首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
《Translational oncology》2021,14(12):101236
Cancer-associated fibroblasts (CAFs) have important roles in promoting cancer development and progression. We previously reported that high expression of sex-determining region Y (SRY)-box9 (SOX9) in oral squamous cell carcinoma (OSCC) cells was positively correlated with poor prognosis. This study developed three-dimensional (3D) in vitro models co-cultured with OSCC cells and CAFs to examine CAF-mediated cancer migration and invasion in vitro and in vivo. Moreover, we performed an immunohistochemical analysis of alpha-smooth muscle actin and SOX9 expression in surgical specimens from 65 OSCC patients. The results indicated that CAFs promote cancer migration and invasion in migration assays and 3D in vitro models. The invading OSCC cells exhibited significant SOX9 expression and changes in the expression of epithelial–mesenchymal transition (EMT) markers, suggesting that SOX9 promotes EMT. TGF-β1 signalling inhibition reduced SOX9 expression and cancer invasion in vitro and in vivo, indicating that TGF-β1-mediated invasion is dependent on SOX9. In surgical specimens, the presence of CAFs was correlated with SOX9 expression in the invasive cancer nests and had a significant impact on regional recurrence. These findings demonstrate that CAFs promote cancer migration and invasion via the TGF-β/SOX9 axis.  相似文献   

2.
《Translational oncology》2022,15(12):101236
Cancer-associated fibroblasts (CAFs) have important roles in promoting cancer development and progression. We previously reported that high expression of sex-determining region Y (SRY)-box9 (SOX9) in oral squamous cell carcinoma (OSCC) cells was positively correlated with poor prognosis. This study developed three-dimensional (3D) in vitro models co-cultured with OSCC cells and CAFs to examine CAF-mediated cancer migration and invasion in vitro and in vivo. Moreover, we performed an immunohistochemical analysis of alpha-smooth muscle actin and SOX9 expression in surgical specimens from 65 OSCC patients. The results indicated that CAFs promote cancer migration and invasion in migration assays and 3D in vitro models. The invading OSCC cells exhibited significant SOX9 expression and changes in the expression of epithelial–mesenchymal transition (EMT) markers, suggesting that SOX9 promotes EMT. TGF-β1 signalling inhibition reduced SOX9 expression and cancer invasion in vitro and in vivo, indicating that TGF-β1-mediated invasion is dependent on SOX9. In surgical specimens, the presence of CAFs was correlated with SOX9 expression in the invasive cancer nests and had a significant impact on regional recurrence. These findings demonstrate that CAFs promote cancer migration and invasion via the TGF-β/SOX9 axis.  相似文献   

3.
Tumor–stroma interactions are critical for epithelial‐derived tumors, and among the stromal cell types, cancer‐associated fibroblasts (CAFs) exhibit multiple functions that fuel growth, dissemination, and drug resistance. However, these interactions remain insufficiently characterized in non‐epithelial tumors such as malignant melanoma. We generated monocultures of melanoma cells and matching CAFs from patients’ metastatic lesions, distinguished by oncogenic drivers and immunoblotting of characteristic markers. RNA sequencing of CAFs revealed a homogenous epigenetic program that strongly resembled the signatures from epithelial cancers, including enrichment for an epithelial‐to‐mesenchymal transition (EMT). Melanoma CAFs in monoculture displayed robust invasive behavior while patient‐derived melanoma monocultures showed very little invasiveness. Instead, melanoma cells showed increased invasion when co‐cultured with CAFs. In turn, CAFs showed increased proliferation when exposed to melanoma conditioned media (CM), mediated in part by melanoma‐secreted transforming growth factor‐alpha that acted on CAFs via the epidermal growth factor receptor. This study provides evidence that bidirectional interactions between melanoma and CAFs regulate progression of metastatic melanoma.  相似文献   

