The SARS-coronavirus papain-like protease: structure, function and inhibition by designed antiviral compounds

Yahira M Báez-Santos, Sarah E St John, Andrew D Mesecar, Yahira M Báez-Santos, Sarah E St John, Andrew D Mesecar

Abstract

Over 10 years have passed since the deadly human coronavirus that causes severe acute respiratory syndrome (SARS-CoV) emerged from the Guangdong Province of China. Despite the fact that the SARS-CoV pandemic infected over 8500 individuals, claimed over 800 lives and cost billions of dollars in economic loss worldwide, there still are no clinically approved antiviral drugs, vaccines or monoclonal antibody therapies to treat SARS-CoV infections. The recent emergence of the deadly human coronavirus that causes Middle East respiratory syndrome (MERS-CoV) is a sobering reminder that new and deadly coronaviruses can emerge at any time with the potential to become pandemics. Therefore, the continued development of therapeutic and prophylactic countermeasures to potentially deadly coronaviruses is warranted. The coronaviral proteases, papain-like protease (PLpro) and 3C-like protease (3CLpro), are attractive antiviral drug targets because they are essential for coronaviral replication. Although the primary function of PLpro and 3CLpro are to process the viral polyprotein in a coordinated manner, PLpro has the additional function of stripping ubiquitin and ISG15 from host-cell proteins to aid coronaviruses in their evasion of the host innate immune responses. Therefore, targeting PLpro with antiviral drugs may have an advantage in not only inhibiting viral replication but also inhibiting the dysregulation of signaling cascades in infected cells that may lead to cell death in surrounding, uninfected cells. This review provides an up-to-date discussion on the SARS-CoV papain-like protease including a brief overview of the SARS-CoV genome and replication followed by a more in-depth discussion on the structure and catalytic mechanism of SARS-CoV PLpro, the multiple cellular functions of SARS-CoV PLpro, the inhibition of SARS-CoV PLpro by small molecule inhibitors, and the prospect of inhibiting papain-like protease from other coronaviruses. This paper forms part of a series of invited articles in Antiviral Research on "From SARS to MERS: 10years of research on highly pathogenic human coronaviruses."

Keywords: 3C-like protease; MERS-CoV; Nsp3; Papain-like protease; SARS-CoV; Ubiquitin.

Copyright © 2014 Elsevier B.V. All rights reserved.

