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

      Extracellular S100A11 Plays a Critical Role in Spread of the Fibroblast Population in Pancreatic Cancers

      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

          The fertile stroma in pancreatic ductal adenocarcinomas (PDACs) has been suspected to greatly contribute to PDAC progression. Since the main cell constituents of the stroma are fibroblasts, there is crosstalking(s) between PDAC cells and surrounding fibroblasts in the stroma, which induces a fibroblast proliferation burst. We have reported that several malignant cancer cells including PDAC cells secrete a pronounced level of S100A11, which in turn stimulates proliferation of cancer cells via the receptor for advanced glycation end products (RAGE) in an autocrine manner. Owing to the RAGE + expression in fibroblasts, the extracellular abundant S100A11 will affect adjacent fibroblasts. In this study, we investigated the significance of the paracrine axis of S100A11–RAGE in fibroblasts for their proliferation activity. In in vitro settings, extracellular S100A11 induced upregulation of fibroblast proliferation. Our mechanistic studies revealed that the induction is through RAGE–MyD88–mTOR–p70 S6 kinase upon S100A11 stimulation. The paracrine effect on fibroblasts is linked mainly to triggering growth but not cellular motility. Thus, the identified pathway might become a potential therapeutic target to suppress PDAC progression through preventing PDAC-associated fibroblast proliferation.

          Related collections

          Most cited references41

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

          Cancer-associated stromal fibroblasts promote pancreatic tumor progression.

          Pancreatic adenocarcinoma is characterized by a dense background of tumor associated stroma originating from abundant pancreatic stellate cells. The aim of this study was to determine the effect of human pancreatic stellate cells (HPSC) on pancreatic tumor progression. HPSCs were isolated from resected pancreatic adenocarcinoma samples and immortalized with telomerase and SV40 large T antigen. Effects of HPSC conditioned medium (HPSC-CM) on in vitro proliferation, migration, invasion, soft-agar colony formation, and survival in the presence of gemcitabine or radiation therapy were measured in two pancreatic cancer cell lines. The effects of HPSCs on tumors were examined in an orthotopic murine model of pancreatic cancer by co-injecting them with cancer cells and analyzing growth and metastasis. HPSC-CM dose-dependently increased BxPC3 and Panc1 tumor cell proliferation, migration, invasion, and colony formation. Furthermore, gemcitabine and radiation therapy were less effective in tumor cells treated with HPSC-CM. HPSC-CM activated the mitogen-activated protein kinase and Akt pathways in tumor cells. Co-injection of tumor cells with HPSCs in an orthotopic model resulted in increased primary tumor incidence, size, and metastasis, which corresponded with the proportion of HPSCs. HPSCs produce soluble factors that stimulate signaling pathways related to proliferation and survival of pancreatic cancer cells, and the presence of HPSCs in tumors increases the growth and metastasis of these cells. These data indicate that stellate cells have an important role in supporting and promoting pancreatic cancer. Identification of HPSC-derived factors may lead to novel stroma-targeted therapies for pancreatic cancer.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            The role of stromal cancer-associated fibroblasts in pancreatic cancer

            Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer generally refractory to conventional treatments. Cancer-associated fibroblasts (CAFs) are cellular components of the desmoplastic stroma characteristic to the tumor that contributes to this treatment resistance. Various markers for CAFs have been explored including palladin and CD146 that have prognostic and functional roles in the pathobiology of PDAC. Mechanisms of CAF-tumor cell interaction have been described including exosomal transfer and paracrine signaling mediated by cytokines such as GM-CSF and IL-6. The role of downstream signaling pathways including JAK/STAT, mTOR, sonic hedge hog (SHH), and NFkB have also been shown to play an important function in PDAC-CAF cross talk. The role of autophagy and other metabolic effects on each cell type within the tumor have also been proposed to play roles in facilitating CAF secretory function and enhancing tumor growth in a low-glucose microenvironment. Targeting the stroma has gained interest with multiple preclinical and clinical trials targeting SHH, JAK2, and methods of either exploiting the secretory capability of CAFs to enhance drug delivery or inhibiting it to prevent its influence on cancer cell chemoresistance. This review summarizes the most recent progress made in understanding stromal formation; its contribution to tumor proliferation, invasion, and metastasis; its role in chemoresistance; and potential therapeutic strategies on the horizon.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Proinflammatory S100 proteins regulate the accumulation of myeloid-derived suppressor cells.

