24
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Breast Cancer Cell Colonization of the Human Bone Marrow Adipose Tissue Niche 1

      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/OBJECTIVES: Bone is a preferred site of breast cancer metastasis, suggesting the presence of tissue-specific features that attract and promote the outgrowth of breast cancer cells. We sought to identify parameters of human bone tissue associated with breast cancer cell osteotropism and colonization in the metastatic niche. METHODS: Migration and colonization patterns of MDA-MB-231-fLuc-EGFP (luciferase-enhanced green fluorescence protein) and MCF-7-fLuc-EGFP breast cancer cells were studied in co-culture with cancellous bone tissue fragments isolated from 14 hip arthroplasties. Breast cancer cell migration into tissues and toward tissue-conditioned medium was measured in Transwell migration chambers using bioluminescence imaging and analyzed as a function of secreted factors measured by multiplex immunoassay. Patterns of breast cancer cell colonization were evaluated with fluorescence microscopy and immunohistochemistry. RESULTS: Enhanced MDA-MB-231-fLuc-EGFP breast cancer cell migration to bone-conditioned versus control medium was observed in 12/14 specimens ( P = .0014) and correlated significantly with increasing levels of the adipokines/cytokines leptin ( P = .006) and IL-1β ( P = .001) in univariate and multivariate regression analyses. Fluorescence microscopy and immunohistochemistry of fragments underscored the extreme adiposity of adult human bone tissues and revealed extensive breast cancer cell colonization within the marrow adipose tissue compartment. CONCLUSIONS: Our results show that breast cancer cells migrate to human bone tissue-conditioned medium in association with increasing levels of leptin and IL-1β, and colonize the bone marrow adipose tissue compartment of cultured fragments. Bone marrow adipose tissue and its molecular signals may be important but understudied components of the breast cancer metastatic niche.

          Related collections

          Most cited references67

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

          Microenvironmental regulation of metastasis.

          Metastasis is a multistage process that requires cancer cells to escape from the primary tumour, survive in the circulation, seed at distant sites and grow. Each of these processes involves rate-limiting steps that are influenced by non-malignant cells of the tumour microenvironment. Many of these cells are derived from the bone marrow, particularly the myeloid lineage, and are recruited by cancer cells to enhance their survival, growth, invasion and dissemination. This Review describes experimental data demonstrating the role of the microenvironment in metastasis, identifies areas for future research and suggests possible new therapeutic avenues.
            Bookmark
            • Record: found
            • Abstract: not found
            • Article: not found

            The distribution of secondary growths in cancer of the breast. 1889.

            S. PAGET (1989)
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Adipose tissue and adipocytes support tumorigenesis and metastasis.

              Adipose tissue influences tumor development in two major ways. First, obese individuals have a higher risk of developing certain cancers (endometrial, esophageal, and renal cell cancer). However, the risk of developing other cancers (melanoma, rectal, and ovarian) is not altered by body mass. In obesity, hypertrophied adipose tissue depots are characterized by a state of low grade inflammation. In this activated state, adipocytes and inflammatory cells secrete adipokines and cytokines which are known to promote tumor development. In addition, the adipocyte mediated conversion of androgens to estrogen specifically contributes to the development of endometrial cancer, which shows the greatest relative risk (6.3-fold) increase between lean and obese individuals. Second, many tumor types (gastric, breast, colon, renal, and ovarian) grow in the anatomical vicinity of adipose tissue. During their interaction with cancer cells, adipocytes dedifferentiate into pre-adipocytes or are reprogrammed into cancer-associated adipocytes (CAA). CAA secrete adipokines which stimulate the adhesion, migration, and invasion of tumor cells. Cancer cells and CAA also engage in a dynamic exchange of metabolites. Specifically, CAA release fatty acids through lipolysis which are then transferred to cancer cells and used for energy production through β-oxidation. The abundant availability of lipids from adipocytes in the tumor microenvironment, supports tumor progression and uncontrolled growth. Given that adipocytes are a major source of adipokines and energy for the cancer cell, understanding the mechanisms of metabolic symbiosis between cancer cells and adipocytes, should reveal new therapeutic possibilities. This article is part of a Special Issue entitled Lipid Metabolism in Cancer. Copyright © 2013 Elsevier B.V. All rights reserved.
                Bookmark

                Author and article information

                Contributors
                Journal
                Neoplasia
                Neoplasia
                Neoplasia (New York, N.Y.)
                Neoplasia Press
                1522-8002
                1476-5586
                13 December 2015
                December 2015
                13 December 2015
                : 17
                : 12
                : 849-861
                Affiliations
                [* ]Department of Pediatrics, 150E Clark Center, 318 Campus Drive, Stanford University School of Medicine, Stanford, CA 94305-5427
                []EMD Millipore Corporation, 14 Research Park Drive, St Charles, MO 63304-5618
                []Department of Immunology, Fairchild Science Building, D033, 299 Campus Drive, Stanford University School of Medicine, Stanford, CA 94305-5124
                [§ ]Department of Biomedical Data Science, Room T101F, Redwood Building, 150 Governor's Lane, Stanford University School of Medicine, Stanford, CA 94305-5405
                []Cell Sciences Imaging Facility, Beckman Center (B050B), Stanford University School of Medicine, Stanford, CA 94305-5301
                [# ]Department of Orthopaedic Surgery, 450 Broadway Street, Pavillion C, 4th Floor, Stanford University School of Medicine, Redwood City, CA 94063-6342
                Author notes
                [* ]Address all correspondence to: Bonnie L. King, Ph.D., Department of Pediatrics, 150E Clark Center, 318 Campus Drive, Stanford University School of Medicine, Stanford, CA 94305-5427.Department of PediatricsStanford University School of Medicine150E Clark Center, 318 Campus DriveStanfordCA94305-5427 bonnie.king@ 123456stanford.edu
                Article
                S1476-5586(15)00142-6
                10.1016/j.neo.2015.11.005
                4688564
                26696367
                91bd5460-d942-435f-9c4f-2b9610c0a6e5
                © 2015 The Authors

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 27 September 2015
                : 5 November 2015
                : 10 November 2015
                Categories
                Article

                Comments

                Comment on this article