Figures

Fig. 1
Fig. 1
Genome and proteome organization of SARS-CoV non-structural proteins highlighting nsp3 domain organization and PLpro cleavage sites. The ∼30 kb genome of SARS-CoV and its associated replicase, structural and accessory proteins are indicated and the sizes of boxes representing each protein are to scale (top of figure). The replicase genes encoded by ORF1a and ORF1b are shaded in grey. The nsp3 multi-domain protein is shown with the amino acids defining the approximate boundaries of each domain indicated underneath. Downstream of the SARS-CoV PLpro cleavage site between nsp2/3 (818/819aa) is a ubiquitin-like domain (Ubl-1), PDB: 2GRI, a N-terminal Glu-rich acidic-domain (AC), ADP-ribose-1″-phosphatase (ADRP) domain (PDB: 2FAV) the SARS unique domain SUD (PDB: 2WCT, 2JZE, 2KAF,), a papain-like protease (PLpro) containing a second UBl-2 domain at the N-terminus (PDB: 2FE8) followed by the nucleic acid binding domain (NAB), PDB: 2K87, the marker domain G2M and four predicted transmembrane domains (TM1–TM4) forming an additional domain containing a metal-binding region (ZF). Finally, the remainder of nsp3 is composed of so-called Y domains (Y1–3), which precede the C-terminal PLpro cleavage sequence at nsp3/4 (2740/2741aa). An alignment of the SARS-CoV PLpro cleavage sequences (right bottom corner) shows a comparison of the P-sites and P′-sites from the nsps to the C-terminal sequences of the cellular proteins ubiquitin (Ub) and ISG15 shown with an isopeptide bond at the P′-sites.
Fig. 2
Fig. 2
Multi-domain architecture of PLpro and its two ubiquitin binding subsites. (a) The SARS-CoV PLpro monomer (PDB: 2FE8) consists of four domains: starting from N- to the C-terminus, the extended UBL, the thumb, the palm and the fingers domain. (b) An overlay of SARS-CoV PLpro (blue) with USP7 (yellow, PDB: 4M5W) displaying the USP fold. (c) A solvent-accessible surface representation of SARS-CoV PLpro is shown in blue. A K48-linked di-Ub molecule (orange) superimposed onto an ISG15 (yellow) molecule is shown with molecules represented as ribbons. The ISG15 structure consists of two tandem UBL folds (Ub1 and Ub2). The two Ub and UBL binding subsites of SARS-CoV PLpro (Ratia et al., 2014) are shown in the solvent accessible surface representation with SUb1 shaded white and SUb2 shaded yellow.
Fig. 3
Fig. 3
Currently proposed sites of action for SARS-CoV PLpro-mediated antagonism of the innate immune response. Viral infection is sensed by RIG-I (RIG-I-like helicase) and MDA-5 (melanoma differentiation-associated protein 5) recognition of viral RNA. Recruitment of adaptor proteins MAVS transduces signals to the downstream kinase complex, which activates the transcription factor, IRF-3 and NF-κB, which coordinates the expression of type I interferons (IFN-β and -α). Type I IFN induces the activation of STAT transcription factors resulting in the expression of ISGs (IFN-stimulated genes) and the establishment of an antiviral state in surrounding cells. PLpro can act on different branches of these pathways by interacting with or recognizing and deISGylating and/or deubiquitinating proteins within these pathways. The net effect of these different functions is to help SARS-CoV evade the host antiviral response via antagonizing the establishment of an antiviral state.
Fig. 4
Fig. 4
Catalytic center of the active site of SARS-CoV PLpro. Unliganded X-ray crystal structure of SARS-CoV PLpro (PDB: 2FE8). The catalytic triad residues, Cys112, His273 and Asp287 are shown along with the oxyanion hole-stabilizing residue Trp107 (right panel). The flexible BL2 loop that interacts with the substrates is indicated as are the zinc ion (purple sphere) and α-helix α4 in the thumb domain. Distances between atoms are given in units of Ångströms (Å) and are indicated by dashed lines.
Fig. 5
Fig. 5
Catalytic cycle and proposed chemical mechanism of the SARS-CoV PLpro catalyzed reaction. Active site residues of the catalytic triad (Cys112, His273, Asp287) and oxyanion hole residue Trp107 are shown in black. The peptide substrate is shown in green and a catalytic water molecule is shown in blue.
Fig. 6
Fig. 6
Examples of cysteine protease inhibitors with covalent “warheads.” The chemical “warhead” groups are shown in boxes and the reactive, electrophilic portions are colored in red, with the corresponding electrophilic “warhead” carbon indicated with an asterisk. The resulting covalent inhibitor–cysteine complexes are shown outside the boxes and the cysteine portions of the covalent adducts are colored in blue.
Fig. 7
Fig. 7
X-ray structure of SARS-CoV PLpro in complex with ubiquitin-aldehyde (Ubal). (a) Pictorial representation of the tetrahedral hemithioaminal intermediate generated upon PLpro–Ubal complex formation, where the Ubal C-terminal residues are shown in orange and the SARS-CoV PLpro residues are shown in black. (b) X-ray crystal structure of PLpro–Ubal complex (PDB: 4MM3 (Ratia et al., 2014)) where the PLpro is shown in blue and the residues His273, Cys112, Asp287, and Trp107 are shown in ball-and-stick and colored according to atom type, and Ubal is shown in orange and colored according to atom type. Distances between atoms are given in units of ångströms (Å) and are indicated by dashed lines.
