Leptin-mediated regulation of ICAM-1 is Rho|[sol]|ROCK dependent and enhances gastric cancer cell migration
Z Dong,ssui fu,xiangbin xu,yan yang,liying du,wei li,S Kan,zhanqing li,xiaochun zhang,L Wang,jin li,hui liu,xueping qu,C Wang
DOI: https://doi.org/10.1038/bjc.2014.70
IF: 9.075
2014-01-01
British Journal of Cancer
Abstract:Gastric cancer (GC) ranks as the second leading cause of cancer-related death in the world (Parkin et al, 2005). The high aggressiveness and rapid metastasis of GC contribute to its high mortality rate (Wang and Chen, 2011). The metastasis involves multiple factors, such as obesity, an important risk factor in GC (Calle et al, 2003; Howard et al, 2010). Adipocytes provide fatty acids for rapid tumour growth, and the dysfunction of lipid metabolism can lead to the pathogenesis of human GC (Song et al, 2012). Leptin is an adipokine of the obesity (ob) gene, and has been known to have roles in the homeostasis of body weight by regulating food intake and energy metabolism (Chan et al, 2006). Recent studies have revealed that leptin is implicated in the tumorigenesis of cancer, such as ovarian cancer, endometrial cancer, GC, and so on (Zhao et al, 2007; Wu et al, 2012; Xu et al, 2013). Leptin exerts its function through leptin receptor, and the use of leptin receptor antagonists has been recognised as a potential therapeutic approach for cancer (Otvos et al, 2011). Our previous study indicates that leptin enhances GC cell invasion by upregulating the expression of membrane type 1-matrix metalloproteinase (MT1-MMP) (Dong et al, 2013). Converging studies reveal that leptin expression is elevated in GC patients, and is also significantly correlated with GC metastasis (Shida et al, 2005; Ishikawa et al, 2006; Zhao et al, 2007). However, the exact effect of leptin on GC metastasis and its underlying mechanism remain poorly understood. Recent findings of upregulation of intercellular adhesion molecule-1 (ICAM-1, also known as CD54) by leptin in allergic inflammation may shed light on how leptin modulates cell migration (Wong et al, 2007). This suggests a link between leptin and ICAM-1. ICAM-1 is a transmembrane glycoprotein of the immunoglobulin superfamily. It binds to two major ligands, lymphocyte function-associated antigen-1 (LFA-1, CD11a/CD18) and macrophage-1 antigen (Mac-1, CD11b/CD18). LFA-1 and Mac-1 expressed on NK cell and macrophages share a common β chain (Diamond et al, 1990; Zimmerman and Blanco, 2008). ICAM-1 has an essential role in cell–cell and cell–extracellular matrix interaction. Binding to LFA-1 and Mac-1, ICAM-1 can facilitate the metastasis of cancer cells by escaping the recognition and attack of immunocytes (Makrilia et al, 2009). Previous studies show that ICAM-1 is associated with more aggressive lesions in prostate cancer (Chen et al, 2012), lung cancer (Grothey et al, 1998), and GC (Maruo et al, 2002). Laboratory models have demonstrated that the upregulation of ICAM-1 can promote tumour cell invasion and metastasis. In addition, overexpressed ICAM-1 may be released from the local cancer cell nests and enter the serum as sICAM-1 (Essick et al, 2008). Elevated serum ICAM-1 level is correlated significantly with serum CA19-9 and CEA in GC (Polychronidis et al, 2003); consistently, suppression of ICAM-1 expression is related to attenuated tumour progression and metastasis (Kaliora et al, 2008; Chen et al, 2012; Yu et al, 2012). Interestingly, both leptin and ICAM-1 are increased after Helicobacter pylori infection (Fan et al, 1995; Azuma et al, 2001). To our knowledge, there is no published data about the effect of leptin on ICAM-1 and its role in GC metastasis. The expression of ICAM-1 may be regulated by a variety of signalling pathways, such as Rho (the small G protein Ras homologue gene family) and its immediate downstream effector Rho-associated coiled-coil-forming protein kinase (ROCK) (Jung et al, 2012). ROCK is ubiquitously expressed serine–threonine protein kinases that are involved in diverse cellular functions, including actin cytoskeleton organisation, cell adhesion, and motility (Burridge and Wennerberg, 2004). Hyperactivation of the Rho/ROCK pathway is known to be linked with more aggressive tumour properties such as metastasis (Sahai and Marshall, 2002). Rho/ROCK may be regulated by some factors in tumours. For example, lysophosphatidic acid triggers focal adhesion assembly through Rho/ROCK pathway in human ovarian cancer cells (Sawada et al, 2002). A previous study reveals that leptin may induce Rho/ROCK pathway (Schram et al, 2011). On the basis of these reports, we hypothesise that Rho/ROCK pathway may be involved in the leptin-mediated expression of ICAM-1. This study has demonstrated that leptin promotes the migration of GC cells by upregulating ICAM-1 expression via the involvement of Rho/ROCK pathway. Targeting ICAM-1 and its regulation pathway might provide preliminary experimental clues for the development of therapies against the metastasis of GC. To test whether leptin-mediated ICAM-1 upregulation was involved in GC cell migration, we first transduced GC cells to overexpress ICAM-1 (pcDNA3.1-ICAM-1). The transfection efficacy was analysed by WB (Figure 4A). Our findings showed that overexpressed ICAM-1 could promote GC cell migration (Figure 4B). Moreover, we performed knockdown of ICAM-1 by siRNA. The knockdown efficiency was evaluated by WB (Figure 4C). As shown in Figure 4D, leptin-induced migration of GC cells was significantly reduced by transient transfection of ICAM-1-siRNA (AGS: 53.9%±3.6%, P=0.020; MKN-45: 42.79%±3.78%, P=0.005). The NC group was transfected with control siRNA. To further explore whether leptin regulated the expression of ICAM-1 through the Rho/ROCK pathway, cells were pretreated with the specific pharmacological inhibitor C3 transferase (Rho inhibitor; Cytoskeleton) at 0.25 μg ml−1 or Y-27632 (ROCK inhibitor; Calbiochem, Nottingham, UK) at 3.3 μM (Schram et al, 2011), followed by leptin treatment. The expression of ICAM-1 was analysed by WB, flow cytometry, and ELISA (for sICAM-1). As shown in Figure 5C, pretreatment with either Rho (C3 transferase) or ROCK (Y-27632) inhibitor prevented the leptin-induced upregulation of ICAM-1 in AGS and MKN-45 cells, and there was no difference between C3 transferase and Y-27632. Although treatment with inhibitors alone resulted in a subtle decrease in the expression of ICAM-1 compared with control, the inhibitors alone did not influence the expression of ICAM-1. A similar trend was also observed with the cell surface ICAM-1 (Figure 5D) and sICAM-1 (Figure 5E). Metastasis, such as lymph node metastasis, is a common cause of death from GC (Wang and Chen, 2011). Leptin has been associated with an increased risk of GC and has critical roles in tumour metastasis (Zhao et al, 2007). However, little is known about the exact effect of leptin on GC cell migration and its underlying mechanism. ICAM-1 is overexpressed in several cancers and implicated in tumour migration (Maruo et al, 2002; Chen et al, 2012). In this study, leptin and ICAM-1 levels were strongly correlated with each other in human GC tissues, as well as with clinical stage and lymph node metastasis, suggesting a causal connection between leptin pathway and ICAM-1. Our results also demonstrated that leptin may enhance the migration of GC cells (AGS and MKN-45) by upregulating ICAM-1 expression in vitro. Moreover, leptin induces the activation of Rho/ROCK pathway, which contributes to the subsequent upregulation of both surface ICAM-1 and sICAM-1. Taken together, our findings indicate that leptin enhances GC migration by increasing ICAM-1 expression through Rho/ROCK pathway. Metastasis is a multi-step process