scholarly journals Overexpression of Extradomain-B Fibronectin is Associated with Invasion of Breast Cancer Cells

Cells ◽  
2020 ◽  
Vol 9 (8) ◽  
pp. 1826
Author(s):  
Amita Vaidya ◽  
Helen Wang ◽  
Victoria Qian ◽  
Hannah Gilmore ◽  
Zheng-Rong Lu

Breast tumor heterogeneity is a major impediment to oncotherapy. Cancer cells undergo rapid clonal evolution, thereby acquiring significant growth and invasive advantages. The absence of specific markers of these high-risk populations precludes efficient therapeutic and diagnostic management of the disease. Given the critical function of tumor microenvironment in the oncogenic circuitry, we sought to determine the expression profile of the extracellular matrix oncoprotein, extradomain-B fibronectin (EDB-FN) in invasive breast cancer. Analyses of TCGA/GTEx databases and immunostaining of clinical samples found a significant overexpression of EDB-FN in breast tumors, which correlated with poor overall survival. Significant upregulation of EDB-FN was observed in invasive cell populations generated from relatively less invasive MCF7 and MDA-MB-468 cells by long-term TGF-β treatment and acquired chemoresistance. Treatment of the invasive cell populations with an AKT inhibitor (MK2206-HCl) reduced their invasive potential, with a concomitant decrease in their EDB-FN expression, partly through the phosphoAKT-SRp55 pathway. EDB-FN downregulation, with direct RNAi of EDB-FN or indirectly through RNAi of SRp55, also resulted in reduced motility of the invasive cell populations, validating the correlation between EDB-FN expression and invasion of breast cancer cells. These data establish EDB-FN as a promising molecular marker for non-invasive therapeutic surveillance of aggressive breast cancer.

2019 ◽  
Author(s):  
Amita M. Vaidya ◽  
Helen Wang ◽  
Victoria Qian ◽  
Zheng-Rong Lu

AbstractBreast tumor heterogeneity is a major impediment to oncotherapy. Tumor cells undergo rapid clonal evolution, thereby acquiring significant growth and invasive advantages. The absence of specific markers of these high-risk tumors precludes efficient therapeutic and diagnostic management of breast cancer. Given the critical function of tumor microenvironment in the oncogenic circuitry, we sought to determine the role of the extracellular matrix oncoprotein, extradomain-B fibronectin (EDB-FN), as a molecular marker of aggressive cancers. High-risk invasive cell lines generated from relatively less invasive MCF7 and MDA-MB-468 breast cancer cells by long-term TGF-β treatment and chemoresistance demonstrated hybrid epithelial-mesenchymal phenotype, enhanced motility, and significantly elevated EDB-FN levels in 2D- and 3D-cultures. To determine if EDB-FN could serve as a therapy-predictive marker, the invasive cell lines were treated with MK2206-HCl, a pan-AKT inhibitor. Phospho-AKT depletion reduced EMT and invasion of the populations, with a concomitant decrease in EDB-FN expression, partly through the phosphoAKT-SRp55 pathway, demonstrating that EDB-FN expression is strongly associated with high-risk breast cancer. EDB-FN is a promising molecular marker for accurate detection, differential diagnosis, and non-invasive therapeutic surveillance of aggressive breast cancer.Summary StatementDynamic changes in invasive properties of breast cancer cells directly influence extradomain-B fibronectin levels, suggesting its potential role as a molecular marker for active surveillance and therapeutic monitoring of breast cancer.


BMC Cancer ◽  
2010 ◽  
Vol 10 (1) ◽  
Author(s):  
Masahiro Uchino ◽  
Hiroko Kojima ◽  
Kenta Wada ◽  
Mika Imada ◽  
Fumitoshi Onoda ◽  
...  

Cancers ◽  
2021 ◽  
Vol 13 (3) ◽  
pp. 480
Author(s):  
Rakshitha Pandulal Miskin ◽  
Janine S. A. Warren ◽  
Abibatou Ndoye ◽  
Lei Wu ◽  
John M. Lamar ◽  
...  