4.
Cancer-associated fibroblasts (CAFs) play crucial roles in tumor progression, given the dependence of cancer cells on stromal support. Therefore, understanding how CAFs communicate with endometrial cancer cell in tumor environment is important for endometrial cancer therapy. Exosomes, which contain proteins and noncoding RNA, are identified as an important mediator of cell–cell communication. However, the function of exosomes in endometrial cancer metastasis remains poorly understood. In the current study we found that CAF-derived exosomes significantly promoted endometrial cancer cell invasion comparing to those from normal fibroblasts (NFs). We identified a significant decrease of miR-148b in CAFs and CAFs-derived exosomes. By exogenously transfect microRNAs, we demonstrated that miR-148b could be transferred from CAFs to endometrial cancer cell through exosomes. In vitro and in vivo studies further revealed that miR-148b functioned as a tumor suppressor by directly binding to its downstream target gene, DNMT1 to suppress endometrial cancer metastasis. In endometrial cancer DNMT1 presented a potential role in enhancing cancer cell metastasis by inducing epithelial–mesenchymal transition (EMT). Therefore, downregulated miR-148b induced EMT of endometrial cancer cell as a result of relieving the suppression of DNMT1. Taken together, these results suggest that CAFs-mediated endometrial cancer progression is partially related to the loss of miR-148b in the exosomes of CAFs and promoting the transfer of stromal cell-derived miR-148b might be a potential treatment to prevent endometrial cancer progression.  相似文献   

5.
Cancer-associated fibroblasts (CAFs) play a crucial role in cancer progression and even initiation. However, the origins of CAFs in various cancer types remain controversial, and one of the important hypothesized origins is through epithelial-mesenchymal transition (EMT) from cancer cells. In this study, we investigated whether the HEp-2 laryngeal cancer cells are able to generate CAFs via EMT during tumor formation, which is now still unknown. The laryngeal xenografted tumor model was established by inoculating the HEp-2 laryngeal cancer cell line in nude mice. Primary cultured CAFs from the tumor nodules and matched normal fibroblasts (NFs) from the adjacent connective tissues were subcultured, purified, and verified by immunofluorescence. Migration, invasion, and proliferation potentials were compared between the CAFs and NFs. A co-culture of CAFs with HEp-2 cells and a co-injection of CAFs with HEp-2 cells in nude mice were performed to examine the cancer-promoting potential of CAFs to further verify their identity. Karyotypic analyses of the CAFs, NFs, and HEp-2 cells were conducted. A co-culture of NFs with HEp-2 cells was also performed to examine the expression of activated markers of CAFs. A pathological examination confirmed that the laryngeal xenografted tumor model was successfully established, containing abundant CAFs. Immunocytochemical staining verified the purities and identities of the CAFs and NFs. Although the CAFs manifested higher migration, invasion, proliferation, and cancer-promoting capacities compared with the NFs, an analysis of chromosomes revealed that both the CAFs and NFs showed typical normal mouse karyotypes. In addition, the NFs co-cultured with HEp-2 cells did not show induced expressions of activated markers of CAFs. Our findings reveal that the CAFs in the HEp-2 established laryngeal xenografted tumor are not of laryngeal cancer origin but of mouse origin, indicating that the HEp-2 laryngeal cancer cells cannot generate their own CAFs via EMT in this model.  相似文献   

6.
Tumor progression requires the communication between tumor cells and tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs) are major components of stromal cells. CAFs contribute to metastasis process through direct or indirect interaction with tumor cells; however, the underlying mechanism is largely unknown. Here, we reported that autophagy was upregulated in lung cancer-associated CAFs compared to normal fibroblasts (NFs), and autophagy was responsible for the promoting effect of CAFs on non-small cell lung cancer (NSCLC) cell migration and invasion. Inhibition of CAFs autophagy attenuated their regulation on epithelial–mesenchymal transition (EMT) and metastasis-related genes of NSCLC cells. High mobility group box 1 (HMGB1) secreted by CAFs mediated CAFs’ effect on lung cancer cell invasion, demonstrated by using recombinant HMGB1, HMGB1 neutralizing antibody, and HMGB1 inhibitor glycyrrhizin (GA). Importantly, the autophagy blockade of CAFs revealed that HMGB1 release was dependent on autophagy. We also found HMGB1 was responsible, at least in part, for autophagy activation of CAFs, suggesting CAFs remain active through an autocrine HMGB1 loop. Further study demonstrated that HMGB1 facilitated lung cancer cell invasion by activating the NFκB pathway. In a mouse xenograft model, the autophagy specific inhibitor chloroquine abolished the stimulating effect of CAFs on tumor growth. These results elucidated an oncogenic function for secretory autophagy in lung cancer-associated CAFs that promotes metastasis potential, and suggested HMGB1 as a novel therapeutic target.Subject terms: Cancer microenvironment, Non-small-cell lung cancer, Metastasis, Translational research  相似文献   

7.