during which cancer cells detach from the primary tumour, migrate, adhere, and invade into the blood or lymphatic vessels. Finally cancer cells extravasate out of the vessel and interact with the target tissue. Although multiple factors are involved in this process, adipocyte provides fatty acids for rapid tumour growth, and the dysfunction of lipid metabolism can lead to the pathogenesis of GC (Song et al, 2012). Leptin, secreted predominantly from adipocytes, may promote GC cell proliferation, suggesting an essential role of leptin in the progression of GC (Pai et al, 2005; Shida et al, 2005). This study has indicated that leptin induces the migration of GC cells (AGS and MKN-45) in a dose- and time-dependent manner, which keeps in line with previous studies that leptin induces migration of endometrial cancer and glioma cells (Yeh et al, 2009; Wu et al, 2012). Further clarifying the mechanisms responsible for leptin-induced GC metastasis is important for better understanding and control of tumour metastasis in patients with GC. Our results revealed that leptin and ICAM-1 were overexpressed in GC tissue and correlated with lymph node metastasis, which is consistent with the previous studies (Maruo et al, 2002; Zhao et al, 2007). Further analysis indicated that leptin was positively correlated with ICAM-1, suggesting a possible link between leptin and ICAM-1 in the progression of GC. This is also supported by the findings that leptin and ICAM-1 are elevated after stimulation with inflammatory cytokines, for example, in patients with H. pylori infection (Fan et al, 1995; Azuma et al, 2001). Previous studies show that ICAM-1 undergoes cleavage at its extracellular stalk domain, and release a soluble form of the protein (sICAM-1), and this process may be mediated by MT1-MMP (MMP14) (Essick et al, 2008). In this study, we found that leptin enhanced the expression of ICAM-1, as well as cell surface ICAM-1 and sICAM-1 in GC cells. Furthermore, we also observed an enhanced expression of MT1-MMP by leptin in GC cells (Dong et al, 2013). Therefore, we speculate that leptin promotes ICAM-1 expression and subsequently enhances the level of sICAM-1 via upregulating MT1-MMP. Increased ICAM-1 may promote the adhesion of cancer cells, leading to tumour growth and mediating cell–extracellular matrix interaction. When metastasis occurs, cancer cells detach from the primary lesion, penetrate through the tissue basement membrane, and invade blood vessels and lymphatic vessels. On the other hand, as the host killer cells (NK cells, LAK cells, and CTLs) and macrophages express LFA-1 and Mac-1, two major receptors of ICAM-1, partially via which they recognise and attack the tumour cells (Diamond et al, 1990), upregulated sICAM-1 may inhibit NK and macrophage activity by competitively binding to LFA-1 and Mac-1 (Koyama, 1994). Thus, it is possible that there is a balance between ICAM-1/sICAM-1 and immunocytes, which can also be explained by the fact that it takes a long time for detectable tumour to form a metastatic lesion. With the development of GC, leptin increased sICAM-1 level, breaking the balance and subsequently resulting in a failure of the host immunocytes to effectively kill the tumour cells, which may have, therefore, protected the tumour cells from elimination and facilitated tumour migration. The hypothesis is well supported by previous findings that serum leptin and sICAM-1 are elevated and positively correlated with the clinical stage and lymph node metastasis of tumour (Maruo et al, 2002; Capelle et al, 2009). Taken together, these results suggest that leptin/ICAM-1/sICAM-1 is implicated in GC metastasis and may thus provide a potential therapeutic target against GC, which needs to be further confirmed by more experiments with animal models. Sunami et al (2000) have shown that lymph node metastasis reduced after successful transfection of ICAM-1 gene into the cancer cells. Because of the enhancement of immune surveillance mechanism, tumour cells might be easily captured and destroyed by the lymphocytes. However, their cell model is different from ours, in addition, these authors did not discuss the production of sICAM-1 in their ICAM-1-overexpressing cells, though previous study from the same group revealed that the survival rate in high sICAM-1 group significantly decreased compared with that in low sICAM-1 group (Nakata et al, 2000). The present study has demonstrated the importance of leptin-induced upregulation of ICAM-1 in GC cell migration and further elucidated the underlying mechanism. It is well known that the activity of small GTPases (including RhoA) is important for the signalling derived from lipid metabolism (Bolick et al, 2005). This study has shown that leptin may activate Rho/ROCK pathway, which is consistent with the previous findings that leptin regulates myocardial matrix remodelling by activating Rho/ROCK (Schram et al, 2011). Further analysis indicates that the inhibitor of either Rho (C3 transferase) or ROCK (Y27632) attenuates leptin-induced ICAM-1 expression, illustrating that the use of ROCK inhibitors might be a potential therapeutic approach. This is also supported by the finding that Rho/ROCK pathway critically regulates the plasticity of metastatic GC (Matsuoka et al, 2011). Notably, the inhibitor treatment in this study failed to completely block the leptin-induced upregulation of ICAM-1, implying a possible contribution from other unidentified leptin-activated pathway(s). Several signalling pathways activated by leptin have been identified in GC, that is, Janus kinase/signal transducers and activators of transcription (JAK/STAT), extracellular signal-regulated kinase 1/2 (ERK1/2), and mitogen-activated protein kinase (MAPK) pathways, as well as phosphatidylinositol 3-kinase (PI3K)/AKT pathways (Pai et al, 2005; Shida et al, 2005). Previous study reveals that leptin upregulates ICAM-1 expression in eosinophils by the combined activation of MAPK and JAK pathways under allergic inflammation (Wong et al, 2007). However, their changes were not examined in this study. Although we cannot exclude the possible effect of these signalling pathways, the upregulation of ICAM-1 induced by leptin in the cells of this study might primarily depend on the RhoA/ROCK pathway. Further experiments will be conducted to test these possibilities. In addition, Rho GTPases are the binary switch between an active GTP-bound form and an inactive GDP-bound form. This process might be mediated by several molecules, such as Rho guanine nucleotide exchange factors (Rossman et al, 2005), NF-κB (Jung et al, 2012), and reactive oxygen species (Jin et al, 2004). Although these mechanisms remain an interesting research topic, they are beyond the scope of the current study. In conclusion, this study has shown that leptin enhances GC cell migration by increasing ICAM-1 expression, which is demonstrated to be largely dependent on the activation of Rho/ROCK signalling pathway. A summarised schematic diagram is shown in Figure 6. Our results may provide preliminary experimental clues for future development of novel therapies against the metastasis of GC. The authors declare no conflict of interest. The project was supported by the China National Natural Science Foundation Projects (Grant No. 81072406, 81271916, 31270971, and 81301506), Research Fund for the Doctoral Program of Higher Education of China (Grant No. 20120131110055), Shandong Province Natural Science Foundation (Grant No. ZR2010HZ004). The authors thank Professor Jian-Jun Wei (Department of Pathology, Robert H. Lurie Comprehensive Cancer Center of Northwestern University) for advice on experimental design and critically reviewing the manuscript.
