29
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      HYAL-2–WWOX–SMAD4 Signaling in Cell Death and Anticancer Response

      review-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

          Hyaluronidase HYAL-2 is a membrane-anchored protein and also localizes, in part, in the lysosome. Recent study from animal models revealed that both HYAL-1 and HYAL-2 are essential for the metabolism of hyaluronan (HA). Hyal-2 deficiency is associated with chronic thrombotic microangiopathy with hemolytic anemia in mice due to over accumulation of high molecular size HA. HYAL-2 is essential for platelet generation. Membrane HYAL-2 degrades HA bound by co-receptor CD44. Also, in a non-canonical signal pathway, HYAL-2 serves as a receptor for transforming growth factor beta (TGF-β) to signal with downstream tumor suppressors WWOX and SMAD4 to control gene transcription. When SMAD4 responsive element is overly driven by the HYAL-2–WWOX–SMAD4 signaling complex, cell death occurs. When rats are subjected to traumatic brain injury, over accumulation of a HYAL-2–WWOX complex occurs in the nucleus to cause neuronal death. HA induces the signaling of HYAL-2–WWOX–SMAD4 and relocation of the signaling complex to the nucleus. If the signaling complex is overexpressed, bubbling cell death occurs in WWOX-expressing cells. In addition, a small synthetic peptide Zfra (zinc finger-like protein that regulates apoptosis) binds membrane HYAL-2 of non-T/non-B spleen HYAL-2 + CD3 CD19 Z lymphocytes and activates the cells to generate memory anticancer response against many types of cancer cells in vivo. Whether the HYAL-2–WWOX–SMAD4 signaling complex is involved is discussed. In this review and opinion article, we have updated the current knowledge of HA, HYAL-2 and WWOX, HYAL-2–WWOX–SMAD4 signaling, bubbling cell death, and Z cell activation for memory anticancer response.

          Related collections

          Most cited references117

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

          Interactions between Hyaluronan and Its Receptors (CD44, RHAMM) Regulate the Activities of Inflammation and Cancer

          The glycosaminoglycan hyaluronan (HA), a major component of extracellular matrices, and cell surface receptors of HA have been proposed to have pivotal roles in cell proliferation, migration, and invasion, which are necessary for inflammation and cancer progression. CD44 and receptor for HA-mediated motility (RHAMM) are the two main HA-receptors whose biological functions in human and murine inflammations and tumor cells have been investigated comprehensively. HA was initially considered to be only an inert component of connective tissues, but is now known as a “dynamic” molecule with a constant turnover in many tissues through rapid metabolism that involves HA molecules of various sizes: high molecular weight HA (HMW HA), low molecular weight HA, and oligosaccharides. The intracellular signaling pathways initiated by HA interactions with CD44 and RHAMM that lead to inflammatory and tumorigenic responses are complex. Interestingly, these molecules have dual functions in inflammations and tumorigenesis. For example, the presence of CD44 is involved in initiation of arthritis, while the absence of CD44 by genetic deletion in an arthritis mouse model increases rather than decreases disease severity. Similar dual functions of CD44 exist in initiation and progression of cancer. RHAMM overexpression is most commonly linked to cancer progression, whereas loss of RHAMM is associated with malignant peripheral nerve sheath tumor growth. HA may similarly perform dual functions. An abundance of HMW HA can promote malignant cell proliferation and development of cancer, whereas antagonists to HA-CD44 signaling inhibit tumor cell growth in vitro and in vivo by interfering with HMW HA-CD44 interaction. This review describes the roles of HA interactions with CD44 and RHAMM in inflammatory responses and tumor development/progression, and how therapeutic strategies that block these key inflammatory/tumorigenic processes may be developed in rodent and human diseases.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            High molecular weight hyaluronan mediates the cancer resistance of the naked mole-rat

