26
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Zinc Downregulates HIF-1α and Inhibits Its Activity in Tumor Cells In Vitro and In Vivo

      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

          Hypoxia inducible factor-1α (HIF-1α) is responsible for the majority of HIF-1-induced gene expression changes under hypoxia and for the “angiogenic switch” during tumor progression. HIF-1α is often upregulated in tumors leading to more aggressive tumor growth and chemoresistance, therefore representing an important target for antitumor intervention. We previously reported that zinc downregulated HIF-1α levels. Here, we evaluated the molecular mechanisms of zinc-induced HIF-1α downregulation and whether zinc affected HIF-1α also in vivo.

          Methodology/Principal Findings

          Here we report that zinc downregulated HIF-1α protein levels in human prostate cancer and glioblastoma cells under hypoxia, whether induced or constitutive. Investigations into the molecular mechanisms showed that zinc induced HIF-1α proteasomal degradation that was prevented by treatment with proteasomal inhibitor MG132. HIF-1α downregulation induced by zinc was ineffective in human RCC4 VHL-null renal carcinoma cell line; likewise, the HIF-1αP402/P564A mutant was resistant to zinc treatment. Similarly to HIF-1α, zinc downregulated also hypoxia-induced HIF-2α whereas the HIF-1β subunit remained unchanged. Zinc inhibited HIF-1α recruitment onto VEGF promoter and the zinc-induced suppression of HIF-1-dependent activation of VEGF correlated with reduction of glioblastoma and prostate cancer cell invasiveness in vitro. Finally, zinc administration downregulated HIF-1α levels in vivo, by bioluminescence imaging, and suppressed intratumoral VEGF expression.

          Conclusions/Significance

          These findings, by demonstrating that zinc induces HIF-1α proteasomal degradation, indicate that zinc could be useful as an inhibitor of HIF-1α in human tumors to repress important pathways involved in tumor progression, such as those induced by VEGF, MDR1, and Bcl2 target genes, and hopefully potentiate the anticancer therapies.

          Related collections

          Most cited references25

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

          HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing.

          HIF (hypoxia-inducible factor) is a transcription factor that plays a pivotal role in cellular adaptation to changes in oxygen availability. In the presence of oxygen, HIF is targeted for destruction by an E3 ubiquitin ligase containing the von Hippel-Lindau tumor suppressor protein (pVHL). We found that human pVHL binds to a short HIF-derived peptide when a conserved proline residue at the core of this peptide is hydroxylated. Because proline hydroxylation requires molecular oxygen and Fe(2+), this protein modification may play a key role in mammalian oxygen sensing.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics.

            Adaptation of cancer cells to their microenvironment is an important driving force in the clonal selection that leads to invasive and metastatic disease. O2 concentrations are markedly reduced in many human cancers compared with normal tissue, and a major mechanism mediating adaptive responses to reduced O2 availability (hypoxia) is the regulation of transcription by hypoxia-inducible factor 1 (HIF-1). This review summarizes the current state of knowledge regarding the molecular mechanisms by which HIF-1 contributes to cancer progression, focusing on (1) clinical data associating increased HIF-1 levels with patient mortality; (2) preclinical data linking HIF-1 activity with tumor growth; (3) molecular data linking specific HIF-1 target gene products to critical aspects of cancer biology and (4) pharmacological data showing anticancer effects of HIF-1 inhibitors in mouse models of human cancer.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis.

              Herein we report that the VEGFR/PDGFR kinase inhibitor sunitinib/SU11248 can accelerate metastatic tumor growth and decrease overall survival in mice receiving short-term therapy in various metastasis assays, including after intravenous injection of tumor cells or after removal of primary orthotopically grown tumors. Acceleration of metastasis was also observed in mice receiving sunitinib prior to intravenous implantation of tumor cells, suggesting possible "metastatic conditioning" in multiple organs. Similar findings with additional VEGF receptor tyrosine kinase inhibitors implicate a class-specific effect for such agents. Importantly, these observations of metastatic acceleration were in contrast to the demonstrable antitumor benefits obtained when the same human breast cancer cells, as well as mouse or human melanoma cells, were grown orthotopically as primary tumors and subjected to identical sunitinib treatments.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS One
                plos
                plosone
                PLoS ONE
                Public Library of Science (San Francisco, USA )
                1932-6203
                2010
                13 December 2010
                : 5
                : 12
                : e15048
                Affiliations
                [1 ]Molecular Oncogenesis Laboratory, Department of Experimental Oncology, National Cancer Institute “Regina Elena”, Rome, Italy
                [2 ]Chair of Endocrinology, Catholic University, Rome, Italy
                [3 ]Department of Oncology and Experimental Medicine, School of Medicine, University “G. d'Annunzio”, Chieti, Italy
                [4 ]Experimental Chemotherapy, National Cancer Institute “Regina Elena”, Rome, Italy
                [5 ]National Research Council, Institute of Neurobiology and Molecular Medicine, Rome, Italy
                [6 ]Cancer Research Center, Chaim Sheba Medical Center, Tel-Hashomer and Sachler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
                [7 ]Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
                Institute for Medical Biomathematics, Israel
                Author notes

                Conceived and designed the experiments: GD LN. Performed the experiments: LN VP RP AA MP AG. Analyzed the data: GD LN AF. Contributed reagents/materials/analysis tools: CL MS GR AF DG. Wrote the paper: GD.

                Article
                PONE-D-10-00110
                10.1371/journal.pone.0015048
                3001454
                21179202
                f87ce73d-7ee2-4e1b-b85d-780e36c14e69
                Nardinocchi et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
                History
                : 30 July 2010
                : 25 October 2010
                Page count
                Pages: 11
                Categories
                Research Article
                Chemistry
                Chemical Biology
                Medicine
                Oncology
                Basic Cancer Research
                Cancer Treatment

                Uncategorized
                Uncategorized

                Comments

                Comment on this article