What problem does this paper attempt to address?
-
Leptin-mediated regulation of ICAM-1 is Rho/ROCK dependent and enhances gastric cancer cell migration
Z Dong,S Fu,X Xu,Y Yang,L Du,W Li,S Kan,Z Li,X Zhang,L Wang,J Li,H Liu,X Qu,C Wang
DOI: https://doi.org/10.1038/bjc.2014.70
IF: 9.075
2014-01-01
British Journal of Cancer
Abstract:Background: Our previous study indicates that leptin enhances gastric cancer (GC) invasion. However, the exact effect of leptin on GC metastasis and its underlying mechanism remain unclear. Intercellular adhesion molecule-1 (ICAM-1), a major molecule in stabilising cell–cell and cell–extracellular matrix interactions, is overexpressed and has crucial roles in tumour metastasis. Methods: Here, we investigated leptin and ICAM-1 expression in GC tissues. Furthermore, we characterised the influence of leptin on ICAM-1 expression in GC cells and elucidated the underlying mechanism. Results: Leptin and ICAM-1 were overexpressed in GC tissues, and a strong positive correlation was observed. They were also related with clinical stage or lymph node metastasis. Furthermore, leptin induced GC cell (AGS and MKN-45) migration by upregulating ICAM-1, and knockdown of ICAM-1 by small interference RNA (siRNA) blocked this process. Cell surface ICAM-1, as well as soluble ICAM-1 (sICAM-1), was also enhanced by leptin. Moreover, leptin increased ICAM-1 expression through Rho/ROCK pathway, which was attenuated by pharmacological inhibition of Rho (C3 transferase) or its downstream effector kinase Rho-associated protein kinase (ROCK) (Y-27632). Conclusions: Our findings indicate that leptin enhances GC cell migration by increasing ICAM-1 through Rho/ROCK pathway, which might provide new insight into the significance of leptin in GC.
-
Leptin-Mediated Regulation of Mt1-Mmp Localization is Kif1b Dependent and Enhances Gastric Cancer Cell Invasion
Zhaogang Dong,Xiaofei Xu,Lutao Du,Yongmei Yang,Huanhuan Cheng,Xin Zhang,Zewu Li,Lili Wang,Juan Li,Hui Liu,Xun Qu,Chuanxin Wang
DOI: https://doi.org/10.1093/carcin/bgt028
2013-01-01
Carcinogenesis
Abstract:Leptin overexpression is closely correlated with gastric cancer (GC) invasion, but its exact effect and the underlying mechanism in tumorigenesis remain poorly understood. Membrane type 1-matrix metalloproteinase (MT1-MMP), a surface-anchored master switch proteinase, is overexpressed and plays crucial roles in tumor invasion. Here, we characterized the influence of leptin on the generation and surface localization of MT1-MMP in GC and elucidated its molecular mechanisms. Our results revealed that leptin promoted GC cell invasion in vitro by upregulating MT1-MMP expression. Furthermore, cell surface biotinylation assay and flow cytometry demonstrated that the surface expression of MT1-MMP was also enhanced by leptin, and knockdown of kinesin family member 1B (KIF1B, a microtubule plus end-directed monomeric motor protein) by small interference RNA inhibited this process. Notably, coimmunoprecipitation analysis indicated that leptin enhanced the interaction of MT1-MMP with KIF1B in a time-dependent manner, which consequently contributed to GC cell invasion. Moreover, leptin increased MT1-MMP or KIF1B expression by the protein kinase B (AKT) pathway and extracellular signal-regulated kinase 1/2 partially participated in this process. However, only AKT was implicated in the leptin-mediated membrane localization of MT1-MMP. Immunohistochemistry analysis revealed that leptin, MT1-MMP and KIF1B are overexpressed in GC tissues, and they positively correlated with clinical stage and lymph node metastasis. These observations indicate that this regulatory network exists in vivo. Taken together, our findings suggest that leptin is an effective intracellular stimulator of MT1-MMP and that leptin-enhanced cell surface localization of MT1-MMP is dependent on KIF1B, which consequently plays a critical role in GC invasion.
