Abstract

BackgroundDefects in tight junctions, gate-keepers of the integrity of the epidermal barrier function, are known to contribute to cancer development. As such, enhancing our understanding of how the expression of proteins involved in these junctions is regulated in cancer, remains a priority. Although the expression of one of these proteins, claudin 1, is down regulated in most invasive human breast cancers (HBC), we have recently shown that high levels of claudin 1, characterized tumors belonging to the very aggressive basal-like breast cancer (BLBC) subtype. In these tumors, the claudin 1 protein, usually localized in the cell membrane, is often mislocalized to the cytoplasm.MethodsTo examine the clinical relevance of this observation, we have generated and analyzed an invasive HBC tissue microarray consisting of 151 breast tumor samples; 79 of which presented a basal-like phenotype (i.e. ER-ve, PR-ve HER2-ve, CK5/6 or EGFR+ve). We also interrogated the outcome of claudin 1 knockdown in a human BLBC cell line, BT-20.ResultsImmunohistochemical analysis of this patient cohort revealed a significant association between high claudin 1 expression and BLBCs in women 55 years of age and older. Interestingly, no significant association was found between claudin 1 and nodal involvement, tumor grade or tumor size. Regression analysis however, showed a significant positive association between claudin 1 and claudin 4, even though claudin 4 did not significantly correlate with patient age. Claudin 1 knockdown in BT-20 cells resulted in decreased cell migration. It also significantly altered the expression of several genes involved in epithelial-mesenchymal-transition (EMT); in particular, SERPINE 1 (PAI1) and SSP1 (osteopontin), known to inhibit EMT and cancer cell migration. Conversely, genes known to maintain EMT through their interaction, SNAIL2, TCF4 and FOXC2 were significantly down regulated.ConclusionsThe association of high claudin 1 protein levels observed in tumors derived from older women with BLBC, suggests that claudin 1 has the potential to serve as a marker which can identify a specific subgroup of patients within the BLBC subtype and thus, further contribute to the characterization of these ill-defined breast cancers. More importantly, our studies strongly suggest that claudin 1 directly participates in promoting breast cancer progression, possibly through the alteration of expression of EMT genes.

Highlights

  • Defects in tight junctions, gate-keepers of the integrity of the epidermal barrier function, are known to contribute to cancer development

  • Whereas the luminal A and B subtypes are characterized by their epithelial phenotypes, hormone sensitivity, mildly invasive capacity and relatively good clinical outcome, the Human epidermal growth factor receptor 2 (HER2)+ and basallike breast cancer (BLBC) subtypes are characterized by their mesenchymal phenotype, insensitivity to hormonal therapy (ER-ve; Progesterone receptor (PR)-ve), enhanced invasiveness and metastatic capacity [2] and poor clinical outcome [3,4,5,6,7]

  • High level of claudin 1 protein is associated with basal-like breast cancer (BLBC) derived from older women Claudin 1 expression was higher in the basal-like tumors compared to the non-basal tumors, confirming the observations made in our previous study [19]

Read more

Summary

Introduction

Gate-keepers of the integrity of the epidermal barrier function, are known to contribute to cancer development. The expression of one of these proteins, claudin 1, is down regulated in most invasive human breast cancers (HBC), we have recently shown that high levels of claudin 1, characterized tumors belonging to the very aggressive basal-like breast cancer (BLBC) subtype. In these tumors, the claudin 1 protein, usually localized in the cell membrane, is often mislocalized to the cytoplasm. The claudins belong to a family of tight junction (TJ) proteins (24 identified to date), that are crucial for the organization of epithelial cell polarity [8] They contribute to the trans-epithelial barrier that controls the transport of ions and small molecules. The claudins are capable of recruiting signaling proteins, thereby regulating various cellular processes including cell growth, differentiation and tumorigenesis [13,14]

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call