Blocking integrin β1 decreases adhesion in chemoresistant urothelial cancer cell lines

  • Authors:
    • Stefan Vallo
    • Jochen Rutz
    • Miriam Kautsch
    • Ria Winkelmann
    • Martin Michaelis
    • Felix Wezel
    • Georg Bartsch
    • Axel Haferkamp
    • Florian Rothweiler
    • Roman A. Blaheta
    • Jindrich Cinatl Jr
  • View Affiliations

  • Published online on: September 4, 2017     https://doi.org/10.3892/ol.2017.6883
  • Pages: 5513-5518
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Treatment failure in metastatic bladder cancer is commonly caused by acquisition of resistance to chemotherapy in association with tumor progression. Since alterations of integrins can influence the adhesive and invasive behaviors of urothelial bladder cancer cell lines, the present study aimed to evaluate the role of integrins in bladder cancer cells with acquired resistance to standard first‑line chemotherapy with gemcitabine, and cisplatin. Therefore, four gemcitabine‑ and four cisplatin‑resistant sublines out of a panel of four parental urothelial bladder cancer cell lines (TCC‑SUP, HT1376, T24, and 5637) were used. Expression of integrin subunits α3, α5, α6, β1, β3, and β4 was detected using flow cytometry. Adhesion and chemotaxis were analyzed. For functional assays, integrin β1 was attenuated with a blocking antibody. In untreated cells, chemotaxis was upregulated in 3/4 gemcitabine‑resistant sublines. In cisplatin‑resistant cells, chemotaxis was enhanced in 2/4 cell lines. Acquired chemoresistance induced the upregulation of integrin β1 in all four tested gemcitabine‑resistant sublines, as well as an upregulation in 3/4 cisplatin‑resistant sublines compared with parental cell lines. Following the inhibition of integrin β1, adhesion to extracellular matrix components was downregulated in 3/4 gemcitabine‑resistant sublines and in all four tested cisplatin‑resistant sublines. Since integrin β1 is frequently upregulated in chemoresistant urothelial cancer cell lines and inhibition of integrin β1 may influence adhesion, further studies are warranted to evaluate integrin β1 as a potential therapeutic target for bladder cancer in vivo.

Introduction

Urothelial cancer of the bladder is the 4th most commonly diagnosed cancer in men worldwide (1). Patients with metastatic disease are often treated with a combination chemotherapy containing gemcitabine and cisplatin as a standard of care (2,3). However, the treatment success is limited, resulting in a median survival of 12–15 months. Treatment failure is commonly caused by acquired resistance after primary response (2,3). Therefore, efficient second line chemotherapies are urgently needed.

Integrins have been identified to play an important role in the development of resistance to chemotherapy in bladder cancer (4). These molecules are transmembrane receptors with two different chains, an α (alpha) and a β (beta) subunit. Integrins are bridges for cell-cell and cell-extracellular matrix (ECM) interactions. Cell-matrix contact plays a fundamental role in the metastatic potential of tumors (5). Alterations of integrin expression may result in an enhanced adhesive behavior in bladder cancer (6). Moreover, the expression patterns of integrin subtypes are known to be important mediators of tumor cell de-differentiation and tumor proliferation (7). Furthermore, it was shown that integrins were involved in the development of metastasis and recurrence of urothelial cancer (6,8,9).

Drug-adapted cancer cell lines have been successfully used to study cancer cell resistance mechanisms (10,11). To reflect the heterogeneity of individual bladder cancer patients and to enable a systematic evaluation of the role of integrins concerning resistance acquisition, we used a panel of 12 urothelial cancer cell lines consisting of 4 parental chemosensitive cell lines and their sublines with acquired resistance to gemcitabine or cisplatin (12,13).

Materials and methods

Drugs

Cisplatin was purchased from Gry-Pharma (Kirchzarten, Germany), gemcitabine from Lilly (Bad Homburg, Germany).