-
Leptin‐mediated suppression of lipoprotein lipase cleavage enhances lipid uptake and facilitates lymph node metastasis in gastric cancer
Jian Xiao,Shuqing Cao,Jiawei Wang,Pengyu Li,Quan Cheng,Xinyi Zhou,Jiacheng Dong,Yuan Li,Xinyu Zhao,Zekuan Xu,Li Yang
DOI: https://doi.org/10.1002/cac2.12583
IF: 15.2825
2024-07-03
Cancer Communications
Abstract:Abstract Background Lymph node metastasis (LNM) is the primary mode of metastasis in gastric cancer (GC). However, the precise mechanisms underlying this process remain elusive. Tumor cells necessitate lipid metabolic reprogramming to facilitate metastasis, yet the role of lipoprotein lipase (LPL), a pivotal enzyme involved in exogenous lipid uptake, remains uncertain in tumor metastasis. Therefore, the aim of this study was to investigate the presence of lipid metabolic reprogramming during LNM of GC as well as the role of LPL in this process. Methods Intracellular lipid levels were quantified using oil red O staining, BODIPY 493/503 staining, and flow cytometry. Lipidomics analysis was employed to identify alterations in intracellular lipid composition following LPL knockdown. Protein expression levels were assessed through immunohistochemistry, Western blotting, and enzyme‐linked immunosorbent assays. The mouse popliteal LNM model was utilized to investigate differences in LNM. Immunoprecipitation and mass spectrometry were employed to examine protein associations. In vitro phosphorylation assays and Phos‐tag sodium dodecyl‐sulfate polyacrylamide gel electrophoresis assays were conducted to detect angiopoietin‐like protein 4 (ANGPTL4) phosphorylation. Results We identified that an elevated intracellular lipid level represents a crucial characteristic of node‐positive (N+) GC and further demonstrated that a high‐fat diet can expedite LNM. LPL was found to be significantly overexpressed in N+ GC tissues and shown to facilitate LNM by mediating dietary lipid uptake within GC cells. Leptin, an obesity‐related hormone, intercepted the effect exerted by ANGPTL4/Furin on LPL cleavage. Circulating leptin binding to the leptin receptor could induce the activation of inositol‐requiring enzyme‐1 (IRE1) kinase, leading to the phosphorylation of ANGPTL4 at the serine 30 residue and subsequently reducing its binding affinity with LPL. Moreover, our research revealed that LPL disrupted lipid homeostasis by elevating intracellular levels of arachidonic acid, which then triggered the cyclooxygenase‐2/prostaglandin E2 (PGE2) pathway, thereby promoting tumor lymphangiogenesis. Conclusions Leptin‐induced phosphorylation of ANGPTL4 facilitates LPL‐mediated lipid uptake and consequently stimulates the production of PGE2, ultimately facilitating LNM in GC.
oncology
-
Overexpressed MAGP1 is Associated with a Poor Prognosis and Promotes Cell Migration and Invasion in Gastric Cancer
Mengjie Wu,Yongfeng Ding,Xiaoxia Jiang,Yanyan Chen,Nan Wu,Linrong Li,Haiyong Wang,Yingying Huang,Nong Xu,Lisong Teng
DOI: https://doi.org/10.3389/fonc.2019.01544
IF: 4.7
2020-01-01
Frontiers in Oncology
Abstract:Gastric cancer (GC) is a frequently occurring malignancy with high mortality rates. However, the underlying mechanism of GC progression is not very clear. The aim of this study is to reveal the inherent molecular mechanism and develop potential therapeutic targets for advanced GC. The microfibril-associated glycoprotein 1 (MAGP1), identified as a potential oncogene, was found upregulated in GC tissues and high MAGP1 expression was associated with aggressive clinicopathological features. Furthermore, the multivariate Cox regression analysis showed that high MAGP1 expression was an independent predictor of poor prognosis (HR = 2.37, 1.07-5.24; P = 0.033). Mechanistically, MAGP1 promoted the migration and invasiveness of GC cells. In addition, the genes co-expressed with MAGP1 were primarily enriched in focal adhesion and PI3K-Akt pathways. MAGP1 overexpression enhanced the phosphorylation of FAK, AKT, and mTOR, whereas its knockdown also inactivated these factors. Furthermore, the AKT inhibitor suppressed the phosphorylation of AKT, FAK, and mTOR in recMAGP1-treated AGS cells, as well as their migration and invasion capacities. Finally, correlation analysis indicated that MAGP1 is involved in AKT signaling in GC, and is clinically relevant. Taken together, MAGP1 is a promising prognostic marker and potential therapeutic target for advanced GC.
-
Interleukin-23 Promotes the Migration and Invasion of Gastric Cancer Cells by Inducing Epithelial-to-mesenchymal Transition Via the STAT3 Pathway.
Xianhui Xu,Changdong Yang,Jun Chen,Junyan Liu,Ping'ang Li,Yan Shi,Peiwu Yu
DOI: https://doi.org/10.1016/j.bbrc.2018.03.144
IF: 3.1
2018-01-01
Biochemical and Biophysical Research Communications
Abstract:Chronic inflammation is associated with all stages of cancer development. Moreover, a proinflammatory microenvironment resulted from chronic inflammation is considered to be an essential component of cancer. Interleukin-23 (IL-23) is a general proinflammatory factor; and is involved in tumor-associated inflammation in gastric cancer (GC). However, the direct effect of IL-23 on GC cells has been rarely reported. The aim of the study was to clarify the direct role of IL-23 in regulating GC progression, and to identify the underlying mechanism. In this study, Positive expression of IL-23R was observed in GC tissues and cell lines by using immunohistochemistry or immunofluorescence. In western blots, the expression of IL-23R was higher in GC tissues compared with adjacent normal tissues. Furthermore, IL-23R positive GC tissues were closely related with larger tumor size and worse T stage and clinical stage. By performing in vitro experiments, we found that IL-23 binding to its receptor promoted the migration and invasion of BGC-823 cells in vitro. Moreover, IL-23 induced the activation of STAT3 and epithelial-to-mesenchymal transition (EMT) in BGC-823 cells. Knocking down STAT3 in BGC-823 cells attenuated the effect of IL-23 on EMT and cell migration and invasion. Taken together, our study has firstly demonstrated the positive expression of IL-23R in human GC tissues and cell lines. IL-23 binding to its receptor promotes the migration and invasion of GC cells by inducing EMT through the STAT3 signaling pathway. This work provides a new mechanism for the oncogenic role of IL-23 on GC progression.
-
Leptin Signaling Enhances Cell Invasion and Promotes the Metastasis of Human Pancreatic Cancer Via Increasing MMP-13 Production.
Yingchao Fan,Yu Gan,Yuling Shen,Xiaojin Cai,Yanfang Song,Fangyu Zhao,Ming Yao,Jianren Gu,Hong Tu
DOI: https://doi.org/10.18632/oncotarget.3878
2015-01-01
Oncotarget
Abstract:Emerging evidence has suggested that leptin, an adipokine related to energy homeostasis, plays a role in cancer growth and metastasis. However, its impact on pancreatic cancer is rarely studied. In this study, we found that leptin's functional receptor Ob-Rb was expressed in pancreatic cancer cell lines. Treatment with leptin enhanced the migration and invasion of pancreatic cancer cells but did not affect the proliferation of human pancreatic cancer cells. Leptin up-regulated the expression of matrix metalloproteinase-13 (MMP-13) via the JAK2/STAT3 signaling pathway. The overexpression of leptin was shown to significantly promote tumor growth and lymph node metastasis in a subcutaneous model and an orthotopic model of human pancreatic cancer, respectively. Furthermore, in human pancreatic cancer tissues, the expression of Ob-Rb was positively correlated with the MMP-13 level. The increased expression of either Ob-Rb or MMP-13 was significantly associated with lymph node metastasis and tended to be associated with the TNM stage in patients with pancreatic cancer. Our findings suggest that leptin enhances the invasion of pancreatic cancer through the increase in MMP-13 production, and targeting the leptin/MMP-13 axis could be an attractive therapeutic strategy for pancreatic cancer.