In the current study, we demonstrate that integrin α3β1 promotes invasive and metastatic traits of triple-negative breast cancer (TNBC) cells through induction of the transcription factor, Brain-2 (Brn-2). We show that RNAi-mediated suppression of α3β1 in MDA-MB-231 cells caused reduced expression of Brn-2 mRNA and protein and reduced activity of the BRN2 gene promoter. In addition, RNAi-targeting of Brn-2 in MDA-MB-231 cells decreased invasion in vitro and lung colonization in vivo, and exogenous Brn-2 expression partially restored invasion to cells in which α3β1 was suppressed. α3β1 promoted phosphorylation of Akt in MDA-MB-231 cells, and treatment of these cells with a pharmacological Akt inhibitor (MK-2206) reduced both Brn-2 expression and cell invasion, indicating that α3β1-Akt signaling contributes to Brn-2 induction. Analysis of RNAseq data from patients with invasive breast carcinoma revealed that high BRN2 expression correlates with poor survival. Moreover, high BRN2 expression positively correlates with high ITGA3 expression in basal-like breast cancer, which is consistent with our experimental findings that α3β1 induces Brn-2 in TNBC cells. Together, our study demonstrates a pro-invasive/pro-metastatic role for Brn-2 in breast cancer cells and identifies a role for integrin α3β1 in regulating Brn-2 expression, thereby revealing a novel mechanism of integrin-dependent breast cancer cell invasion.


2020 ◽  
Author(s):  
Ying Liu ◽  
Dandan Wang ◽  
Mengxia Lei ◽  
Jiayi Gao ◽  
Yuqing Cui ◽  
...  

Abstract Background γ-aminobutyric acid type A (GABAA) receptor-associated protein (GABARAP) is rarely studied in tumor progression. Here, the authors investigated the expression and significance of GABARAP in breast cancer. Method: A large group of clinical samples was assessed to detect GABARAP expression and its associations with clinicopathological features and prognosis. Gain- and loss-of-function experiments in cell lines and mouse xenograft models were performed to elucidate the function and underlying mechanisms of GABARAP-regulated tumor progression. Results We analyzed GABARAP levels in clinical breast cancer samples and cell lines and confirmed that GABARAP was negatively correlated with advanced clinicopathologic features, such as tumor size (P = 0.025) and TNM stage (P = 0.001). Importantly, patients with low GABARAP levels had a poor prognosis (p = 0.0047). Functionally, our data revealed that GABARAP can inhibit proliferation, migration and invasion in vitro and in vivo. Importantly, low levels of GABARAP induced epithelial-mesenchymal transition (EMT), one of the most important mechanisms for the promotion of tumor metastasis, in breast cancer cells. Mechanistically, low levels of GABARAP increased the levels of p-AKT (S473) and p-mTOR (S2448), and a specific AKT pathway inhibitor reversed the downregulation of GABARAP-induced tumor progression. In clinical breast cancer specimens, immunohistochemistry (IHC) revealed that the distribution and intensity of GABARAP expression were negatively correlated with those of matrix metalloproteinase (MMP) 2 (P = 0.0013) and MMP14 (P = 0.019). Conclusions Collectively, these data indicated that GABARAP suppressed the malignant behaviors of breast cancer cells, illuminating that the possible mechanism acts via the AkT/mTOR pathway. Targeting GABARAP may provide a potential diagnosis and treatment strategy for breast cancer.


Author(s):  
xiaohong wang ◽  
kai cheng ◽  
guoqiang zhang ◽  
yue yu ◽  
song liu ◽  
...  