Background

It is supposed that fibroblasts present in tumour microenvironment increase cancer invasiveness and its ability to metastasize but the mechanisms have not been clearly defined yet. Thus, the current study was designed to assess changes in gene expression in five various cancer cell lines grown as a co-culture with the carcinoma-associated fibroblasts (CAFs) in vitro.

Results

A carcinoma-associated fibroblast cell line was isolated from a canine mammary cancer. Then, a co-culture of cancer cells with the CAFs was established and maintained for 72 hrs. Having sorted the cells, a global gene expression in cancer cells using DNA microarrays was examined. The analysis revealed an up-regulation of 100 genes and a down-regulation of 106 genes in the cancer cells grown as a co-culture with the CAFs in comparison to control conditions. The PANTHER binomial statistics tool was applied to determine statistically over-manifested pathways (p < 0.05). Bulk of the up-regulated genes are involved in the adhesion, the angiogenesis, the epithelial-mesenchymal transition (EMT) and generally take part in the developmental processes. These results were further confirmed using real-time qPCR. Moreover, a wound-healing assay and growth characteristics on Matrigel matrix showed that CAFs increase cancer cell migration and matrix invasion.

Conclusion

The results of the current study showed that the co-culturing of cancer cells and the CAFs caused significant changes to the cancer gene expression. The presence of the CAFs in a microenvironment of cancer cells promotes adhesion, angiogenesis and EMT.  相似文献   

