15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Perivascular tenascin C triggers sequential activation of macrophages and endothelial cells to generate a pro-metastatic vascular niche in the lungs

      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

          Disseminated cancer cells frequently lodge near vasculature in secondary organs. However, our understanding of the cellular crosstalk invoked at perivascular sites is still rudimentary. Here, we identify intercellular machinery governing formation of a pro-metastatic vascular niche during breast cancer colonization in the lung. We show that specific secreted factors, induced in metastasis-associated endothelial cells (ECs), promote metastasis in mice by enhancing stem cell properties and the viability of cancer cells. Perivascular macrophages, activated via tenascin C (TNC) stimulation of Toll-like receptor 4 (TLR4), were shown to be crucial in niche activation by secreting nitric oxide (NO) and tumor necrosis factor (TNF) to induce EC-mediated production of niche components. Notably, this mechanism was independent of vascular endothelial growth factor (VEGF), a key regulator of EC behavior and angiogenesis. However, targeting both macrophage-mediated vascular niche activation and VEGF-regulated angiogenesis resulted in added potency to curb lung metastasis in mice. Together, our findings provide mechanistic insights into the formation of vascular niches in metastasis.

          Abstract

          Hongu et al. find that perivascular macrophages stimulate activation of the pro-metastatic vascular niche via tenascin C stimulation of TLR4 and show that combined TLR4 and VEGF inhibition prevents TNC-mediated metastatic vascular activity.

          Related collections

          Most cited references66

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists

          Functional analysis of large gene lists, derived in most cases from emerging high-throughput genomic, proteomic and bioinformatics scanning approaches, is still a challenging and daunting task. The gene-annotation enrichment analysis is a promising high-throughput strategy that increases the likelihood for investigators to identify biological processes most pertinent to their study. Approximately 68 bioinformatics enrichment tools that are currently available in the community are collected in this survey. Tools are uniquely categorized into three major classes, according to their underlying enrichment algorithms. The comprehensive collections, unique tool classifications and associated questions/issues will provide a more comprehensive and up-to-date view regarding the advantages, pitfalls and recent trends in a simpler tool-class level rather than by a tool-by-tool approach. Thus, the survey will help tool designers/developers and experienced end users understand the underlying algorithms and pertinent details of particular tool categories/tools, enabling them to make the best choices for their particular research interests.
            Bookmark
            • 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: found
              • Article: found

              The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups

              The elucidation of breast cancer subgroups and their molecular drivers requires integrated views of the genome and transcriptome from representative numbers of patients. We present an integrated analysis of copy number and gene expression in a discovery and validation set of 997 and 995 primary breast tumours, respectively, with long-term clinical follow-up. Inherited variants (copy number variants and single nucleotide polymorphisms) and acquired somatic copy number aberrations (CNAs) were associated with expression in ~40% of genes, with the landscape dominated by cis- and trans-acting CNAs. By delineating expression outlier genes driven in cis by CNAs, we identified putative cancer genes, including deletions in PPP2R2A, MTAP and MAP2K4. Unsupervised analysis of paired DNA–RNA profiles revealed novel subgroups with distinct clinical outcomes, which reproduced in the validation cohort. These include a high-risk, oestrogen-receptor-positive 11q13/14 cis-acting subgroup and a favourable prognosis subgroup devoid of CNAs. Trans-acting aberration hotspots were found to modulate subgroup-specific gene networks, including a TCR deletion-mediated adaptive immune response in the ‘CNA-devoid’ subgroup and a basal-specific chromosome 5 deletion-associated mitotic network. Our results provide a novel molecular stratification of the breast cancer population, derived from the impact of somatic CNAs on the transcriptome.
                Bookmark

                Author and article information

                Contributors
                t.oskarsson@dkfz-heidelberg.de , Thordur.Oskarsson@moffitt.org
                Journal
                Nat Cancer
                Nat Cancer
                Nature Cancer
                Nature Publishing Group US (New York )
                2662-1347
                25 April 2022
                25 April 2022
                2022
                : 3
                : 4
                : 486-504
                Affiliations
                [1 ]GRID grid.482664.a, Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), ; Heidelberg, Germany
                [2 ]GRID grid.7497.d, ISNI 0000 0004 0492 0584, Division of Stem Cells and Cancer, , German Cancer Research Center, ; Heidelberg, Germany
                [3 ]GRID grid.7700.0, ISNI 0000 0001 2190 4373, Faculty of Biosciences, , University of Heidelberg, ; Heidelberg, Germany
                [4 ]GRID grid.7700.0, ISNI 0000 0001 2190 4373, Institute of Pathology, , University of Heidelberg, ; Heidelberg, Germany
                [5 ]GRID grid.411760.5, ISNI 0000 0001 1378 7891, Mildred Scheel Early Career Center, , University Hospital of Würzburg, ; Würzburg, Germany
                [6 ]GRID grid.7497.d, ISNI 0000 0004 0492 0584, DNA Vector Laboratory, , German Cancer Research Center, ; Heidelberg, Germany
                [7 ]GRID grid.509524.f, DKFZ-ZMBH Alliance, ; Heidelberg, Germany
                [8 ]GRID grid.7497.d, ISNI 0000 0004 0492 0584, German Cancer Consortium, ; Heidelberg, Germany
                [9 ]GRID grid.468198.a, ISNI 0000 0000 9891 5233, Present Address: Department of Molecular Oncology and Cancer Biology and Evolution Program, , H. Lee Moffitt Cancer Center and Research Institute, ; Tampa, FL USA
                Author information
                http://orcid.org/0000-0002-7906-5960
                http://orcid.org/0000-0002-6212-3466
                http://orcid.org/0000-0003-2836-6699
                http://orcid.org/0000-0002-0983-6969
                Article
                353
                10.1038/s43018-022-00353-6
                9046090
                35469015
                39dcc9fd-7b92-42d7-848e-d5578d0e3492
                © The Author(s) 2022

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as 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 images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 2 October 2020
                : 24 February 2022
                Funding
                Funded by: - International tenure track program of University of Tsukuba, Japan. - Overseas research fellowship from the Uehara memorial foundation, Japan.
                Funded by: Scholarship from the Helmholtz International Graduate School for Cancer Research
                Funded by: FundRef https://doi.org/10.13039/501100005941, Dietmar Hopp Stiftung;
                Award ID: N/A
                Award Recipient :
                Categories
                Article
                Custom metadata
                © The Author(s), under exclusive licence to Springer Nature America, Inc. 2022

                breast cancer,cancer microenvironment,metastasis,cancer
                breast cancer, cancer microenvironment, metastasis, cancer

                Comments

                Comment on this article