Cell lines

The cell lines 5637, T24, HT1376, and TCC-SUP were obtained from the American Type Culture Collection (Manassas, VA, USA). The following drug-resistant sublines were established by continuous exposure to increasing drug concentrations as described previously (12,14) and are part of the Resistant Cancer Cell Line (RCCL) collection (http://www.kent.ac.uk/stms/cmp/RCCL/RCCLabout.html): 5637rCDDP1000 (cisplatin-resistant, 1,000 ng cisplatin/ml), 5637rGEMCI20 (gemcitabine-resistant, 20 ng gemcitabine/ml), T24rCDDP1000, T24rGEMCI20, HT1376rCDDP1000, HT1376rGEMCI20, TCC-SUPrCDDP1000, and TCC-SUPrGEMCI20. All cell lines were grown in Iscove's modified Dulbecco's medium supplemented with 10% fetal calf serum (FCS; Gibco, Karlsruhe, Germany), 100 IU/ml penicillin, and 100 µg/ml streptomycin at 37°C. Cell line authentication was performed by short tandem repeat profiling.

Cell adhesion to extracellular matrix components

24-well plates were coated with extracellular matrix components (Matrigel; Corning, Amsterdam, The Netherlands) overnight. Plates were washed with 1% bovine serum albumin (BSA) in phosphate buffered saline (PBS) to block nonspecific cell adhesion. Thereafter, 0.5×106 tumor cells were added to each well for 60 min. Subsequently, non-adherent tumor cells were washed off. The adherent cells were fixed with 1% glutaraldehyde and counted in five different fields using a microscope (20× objective) to calculate the mean cellular adhesion rate.

Chemotaxis

Serum induced cell migration was examined using 6-well transwell chambers (Greiner, Frickenhausen, Germany) with 8 µm pores. To evaluate cell migration, cells were placed in the upper chamber for 20 h in serum-free medium. The lower chamber contained 10% serum. After incubation, the upper surface of the transwell membrane was wiped gently with a cotton swab to remove non-migrating cells. Cells migrating to the lower surface of the membrane were stained using hematoxylin and counted. Cells migrating into the lower chamber were counted separately under the microscope.

Blocking study

Cells were preincubated for 60 min with a function-blocking anti-integrin β1 monoclonal antibody (20 mg/l) (MAB 2253Z; MerckMillipore, Darmstadt, Germany). Controls remained untreated. Adhesion and chemotaxis was tested as indicated above.

Flow cytometry

Cells were washed in blocking solution (PBS, 0.5% BSA) and then incubated for 60 min at 4°C with phycoerythrin (PE)-conjugated monoclonal antibodies directed against the following integrin subtypes: Anti-α3 (IgG1; clone C3II.1), anti-α5 (IgG1; clone IIA1), anti-α6 (IgG2b, clone MP 4F10), anti-β1 (IgG1; clone MAR4), anti-β3 (IgG1; clone VI-PL2) or anti-β4 (IgG2a; clone 439-9B; all: BD Biosciences, Heidelberg, Germany). Integrin expression was measured by flow cytometry (FACSCalibur; BD Biosciences, Heidelberg, Germany). Mouse IgG1-PE (MOPC-21) or mouse IgG2a-PE (G155-178; all: BD Biosciences) antibodies were used as isotype control.

Immunohistochemistry

33 cases of invasive and non-invasive bladder cancers as well es corresponding normal urothelium were taken from the archive of the Dr. Senckenberg Institute of Pathology in Frankfurt. Tissue sections were stained for Integrin β1, (D2E5) Rabbit mAb, Cell Signaling Technology (Waltham, MA, USA), dilution 1:100. In brief, 4 µm sections were cut and pretreated with Trilogy™, Cell Marcque (Rocklin, CA, USA), incubated with the antibody, antigen retrieval was performed at pH 6 in a microwave oven using the Peroxidase-FLEX EnVision kit (Dako, Jena, Germany).

A pathologist, who was blinded to clinical history and therapeutic response, scored the immunohistochemical staining using a five-stage staining score: 0=negative; 1=weak; 2=moderate; 3=strong; 4=very strong.

Images were acquired using a digital slide scanner (ScanScope XT; Aperio, Vista, CA, USA).

Statistical analysis

Results are expressed as mean ± SD of at least three independent experiments. For statistical analysis student's t-test, analysis of variance (ANOVA), and Student-Newman-Keuls-Test were performed whenever applicable. P<0.05 was considered to indicate a statistically significant difference.

Results

Influence of acquired resistance on adhesion to extracellular matrix components

In untreated cells, adhesion to extracellular matrix components was decreased in 2 of 4 gemcitabine-resistant sublines (HT1376rGEMCI20 and TCC-SUPrGEMCI20) and upregulated in 2 of 4 cell lines (T24rGEMCI20 and 5637rGEMCI20) compared to parental cells. In cisplatin-resistant sublines, adhesion was decreased in 1 of 4 cisplatin-resistant sublines (TCC-SUPrCDDP1000) and enhanced in 2 cell lines (HT1376rCDDP1000 and 5637rCDDP1000) (Fig. 1).