8.
Epithelial to mesenchymal transitions (EMT) have been suggested to be crucial during epithelial cancer cell invasion. However, in a three-dimensional “organotypic” invasion assay squamous cell carcinoma (SCC) cells that retain epithelial characteristics “hitch a ride” with carcinoma associated fibroblasts (CAFs) in order to collectively invade. Thus epithelial cancer cells can utilise the mesenchymal characteristics of CAFs without the need to undergo EMT themselves. This work provides new insight in cancer cell invasion and shows a new role for CAFs as a target for an anti-invasive therapy.Key words: collective invasion, carcinoma associated fibroblast, extracellular matrix, matrix metalloproteinases, RhoCancer cell invasion and metastasis are the main causes of mortality in cancer patients. Understanding how cancer cells move and invade within the surrounding tissue is therefore a key issue. Stromal fibroblasts within a tumor play a crucial role in cancer cell proliferation, survival, angiogenesis as well as invasion (reviewed in ref. 1). In many cases stromal CAFs are able to produce a wide range of growth factors and cytokines that modulate tumor growth and invasion.2,3 Their influence in cancer cell invasion and metastasis can also be mediated through the production of MMP''s that promote extra-cellular matrix degradation.4It has recently been shown that CAFs can play an unexpected role in SCC invasion.5 In a 3D ‘organotypic’ model of invasion that recreates the epidermal/dermal environment CAFs promote the collective invasion of SCC cells.6 3D time-lapse confocal microscopy imaging showed that CAFs were always the leading cell of the invading cohort with the SCC cells following behind. These cohorts closely resembled invading clusters of SCC cells observed in human cancer samples.7 CAFs promoted SCC cells collective invasion by remodelling the matrix and making a path that SCC cells can use to invade. This process is clearly shown in Figure 1: a CAF (in red) leads the invasion of a collective chain of SCC cells (green) and makes a path in the surrounding matrix, visualized in grey using confocal reflectance microscopy. Two key experiments helped to understand the role of fibroblasts in this system. Firstly, the separation of the two cell populations by a thin layer of gel without fibroblasts completely abolished SCC invasion and so ruled out the possibility of long distance chemoattractant molecules inducing SCC invasion. Secondly, SCC cells were able to invade into a gel which had previously been remodelled by CAFs that had subsequently been removed. Together these experiments showed that tracks made by the fibroblasts are essential and sufficient to promote collective carcinoma cells invasion. Heterotypic cell contact between both populations was not required, as SCC cells can invade using tracks made by the CAFs even if the CAFs have been removed.Open in a separate windowFigure 1Collective invasion of carcinoma cells led by fibroblast. Confocal time-lapse imaging of carcinoma associated fibroblast (red) leading the way of an invading chain of SCC cells (green) and making path into the surrounding matrix (grey). Panel is 80 x 80 mm and spans 300 minutes, scale are 20 um.Interestingly, inhibition of Rho/ROCK signalling to the actomyosin cytoskeleton or MMPs using small molecule inhibitors blocked SCC invasion even when only CAFs where targeted. Blocking these pathways in carcinoma cells had little or no effect on their invasion. Moreover, inhibition of Rho function specifically in CAFs did not block their invasion into matrices but prevented SCC cells from following. These experiments showed the role of Rho/ROCK and MMPs molecular pathways in track generation by the CAFs and that targeting these pathways in CAFs, but not SCC cells, is critical for preventing cancer invasion. Strikingly, blockade of protease function after CAFs had remodelled the ECM had little effect on the ability of SCC cells to invade. This could explain the relative poor results obtained using MMPs inhibitors as anti-invasive therapies.8 Rho/ROCK function was dispensable in SCC cells; however, depletion of the small GTPase Cdc42 and its effector MRCK disrupted the acto-myosin cortex of carcinoma cells and blocked their capacity to invade in response to CAFs.In order to invade and metastasise, carcinoma cells can switch from an epithelial state to a more mesenchymal phenotype.9 This process, called EMT, allows epithelial cancer cells to adapt their behaviour and confers the capacity to remodel the ECM on the cancer cells.10 However, in patient tissue samples, it has been observed that carcinoma cells can invade without undergoing an EMT, these cancer cells do not upregulate mesenchymal markers and retain cell to cell contact during their invasion.11 This work explains how carcinoma cells that have not undergone EMT could invade a 3D matrix. These cells use the mesenchymal characteristics of the stromal fibroblasts to remodel the ECM and consequently follow behind invading fibroblasts. In tumours of mesenchymal origin CAFs are not required for invasion; work from Friedl and colleagues, clearly shows that HT1080 fibrosarcoma cells could lead collectively invading chains of cancer cells The authors showed how the leading cell of the collective chain remodels collagen fibres into tracks as it invades through the action of MT1-MMP (MMP14).12In normal conditions, epithelial cells and dermal fibroblasts are in complete homeostasis and separated by a basement membrane (Fig. 2A). In addition, normal dermal fibroblasts are unable to promote SCC invasion. Understanding how CAFs are activated will be an important step forward. A desmoplastic response is observed in many tumours indicating a change in behaviour of fibroblasts.13 During wound healing or fibrosis, fibroblasts are in an active state that has been suggested to be similar to cancer activation.14 TGFβ has been shown to be a key player in fibroblasts activation and could support cancer progression.15 However, TGFβ was not responsible for SCC cells invasion since a TGFβ inhibitor had no effect in carcinoma cells collective invasion induced by the CAFs in the 3D invasion assay (Cedric Gaggioli and Steven Hooper, unpublished data). Interestingly, a probe that binds only to the active form of the small GTPase Rho showed that the activity of this protein was increased in CAFs compared to normal fibroblasts in tissue samples. Elevated expression of α5 integrin was also present in these cells and this has been implicated in Rho activation in a number of systems.1618 Consistent with this observation, depletion of integrin a5 in CAFs reduced their ability to promote the invasion of SCC cells. Alternatively, CAFs could also be derived from endothelial cells through a process called endothelial to mesenchymal transition19 (EndMT), or from cancer cells through EMT.20 These processes could be responsible for CAFs generation in the tumor stroma resulting in matrix remodelling and tracks generation in order for the carcinoma cells to collectively invade the surrounding tissue and metastasize (Fig. 2B).Open in a separate windowFigure 2Model of carcinoma cells collective invasion. (A) Schematic representation of a normal epithelium. Epithelial cells (light blue) and normal fibroblasts (pink) are separated by a basal membrane and are in a perfect homeostasis. Cross talk between both cell types occurs through adhesion and chemokine secretion. (B) Schematic representation of carcinoma cells collective invasion. CAFs (red) take the lead of a collective invading chain of SCC cells (brown). Invasion of CAFs is MMPs dependent but Rho/ROCK independent. However, track generation by CAFs is Rho/ROCK/MLC dependent. SCC cells require the small GTPase Cdc42 and its effector MRCK in order to collectively invade trough those tracks (black).This study opens a new field of investigation for collective cancer cell invasion. This work highlights carcinoma associated fibroblasts as new therapeutic targets which will be a new direction in cancer cell invasion and metastasis therapy.  相似文献   

