencing, characterized by DCLK1 reduction was rescued by re-expression of YY1 at protein levels (Fig. 5D). Moreover, RT-PCR was used to determine the expression of miR-124-3p and miR-188-5p. As results, the level of miR-124-3p and miR-188-5p were significantly increased by YY1 silencing which can be strongly reduced by enforced expression of HA-YY1 (Fig. 5E).
In order to address our hypothesis that YAP1 inhibit miR-124-3p and miR-188-5p expression by recruiting YY1, co-immunoprecipitation analysis was performed in HCT116 cells, and we found that endogenous YAP1 could physically interact with endogenous YY1 in HCT116 cells. Moreover, when HCT116 cells were transfected with flag- YAP5SA and HA-YY1, YAP1 and YY1 was found to interact in the nucleus by exogenously overexpression using Co-IP assay (Fig. 5F). Consistently, immunofluorescence analysis further showed the nuclear co-localization of YAP1 and YY1 in CRC cells (Fig. 5G). Moreover, luciferase reporter assay showed that the promoter activities of miR-124-3p and miR-188-5p were robustly enhanced compared with control vector of pGL3-baisc, which was significantly hampered upon YY1 overexpression. (Fig. 5H). Taken together, these results unveiled that YAP1 inhibit miR-124-3p and miR-188-5p expression by recruiting YY1 to the promoter, therefore, YAP1/YY1 cooperate to regulate EMT plasticity, stemness and metastasis in CRC cells.
YAP1 inhibitor suppresses CXCL12/CXCR7-induced EMT and tumor metastasis in vitro and in vivo
In light of the crucial role of YAP1 in regulating EMT by repressing miR-124-3p and miR-188-5p via interacting with YY1, we wonder if YAP1 inhibitor verteporfin could blunt CXCL12/CXCR7-induced EMT and distant metastasis. As shown in Fig.6A, CXCL12/CXCR7 biased activation strongly upregulated the expression of DCLK1, which was attenuated by verteporfin. We further detect the effects of verteporfin on distant metastasis when injected with HCT116 cells overexpressing CXCR7 via tail veins. The results indicated that CXCR7 overexpression facilitated more distant metastasis such as lung metastasis, which was greatly impaired by verteporfin. The representative Hematoxylin and Eosin (HE) staining of metastatic nodules in lungs were illustrated in Fig. 6B.
To investigate the role of YAP1 in colitis-associated carcinogenesis and progression upon CXCL12/CXCR7 biased activation in vivo, wild type (WT) and villin-CXCR7 transgenic mice (villin-CXCR7) were treated with AOM and DSS for 3 cycles as described in methods. We found that AOM/DSS exposure seriously aggravated colonic inflammation and tumor burden in villin-CXCR7mice compared with WT mice, as indicated by the larger size of colonic adenocarcinoma. Importantly, Verteporfin, a YAP1 inhibitor which disrupts YAP-TEAD interactions, led to a significant reduction of the colonic adenocarcinomas (Fig. 6C). To investigate whether YAP1 inhibitor hindered EMT process in CRC by regulation of miR-124-3p and miR-188-5p in vivo, RT-qPCR analysis indicated that pharmacological inactivation of YAP1 with verteporfin reversed the repression of miRNAs in AOM/DSS induced villin-CXCR7 mice (Fig. 6D). Further IHC and Western blot analysis showed that vimentin and DCLK1 were highly expressed in colonic adenocarcinoma of villin-CXCR7 mice, which was abrogated by Verteporfin (Fig. 6E, 6F). These results suggest that YAP1 inhibitor verteporfin recapitulates the anti-tumorigenesis and anti-metastasis of YAP1 depletion upon the activation of CXCL12/CXCR7, highlighting the therapeutic potential of targeting YAP1 in the control of CRC progression and metastasis.
CXCL12/CXCR7/β-arr1-induced YAP1 nuclear translocation is associated with EMT and metastasis in human CRC tissues
To extend current knowledge to colorectal cancer patients, we collected 22 pairs of human CRC specimens and adjacent normal colon tissues for immunohistochemistry, Western Blot and RT-PCR analysis. As shown in Fig. 7A, CXCR7 was highly expressed in human CRC tissues compared with normal colon tissues, particularly with higher expression in metastatic CRC than non-metastatic counterpart. YAP1 was remarkably overexpressed in nuclei in CRC tissues whereas predominantly expressed in cytoplasm in adjacent normal colon tissues. More importantly, there was also a higher expression of nuclear YAP1 in metastatic CRC than non-metastatic counterpart. Similarly, the expression of vimentin was higher in CRC particularly metastatic CRC tissues compared with adjacent normal colon tissue (Fig. 7A). Further protein analysis exhibited prominently high expression of YAP1 and DCLK1 in CRC tissues compared with adjacent normal tissues (Fig. 7B). To explore the potential link between YAP1, vimentin, DCLK1 and upstream CXCR7, we used public accessible online tool GEPIA (http://GEPIA.cancer-pku.cn/index.html). It revealed a significant positive correlation of YAP1 and vimentin (R = 0.24, p < 0.001). The expression of YAP1 and DCLK1 also displays a potent correlation (R = 0.27, p < 0.001) (Fig.7C). Notably, the expression levels of CXCR7 strongly correlated with those of YAP1 (R = 0.33, p < 0.001) in CRC tissues. miR-124-3p expression were significantly reduced in human CRC tissues and was statistically significant negatively correlated with the expression of Vimentin at mRNA levels (Pearson R = -0.3386, p = 0.0326) (Fig. 7D, 7E), highlighting that miR-124-3p functions as a tumor suppressive miRNA in CRC tissues. Taken together, these data suggest that CXCL12/CXCR7/β-arr1 biased activation triggered YAP1 nuclear translocation, which contributes to EMT and CRC metastasis by repressing miR-124-3p and miR-188-5p in clinical CRC specimens (Fig.8).