11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      MicroRNA-124 enhances response to radiotherapy in human epidermal growth factor receptor 2-positive breast cancer cells by targeting signal transducer and activator of transcription 3

      research-article
      1 , 2
      Croatian Medical Journal
      Croatian Medical Schools

      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

          Aim

          To determine whether microRNA (miR)-124 enhances the response to radiotherapy in human epidermal growth factor receptor 2 (HER2)-positive breast cancer cells by targeting signal transducer and activator of transcription 3 (Stat3).

          Methods

          miR-29b expression was measured in 80 pairs of breast tumor samples and adjacent normal tissues collected between January 2013 and July 2014. Activity changes of 50 canonical signaling pathways upon miR-124 overexpression were determined using Cignal Signal Transduction Reporter Array. Target gene of miR-124 was determined using Targetscan and validated by Western blotting and dual-luciferase assay. Cell death rate was assessed by propidium iodide (PI)/ Annexin V staining followed by flow cytometry analysis. Stat3 and miR-124 expression was further measured in 10 relapsed (non-responder) and 10 recurrence-free HER2-positive breast cancer patients.

          Results

          MiR-124 expression was down-regulated in HER2 positive breast cancers compared with normal tissues, and was negatively associated with tumor size. MiR-124 overexpression in HER2 positive breast cancer cell line SKBR3 significantly reduced the activity of Stat3 signaling pathway compared with control transfection ( P < 0.001). Bioinformatic prediction and function assay suggested that miR-124 directly targeted Stat3, which is a key regulator of HER2 expression. MiR-124 overexpression down-regulated Stat3 and potently enhanced cell death upon irradiation. Consistently, chemical inhibitor of Stat3 also sensitized HER2-positive breast cancer cells to irradiation. Moreover, increased Stat3 expression and reduced miR-124 expression were associated with a poor response to radiotherapy in HER2-positive breast cancers.

          Conclusions

          Weak miR-124 expression might enhance Stat3 expression and radiotherapy resistance in HER2-positive breast cancer cells.

          Related collections

          Most cited references16

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

          Breast cancer bone metastasis mediated by the Smad tumor suppressor pathway.

          TGF-beta can signal by means of Smad transcription factors, which are quintessential tumor suppressors that inhibit cell proliferation, and by means of Smad-independent mechanisms, which have been implicated in tumor progression. Although Smad mutations disable this tumor-suppressive pathway in certain cancers, breast cancer cells frequently evade the cytostatic action of TGF-beta while retaining Smad function. Through immunohistochemical analysis of human breast cancer bone metastases and functional imaging of the Smad pathway in a mouse xenograft model, we provide evidence for active Smad signaling in human and mouse bone-metastatic lesions. Genetic depletion experiments further demonstrate that Smad4 contributes to the formation of osteolytic bone metastases and is essential for the induction of IL-11, a gene implicated in bone metastasis in this mouse model system. Activator protein-1 is a key participant in Smad-dependent transcriptional activation of IL-11 and its overexpression in bone-metastatic cells. Our findings provide functional evidence for a switch of the Smad pathway, from tumor-suppressor to prometastatic, in the development of breast cancer bone metastasis.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            A pathway-based classification of human breast cancer.

            The hallmark of human cancer is heterogeneity, reflecting the complexity and variability of the vast array of somatic mutations acquired during oncogenesis. An ability to dissect this heterogeneity, to identify subgroups that represent common mechanisms of disease, will be critical to understanding the complexities of genetic alterations and to provide a framework to develop rational therapeutic strategies. Here, we describe a classification scheme for human breast cancer making use of patterns of pathway activity to build on previous subtype characterizations using intrinsic gene expression signatures, to provide a functional interpretation of the gene expression data that can be linked to therapeutic options. We show that the identified subgroups provide a robust mechanism for classifying independent samples, identifying tumors that share patterns of pathway activity and exhibit similar clinical and biological properties, including distinct patterns of chromosomal alterations that were not evident in the heterogeneous total population of tumors. We propose that this classification scheme provides a basis for understanding the complex mechanisms of oncogenesis that give rise to these tumors and to identify rational opportunities for combination therapies.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              miR-124 inhibits STAT3 signaling to enhance T cell-mediated immune clearance of glioma.

              miRNAs (miR) have been shown to modulate critical gene transcripts involved in tumorigenesis, but their role in tumor-mediated immunosuppression is largely unknown. On the basis of miRNA gene expression in gliomas using tissue microarrays, in situ hybridization, and molecular modeling, miR-124 was identified as a lead candidate for modulating STAT3 signaling, a key pathway mediating immunosuppression in the tumor microenvironment. miR-124 is absent in all grades and pathologic types of gliomas. Upon upregulating miR-124 in glioma cancer stem cells (gCSC), the STAT3 pathway was inhibited, and miR-124 reversed gCSC-mediated immunosuppression of T-cell proliferation and induction of forkhead box P3 (Foxp3)(+) regulatory T cells (Treg). Treatment of T cells from immunosuppressed glioblastoma patients with miR-124 induced marked effector response including upregulation of interleukin (IL)-2, IFN-γ, and TNF-α. Both systemic administration of miR-124 or adoptive miR-124-transfected T-cell transfers exerted potent anti-glioma therapeutic effects in clonotypic and genetically engineered murine models of glioblastoma and enhanced effector responses in the local tumor microenvironment. These therapeutic effects were ablated in both CD4(+)- and CD8(+)-depleted mice and nude mouse systems, indicating that the therapeutic effect of miR-124 depends on the presence of a T-cell-mediated antitumor immune response. Our findings highlight the potential application of miR-124 as a novel immunotherapeutic agent for neoplasms and serve as a model for identifying miRNAs that can be exploited as immunotherapeutics. ©2013 AACR.
                Bookmark

                Author and article information

                Journal
                Croat Med J
                Croat. Med. J
                CMJ
                Croatian Medical Journal
                Croatian Medical Schools
                0353-9504
                1332-8166
                October 2016
                : 57
                : 5
                : 457-464
                Affiliations
                [1 ]Department of General Surgery, the First Affiliated Hospital of Nanchang University, Nanchang, China
                [2 ]Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, China
                Author notes
                Correspondence to:
Jianping Xiong
17 Yongwaizheng rd
Nanchang
Jiangxi, China 330006
 jianpingxiong_26@ 123456126.com
                Article
                CroatMedJ_57_0457
                10.3325/cmj.2016.57.457
                5141458
                27815936
                81197e18-4ad1-482a-9c4f-e53234ca2f93
                Copyright © 2016 by the Croatian Medical Journal. All rights reserved.

                This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                : 14 June 2016
                : 10 October 2016
                Categories
                Basic Science

                Medicine
                Medicine

                Comments

                Comment on this article