-
CRIP1 Reshapes the Gastric Cancer Microenvironment to Facilitate Development of Lymphatic Metastasis
Zhonghua Wu,Bicheng Qu,Minxian Yuan,Jingjing Liu,Cen Zhou,Mingwei Sun,Zhexu Guo,Yaqing Zhang,Yongxi Song,Zhenning Wang
DOI: https://doi.org/10.1002/advs.202303246
IF: 15.1
2023-07-08
Advanced Science
Abstract:Cysteine‐rich intestinal protein‐1 (CRIP1) can interact with the cAMP responsive element binding protein 1 (CREB1) and enhance CREB1‐mediated VEGFC and CCL5 transcription. Secretion of VEGFC can induce lymphangiogenesis, while increased CCL5 in tumor microenvironment can form a concentration gradient to recruit M2 TAM, then promote TNF‐α secretion to increase lymphatic permeability. Through the reciprocal effects of VEGFC and CCL5, CRIP1 can promote lymphangiogenesis and lymphatic metastasis development in gastric cancer. Lymphangiogenesis in tumors provides an auxiliary route for cancer cell invasion to drainage lymph nodes, facilitating the development of lymphatic metastasis (LM). However, the mechanisms governing tumor lymphangiogenesis and lymphatic permeability in gastric cancer (GC) remain largely unknown. Here, the unprecedented role and mechanism of cysteine‐rich intestinal protein‐1 (CRIP1) in mediating the development of GC LM is uncovered. A series of assays are performed to identify downstream targets of CRIP1, and rescue experiments are performed to confirm the effects of this regulatory axis on LM. CRIP1 overexpression facilitates LM in GC by promoting lymphangiogenesis and lymphatic vessel permeability. CRIP1 promotes phosphorylation of cAMP responsive element binding protein 1(CREB1), which then mediates vascular endothelial growth factor C (VEGFC) expression necessary for CRIP1‐induced lymphangiogenesis and transcriptionally promotes C‐C motif chemokine ligand 5 (CCL5) expression. CCL5 recruits macrophages to promote tumor necrosis factor alpha (TNF‐α) secretion, eventually enhancing lymphatic permeability. The study highlights CRIP1 regulates the tumor microenvironment to promote lymphangiogenesis and LM in GC. Considering the current limited understanding of LM development in GC, these pathways provide potential targets for future therapeutics.
materials science, multidisciplinary,nanoscience & nanotechnology,chemistry
-
Lymphatic Endothelial Cell-Secreted Cxcl1 Stimulates Lymphangiogenesis and Metastasis of Gastric Cancer
Jianbo Xu,Changhua Zhang,Yulong He,Hui Wu,Zhao Wang,Wu Song,Wen Li,Weiling He,Shirong Cai,Wenhua Zhan
DOI: https://doi.org/10.1002/ijc.26035
2011-01-01
International Journal of Cancer
Abstract:Lymph node metastasis is a significant factor in gastric cancer prognosis. It is well known that cancer cells secrete lymphangiogenic factors, thereby promoting lymphangiogenesis. However, the effects of lymphatic endothelial cell (LEC)-secreted factors on the process of lymphangiogenesis and tumor cell metastasis remain unclear. We established an animal model and successfully isolated LECs from afferent lymph vessels of sentinel lymph nodes (SLNs) in animal models. A microarray analysis was performed to characterize gene expression profile in afferent LECs induced by metastatic cancer cells. There were significant differences in 846 genes between normal LECs and LECs with lymph node metastasis. Among these genes, we found that expression of CXCL1 was upregulated, which was confirmed by quantitative reverse-transcriptase polymerase chain reaction. In a coculture system, gastric cancer cells induced CXCL1 secretion from LECs, which was associated with the NF-κB pathway. CXCL1 stimulated LECs migration and tube formation involving FAK-ERK1/2-RhoA activation and reorganization of F-actin. In human gastric cancer specimens, CXCR2 expression was positively correlated with TNM (Tumor, node, metastasis) stage and lymphatic vessel density. These results suggested that LECs of afferent SLNs had specific expression profiles, which were distinct from those of normal lymphatic vessels and appeared to promote metastasis. The expression pattern described in our study, including CXCL1 in LECs and its receptor CXCR2 in cancer cells, offers a promising therapeutic target for gastric cancer.
-
ICAM1 (CD54) Contributes to the Metastatic Capacity of Gastric Cancer Stem Cells
José Manuel Tinajero-Rodríguez,Lizbeth Ramírez-Vidal,Jared Becerril-Rico,Eduardo Alvarado-Ortiz,Dámaris P Romero-Rodríguez,Fernando López-Casillas,Daniel Hernández-Sotelo,Fernando Fernández-Ramírez,Adriana Contreras-Paredes,Elizabeth Ortiz-Sánchez
DOI: https://doi.org/10.3390/ijms25168865
2024-08-14
Abstract:Gastric cancer is the fourth leading cause of cancer deaths worldwide. The presence of chemoresistant cells has been used to explain this high mortality rate. These higher tumorigenic and chemoresistant cells involve cancer stem cells (CSCs), which have the potential for self-renewal, a cell differentiation capacity, and a greater tumorigenic capacity. Our research group identified gastric cancer stem cells (GCSCs) with the CD24+CD44+CD326+ICAM1+ immunophenotype isolated from gastric cancer patients. Interestingly, this GCSC immunophenotype was absent in cells isolated from healthy people, who presented a cell population with a CD24+CD44+CD326+ immunophenotype, lacking ICAM1. We aimed to explore the role of ICAM1 in these GCSCs; for this purpose, we isolated GCSCs from the AGS cell line and generated a GCSC line knockout for ICAM1 using CRISPR/iCas9, which we named GCSC-ICAM1KO. To assess the role of ICAM1 in the GCSCs, we analyzed the migration, invasion, and chemoresistance capabilities of the GCSCs using in vitro assays and evaluated the migratory, invasive, and tumorigenic properties in a zebrafish model. The in vitro analysis showed that ICAM1 regulated STAT3 activation (pSTAT3-ser727) in the GCSCs, which could contribute to the ability of GCSCs to migrate, invade, and metastasize. Interestingly, we demonstrated that the GCSC-ICAM1KO cells lost their capacity to migrate, invade, and metastasize, but they exhibited an increased resistance to a cisplatin treatment compared to their parental GCSCs; the GCSC-ICAM1KO cells also exhibited an increased tumorigenic capability in vivo.
