46
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Cross-Species Array Comparative Genomic Hybridization Identifies Novel Oncogenic Events in Zebrafish and Human Embryonal Rhabdomyosarcoma

      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

          Human cancer genomes are highly complex, making it challenging to identify specific drivers of cancer growth, progression, and tumor maintenance. To bypass this obstacle, we have applied array comparative genomic hybridization (array CGH) to zebrafish embryonal rhabdomyosaroma (ERMS) and utilized cross-species comparison to rapidly identify genomic copy number aberrations and novel candidate oncogenes in human disease. Zebrafish ERMS contain small, focal regions of low-copy amplification. These same regions were commonly amplified in human disease. For example, 16 of 19 chromosomal gains identified in zebrafish ERMS also exhibited focal, low-copy gains in human disease. Genes found in amplified genomic regions were assessed for functional roles in promoting continued tumor growth in human and zebrafish ERMS – identifying critical genes associated with tumor maintenance. Knockdown studies identified important roles for Cyclin D2 (CCND2), Homeobox Protein C6 (HOXC6) and PlexinA1 (PLXNA1) in human ERMS cell proliferation. PLXNA1 knockdown also enhanced differentiation, reduced migration, and altered anchorage-independent growth. By contrast, chemical inhibition of vascular endothelial growth factor (VEGF) signaling reduced angiogenesis and tumor size in ERMS-bearing zebrafish. Importantly, VEGFA expression correlated with poor clinical outcome in patients with ERMS, implicating inhibitors of the VEGF pathway as a promising therapy for improving patient survival. Our results demonstrate the utility of array CGH and cross-species comparisons to identify candidate oncogenes essential for the pathogenesis of human cancer.

          Author Summary

          Cancer is a complex genetic disease that is often associated with regional gains and losses of genomic DNA segments. These changes result in aberrant gene expression and drive continued tumor growth. Because amplified and deleted DNA segments tend to span large regions of chromosomes, it has been challenging to identify the genes that are required for continued tumor growth and progression. Array comparative genomic hybridization (array CGH) is an effective technology in identifying abnormal copy number variations in cancer genomes. In this study, array CGH was used in a zebrafish model of embryonal rhabdomyosarcoma - a pediatric muscle tumor. Our work shows that the zebrafish cancer genome contains a small number of recurrent DNA copy number changes, which are also commonly amplified in the human disease. Moreover, these chromosomal regions are small, facilitating rapid identification of candidate oncogenes. A subset of genes identified in zebrafish array CGH was prioritized for functional characterization in human ERMS, identifying evolutionarily conserved pathways that regulate proliferation, migration, differentiation, and neovascularization. Our results demonstrate the broad utility of cross-species array CGH comparisons of human and zebrafish cancer and provide a much needed discovery platform for identifying critical cancer-causing genes in a wide range of malignancies.

          Related collections

          Most cited references39

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

          Genetic interaction of PGE2 and Wnt signaling regulates developmental specification of stem cells and regeneration.

          Interactions between developmental signaling pathways govern the formation and function of stem cells. Prostaglandin (PG) E2 regulates vertebrate hematopoietic stem cells (HSC). Similarly, the Wnt signaling pathway controls HSC self-renewal and bone marrow repopulation. Here, we show that wnt reporter activity in zebrafish HSCs is responsive to PGE2 modulation, demonstrating a direct interaction in vivo. Inhibition of PGE2 synthesis blocked wnt-induced alterations in HSC formation. PGE2 modified the wnt signaling cascade at the level of beta-catenin degradation through cAMP/PKA-mediated stabilizing phosphorylation events. The PGE2/Wnt interaction regulated murine stem and progenitor populations in vitro in hematopoietic ES cell assays and in vivo following transplantation. The relationship between PGE2 and Wnt was also conserved during regeneration of other organ systems. Our work provides in vivo evidence that Wnt activation in stem cells requires PGE2, and suggests the PGE2/Wnt interaction is a master regulator of vertebrate regeneration and recovery.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Semaphorins command cells to move.