Fig. 8
Fig. 8
SARS-CoV PLpro inhibitors identified through small-scale screening efforts. Positions within each molecule that are susceptible to covalent modification through nucleophilic attack (“warhead” positions) are indicated in red where the electrophilic carbons are indicated with an asterisk. (a) Inhibitors identified via yeast-based screen (Frieman et al., 2011), (b) thiopurine inhibitors (Chen et al., 2009, Chou et al., 2008), (c) natural product inhibitors (Cho et al., 2013, Park et al., 2012a, Park et al., 2012b), where the most potent natural product of each class is shown and the IC50 detailed.
Fig. 9
Fig. 9
Discovery and design pipeline for SARS-CoV PLpro inhibitors 24 and 15g. Results of the primary high-throughput screen identified hit compounds 7724772 (a1) and 6577871 (a2). SAR diagrams for analogs of 7724772 (b1) and 6577871 (b2) (Ghosh et al., 2010, Ratia et al., 2008). Structures of potent, first-generation leads 24 (c1) and 15g (c2). X-ray crystal structures of 24 (d1) (PDB: 3E9S) and 15g (d2) (PDB: 3MJ5) bound to SARS-CoV PLpro highlighting the residues involved in binding each inhibitor. Hydrogen bonds are shown as dashed lines.
Fig. 10
Fig. 10
Discovery and design pipeline of compounds 3k, 3j, 3e, and 5c. (a) Initial SARS-CoV PLpro inhibitor leads, 15g and 24, (b) SARs derived from small 15g analog library, (c) top SARS-CoV PLpro inhibitor candidates, 3k, 3j, 3e, and 5c (Baez-Santos et al., 2014a).
Fig. 11
Fig. 11
X-ray crystal structures of SARS-CoV PLpro in complex with inhibitors 3k and 3j and comparison to 15g. (a) An overlay of SARS-CoV PLpro (blue cartoon, gray surface) in complex with 3k (orange stick, PDB: 4OW0) and PLpro (cyan cartoon, gray surface) in complex with 3j (pink sticks, PDB: 4OVZ) depicting amino acids important for inhibitor binding (Baez-Santos et al., 2014a). (b) An overlay of PLpro–3k (PLpro in blue cartoon, 3k in orange sticks), PLpro–3j (PLpro in cyan cartoon, 3j in pink sticks) and PLpro–15g (green cartoon and sticks, (PDB: 3MJ5, (Ghosh et al., 2010)) complexes.
Fig. 12
Fig. 12
X-ray crystal structures of SARS-CoV PLpro in different asymmetric units. PLpro is shown as a ribbon representation with each monomer within an asymmetric unit colored in a different shade of blue. The BL2 loop is highlighted in red and the zinc ion is shown as a pink sphere. (a) The crystallographic trimer of unliganded PLpro (PDB: 2FE8, (Ratia et al., 2006)) is shown in the left panel and a structural superposition of the three monomers is shown in the right panel. (b) The crystallographic dimer of the SARS-CoV PLpro–15g inhibitor complex (PDB: 3MJ5, (Ghosh et al., 2010)) is shown in the left panel and a structural superposition of the two monomers is shown in the right panel. Compound 15g is shown in orange. (c) The crystallographic monomer of the SARS-CoV PLpro–24 inhibitor complex (PDB: 3E9S, (Ratia et al., 2008)) is shown in the left panel. Compound 24 is shown in green. The right panel depicts the structural superposition of the SARS-CoV PLpro–15g and SARS-CoV PLpro–24 inhibitor complexes. (D) The crystallographic dimer of the SARS-CoV PLpro–3k inhibitor complex (PDB: 4OW0, (Baez-Santos et al., 2014a)) is shown in the left panel and a structural superposition of the two monomers in the asymmetric unit is shown in the right panel.
Fig. 13
Fig. 13
X-ray crystal structures of SARS-CoV PLpro in complex with ubiquitin. (a) An overlay of the crystallographic monomer from the two PLpro–Ub complex (PDB: 4M0W, (Chou et al., 2014) and 4MM3 (Ratia et al., 2014)) crystal structures. The PLpro–Ubal complex is shown in blue with Ubal in orange and the PLproC112S–Ub complex is shown in light blue with Ub in yellow. (b) The conformational difference of the BL2 loop and the different orientations of PLpro BL2 residues and the C-terminal residues (72–76) of Ub (italics labels). (c) The different orientations of the active site residues with the distances between the atoms indicated in angstrom (black for PLpro–Ubal and red for PLproC112S–Ub).
Fig. 14
Fig. 14
Structural comparisons of the active sites of SARS-CoV PLpro, HCoV-NL63 PLP2 and MERS-CoV PLpro. The X-ray structure of SARS-CoV PLpro in complex with compound 3k (orange ball and sticks, PDB: 4OW0) is shown superimposed with homology models of MERS-CoV PLpro and HCoV-NL63 PLP2 (green). The amino-acid residues important for SARS-CoV PLpro–inhibitor interactions are shown (blue front) along with the predicted corresponding amino acid in HCoV-NL63 PLP2 (green font) and MERS-CoV PLpro (black font). Highlighted in bold are the non-conserved substitutions in MERS-CoV PLpro sequence. Shown at the bottom of the figure is a comparison between SARS-CoV, HCoV-NL63 and MERS-CoV PLpro’s amino acid composition of the BL2 β-turn/loop (highlighted with an arrow). This loop is essential for the inhibitor induced fit mechanism of association of compound 3k and related inhibitors with SARS-CoV PLpro. This figure has been adapted from Baez-Santos et al. (2014b).