Abstract Background: Exosomes have been shown to be associated with chemotherapy resistance transmission between cancer cells. However, the cargo and function of exosomes changed in response to doxorubicin remains unclear. Methods: We compared proteome profiles of exosomes extracted from the supernatant of MCF-7(S/Exo) and MCF-7/ADR(A/Exo) cells. We confirmed the differential expression of the candidate target-exosomic-CD44 by immune gold staining and western blot. We further studied the changes of chemosensitivity and CD44 expression in MCF-7 cells co-incubated with A/Exo. We analyzed the levels of exosomal CD44 from patient plasma, and compared the sensitivity and specificity of exosomic CD44 and plasma CD44 on diagnosis of chemoresistance. We modified the MCF-7-derived exosomes loaded with siRNA against CD44 to observe the effects of targeting reduced CD44 expression in lumimal A breast cancer cells. Results: DOX increased the exosomes release from MCF-7/ADR cells and the exosomes mediated proteins intercellular transfer in breast cancer chemoresistance regulation. The candidate target of CD44 in A/Exo was much higher than in S/Exo and the increase levels of exosomic CD44 (21.65-fold) was much higher than cellular CD44 (6.55-fold). The same results were obtained in clinical samples. Exosome-siRNA targeted CD44 (Exos-siCD44) could efficiently targeted to silence its expression. When co-cultured on Exos-siCD44, breast cancer cells exhibited reduced cell proliferation and enhanced susceptibility to DOX and the same phenomenon was observed in mice. Conclusion:Drug-resistant breast cancer cells spread resistance capacity to sensitive ones by releasing exosomes to transfer proteins in intercellular.


2020 ◽  
Author(s):  
Ying Liu ◽  
Dandan Wang ◽  
Mengxia Lei ◽  
Jiayi Gao ◽  
Yuqing Cui ◽  
...  

Abstract Background: Recent studies document that γ-aminobutyric acid type A (GABAA) receptor-associated protein (GABARAP) plays an important role in cancer autophagy. However, little is known about its role in tumor invasion, migration and metastasis. Here, the authors investigated the expression and significance of GABARAP in breast cancer. Method: A large group of clinical samples was assessed to detect GABARAP expression and its associations with clinicopathological features and prognosis. Gain- and loss-of-function experiments in cell lines and mouse xenograft models were performed to elucidate the function and underlying mechanisms of GABARAP-regulated tumor progression. Results: We analyzed GABARAP levels in clinical breast cancer samples and cell lines and confirmed that GABARAP was negatively correlated with advanced clinicopathologic features, such as tumor size (P=0.025) and TNM stage (P=0.001). Importantly, patients with low GABARAP levels had a poor prognosis (p = 0.0047). Functionally, our data revealed that GABARAP can inhibit proliferation, migration and invasion in vitro and in vivo. Importantly, low levels of GABARAP induced epithelial-mesenchymal transition (EMT), one of the most important mechanisms for the promotion of tumor metastasis, in breast cancer cells. Mechanistically, low levels of GABARAP increased the levels of p-AKT (S473) and p-mTOR (S2448), and a specific AKT pathway inhibitor reversed the downregulation of GABARAP-induced tumor progression. In clinical breast cancer specimens, immunohistochemistry (IHC) revealed that the distribution and intensity of GABARAP expression were negatively correlated with those of matrix metalloproteinase (MMP) 2 (P=0.0013) and MMP14 (P=0.019). Conclusions: Collectively, these data indicated that GABARAP suppressed the malignant behaviors of breast cancer cells, illuminating that the possible mechanism acts via the AkT/mTOR pathway. Targeting GABARAP may provide a potential diagnosis and treatment strategy for breast cancer.


2006 ◽  
Vol 78 ◽  
pp. S93
Author(s):  
S. Fu ◽  
Q.W. Zeng ◽  
Y. Sun ◽  
Q. Zhang ◽  
Y.H. Shao ◽  
...  

2013 ◽  
Vol 452 (3) ◽  
pp. 499-508 ◽  
Author(s):  
Eeva M. Sommer ◽  
Hannah Dry ◽  
Darren Cross ◽  
Sylvie Guichard ◽  
Barry R. Davies ◽  
...  