            The naked mole-rat displays exceptional longevity, with a maximum lifespan exceeding 30 years 1–3 . This is the longest reported lifespan for a rodent species and is especially striking considering the small body mass of the naked mole-rat. In comparison, a similarly sized house mouse has a maximum lifespan of 4 years 4,5 . In addition to their longevity, naked mole-rats show an unusual resistance to cancer. Multi-year observations of large naked mole-rat colonies did not detect a single incidence of cancer 2,6 . Here we identify a mechanism responsible for the naked mole-rat’s cancer resistance. We found that naked mole-rat fibroblasts secrete extremely high molecular weight hyaluronan (HA), which is over five times larger than human or mouse HA. This high molecular weight HA accumulates abundantly in naked mole rat tissues due to the decreased activity of HA-degrading enzymes and a unique sequence of hyaluronan synthase 2 (HAS2). Furthermore, the naked mole-rat cells are more sensitive to HA signaling, as the naked mole rat cells have a higher affinity to HA than the mouse or human cells. Perturbation of the signaling pathways sufficient for malignant transformation of mouse fibroblasts fails to transform naked mole-rat cells. However, once high molecular weight HA is removed by either knocking down HAS2 or overexpressing the HA-degrading enzyme, Hyal2, naked mole-rat cells become susceptible to malignant transformation and readily form tumors in mice. We speculate that naked mole-rats have evolved a higher concentration of HA in the skin to provide skin elasticity needed for life in underground tunnels. This trait may have then been co-opted to provide cancer resistance and longevity to this species.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Signaling Receptors for TGF-β Family Members.

              Transforming growth factor β (TGF-β) family members signal via heterotetrameric complexes of type I and type II dual specificity kinase receptors. The activation and stability of the receptors are controlled by posttranslational modifications, such as phosphorylation, ubiquitylation, sumoylation, and neddylation, as well as by interaction with other proteins at the cell surface and in the cytoplasm. Activation of TGF-β receptors induces signaling via formation of Smad complexes that are translocated to the nucleus where they act as transcription factors, as well as via non-Smad pathways, including the Erk1/2, JNK and p38 MAP kinase pathways, and the Src tyrosine kinase, phosphatidylinositol 3'-kinase, and Rho GTPases.
                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
                06 December 2016
                2016
                : 4
                : 141
                Affiliations
                [1] 1Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University Tainan, Taiwan
                [2] 2Department of Neurosurgery, Mackay Memorial Hospital, Mackay Medicine, Nursing and Management College, and Graduate Institute of Injury Prevention and Control, Taipei Medical University Taipei, Taiwan
                [3] 3Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University Tainan, Taiwan
                [4] 4Institute of Molecular Medicine, College of Medicine, National Cheng Kung University Tainan, Taiwan
                [5] 5Advanced Optoelectronic Technology Center, National Cheng Kung University Tainan, Taiwan
                [6] 6Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University Tainan, Taiwan
                [7] 7Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities Staten Island, NY, USA
                [8] 8Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University Taichung, Taiwan
                Author notes

                Edited by: Simon Rousseau, McGill University, Canada

                Reviewed by: Uwe Lendeckel, University of Greifswald, Germany; Carl-Henrik Heldin, Uppsala University, Sweden

                *Correspondence: Nan-Shan Chang changns@ 123456mail.ncku.edu.tw

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

                †These authors have contributed equally to this work.

                Article
                10.3389/fcell.2016.00141
                5138198
                27999774
                b0df8a81-b0cd-474e-afe7-93eb8c8a163c
                Copyright © 2016 Hsu, Chiang, Sze, Su, Yap, Lee, Kuo and Chang.

                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) or licensor 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
                : 07 September 2016
                : 18 November 2016
                Page count
                Figures: 2, Tables: 0, Equations: 0, References: 136, Pages: 11, Words: 10156
                Funding
                Funded by: U.S. Department of Defense 10.13039/100000005
                Award ID: W81XWH-08–1-0682
                Funded by: National Health Research Institutes 10.13039/501100004737
                Award ID: NHRI-EX99–9704BI
                Categories
                Cell and Developmental Biology
                Review

                zfra,cancer,hyaluronan,hyaluronidase,prevention,treatment,hyal-2,z cell
                zfra, cancer, hyaluronan, hyaluronidase, prevention, treatment, hyal-2, z cell

                Comments

                Comment on this article