-
LETM1 Promotes Gastric Cancer Cell Proliferation, Migration, and Invasion Via the PI3K/Akt Signaling Pathway
Yunfeng Zhang,Lele Chen,Yifan Cao,Si Chen,Chao Xu,Jun Xing,Kaiguang Zhang
DOI: https://doi.org/10.5230/jgc.2020.20.e12
2020-01-01
Journal of the Korean Gastric Cancer Association
Abstract:Purpose Globally, there is a high incidence of gastric cancer (GC). Leucine zipper-EF-hand containing transmembrane protein 1 (LETM1) is reported to play a vital role in several human malignancies. However, there is limited understanding of the role of LETM1 in GC. This study aims to investigate the effects of LETM1 on proliferation, migration, and invasion of GC cells. Materials and Methods The expression levels of LETM1 in the normal gastric mucosal epithelial cells (GES-1) and GC cells were analyzed by quantitative real-time polymerase chain reaction and western blotting. CCK-8, wound healing, and Transwell invasion assays were performed to evaluate the effect of LETM1 knockdown or overexpression on the proliferation, migration, and invasion of the GC cells, respectively. Additionally, the effect of LETM1 knockdown or overexpression on GC cell apoptosis was determined by flow cytometry. Furthermore, the effect of LETM1 knockdown or overexpression on the expression levels of PI3K/Akt signaling pathway-related proteins was evaluated by western blotting. Results The GC cells exhibited markedly higher mRNA and protein expression levels of LETM1 than the GES-1 cells. Additionally, the knockdown of LETM1 remarkably suppressed the GC cell proliferation, migration, and invasion, and promoted the apoptosis of GC cells, which were reversed upon LETM1 overexpression. Furthermore, the western blotting analysis indicated that LETM1 facilitates GC progression via the PI3K/Akt signaling pathway. Conclusions LETM1 acts as an oncogenic gene to promote GC cell proliferation, migration, and invasion via the PI3K/Akt signaling pathway. Therefore, LETM1 may be a potential target for GC diagnosis and treatment.
-
Apolipoprotein C1 regulates gastric cancer progression by inducing epithelial-mesenchymal transition via the JAK/STAT pathway
Jie Zhang,Zhi Yang,Zijian Li,Ruiqing Li,Da Li,Min Feng,Wenxian Guan,Xinnong Liu
DOI: https://doi.org/10.21203/rs.3.rs-2659063/v1
2023-01-01
Abstract:Abstract This study aimed to determine the role of apolipoprotein C1 (APOC1) in GC (gastric cancer), as well as elucidate the mechanism of its effects. We studied the relationship between clinicopathological characteristics and APOC1 expression in 127 patients with GC, and determined the effects of APOC1 on GC cell apoptosis, proliferation, migration, invasion, and metastasis. We analyzed markers of epithelial-mesenchymal transition (EMT) and JAK/STAT signaling protein expression, and determined the effects of the STAT3 activator colivelin on APOC1-knockdown GC cells. High APOC1 expression was observed in GC tissues and cells; the level correlated with GC differentiation degree, T stage, and TNM stage. There was an association between high APOC1 expression and poor prognosis. APOC1 knockdown induced GC cell apoptosis and inhibited EMT, migration, invasion, and proliferation in vitro , besides reduced the weight and volume of subcutaneous xenotransplanted tumors and the number of hepatic metastatic foci in vivo in a mouse model. APOC1 knockdown caused inhibition of phosphorylated-STAT3 protein expression, upregulation of E-cadherin, and downregulation of N-cadherin and vimentin in GC cells; these effects were reversed by JAK/STAT reactivation. Thus, APOC1 participates in EMT and regulates apoptosis, invasion, migration, as well as metastasis of gastric cancer via the JAK1/STAT3 pathway.
-
Gallbladder Cancer Cell-Derived Exosome-Mediated Transfer of Leptin Promotes Cell Invasion and Migration by Modulating STAT3-Mediated M2 Macrophage Polarization
Songling Zhao,Yunxia Liu,Linhai He,Yuehua Li,Ke Lin,Qiang Kang,Lixin Liu,Hao Zou
DOI: https://doi.org/10.1155/2022/9994906
2022-01-24
Analytical Cellular Pathology
Abstract:Tumor-associated macrophage (TAM) is a major component of tumor microenvironment (TME) and plays critical role in the progression of cancer metastasis. However, TAM-mediated regulation in gallbladder cancer (GBC) has not been fully characterized. Here, we found that exosomes derived from GBC cell polarized macrophage to M2 phenotype, which then facilitated the invasion and migration of GBC cells. We discovered that leptin was enriched in GBC cell-derived exosomes. Exosomal leptin levels promoted invasion and migration of GBC-SD cells. The inhibition of leptin not only attenuated M2 macrophage of polarization but also inhibited the invasive and migratory ability of GBC cell. In addition, GBC-SD cell-derived exosomal leptin induced M2 polarization of macrophage via activation of STAT3 signal pathway. Taken together, our results suggested that GBC cells secrete exosome-enclosed leptin facilitated cell invasion and migration via polarizing TAM.
oncology,pathology,cell biology
-
Fut3 Promotes Gastric Cancer Cell Migration by Synthesizing Lea on Itga6 and Glg1, Affecting Adhesion and Vesicle Distribution
Fei Wu,Li Cao,Jinyuan Zhang,Shuang Cai,Huizi Wu,Jiyu Miao,Lingyu Zhao,Changan Zhao,Xiaofei Wang,Muhammad Anas Ramzan,Sadiq Ali,Feng Wu,Lei Ni,Liying Liu,Yannan Qin,Chen Huang
DOI: https://doi.org/10.1016/j.lfs.2024.123193
IF: 6.78
2024-01-01
Life Sciences
Abstract:AIMS:Lewis antigen plays an important role in the progression of gastric cancer (GC), FUT3 is a key enzyme in the synthesis of Lewis antigen, but the molecular mechanism of its promotion of GC progression remains unclear. MAIN METHODS:We used Lea-antibody capturing coupled with mass spectrometry to identify the target proteins of FUT3, immunofluorescence (IF), molecular biology and cell function experiments were conducted to clarify the molecular mechanism of FUT3 promoting the migration and invasion of GC cells by regulating Lea glycosylation on ITGA6 and GLG1. KEY FINDINGS:FUT3 promote migration and invasion of GC cells. FUT3 silencing in GC cells led to the aggregation of integrin α6β4 on the plasma membrane, associated with focal adhesion and hemidesmosome, and decreased GLG1 distribution in cellular vesicles. IGP analysis revealed Lea structure in 10 N-glycans of 2 glycosites for ITGA6 and 31 N-glycans of 4 glycosites for GLG1. Silencing ITGA6 promoted migration and invasion, while silencing GLG1 inhibited these processes in GC cells, regulated by FUT3-mediated Lea synthesis. SIGNIFICANCE:In conclusion, FUT3's promotion of GC cell migration and invasion is attributed to Lea synthesis on ITGA6, impacting cell adhesion, and on GLG1, influencing distribution in intracellular vesicles. These findings may contribute to developing novel therapeutic targets for inhibiting or controlling the metastatic behavior of GC cells and enhancing our understanding of Lea's role in regulating protein functions.