Influence of acquired chemoresistance on chemotaxis

Chemotaxis was enhanced in 3 of 4 gemcitabine-resistant urothelial cancer cell lines (gemcitabine-resistant sublines of TCC-SUP, HT1376 and T24). In cisplatin-resistant sublines, chemotaxis was enhanced in HT1376rCDDP1000 and T24rCDDP1000 compared to parental cell lines (Fig. 2).

Differential expression of cell surface integrins

Expression of integrins on the cell surface was analyzed by flow cytometry (Fig. 3). In gemcitabine-resistant sublines, the expression of integrin α3 was enhanced in 3 sublines (gemcitabine-resistant sublines of T24, 5637, and TCC-SUP) and diminished in HT1376rGEMCI20 compared to parental cell lines. Integrin β1 expression was upregulated in all gemcitabine-resistant sublines compared to parental cells. Integrin β4 expression was enhanced in TCC-SUPrGEMCI20 and diminished in T24rGEMCI20 and in 5637rGEMCI20.

Comparing cisplatin-resistant cell lines, Integrin α3 expression was upregulated in 3 of 4 sublines (cisplatin-resistant sublines of T24, 5637, and TCC-SUP) and downregulated in HT1376rCDDP1000. Integrin α5 was upregulated in 3 of 4 sublines (cisplatin-resistant sublines of 5637, TCC-SUP, and HT1376). Integrin β1 expression was upregulated in 3 of 4 sublines (cisplatin-resistant sublines of HT1376, T24, and 5637) and downregulated in TCC-SUPrCDDP1000. Integrin β4 was upregulated in HT1376rCDDP1000 and downregulated in all other tested sublines (cisplatin-resistant sublines of T24, 5637, and TCC-SUP).

Immunohistochemical staining of integrin β1

In non-malignant tissue samples, integrin β1 was only visible in the basal layer of the urothelium. The medium staining score for normal urothelium was between ‘negative’ to ‘weak’ (0.88±0.33). In bladder cancer samples, the mean integrin β1 staining score was between ‘moderate’ and ‘strong’ (2.48±1.42). There was no significant difference of integrin β1 staining between low grade tumors or high grade tumors and no significant difference between non-muscle invasive tumors and muscle invasive tumors (Fig. 4).

Influence of blocking integrin β1 on adhesion and chemotaxis

Functional blocking of integrin β1 resulted in a reduced adhesion in 2 of 4 parental urothelial cancer cell lines (HT1376 and T24). In gemcitabine-resistant cells, adhesion was downregulated in 3 of 4 cell lines (gemcitabine-resistant sublines of HT1376, T24 and TCC-SUP). In cisplatin-resistant cells, adhesion was downregulated in all 4 tested cell lines (Fig. 5). We could not detect an influence on chemotaxis after blocking integrin β1 (Fig. 6).

Discussion

In the present study, we used a well-established panel of urothelial cancer cell lines with acquired resistance to gemcitabine or cisplatin, the standard therapeutics for metastatic urothelial cancer of the bladder (2,3). Cell line panels seem to be necessary to reflect the heterogeneity of different patient-derived cancer cell lines. Although the complex scenario of metastatic colonization is not fully understood, there is strong evidence that alterations of tumor-matrix contact are necessary to allow motile crawling into the surrounding tissue (15).

In several cancer cells, resistance to gemcitabine seems to be connected with integrins and associated proteins (1618). In addition, resistance to cytotoxic drugs and proliferation regulation was shown to be dependent on extracellular matrix proteins (19). In this study, acquisition of resistance to gemcitabine or cisplatin showed a changed adhesive behavior with some resistant sublines showing an enhanced adhesive behavior and other resistant sublines being less adhesive (Fig. 1). In contrast, the influence on chemotaxis was more uniform, with 5 of 8 sublines showing an enhanced chemotaxis and no subline with a significantly diminished chemotaxis after acquisition of resistance (Fig. 2). This is in line with Ploenes et al who reported about an enhanced chemotaxis in lung cancer cell lines with an increased chemoresistance (20).