The majority of human cancers harbour mutations promoting activation of the Akt protein kinase, and Akt inhibitors are being evaluated in clinical trials. An important question concerns the understanding of the innate mechanisms that confer resistance of tumour cells to Akt inhibitors. SGK (serum- and glucocorticoid-regulated kinase) is closely related to Akt and controlled by identical upstream regulators {PI3K (phosphoinositide 3-kinase), PDK1 (phosphoinositide-dependent kinase 1) and mTORC2 [mTOR (mammalian target of rapamycin) complex 2]}. Mutations that trigger activation of Akt would also stimulate SGK. Moreover, Akt and SGK possess analogous substrate specificities and are likely to phosphorylate overlapping substrates to promote proliferation. To investigate whether cancers possessing high SGK activity could possess innate resistance to Akt-specific inhibitors (that do not target SGK), we analysed SGK levels and sensitivity of a panel of breast cancer cells towards two distinct Akt inhibitors currently in clinical trials (AZD5363 and MK-2206). This revealed a number of Akt-inhibitor-resistant lines displaying markedly elevated SGK1 that also exhibited significant phosphorylation of the SGK1 substrate NDRG1 [N-Myc (neuroblastoma-derived Myc) downstream-regulated gene 1]. In contrast, most Akt-inhibitor-sensitive cell lines displayed low/undetectable levels of SGK1. Intriguingly, despite low SGK1 levels, several Akt-inhibitor-sensitive cells showed marked NDRG1 phosphorylation that was, unlike in the resistant cells, suppressed by Akt inhibitors. SGK1 knockdown markedly reduced proliferation of Akt-inhibitor-resistant, but not -sensitive, cells. Furthermore, treatment of Akt-inhibitor-resistant cells with an mTOR inhibitor suppressed proliferation and led to inhibition of SGK1. The results of the present study suggest that monitoring SGK1 levels as well as responses of NDRG1 phosphorylation to Akt inhibitor administration could have a use in predicting the sensitivity of tumours to compounds that target Akt. Our findings highlight the therapeutic potential that SGK inhibitors or dual Akt/SGK inhibitors might have for treatment of cancers displaying elevated SGK activity.


2021 ◽  
Author(s):  
Catharina Melzer ◽  
Juliane von der Ohe ◽  
Tianjiao Luo ◽  
Ralf Hass

Abstract Background: A variety of different tumors including breast cancer cells can closely interact with mesenchymal stroma/stem-like cells (MSC) in the tumor microenvironment eventually resulting in cell fusion and formation of new hybrid cancer cell populations displaying altered properties. Methods: Lentiviral-transduced MDA-MB-231 cherry breast cancer cells and MSC GFP were co-cultured and a resulting hybrid cancer cell population (MDA-MSC-hyb5) was isolated. Characterization was performed for marker expression and short tandem repeat (STR) fragment analysis compared to the parental cells. Moreover, in vivo tumor development and metastatic capacity of MDA-MSC-hyb5 was studied and unique properties were analyzed by RNA microarray expression analyses compared to other breast cancer hybrid populations. Potential chemotherapeutic sensitivity was carried out in tumor explant cultures of MDA-MSC-hyb5 cells. Results: Direct cellular interactions of MDA-MB-231 cherry breast cancer cells with human MSC GFP in a co-culture model resulted in spontaneous cell fusion by generation of MDA-MSC-hyb5 cherry GFP breast cancer hybrid cells. Proliferative capacity of MDA-MSC-hyb5 cells was about 1.8-fold enhanced when compared to the parental MDA-MB-231 cherry breast cancer cells. In contrast to a spontaneous MDA-MB-231 cherry -induced tumor development in vivo within 18.8 days MDA-MSC-hyb5 cells initially remained quiescent in a dormancy-like state. At distinct time points up to about a half year later after injection NODscid mice started to develop MDA-MSC-hyb5 cell-induced tumors. Following tumor initiation, formation of metastases in various different organs occurred rapidly within about 10.5 days. Changes in gene expression levels were evaluated by RNA-microarray analysis and revealed certain increase in dormancy-associated transcripts in MDA-MSC-hyb5. Chemotherapeutic responsiveness of MDA-MSC-hyb5 cells was partially enhanced as compared to MDA-MB-231 cells, however, some resistance e.g. for taxol was detectable in cancer hybrid cells. Moreover, drug response partially changed during tumor development of MDA-MSC-hyb5 cells suggesting unstable in vivo phenotypes of MDA-hyb5 cells with increased tumor heterogeneity. Conclusions: The spontaneous formation of cancer hybrid cell populations like MDA-MSC-hyb5 by cell fusion contributes to tumorigenic diversification by acquisition of new properties such as altered chemotherapeutic responsiveness. The unique tumor dormancy of MDA-MSC-hyb5 cells not observed in other breast cancer hybrid cells so far markedly increases tumor heterogeneity.


Sign in / Sign up

Export Citation Format

Share Document