-
Cancer-Associated Fibroblast-Secreted Exosomes Promote Gastric Cancer Cell Migration and Invasion via the IL-32/ESR1 Axis
Lifeng Shang,Xinli Chen,Tianyu Zhu,Shujing Chong,Haiwang Liu,Wei Huang,Weibo Fu,Hao She,Xin Shen
DOI: https://doi.org/10.1007/s12010-023-04782-6
2024-01-06
Applied Biochemistry and Biotechnology
Abstract:Exosomes secreted by cancer-associated fibroblasts (CAFs) play a critical part in cancer progression. This study aimed to explore the effects of CAF-exosomes on gastric cancer (GC) cell metastasis. AGS and HGC-27 cells were treated with exosomes and cell viability, migration, and invasion were evaluated using Cell-Counting Kit-8 and Transwell assays. Exosome-regulated mRNAs were explored using quantitative real-time PCR. The relationship between interleukin (IL)32 and estrogen receptor 1 (ESR1) was evaluated using co-immunoprecipitation and dual-luciferase reporter assays. The results of this study show that CAF-derived exosomes promote GC cell viability, migration, and invasion. Exosome treatment increased the levels of IL32, which interacted with ESR1 and negatively regulated ESR1 levels. Rescue experiments were conducted to demonstrate that CAF-exosomes promoted biological behaviors of GC cells by upregulating IL32 and downregulating ESR1 expression. In conclusion, CAF-derived exosomes promote GC cell viability, migration, and invasion by elevating the IL32/ESR1 axis, suggesting a novel strategy for metastatic GC treatment.
biotechnology & applied microbiology,biochemistry & molecular biology
-
Apolipoprotein C‐II induces EMT to promote gastric cancer peritoneal metastasis via PI3K/AKT/mTOR pathway
Chao Wang,Zhi Yang,En Xu,Xiaofei Shen,Xingzhou Wang,Zijian Li,Heng Yu,Kai Chen,Qiongyuan Hu,Xuefeng Xia,Song Liu,Wenxian Guan
DOI: https://doi.org/10.1002/ctm2.522
IF: 8.554
2021-08-01
Clinical and Translational Medicine
Abstract:BackgroundPeritoneal metastasis (PM) occurs frequently in patients with gastric cancer (GC) and confers poor survival. Lipid metabolism acts as a non-negligible regulator in epithelial–mesenchymal transition (EMT), which is crucial for the metastasis of GC. As apolipoprotein C2 (APOC2) is a key activator of lipoprotein lipase for triglyceride metabolism, the exact mechanism of APOC2 remains largely unknown in GC. MethodsTandem mass tags identified differentially expressed proteins between human PM and GC tissues, and showed that APOC2 overexpressed in PM tissues, which was further confirmed by immunoblotting, immunohistochemistry, and ELISA. Global gene expression changes were identified in APOC2 knockdown cells via RNA-sequencing. The role of APOC2 in lipid metabolism of GC cells was assessed via the Seahorse XF analyzer and lipid staining assays. The biological role of APOC2 in GC cells was determined by 3D Spheroid invasion, apoptosis, colony formation, wound healing, transwell assay, and mouse models. The interaction between APOC2 and CD36 was analyzed by co-immunoprecipitation and biolayer interferometry. The underlying mechanisms were investigated using western blot technique. ResultsAPOC2 overexpressed in GC PM tissues. Upregulation of APOC2 correlated with a poor prognosis in GC patients. APOC2 promoted GC cell invasion, migration, and proliferation via CD36-mediated PI3K/AKT/mTOR signaling activation. Furthermore, APOC2-CD36 axis upregulated EMT markers of GC cells via increasing the phosphorylation of PI3K, AKT, and mTOR. Knockdown either APOC2 or CD36 inhibited the malignant phenotype of cancer cells, and delayed GC PM progression in murine GC models. ConclusionAPOC2 cooperates with CD36 to induce EMT to promote GC PM via PI3K/AKT/mTOR pathway. APOC2-CD36 axis may be a potential target for the treatment of aggressive GC.
oncology,medicine, research & experimental
-
RBMS1 promotes gastric cancer metastasis through autocrine IL-6/JAK2/STAT3 signaling
Mengyuan Liu,Heming Li,Huijing Zhang,Huan Zhou,Taiwei Jiao,Mingliang Feng,Fangjian Na,Mingjun Sun,Mingfang Zhao,Lei Xue,Lu Xu
DOI: https://doi.org/10.1038/s41419-022-04747-3
2022-01-01
Abstract:Metastasis is the most important reason for the poor prognosis of gastric cancer (GC) patients, and the mechanism urgently needs to be clarified. Here, we explored a prognostic model for the estimation of tumor-associated mortality in GC patients and revealed the RNA-binding protein RBMS1 as a candidate promoter gene for GC metastasis by analyzing GOBO and Oncomine high-throughput sequencing datasets for 408 GC patients. Additionally, RBMS1 was observed with overexpression in 85 GC patient clinical specimens by IHC staining and further be verified its role in GC metastasis via inducing EMT process both in in vitro and in vivo experiments. Moreover, we identified that IL-6 was predicted to be one of the most significant upstream cytokines in the RBMS1 overexpression gene set based on the Ingenuity Pathway Analysis (IPA) algorithm. Most importantly, we also revealed that RBMS1 could promote migration and invasion through IL6 transactivation and JAK2/STAT3 downstream signaling pathway activation by influencing histone modification in the promoter regions after binding with the transcription factor MYC in the HGC-27 and SGC-7901 GC cell lines. Hence, we shed light on the potential molecular mechanisms of RBMS1 in the promotion of GC metastasis, which suggests that RBMS1 may be a potential therapeutic target for GC patients.
