5
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Protein kinase Ds promote tumor angiogenesis through mast cell recruitment and expression of angiogenic factors in prostate cancer microenvironment

      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

          Mast cells are being increasingly recognized as critical components in the tumor microenvironment. Protein Kinase D (PKD) is essential for the progression of prostate cancer, but its role in prostate cancer microenvironment remains poorly understood.

          Methods

          The expression of PKD, mast cells and microvessel density were examined by IHC. The clinical significance was determined by statistical analyses. The biological function of PKD and the underlying mechanisms were investigated using in vitro and in vivo models.

          Results

          PKD2/3 contributed to MCs recruitment and tumor angiogenesis in the prostate cancer microenvironment. Clinical data showed that increased activation of PKD at Ser744/748 in prostate cancer was correlated with mast cell infiltration and microvascular density. PKD2/3 silencing of prostate cancer cells markedly decreased MCs migration and tube formation of HUVEC cells. Moreover, PKD2/3 depletion not only reduced SCF, CCL5 and CCL11 expression in prostate cancer cells but also inhibited angiogenic factors in MCs. Conversely, exogenous SCF, CCL5 and CCL11 reversed the effect on MCs migration inhibited by PKD2/3 silencing. Mechanistically, PKD2/3 interacted with Erk1/2 and activated Erk1/2 or NF-κB signaling pathway, leading to AP-1 or NF-κB binding to the promoter of scf, ccl5 and ccl11. Finally, PKD-specific inhibitor significantly reduced tumor volume and tumor growth in mice bearing RM-1 prostate cancer cells, which was attributed to attenuation of mast cell recruitment and tumor angiogenesis.

          Conclusions

          These results demonstrate a novel PKDs function that contributes to tumor angiogenesis and progression through mast cells recruitment in prostate cancer microenvironment.

          Electronic supplementary material

          The online version of this article (10.1186/s13046-019-1118-y) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references45

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

          The Diagnosis and Treatment of Prostate Cancer: A Review.

          Prostate cancer is the most common cancer diagnosis made in men with more than 160 000 new cases each year in the United States. Although it often has an indolent course, prostate cancer remains the third-leading cause of cancer death in men.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Mast cells as "tunable" effector and immunoregulatory cells: recent advances.

            This review focuses on recent progress in our understanding of how mast cells can contribute to the initiation, development, expression, and regulation of acquired immune responses, both those associated with IgE and those that are apparently expressed independently of this class of Ig. We emphasize findings derived from in vivo studies in mice, particularly those employing genetic approaches to influence mast cell numbers and/or to alter or delete components of pathways that can regulate mast cell development, signaling, or function. We advance the hypothesis that mast cells not only can function as proinflammatory effector cells and drivers of tissue remodeling in established acquired immune responses, but also may contribute to the initiation and regulation of such responses. That is, we propose that mast cells can also function as immunoregulatory cells. Finally, we show that the notion that mast cells have primarily two functional configurations, off (or resting) or on (or activated for extensive mediator release), markedly oversimplifies reality. Instead, we propose that mast cells are "tunable," by both genetic and environmental factors, such that, depending on the circumstances, the cell can be positioned phenotypically to express a wide spectrum of variation in the types, kinetics, and/or magnitude of its secretory functions.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Inflammatory mast cells up-regulate angiogenesis during squamous epithelial carcinogenesis.

              Expression of HPV16 early region genes in basal keratinocytes of transgenic mice elicits a multistage pathway to squamous carcinoma. We report that infiltration by mast cells and activation of the matrix metalloproteinase MMP-9/gelatinase B coincides with the angiogenic switch in premalignant lesions. Mast cells infiltrate hyperplasias, dysplasias, and invasive fronts of carcinomas, but not the core of solid tumors, where they degranulate in close apposition to capillaries and epithelial basement membranes, releasing mast-cell-specific serine proteases MCP-4 (chymase) and MCP-6 (tryptase). MCP-6 is shown to be a mitogen for dermal fibroblasts that proliferate in the reactive stroma, whereas MCP-4 can activate progelatinase B and induce hyperplastic skin to become angiogenic in an in vitro bioassay. Notably, premalignant angiogenesis is abated in a mast-cell-deficient (KITW/KITWWv) HPV16 transgenic mouse. The data indicate that neoplastic progression in this model involves exploitation of an inflammatory response to tissue abnormality. Thus, regulation of angiogenesis during squamous carcinogenesis is biphasic: In hyperplasias, dysplasias, and invading cancer fronts, inflammatory mast cells are conscripted to reorganize stromal architecture and hyperactivate angiogenesis; within the cancer core, upregulation of angiogenesis factors in tumor cells apparently renders them self-sufficient at sustaining neovascularization.
                Bookmark

                Author and article information

                Contributors
                xushi123@gmail.com
                sabrina.kb.ryusuke@hotmail.com
                zengfy@126.com
                xiaoyukaoyan0103@163.com
                gwjabout@163.com
                chenlipingchan@163.com
                yunturn@126.com
                15818124098@163.com
                qjw1@pitt.edu
                fandeng@smu.edu.cn
                Journal
                J Exp Clin Cancer Res
                J. Exp. Clin. Cancer Res
                Journal of Experimental & Clinical Cancer Research : CR
                BioMed Central (London )
                0392-9078
                1756-9966
                6 March 2019
                6 March 2019
                2019
                : 38
                : 114
                Affiliations
                [1 ]ISNI 0000 0000 8877 7471, GRID grid.284723.8, Department of Cell Biology, School of Basic Medical Sciences, , Southern Medical University, ; Guangzhou, 510515 China
                [2 ]ISNI 0000 0000 8877 7471, GRID grid.284723.8, Department of Clinical Laboratory, The Fifth Affiliated Hospital, , Southern Medical University, ; Guangzhou, 510900 China
                [3 ]ISNI 0000 0004 1936 9000, GRID grid.21925.3d, Department of Pharmacology and Chemical Biology, , University of Pittsburgh School of Medicine, ; Pittsburgh, PA 15261 USA
                [4 ]ISNI 0000 0000 8653 1072, GRID grid.410737.6, Present address: Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, , Guangzhou Medical University, ; Guangzhou, 510623 China
                Article
                1118
                10.1186/s13046-019-1118-y
                6404326
                30841931
                8066550b-d3eb-46a2-a44e-958e29f4f636
                © The Author(s). 2019

                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
                : 29 October 2018
                : 22 February 2019
                Funding
                Funded by: National Natural Science Foundation of China
                Award ID: 81272852
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100001809, National Natural Science Foundation of China;
                Award ID: 81472407
                Award ID: 81772761
                Award ID: 81672540
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100010902, Joint Research Fund for Overseas Chinese Scholars and Scholars in Hong Kong and Macao;
                Award ID: 81328020
                Award Recipient :
                Funded by: Science and Technology Project of Guangzhou
                Award ID: 210707010303
                Award ID: 201607010351
                Award Recipient :
                Categories
                Research
                Custom metadata
                © The Author(s) 2019

                Oncology & Radiotherapy
                protein kinase d(pkd),mast cells(mcs),angiogenesis,prostate cancer
                Oncology & Radiotherapy
                protein kinase d(pkd), mast cells(mcs), angiogenesis, prostate cancer

                Comments

                Comment on this article