            Semaphorins are secreted or transmembrane proteins that regulate cell motility and attachment in axon guidance, vascular growth, immune cell regulation and tumour progression. The main receptors for semaphorins are plexins, which have established roles in regulating Rho-family GTPases. Recent work shows that plexins can also influence R-Ras, which, in turn, can regulate integrins. Such regulation is probably a common feature of semaphorin signalling and contributes substantially to our understanding of semaphorin biology.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Effects of RAS on the genesis of embryonal rhabdomyosarcoma.

              Embryonal rhabdomyosarcoma (ERMS) is a devastating cancer with specific features of muscle differentiation that can result from mutational activation of RAS family members. However, to date, RAS pathway activation has not been reported in a majority of ERMS patients. Here, we have created a zebrafish model of RAS-induced ERMS, in which animals develop externally visible tumors by 10 d of life. Microarray analysis and cross-species comparisons identified two conserved gene signatures found in both zebrafish and human ERMS, one associated with tumor-specific and tissue-restricted gene expression in rhabdomyosarcoma and a second comprising a novel RAS-induced gene signature. Remarkably, our analysis uncovered that RAS pathway activation is exceedingly common in human RMS. We also created a new transgenic coinjection methodology to fluorescently label distinct subpopulations of tumor cells based on muscle differentiation status. In conjunction with fluorescent activated cell sorting, cell transplantation, and limiting dilution analysis, we were able to identify the cancer stem cell in zebrafish ERMS. When coupled with gene expression studies of this cell population, we propose that the zebrafish RMS cancer stem cell shares similar self-renewal programs as those found in activated satellite cells.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS Genet
                PLoS Genet
                plos
                plosgen
                PLoS Genetics
                Public Library of Science (San Francisco, USA )
                1553-7390
                1553-7404
                August 2013
                August 2013
                29 August 2013
                : 9
                : 8
                : e1003727
                Affiliations
                [1 ]Division of Molecular Pathology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
                [2 ]Harvard Stem Cell Institute, Boston, Massachusetts, United States of America
                [3 ]Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
                [4 ]Harvard Medical School, Boston, Massachusetts, United States of America
                [5 ]Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
                [6 ]Department of Pathology and Cell Biology, College of Medicine, University of Southern Florida, Tampa, Florida, United States of America
                [7 ]Department of Biology, Portland State University, Portland, Oregon, United States of America
                [8 ]Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
                [9 ]Pediatric Cancer Biology Program, Department of Pediatrics, Oregon Health and Science University, Portland, Oregon, United States of America
                Stanford University School of Medicine, United States of America
                Author notes

                The authors have declared that no competing interests exist.

                Conceived and designed the experiments: EYC KPD KHB CL DML SR. Performed the experiments: EYC KPD KHB RC EE JYHC MSI JB. Analyzed the data: EYC KPD KHB SR DML CL CK MSI GPN. Contributed reagents/materials/analysis tools: EYC KPD KHB EE SR. Wrote the paper: EYC KPD KHB DML.

                Article
                PGENETICS-D-13-00917
                10.1371/journal.pgen.1003727
                3757044
                24009521
                66768dee-fcec-4b62-94e9-6e7affb412c0
                Copyright @ 2013

                This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

                History
                : 5 April 2013
                : 1 July 2013
                Page count
                Pages: 15
                Funding
                DML was supported by NIH grants K01 AR055619, RO1 CA154923, and R21 CA156056, the Alex's Lemonade Stand Foundation (ALSF), the American Cancer Society, and the Harvard Stem Cell Institute. EYC was supported by NIH K08 AR063165-01 and St. Baldrick's Foundation Scholar Award. KHB was supported by Grant K99/R00 ES018892 (NIH–National Institute of Environmental Health Sciences). MSI was supported by an “A” ALSF ward. CL is supported by Grants 5R01CA111560 (NIH–National Cancer Institute) and 5P41HG004421 (NIH–National Human Genome Research Institute). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript
                Categories
                Research Article

                Genetics
                Genetics

                Comments

                Comment on this article