96
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The voltage gated Ca 2+-channel Ca v3.2 and therapeutic responses in breast cancer

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          Understanding the cause of therapeutic resistance and identifying new biomarkers in breast cancer to predict therapeutic responses will help optimise patient care. Calcium (Ca 2+)-signalling is important in a variety of processes associated with tumour progression, including breast cancer cell migration and proliferation. Ca 2+-signalling is also linked to the acquisition of multidrug resistance. This study aimed to assess the expression level of proteins involved in Ca 2+-signalling in an in vitro model of trastuzumab-resistance and to assess the ability of identified targets to reverse resistance and/or act as potential biomarkers for prognosis or therapy outcome.

          Methods

          Expression levels of a panel of Ca 2+-pumps, channels and channel regulators were assessed using RT-qPCR in resistant and sensitive age-matched SKBR3 breast cancer cells, established through continuous culture in the absence or presence of trastuzumab. The role of Ca v3.2 in the acquisition of trastuzumab-resistance was assessed through pharmacological inhibition and induced overexpression. Levels of Ca v3.2 were assessed in a panel of non-malignant and malignant breast cell lines using RT-qPCR and in patient samples representing different molecular subtypes (PAM50 cohort). Patient survival was also assessed in samples stratified by Ca v3.2 expression (METABRIC and KM-Plotter cohort).

          Results

          Increased mRNA of Ca v3.2 was a feature of both acquired and intrinsic trastuzumab-resistant SKBR3 cells. However, pharmacological inhibition of Ca v3.2 did not restore trastuzumab-sensitivity nor did Ca v3.2 overexpression induce the expression of markers associated with resistance, suggesting that Ca v3.2 is not a driver of trastuzumab-resistance. Ca v3.2 levels were significantly higher in luminal A, luminal B and HER2-enriched subtypes compared to the basal subtype. High levels of Ca v3.2 were associated with poor outcome in patients with oestrogen receptor positive (ER+) breast cancers, whereas Ca v3.2 levels were correlated positively with patient survival after chemotherapy in patients with HER2-positive breast cancers.

          Conclusion

          Our study identified elevated levels of Ca v3.2 in trastuzumab-resistant SKBR3 cell lines. Although not a regulator of trastuzumab-resistance in HER2-positive breast cancer cells, Ca v3.2 may be a potential differential biomarker for survival and treatment response in specific breast cancer subtypes. These studies add to the complex and diverse role of Ca 2+-signalling in breast cancer progression and treatment.

          Electronic supplementary material

          The online version of this article (doi:10.1186/s12935-016-0299-0) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references46

          • Record: found
          • Abstract: found
          • Article: not found

          The HER-2 receptor and breast cancer: ten years of targeted anti-HER-2 therapy and personalized medicine.

          The human epidermal growth factor receptor (HER-2) oncogene encodes a transmembrane tyrosine kinase receptor that has evolved as a major classifier of invasive breast cancer and target of therapy for the disease. The validation of the general prognostic significance of HER-2 gene amplification and protein overexpression in the absence of anti-HER-2 targeted therapy is discussed in a study of 107 published studies involving 39,730 patients, which produced an overall HER-2-positive rate of 22.2% and a mean relative risk for overall survival (OS) of 2.74. The issue of HER-2 status in primary versus metastatic breast cancer is considered along with a section on the features of metastatic HER-2-positive disease. The major marketed slide-based HER-2 testing approaches, immunohistochemistry, fluorescence in situ hybridization, and chromogenic in situ hybridization, are presented and contrasted in detail against the background of the published American Society of Clinical Oncology-College of American Pathologists guidelines for HER-2 testing. Testing issues, such as the impact of chromosome 17 polysomy and local versus central HER-2 testing, are also discussed. Emerging novel HER-2 testing techniques, including mRNA-based testing by real-time polymerase chain reaction and DNA microarray methods, HER-2 receptor dimerization, phosphorylated HER-2 receptors, and HER-2 status in circulating tumor cells, are also considered. A series of biomarkers potentially associated with resistance to trastuzumab is discussed with emphasis on the phosphatase and tensin homologue deleted on chromosome ten/Akt and insulin-like growth factor receptor pathways. The efficacy results for the more recently approved small molecule HER-1/HER-2 kinase inhibitor lapatinib are also presented along with a more limited review of markers of resistance for this agent. Additional topics in this section include combinations of both anti-HER-2 targeted therapies together as well as with novel agents including bevacizumab, everolimus, and tenespimycin. A series of novel HER-2-targeting agents is also presented, including pertuzumab, ertumaxomab, HER-2 vaccines, and recently discovered tyrosine kinase inhibitors. Biomarkers predictive of HER-2 targeted therapy toxicity are included, and the review concludes with a consideration of HER-2 status in the prediction of response to non-HER-2 targeted treatments including hormonal therapy, anthracyclines, and taxanes.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The Caco-2 cell line as a model of the intestinal barrier: influence of cell and culture-related factors on Caco-2 cell functional characteristics.