9.
Cancer-associated fibroblasts (CAFs) have important roles in promoting cancer development and progression. We previously reported that high expression of sex-determining region Y (SRY)-box9 (SOX9) in oral squamous cell carcinoma (OSCC) cells was positively correlated with poor prognosis. This study developed three-dimensional (3D) in vitro models co-cultured with OSCC cells and CAFs to examine CAF-mediated cancer migration and invasion in vitro and in vivo. Moreover, we performed an immunohistochemical analysis of alpha-smooth muscle actin and SOX9 expression in surgical specimens from 65 OSCC patients. The results indicated that CAFs promote cancer migration and invasion in migration assays and 3D in vitro models. The invading OSCC cells exhibited significant SOX9 expression and changes in the expression of epithelial–mesenchymal transition (EMT) markers, suggesting that SOX9 promotes EMT. TGF-β1 signalling inhibition reduced SOX9 expression and cancer invasion in vitro and in vivo, indicating that TGF-β1-mediated invasion is dependent on SOX9. In surgical specimens, the presence of CAFs was correlated with SOX9 expression in the invasive cancer nests and had a significant impact on regional recurrence. These findings demonstrate that CAFs promote cancer migration and invasion via the TGF-β/SOX9 axis.  相似文献   

10.
11.
12.
13.
ObjectiveCancer-associated fibroblasts (CAFs) function as a crucial factor in tumor progression by carrying exosomes to neighboring cells. This study was assigned to expound the underlying mechanism of CAFs-derived exosomal miR-210 in non-small cell lung cancer (NSCLC) progression.MethodCAFs and normal fibroblasts (NFs) were isolated and identified. Exosomes secreted from CAFs and NFs were isolated to analyze their effects on tumor volume and epithelial-mesenchymal transition (EMT). Exosomal miR-210 expression level was measured. The effects of exosomal miR-210 and UPF1 on cell viability, EMT, PTEN/PI3K/AKT signal pathway were determined. Dual-luciferase reporter gene assay was utilized to validate the binding of UPF1 to miR-210.ResultsCAFs-derived exosomes (CAFs-exo) were successfully extracted and proven to be uptake by lung cancer cells. Up-regulated expression level of miR-210 was found in CAFs-exo, which was then proved to enhance cell migration, proliferation, invasion abilities and EMT in NSCLC cells. Overexpression of miR-210 can also inhibit UPF1 and PTEN, but activate the PTEN/PI3K/AKT pathway. UPF1 was a target gene of miR-210. MiR-210 can up-regulate UPF1 expression level to activate PTEN/PI3K/AKT pathway.ConclusionMiR-210 secreted by CAFs-exo could promote EMT by targeting UPF1 and activating PTEN/PI3K/AKT pathway, thereby promoting NSCLC migration and invasion.  相似文献   

14.
Interactions occurring between malignant cells and the stromal microenvironment heavily influence tumor progression. We investigated whether this cross-talk affects some molecular and functional aspects specifically correlated with the invasive phenotype of breast tumor cells (i.e. adhesion molecule expression, membrane fluidity, migration) by co-culturing mammary cancer cells exhibiting different degrees of metastatic potential (MDA-MB-231>MCF-7) with fibroblasts isolated from breast healthy skin (normal fibroblasts, NFs) or from breast tumor stroma (cancer-associated fibroblasts, CAFs) in 2D or 3D (nodules) cultures. Confocal immunofluorescence analysis of the epithelial adhesion molecule E-cadherin on frozen nodule sections demonstrated that NFs and CAFs, respectively, induced or inhibited its expression in MCF-7 cells. An increase in the mesenchymal adhesion protein N-cadherin was observed in CAFs, but not in NFs, as a result of the interaction with both kinds of cancer cells. CAFs, in turn, promoted N-cadherin up-regulation in MDA-MB-231 cells and its de novo expression in MCF-7 cells. Beyond promotion of “cadherin switching”, another sign of the CAF-triggered epithelial-mesenchymal transition (EMT) was the induction of vimentin expression in MCF-7 cells. Plasma membrane labeling of monolayer cultures with the fluorescent probe Laurdan showed an enhancement of the membrane fluidity in cancer cells co-cultured with NFs or CAFs. An increase in lipid packing density of fibroblast membranes was promoted by MCF-7 cells. Time-lapsed cell tracking analysis of mammary cancer cells co-cultured with NFs or CAFs revealed an enhancement of tumor cell migration velocity, even with a marked increase in the directness induced by CAFs.Our results demonstrate a reciprocal influence of mammary cancer and fibroblasts on various adhesiveness/invasiveness features. Notably, CAFs'' ability to promote EMT, reduction of cell adhesion, increase in membrane fluidity, and migration velocity and directness in mammary cancer cells can be viewed as an overall progression- and invasion-promoting effect.  相似文献   