-
H3K18 lactylation-mediated VCAM1 expression promotes gastric cancer progression and metastasis via AKT-mTOR-CXCL1 axis
Yupeng Zhao,Jiang Jiang,Peng Zhou,Kaiyuan Deng,Ziyuan Lui,Mengqi Yang,Xiao Yang,Jianfang Li,Ranran Li,Jiazeng Xia
DOI: https://doi.org/10.1016/j.bcp.2024.116120
IF: 6.1
2024-03-10
Biochemical Pharmacology
Abstract:The role of the Immunoglobulin Superfamily (IgSF) as adhesion molecules in orchestrating inflammation is pivotal, yet its specific involvement in gastric cancer (GC) remains unknown. We analyzed IgSF components and discerned conspicuously elevated VCAM1 expression in GC, correlating with a poor prognosis. Remarkably, VCAM1 enhances GC cell proliferation and migration by activating AKT-mTOR signaling. Moreover, lactate in the tumor microenvironment (TME) promotes dynamic lactylation of H3K18 (H3K18la), leading to transcriptional activation of VCAM1 in GC cells. Furthermore, VCAM1 actively mediates intercellular communication in the TME. AKT-mTOR-mediated CXCL1 expression is increased by VCAM1, facilitating the recruitment of human GC-derived mesenchymal stem cells (hGC-MSCs), thereby fostering immunesuppression and accelerating cancer progression. In summary, H3K18 lactylation upregulated VCAM1 transcription, which activated AKT-mTOR signaling, and promoted tumor cell proliferation, EMT Transition and tumor metastasis. VCAM1 upregulated CXCL1 expression by AKT-mTOR pathway, so as to facilitate hGC-MSCs and M2 macrophage recruitment and infiltration. These findings provide novel therapeutic targets for GC.
pharmacology & pharmacy
-
Circulating Galectin-3 Promotes Metastasis by Modifying Muc1 Localization on Cancer Cell Surface
Qicheng Zhao,Xiuli Guo,Gerard B. Nash,Philip C. Stone,John Hilkens,Jonathan M. Rhodes,Lu-Gang Yu
DOI: https://doi.org/10.1158/0008-5472.can-09-1096
IF: 11.2
2009-01-01
Cancer Research
Abstract:Abstract Adhesion of circulating tumor cells to the blood vessel endothelium is a critical step in cancer metastasis. We show in this study that galectin-3, the concentration of which is greatly increased in the circulation of cancer patients, increases cancer cell adhesion to macrovascular and microvascular endothelial cells under static and flow conditions, increases transendothelial invasion, and decreases the latency of experimental metastasis in athymic mice. These effects of galectin-3 are shown to be a consequence of its interaction with cancer-associated MUC1, which breaks the “protective shield” of the cell-surface MUC1 by causing MUC1 polarization, leading to exposure of smaller cell-surface adhesion molecules/ligands including CD44 and ligand(s) for E-selectin. Thus, the interaction in the bloodstream of cancer patients between circulating galectin-3 and cancer cells expressing MUC1 bearing the galectin-3 ligand TF (Galβ1,3GalNAc-) promotes metastasis. This provides insight into the molecular regulation of metastasis and has important implications for the development of novel therapeutic strategies for prevention of metastasis. [Cancer Res 2009;69(17):6799–806]
-
Gastrin stimulates pancreatic cancer cell directional migration by activating the Gα12/13–RhoA–ROCK signaling pathway
Ganggang Mu,Qianshan Ding,Hongyan Li,Li Zhang,Lingli Zhang,Ke He,Lu Wu,Yunchao Deng,Dongmei Yang,Lianlian Wu,Ming Xu,Jie Zhou,Honggang Yu
DOI: https://doi.org/10.1038/s12276-018-0081-6
2018-05-01
Abstract:The mechanism by which gastrin promotes pancreatic cancer cell metastasis is unclear. The process of directing polarized cancer cells toward the extracellular matrix is principally required for invasion and distant metastasis; however, whether gastrin can induce this process and its underlying mechanism remain to be elucidated. In this study, we found that gastrin-induced phosphorylation of paxillin at tyrosine 31/118 and RhoA activation as well as promoted the metastasis of PANC-1 cancer cells. Depletion of Gα12 and Gα13 inhibited the phosphorylation of paxillin and downstream activation of GTP-RhoA, blocked the formation and aggregation of focal adhesions and facilitated polarization of actin filaments induced by gastrin. Suppression of RhoA and ROCK also exhibited identical results. Selective inhibition of the CCKBR–Gα12/13–RhoA–ROCK signaling pathway blocked the reoriented localization of the Golgi apparatus at the leading edge of migrated cancer cells. YM022 and Y-27632 significantly suppressed hepatic metastasis of orthotic pancreatic tumors induced by gastrin in vivo. Collectively, we demonstrate that gastrin promotes Golgi reorientation and directional polarization of pancreatic cancer cells by activation of paxillin via the CCKBR–Gα12/13–RhoA–ROCK signal pathway.
biochemistry & molecular biology,medicine, research & experimental
-
CXCL5 Promotes Gastric Cancer Metastasis by Inducing Epithelial-Mesenchymal Transition and Activating Neutrophils
Zheying Mao,Jiahui Zhang,Yinghong Shi,Wei Li,Hui Shi,Runbi Ji,Fei Mao,Hui Qian,Wenrong Xu,Xu Zhang
DOI: https://doi.org/10.1038/s41389-020-00249-z
2020-01-01
Oncogenesis
Abstract:Deregulated expression of chemokines in tumor microenvironment contributes to tumor metastasis by targeting distinct cells. Epithelial-derived neutrophil-activating peptide-78 (ENA78/CXCL5) is upregulated in many cancers and involved in tumor progression. The role and underlying mechanism of CXCL5 in gastric cancer (GC) metastasis remain unclear. In this study, we reported that the expression of CXCL5 was elevated in tumor tissues and positively associated with lymphatic metastasis and tumor differentiation. Stimulation by recombinant human CXCL5 (rhCXCL5) induced epithelial-mesenchymal transition (EMT) in GC cells through the activation of ERK pathway, which enhanced their migration and invasion abilities. The culture supernatant from tumor tissues also enhanced the migration and invasion abilities of GC cells, however, this effect was reversed by pre-treatment with CXCL5 neutralizing antibody. Further studies showed that rhCXCL5 could induce the expression of IL-6 and IL-23 in neutrophils through the activation of ERK and p38 signaling pathways, which in turn facilitated GC cell migration and invasion. The culture supernatant from tumor tissues showed similar effects on neutrophils in a CXCL5-dependent manner. Blockade of IL-6 and IL-23 with neutralizing antibodies reversed the induction of EMT and the increased migration and invasion abilities in GC cells by CXCL5-activated neutrophils. Moreover, CXCL5 activated neutrophils could promote gastric cancer metastasis in vivo. Taken together, our results indicate that CXCL5 acts on gastric cancer cells to induce EMT and mediates pro-tumor activation of neutrophils, which synergistically promotes the metastatic ability of GC cells.