            The human intestinal Caco-2 cell line has been extensively used over the last twenty years as a model of the intestinal barrier. The parental cell line, originally obtained from a human colon adenocarcinoma, undergoes in culture a process of spontaneous differentiation that leads to the formation of a monolayer of cells, expressing several morphological and functional characteristics of the mature enterocyte. Culture-related conditions were shown to influence the expression of these characteristics, in part due to the intrinsic heterogeneity of the parental cell line, leading to selection of sub-populations of cells becoming prominent in the culture. In addition, several clonal cell lines have been isolated from the parental line, exhibiting in general a more homogeneous expression of differentiation traits, while not always expressing all characteristics of the parental line. Culture-related conditions, as well as the different Caco-2 cell lines utilized in different laboratories, often make it extremely difficult to compare results in the literature. This review is aimed at summarizing recent, or previously unreviewed, data from the literature on the effects of culture-related factors and the influence of line sub-types (parental vs. different clonal lines) on the expression of differentiation traits important for the use of Caco-2 cells as a model of the absorptive and defensive properties of the intestinal mucosa. Since the use of Caco-2 cells has grown exponentially in recent years, it is particularly important to highlight these methodological aspects in order to promote the standardization and optimisation of this intestinal model.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Orai1 and STIM1 are critical for breast tumor cell migration and metastasis.

              Tumor metastasis is the primary cause of death of cancer patients. Understanding the molecular mechanisms underlying tumor metastasis will provide potential drug targets. We report here that Orai1 and STIM1, both of which are involved in store-operated calcium entry, are essential for breast tumor cell migration in vitro and tumor metastasis in mice. Reduction of Orai1 or STIM1 by RNA interference in highly metastatic human breast cancer cells or treatment with a pharmacological inhibitor of store-operated calcium channels decreased tumor metastasis in animal models. Our data demonstrate a role for Orai1 and STIM1 in tumor metastasis and suggest store-operated calcium entry channels as potential cancer therapeutic targets.
                Bookmark

                Author and article information

                Contributors
                epera@me.com
                e.kaemmerer@uq.edu.au
                michael.milevskiy@uqconnect.edu.au
                k.yapa@uq.edu.au
                jake.odonnell@uq.edu.au
                melissa.brown@uq.edu.au
                f.simpson@uq.edu.au
                a.peters1@uq.edu.au
                sarahrt@uq.edu.au
                gregm@uq.edu.au
                Journal
                Cancer Cell Int
                Cancer Cell Int
                Cancer Cell International
                BioMed Central (London )
                1475-2867
                31 March 2016
                31 March 2016
                2016
                : 16
                : 24
                Affiliations
                [ ]The School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, 20 Cornwall St, Woolloongabba, Brisbane, QLD Australia
                [ ]Mater Research Institute, The University of Queensland, Brisbane, QLD Australia
                [ ]Translational Research Institute, Brisbane, QLD Australia
                [ ]The School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD Australia
                [ ]Diamantina Institute, The University of Queensland, Brisbane, QLD Australia
                Article
                299
                10.1186/s12935-016-0299-0
                4815142
                27034617
                21b2fc73-3b2d-4929-ad3b-87add3ba41f8
                © Pera et al. 2016

                Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

                History
                : 4 February 2016
                : 22 March 2016
                Funding
                Funded by: NHMRC
                Award ID: 1079672
                Award Recipient :
                Categories
                Primary Research
                Custom metadata
                © The Author(s) 2016

                Oncology & Radiotherapy
                breast cancer,trastuzumab-resistance,calcium-signalling,cav3.2 (cacna1h),therapeutic response,biomarker

                Comments

                Comment on this article