Since integrins seem to be involved in the development of resistance to chemotherapy in bladder cancer (4) and alterations of integrin expression change adhesive and invasive behavior of bladder cancer cells (6), we aimed to elucidate the role of integrins in this context.

Integrin α3 might be involved in resistance acquisition, since it was upregulated in 3 of 4 gemcitabine-resistant and also in 3 of 4 cisplatin-resistant sublines in this study (Fig. 3). Litynska et al (21) tried to analyze the role of integrin α3 in bladder cancer by blocking its function. They described that adhesion was up- or downregulated after blocking integrin α3 depending on the tested cell line. The cell line specific effects that can be triggered after acquisition of resistance show the heterogeneity between independent cell lines and underline the importance of using a panel of cell lines for a better interpretation.

It was reported that integrin α5 contributes to a more malignant phenotype in urothelial bladder cancer (22). In our study, this integrin subunit was overexpressed in most of the tested chemoresistant sublines what might underline the more malignant phenotype of the chemoresistant sublines (Fig. 3).

We observed that most chemoresistant sublines showed a diminished expression of integrin β4 compared to their parental counterparts (Fig. 3). Therefore, a downregulation of integrin β4 could be connected with a more malignant behavior. This is in line with reports that an overexpression of integrin β4 inhibits growth and migration in bladder cancer cell lines and plays an anti-tumoral role (23,24).

In all gemcitabine-resistant and in 3 of 4 cisplatin-resistant sublines, surface expressed integrin β1 was upregulated compared to parental cell lines (Fig. 3). Since chemotaxis was frequently enhanced after acquisition of resistance, these results might support the conclusions of Chakraborty et al (25) who postulated that blockade of β1-integrin with a specific antibody could result in alteration of multiple signaling pathways related to adhesion and migration. Interestingly, Zhang and coworkers showed that they could reverse chemoresistance to mitomycin c by blocking integrin β1 (4). Integrin β1 was overexpressed in most of the tested sublines and it was reported to contribute to a more malignant phenotype in urothelial bladder cancer (4,25). We could confirm in this study that overexpression of integrin β1 is associated with a malignant phenotype since we detected a stronger expression in malignant tissue samples compared to normal urothelium. Nevertheless, there was no different expression comparing low grade with high grade tumors or between non-muscle invasive bladder cancer and muscle invasive bladder cancer (Fig. 4).

To further analyze the role of integrin β1, we suppressed the function of integrin β1 and meassured adhesion and chemotaxis afterwards. There was an influence on adhesion after blocking integrin β1 with a reduced adhesion in 2 of 4 parental and 3 of 4 gemcitabine-resistant sublines. In cisplatin-resistant cells, adhesion was even downregulated in all 4 tested cell lines (Fig. 5).

We could not show an influence on chemotaxis after blocking integrin β1 (Fig. 6). If there is no influence on chemotaxis or if the used transwell migration assay is not able to reflect the impact on chemotaxis is not clear. An explanation for the latter might be that effects that influence invasion after blocking integrin β1 are delayed and therefore not detectable with the used transwell migration assay.

Discussing the role of different integrins and the influence on adhesive and invasive behavior, we should be aware that differentially guided adhesive behavior of different tumor cell lines has been previously observed (21). Blocking integrin β1 inhibited cell-matrix interactions in HCV29 and BC3726 cell lines, whereas binding of the bladder cancer cell lines T24 and Hu456 was enhanced (21). Each cell line therefore may possess a characteristic receptor set and long-term treatment with chemotherapy may influence integrin subfamilies differently. Therefore systematic analysis of cell line panels is fundamental (6).

We did not investigate the relevance of each integrin member in detail that was used here. To provide a complete picture of the role of integrin subtypes in gemcitabine- and cisplatin-resistant bladder cancer ongoing studies are necessary. Particularly, blocking experiments using integrins α3, α5, and β4 would be of interest. In addition, these findings are limited to bladder cancer cell lines and bladder tissue. The role of integrins in chemoresistant bladder cancer should be further evaluated in vivo in an animal model.

Overall, evidence is presented here that acquired resistance to gemcitabine or cisplatin frequently enhances chemotaxis, what might be a surrogate for an increased invasive behavior in chemoresistant bladder cancer cell lines. Since overexpression of integrin β1 seems to be frequently upregulated in chemoresistant urothelial bladder cancer cell lines, further in vivo studies should evaluate downregulation of integrin β1 as a potential therapeutic target especially in chemotherapy refractory cases.

