36
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Structural Analysis of Specific Metal Chelating Inhibitor Binding to the Endonuclease Domain of Influenza pH1N1 (2009) Polymerase

      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

          It is generally recognised that novel antiviral drugs, less prone to resistance, would be a desirable alternative to current drug options in order to be able to treat potentially serious influenza infections. The viral polymerase, which performs transcription and replication of the RNA genome, is an attractive target for antiviral drugs since potent polymerase inhibitors could directly stop viral replication at an early stage. Recent structural studies on functional domains of the heterotrimeric polymerase, which comprises subunits PA, PB1 and PB2, open the way to a structure based approach to optimise inhibitors of viral replication. In particular, the unique cap-snatching mechanism of viral transcription can be inhibited by targeting either the PB2 cap-binding or PA endonuclease domains. Here we describe high resolution X-ray co-crystal structures of the 2009 pandemic H1N1 (pH1N1) PA endonuclease domain with a series of specific inhibitors, including four diketo compounds and a green tea catechin, all of which chelate the two critical manganese ions in the active site of the enzyme. Comparison of the binding mode of the different compounds and that of a mononucleotide phosphate highlights, firstly, how different substituent groups on the basic metal binding scaffold can be orientated to bind in distinct sub-pockets within the active site cavity, and secondly, the plasticity of certain structural elements of the active site cavity, which result in induced fit binding. These results will be important in optimising the design of more potent inhibitors targeting the cap-snatching endonuclease activity of influenza virus polymerase.

          Author Summary

          The 2009 influenza pandemic, the on-going potential threat of highly pathogenic H5N1 avian strains and the widespread occurrence of resistance to current anti-influenza drugs targeting the neuraminidase or the M2 ion channel, all highlight the need for alternative therapeutic options to treat serious influenza infections in the absence of protection by vaccination. The viral polymerase, which performs transcription and replication of the RNA genome, is an attractive target for novel antiviral drugs since potent polymerase inhibitors will directly stall replication. The heterotrimeric polymerase performs transcription by a unique cap-snatching mechanism, which involves host pre-mRNA cap-binding and endonucleolytic cleavage by the PB2 and PA subunits respectively. Crystal structures of both the PB2 cap-binding and PA nuclease domains are now available allowing structure-guided optimisation of cap-snatching inhibitors. Here we present a series of co-crystal structures of the 2009 pandemic H1N1 PA endonuclease domain that reveal the binding mode of several known endonuclease inhibitors. All inhibitors chelate the two manganese ions in the active site of the nuclease but different extensions to the metal binding scaffold bind in distinct sub-pockets of the active site cavity. These results highlight the value of structure-based approaches to the development of more potent influenza polymerase inhibitors.

          Related collections

          Most cited references33

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

          Automatic processing of rotation diffraction data from crystals of initially unknown symmetry and cell constants

          W Kabsch (1993)
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The cap-snatching endonuclease of influenza virus polymerase resides in the PA subunit.

            The influenza virus polymerase, a heterotrimer composed of three subunits, PA, PB1 and PB2, is responsible for replication and transcription of the eight separate segments of the viral RNA genome in the nuclei of infected cells. The polymerase synthesizes viral messenger RNAs using short capped primers derived from cellular transcripts by a unique 'cap-snatching' mechanism. The PB2 subunit binds the 5' cap of host pre-mRNAs, which are subsequently cleaved after 10-13 nucleotides by the viral endonuclease, hitherto thought to reside in the PB2 (ref. 5) or PB1 (ref. 2) subunits. Here we describe biochemical and structural studies showing that the amino-terminal 209 residues of the PA subunit contain the endonuclease active site. We show that this domain has intrinsic RNA and DNA endonuclease activity that is strongly activated by manganese ions, matching observations reported for the endonuclease activity of the intact trimeric polymerase. Furthermore, this activity is inhibited by 2,4-dioxo-4-phenylbutanoic acid, a known inhibitor of the influenza endonuclease. The crystal structure of the domain reveals a structural core closely resembling resolvases and type II restriction endonucleases. The active site comprises a histidine and a cluster of three acidic residues, conserved in all influenza viruses, which bind two manganese ions in a configuration similar to other two-metal-dependent endonucleases. Two active site residues have previously been shown to specifically eliminate the polymerase endonuclease activity when mutated. These results will facilitate the optimisation of endonuclease inhibitors as potential new anti-influenza drugs.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Antiviral effect of catechins in green tea on influenza virus.