              Chronic inflammation is a complex process that promotes carcinogenesis and tumor progression; however, the mechanisms by which specific inflammatory mediators contribute to tumor growth remain unclear. We and others recently demonstrated that the inflammatory mediators IL-1beta, IL-6, and PGE(2) induce accumulation of myeloid-derived suppressor cells (MDSC) in tumor-bearing individuals. MDSC impair tumor immunity and thereby facilitate carcinogenesis and tumor progression by inhibiting T and NK cell activation, and by polarizing immunity toward a tumor-promoting type 2 phenotype. We now show that this population of immature myeloid cells induced by a given tumor share a common phenotype regardless of their in vivo location (bone marrow, spleen, blood, or tumor site), and that Gr1(high)CD11b(high)F4/80(-)CD80(+)IL4Ralpha(+/-)Arginase(+) MDSC are induced by the proinflammatory proteins S100A8/A9. S100A8/A9 proteins bind to carboxylated N-glycans expressed on the receptor for advanced glycation end-products and other cell surface glycoprotein receptors on MDSC, signal through the NF-kappaB pathway, and promote MDSC migration. MDSC also synthesize and secrete S100A8/A9 proteins that accumulate in the serum of tumor-bearing mice, and in vivo blocking of S100A8/A9 binding to MDSC using an anti-carboxylated glycan Ab reduces MDSC levels in blood and secondary lymphoid organs in mice with metastatic disease. Therefore, the S100 family of inflammatory mediators serves as an autocrine feedback loop that sustains accumulation of MDSC. Since S100A8/A9 activation of MDSC is through the NF-kappaB signaling pathway, drugs that target this pathway may reduce MDSC levels and be useful therapeutic agents in conjunction with active immunotherapy in cancer patients.
                Bookmark

                Author and article information

                Journal
                Oncol Res
                Oncol Res
                OR
                Oncology Research
                Cognizant Communication Corporation (Elmsford, NY )
                0965-0407
                1555-3906
                2019
                21 June 2019
                : 27
                : 6
                : 713-727
                Affiliations
                [1]*Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama, Japan
                [2]†Faculty of Science and Technology, Division of Molecular Science, Gunma University , Kiryu, Gunma, Japan
                [3]‡Department of Biochemistry, Kawasaki Medical School , Kurashiki, Okayama, Japan
                [4]§Faculty of Medicine, Udayana University , Denpasar, Bali, Indonesia
                [5]¶Department of Clinical Oncology, Kawasaki Medical School , Kurashiki, Okayama, Japan
                [6]#Department of Pathology, Kawasaki Medical School , Kurashiki, Okayama, Japan
                [7]**Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama, Japan
                [8]††Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama, Japan
                [9]‡‡Department of Interdisciplinary Science and Engineering in Health Systems, Okayama University , Okayama, Japan
                [10]§§Division of Molecular and Cellular Pathology, Niigata University Graduate School of Medical and Dental Sciences , Niigata, Japan
                [11]¶¶Department of Pediatrics, Dr. Sardjito Hospital/Faculty of Medicine, Universitas Gadjah Mada , Yogyakarta, Indonesia
                [12]##Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama, Japan
                [13]***Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences , Kanazawa, Ishikawa, Japan
                Author notes

                1These authors provided equal contribution to this work.

                Address correspondence to Masakiyo Sakaguchi, Ph.D., Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan. Tel: +81-86-235-7395; Fax: +81-86-235-7400; E-mail: masa-s@ 123456md.okayama-u.ac.jp
                Article
                OR1355
                10.3727/096504018X15433161908259
                7848439
                30850029
                90a90a44-615a-4e3f-9ad0-5cddf78d2875
                Copyright © 2019 Cognizant, LLC.

                This article is licensed under a Creative Commons Attribution-NonCommercial NoDerivatives 4.0 International License.

                History
                Page count
                Figures: 8, Tables: 0, References: 41, Pages: 15
                Categories
                Article

                s100a11,pancreatic cancer,fibroblasts,rage,cancer microenvironment

                Comments

                Comment on this article