Acknowledgements

The work was supported by the charity Hilfe für krebskranke Kinder Frankfurt e.V., its trust Frankfurter Stiftung für krebskranke Kinder, the Patenschaftsmodell program of the University Hospital Frankfurt and the Kent Cancer Trust.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Pectasides D, Pectasides M and Economopoulos T: Systemic chemotherapy in locally advanced and/or metastatic bladder cancer. Cancer Treat Rev. 32:456–470. 2006. View Article : Google Scholar : PubMed/NCBI

3 

von der Maase H, Sengelov L, Roberts JT, Ricci S, Dogliotti L, Oliver T, Moore MJ, Zimmermann A and Arning M: Long-term survival results of a randomized trial comparing gemcitabine plus cisplatin, with methotrexate, vinblastine, doxorubicin, plus cisplatin in patients with bladder cancer. J Clin Oncol. 23:4602–4608. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Zhang CJ, Shen ZJ, Pan CW, Zhong S, Li T and Zhang MG: Engagement of integrinβ1 induces resistance of bladder cancer cells to mitomycin-C. Urology. 79:638–643. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Canel M, Serrels A, Frame MC and Brunton VG: E-cadherin-integrin crosstalk in cancer invasion and metastasis. J Cell Sci. 126:393–401. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Juengel E, dos Santos Meyer S, Schneider T, Makarevic J, Hudak L, Bartsch G, Haferkamp A, Wiesner C and Blaheta RA: HDAC inhibition suppresses bladder cancer cell adhesion to collagen under flow conditions. Exp Biol Med (Maywood). 238:1297–1304. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Danen EH: Integrins: Regulators of tissue function and cancer progression. Curr Pharm Des. 11:881–891. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Yamasaki M, Soda S, Sakakibara Y, Suiko M and Nishiyama K: The importance of 1,2-dithiolane structure in α-lipoic acid for the downregulation of cell surface β1-integrin expression of human bladder cancer cells. Biosci Biotechnol Biochem. 78:1939–1942. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Behnsawy HM, Miyake H, Abdalla MA, Sayed MA, Ahmed Ael-F and Fujisawa M: Expression of integrin proteins in non-muscle-invasive bladder cancer: Significance of intravesical recurrence after transurethral resection. BJU Int. 107:240–246. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Sharma SV, Haber DA and Settleman J: Cell line-based platforms to evaluate the therapeutic efficacy of candidate anticancer agents. Nat Rev Cancer. 10:241–253. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Domingo-Domenech J, Vidal SJ, Rodriguez-Bravo V, Castillo-Martin M, Quinn SA, Rodriguez-Barrueco R, Bonal DM, Charytonowicz E, Gladoun N, de la Iglesia-Vicente J, et al: Suppression of acquired docetaxel resistance in prostate cancer through depletion of notch- and hedgehog-dependent tumor-initiating cells. Cancer Cell. 22:373–388. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Vallo S, Michaelis M, Rothweiler F, Bartsch G, Gust KM, Limbart DM, Rödel F, Wezel F, Haferkamp A and Cinatl J Jr: Drug-resistant urothelial cancer cell lines display diverse sensitivity profiles to potential second-line therapeutics. Transl Oncol. 8:210–216. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Mani J, Vallo S, Rakel S, Antonietti P, Gessler F, Blaheta R, Bartsch G, Michaelis M, Cinatl J, Haferkamp A and Kögel D: Chemoresistance is associated with increased cytoprotective autophagy and diminished apoptosis in bladder cancer cells treated with the BH3 mimetic (−)-Gossypol (AT-101). BMC Cancer. 15:2242015. View Article : Google Scholar : PubMed/NCBI

14 

Michaelis M, Rothweiler F, Barth S, Cinatl J, van Rikxoort M, Löschmann N, Voges Y, Breitling R, von Deimling A, Rödel F, et al: Adaptation of cancer cells from different entities to the MDM2 inhibitor nutlin-3 results in the emergence of p53-mutated multi-drug resistant cancer cells. Cell Death Dis. 2:e2432011. View Article : Google Scholar : PubMed/NCBI

15 

Yilmaz M and Christofori G: Mechanisms of motility in metastasizing cells. Mol Cancer Res. 8:629–642. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Huanwen W, Zhiyong L, Xiaohua S, Xinyu R, Kai W and Tonghua L: Intrinsic chemoresistance to gemcitabine is associated with constitutive and laminin-induced phosphorylation of FAK in pancreatic cancer cell lines. Mol Cancer. 8:1252009. View Article : Google Scholar : PubMed/NCBI