              Polyphenolic compound catechins ((-)-epigallocatechin gallate (EGCG), (-)-epicatechin gallate (ECG) and (-)-epigallocatechin (EGC)) from green tea were evaluated for their ability to inhibit influenza virus replication in cell culture and for potentially direct virucidal effect. Among the test compounds, the EGCG and ECG were found to be potent inhibitors of influenza virus replication in MDCK cell culture and this effect was observed in all influenza virus subtypes tested, including A/H1N1, A/H3N2 and B virus. The 50% effective inhibition concentration (EC50) of EGCG, ECG, and EGC for influenza A virus were 22-28, 22-40 and 309-318 microM, respectively. EGCG and ECG exhibited hemagglutination inhibition activity, EGCG being more effective. However, the sensitivity in hemagglutination inhibition was widely different among three different subtypes of influenza viruses tested. Quantitative RT-PCR analysis revealed that, at high concentration, EGCG and ECG also suppressed viral RNA synthesis in MDCK cells whereas EGC failed to show similar effect. Similarly, EGCG and ECG inhibited the neuraminidase activity more effectively than the EGC. The results show that the 3-galloyl group of catechin skeleton plays an important role on the observed antiviral activity, whereas the 5'-OH at the trihydroxy benzyl moiety at 2-position plays a minor role. The results, along with the HA type-specific effect, suggest that the antiviral effect of catechins on influenza virus is mediated not only by specific interaction with HA, but altering the physical properties of viral membrane.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS Pathog
                PLoS Pathog
                plos
                plospath
                PLoS Pathogens
                Public Library of Science (San Francisco, USA )
                1553-7366
                1553-7374
                August 2012
                August 2012
                2 August 2012
                : 8
                : 8
                : e1002831
                Affiliations
                [1 ]European Molecular Biology Laboratory, Grenoble Outstation, BP181, Grenoble, France
                [2 ]Unit of Virus Host-Cell Interactions, UJF-EMBL-CNRS, UMI 3265, BP181, Grenoble, France
                [3 ]Savira pharmaceuticals Gmbh, Vienna, Austria
                Johns Hopkins University - Bloomberg School of Public Health, United States of America
                Author notes

                ¤: a Current address: Max-Planck Institute for Biochemistry, Martinsried, Germany

                ¤: b Current address: European Synchrotron Radiation Facility, BP 220, Grenoble, France

                Conceived and designed the experiments: EK CZ AW OHJS RWHR SC. Performed the experiments: EK CZ AW SC. Analyzed the data: EK CZ AW SC. Contributed reagents/materials/analysis tools: EK CZ AW. Wrote the paper: EK AW SC.

                Article
                PPATHOGENS-D-12-00745
                10.1371/journal.ppat.1002831
                3410856
                22876177
                1fa00478-241c-435f-8483-e8d219aac039
                Kowalinski 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
                : 26 March 2012
                : 15 June 2012
                Page count
                Pages: 14
                Categories
                Research Article
                Biology
                Biochemistry
                Drug Discovery
                Enzymes
                Small Molecules
                Biophysics
                Biomacromolecule-Ligand Interactions
                Microbiology
                Virology
                Chemistry
                Medicinal Chemistry

                Infectious disease & Microbiology
                Infectious disease & Microbiology

                Comments

                Comment on this article