15.
MiR-21 is known to play an important role in the development and progression, including migration and invasion, of many malignancies including breast cancer. Accumulating evidence suggest that the induction of epithelial-mesenchymal transition (EMT) phenotype and acquisition of cancer stem cell (CSC) characteristics are highly interrelated, and contribute to tumorigenesis, tumor progression, metastasis, and relapse. The molecular mechanisms underlying EMT and CSC characteristics during miR-21 contributes to cell migration and invasion of breast cancer are poorly understood. Therefore, we established miR-21 re-expressing breast cancer MCF-7 (MCF-7/miR-21) cells, which showed increasing cell growth, migration and invasion, self-renewal and clonogenicity. Our data showed that re-expression of miR-21 induced the acquisition of EMT phenotype by activation of mesenchymal cell markers (N-cadherin, Vimentin, α-SMA) and inhibition of epithelial cell marker (E-cadherin) in MCF-7/miR-21 cells, which consistent with increased cell subpopulation expressing CSC surface markers (ALDH1(+) and CD44(+)/CD24(-/low)) and the capacity of sphereforming (mammospheres). Our results demonstrated that re-expression of miR-21 is responsible for migration and invasion by activating the EMT process and enhancing the characteristics of CSCs in MCF-7 cells.  相似文献   

16.
17.
The involvement of the tumor stromal cells in acquired resistance of non-small cell lung cancers (NSCLCs) to tyrosine kinase inhibitors (TKIs) has previously been reported, but the precise mechanism remains unclear. In the present study, we investigated the role and mechanism underlying Cancer-associated fibroblasts (CAFs) in TKI resistance of NSCLCs. In vitro and in vivo experiments showed that HCC827 and PC9 cells, non-small cell lung cancer cells with EGFR-activating mutations, became resistant to the EGFR-TKI gefitinib when cultured with CAFs isolated from NSCLC tissues. Moreover, we showed that CAFs could induce epithelial-mesenchymal transition (EMT) phenotype of HCC827 and PC9 cells, with an associated change in the expression of epithelial to mesenchymal transition markers. Using proteomics-based method, we identified that CAFs significantly increased the expression of the Annexin A2 (ANXA2). More importantly, knockdown of ANXA2 completely reversed EMT phenotype and gefitinib resistance induced by CAFs. Furthermore, we found that CAFs increased the expression and phosphorylation of ANXA2 by secretion of growth factors HGF and IGF-1 and by activation of the corresponding receptors c-met and IGF-1R. Dual inhibition of HGF/c-met and IGF-1/IGF-1R pathways could significantly suppress ANXA2, and markedly reduced CAFs-induced EMT and gefitinib resistance. Taken together, these findings indicate that CAFs promote EGFR-TKIs resistance through HGF/IGF-1/ANXA2/EMT signaling and may be an ideal therapeutic target in NSCLCs with EGFR-activating mutations.  相似文献   

18.
Metastatic dissemination requires carcinoma cells to detach from the primary tumor and invade through the basement membrane. To acquire these characteristics, epithelial tumor cells undergo epithelial-to-mesenchymal transitions (EMT), whereby cells lose polarity and E-cadherin-mediated cell-cell adhesion. Post-EMT cells have also been shown, or assumed, to be more migratory; however, there have been contradictory reports on an immortalized human mammary epithelial cell line (HMLE) that underwent EMT. In the context of carcinoma-associated EMT, it is not yet clear whether the change in migration and invasion must be positively correlated during EMT or whether enhanced migration is a necessary consequence of having undergone EMT. Here, we report that pre-EMT rat prostate cancer (PC) and HMLE cells are more migratory than their post-EMT counterparts. To determine a mechanism for increased epithelial cell migration, gene expression analysis was performed and revealed an increase in epidermal growth factor receptor (EGFR) expression in pre-EMT cells. Indeed, inhibition of EGFR in PC epithelial cells slowed migration. Importantly, while post-EMT PC and HMLE cell lines are less migratory, both remain invasive in vitro and, for PC cells, in vivo. Our study demonstrates that enhanced migration is not a phenotypic requirement of EMT, and migration and invasion can be uncoupled during carcinoma-associated EMT.  相似文献   

19.
20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号