17 

Jia Z: Role of integrin-linked kinase in drug resistance of lung cancer. Onco Targets Ther. 8:1561–1565. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Duxbury MS, Ito H, Benoit E, Waseem T, Ashley SW and Whang EE: RNA interference demonstrates a novel role for integrin-linked kinase as a determinant of pancreatic adenocarcinoma cell gemcitabine chemoresistance. Clin Cancer Res. 11:3433–3438. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Miyamoto H, Murakami T, Tsuchida K, Sugino H, Miyake H and Tashiro S: Tumor-stroma interaction of human pancreatic cancer: Acquired resistance to anticancer drugs and proliferation regulation is dependent on extracellular matrix proteins. Pancreas. 28:38–44. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Ploenes T, Scholtes B, Krohn A, Burger M, Passlick B, Müller-Quernheim J and Zissel G: CC-chemokine ligand 18 induces epithelial to mesenchymal transition in lung cancer A549 cells and elevates the invasive potential. PLoS One. 8:e530682013. View Article : Google Scholar : PubMed/NCBI

21 

Lityńska A, Przybyło M, Pocheć E and Laidler P: Adhesion properties of human bladder cell lines with extracellular matrix components: The role of integrins and glycosylation. Acta Biochim Pol. 49:643–650. 2002.PubMed/NCBI

22 

Saito T, Kimura M, Kawasaki T, Sato S and Tomita Y: Correlation between integrin alpha 5 expression and the malignant phenotype of transitional cell carcinoma. Br J Cancer. 73:327–331. 1996. View Article : Google Scholar : PubMed/NCBI

23 

Kim SY, Bachman NJ, Nair TS, Goldsmith S, Liebert M, Grossman HB, Lomax MI and Carey TE: Beta 4 integrin transfection of UM-UC-2 (human bladder carcinoma) cells: Stable expression of a spontaneous cytoplasmic truncation mutant with rapid loss of clones expressing intact beta 4. Cancer Res. 57:38–42. 1997.PubMed/NCBI

24 

Harabayashi T, Kanai Y, Yamada T, Sakamoto M, Ochiai A, Kakizoe T, Koyanagi T and Hirohashi S: Reduction of integrin beta4 and enhanced migration on laminin in association with intraepithelial spreading of urinary bladder carcinomas. J Urol. 161:1364–1371. 1999. View Article : Google Scholar : PubMed/NCBI

25 

Chakraborty A, White SM and Guha S: Granulocyte colony-stimulating receptor promotes beta1-integrin-mediated adhesion and invasion of bladder cancer cells. Urology. 68:208–213. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 14 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Vallo S, Rutz J, Kautsch M, Winkelmann R, Michaelis M, Wezel F, Bartsch G, Haferkamp A, Rothweiler F, Blaheta RA, Blaheta RA, et al: Blocking integrin β1 decreases adhesion in chemoresistant urothelial cancer cell lines. Oncol Lett 14: 5513-5518, 2017
APA
Vallo, S., Rutz, J., Kautsch, M., Winkelmann, R., Michaelis, M., Wezel, F. ... Cinatl Jr, J. (2017). Blocking integrin β1 decreases adhesion in chemoresistant urothelial cancer cell lines. Oncology Letters, 14, 5513-5518. https://doi.org/10.3892/ol.2017.6883
MLA
Vallo, S., Rutz, J., Kautsch, M., Winkelmann, R., Michaelis, M., Wezel, F., Bartsch, G., Haferkamp, A., Rothweiler, F., Blaheta, R. A., Cinatl Jr, J."Blocking integrin β1 decreases adhesion in chemoresistant urothelial cancer cell lines". Oncology Letters 14.5 (2017): 5513-5518.
Chicago
Vallo, S., Rutz, J., Kautsch, M., Winkelmann, R., Michaelis, M., Wezel, F., Bartsch, G., Haferkamp, A., Rothweiler, F., Blaheta, R. A., Cinatl Jr, J."Blocking integrin β1 decreases adhesion in chemoresistant urothelial cancer cell lines". Oncology Letters 14, no. 5 (2017): 5513-5518. https://doi.org/10.3892/ol.2017.6883