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

      An FGFR/AKT/SOX2 Signaling Axis Controls Pancreatic Cancer Stemness

      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

          Cancer stemness is associated with high malignancy and low differentiation, as well as therapeutic resistance of tumors including pancreatic ductal adenocarcinoma (PDAC). Fibroblast growth factors (FGFs) exert pleiotropic effects on a variety of cellular processes and functions including embryonic stem cell pluripotency and cancer cell stemness via the activation of four tyrosine kinase FGF receptors (FGFRs). FGF ligands have been a major component of the cocktail of growth factors contained in the cancer stemness-inducing (CSI) and organoid culture medium. Although FGF/FGFR signaling has been hypothesized to maintain cancer stemness, its function in this process is still unclear. We report that inhibition of FGF/FGFR signaling impairs sphere-forming ability of PDAC in vitro, and knocking down FGFR1 and FGFR2 decreased their tumorigenesis abilities in vivo. Mechanistically, we demonstrated that SOX2 is down-regulated upon loss of FGFR signaling. The overexpression of SOX2 in SOX2-negative cells, which normally do not display stemness capabilities, is sufficient to induce spheroid formation. Additionally, we found that AKT phosphorylation was reduced upon FGFR signaling inhibition. The inhibition of AKT using specific pharmacological inhibitors in the context of CSI medium leads to the loss of spheroid formation associated with loss of SOX2 nuclear expression and increased degradation. We demonstrate that an FGFR/AKT/SOX2 axis controls cancer stemness in PDAC and therefore may represent an important therapeutic target in the fight against this very aggressive form of cancer.

          Related collections

          Most cited references51

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

          Identification of pancreatic cancer stem cells.

          Emerging evidence has suggested that the capability of a tumor to grow and propagate is dependent on a small subset of cells within a tumor, termed cancer stem cells. Although data have been provided to support this theory in human blood, brain, and breast cancers, the identity of pancreatic cancer stem cells has not been determined. Using a xenograft model in which primary human pancreatic adenocarcinomas were grown in immunocompromised mice, we identified a highly tumorigenic subpopulation of pancreatic cancer cells expressing the cell surface markers CD44, CD24, and epithelial-specific antigen (ESA). Pancreatic cancer cells with the CD44(+)CD24(+)ESA(+) phenotype (0.2-0.8% of pancreatic cancer cells) had a 100-fold increased tumorigenic potential compared with nontumorigenic cancer cells, with 50% of animals injected with as few as 100 CD44(+)CD24(+)ESA(+) cells forming tumors that were histologically indistinguishable from the human tumors from which they originated. The enhanced ability of CD44(+)CD24(+)ESA(+) pancreatic cancer cells to form tumors was confirmed in an orthotopic pancreatic tail injection model. The CD44(+)CD24(+)ESA(+) pancreatic cancer cells showed the stem cell properties of self-renewal, the ability to produce differentiated progeny, and increased expression of the developmental signaling molecule sonic hedgehog. Identification of pancreatic cancer stem cells and further elucidation of the signaling pathways that regulate their growth and survival may provide novel therapeutic approaches to treat pancreatic cancer, which is notoriously resistant to standard chemotherapy and radiation.
            Bookmark
            • Record: found
            • Abstract: not found
            • Article: not found

            Applying the principles of stem-cell biology to cancer.

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

              KRAS: The Critical Driver and Therapeutic Target for Pancreatic Cancer

              RAS genes (HRAS, KRAS, and NRAS) comprise the most frequently mutated oncogene family in human cancer. With the highest RAS mutation frequencies seen with the top three causes of cancer deaths in the United States (lung, colorectal, and pancreatic cancer), the development of anti-RAS therapies is a major priority for cancer research. Despite more than three decades of intense effort, no effective RAS inhibitors have yet to reach the cancer patient. With bitter lessons learned from past failures and with new ideas and strategies, there is renewed hope that undruggable RAS may finally be conquered. With the KRAS isoform mutated in 84% of all RAS-mutant cancers, we focus on KRAS. With a near 100% KRAS mutation frequency, pancreatic ductal adenocarcinoma (PDAC) is considered the most RAS-addicted of all cancers. We review the role of KRAS as a driver and therapeutic target in PDAC.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Cell Dev Biol
                Front Cell Dev Biol
                Front. Cell Dev. Biol.
                Frontiers in Cell and Developmental Biology
                Frontiers Media S.A.
                2296-634X
                07 May 2020
                2020
                : 8
                : 287
                Affiliations
                [1] 1School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University , Wenzhou, China
                [2] 2Institute of Life Sciences, Wenzhou University , Wenzhou, China
                [3] 3Center for Precision Medicine, The First Affiliated Hospital of Wenzhou Medical University , Wenzhou, China
                [4] 4Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic , Rochester, MN, United States
                [5] 5Cardio-Pulmonary Institute, Member of the German Lung Center, Justus Liebig University Giessen , Giessen, Germany
                Author notes

                Edited by: Nan-Shan Chang, National Cheng Kung University, Taiwan

                Reviewed by: Raj Ladher, National Centre for Biological Sciences, India; Michael Blank, Bar-Ilan University, Israel; Angie Rizzino, University of Nebraska Medical Center, United States

                *Correspondence: Xiaokun Li, Xiaokunli@ 123456wmu.edu.cn

                ORCID: Jin-San Zhang, orcid.org/0000-0002-4436-9593

                This article was submitted to Signaling, a section of the journal Frontiers in Cell and Developmental Biology

                Article
                10.3389/fcell.2020.00287
                7221133
                32457900
                6532ddfe-5a2c-492b-a844-cd472c31e52b
                Copyright © 2020 Quan, Guo, Liu, Yang, Bai, Wang, Cai, Han, Lv, Ding, Billadeau, Lou, Bellusci, Li and Zhang.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 10 December 2019
                : 02 April 2020
                Page count
                Figures: 6, Tables: 2, Equations: 0, References: 68, Pages: 15, Words: 0
                Categories
                Cell and Developmental Biology
                Original Research

                fgfr,sox2,pancreatic cancer,stemness,sphere-formation assay
                fgfr, sox2, pancreatic cancer, stemness, sphere-formation assay

                Comments

                Comment on this article