References

    1. Angelini M.M., Akhlaghpour M., Neuman B.W., Buchmeier M.J. Severe acute respiratory syndrome coronavirus nonstructural proteins 3, 4 and 6 induce double-membrane vesicles. MBio. 2013;4
    1. Baez-Santos Y.M. University of Illinois; Chicago: 2012. Insight into the Substrate Specificity and Inhibition of Human Coronavirus Papain-Like Proteases, Pharmacy. pp. 1–230.
    1. Baez-Santos Y.M., Barraza S.J., Wilson M.W., Agius M.P., Mielech A.M., Davis N.M., Baker S.C., Larsen S.D., Mesecar A.D. X-ray structural and biological evaluation of a series of potent and highly selective inhibitors of human coronavirus papain-like proteases. J. Med. Chem. 2014;57:2393–2412.
    1. Baez-Santos Y.M., Mielech A.M., Deng X., Baker S., Mesecar A.D. Catalytic function and substrate specificity of the PLpro domain of nsp3 from the Middle East Respiratory Syndrome Coronavirus (MERS-CoV) J. Virol. 2014
    1. Barrett A.J., Rawlings N.D., Woessner J.F. Handbook of Proteolytic Enzymes. third ed. vol. 1. Elsevier Science; 2012. (Catalytic Mechanisms of Cysteine Peptidases). (chapter 404)
    1. Barretto N., Jukneliene D., Ratia K., Chen Z., Mesecar A.D., Baker S.C. The papain-like protease of severe acute respiratory syndrome coronavirus has deubiquitinating activity. J. Virol. 2005;79:15189–15198.
    1. Barretto N., Jukneliene D., Ratia K., Chen Z., Mesecar A.D., Baker S.C. Deubiquitinating activity of the SARS-CoV papain-like protease. Adv. Exp. Med. Biol. 2006;581:37–41.
    1. Borodovsky A., Kessler B.M., Casagrande R., Overkleeft H.S., Wilkinson K.D., Ploegh H.L. A novel active site-directed probe specific for deubiquitylating enzymes reveals proteasome association of USP14. EMBO J. 2001;20:5187–5196.
    1. Borodovsky A., Ovaa H., Kolli N., Gan-Erdene T., Wilkinson K.D., Ploegh H.L., Kessler B.M. Chemistry-based functional proteomics reveals novel members of the deubiquitinating enzyme family. Chem. Biol. 2002;9:1149–1159.
    1. Bredenbeek P.J., Pachuk C.J., Noten A.F., Charite J., Luytjes W., Weiss S.R., Spaan W.J. The primary structure and expression of the second open reading frame of the polymerase gene of the coronavirus MHV-A59: a highly conserved polymerase is expressed by an efficient ribosomal frameshifting mechanism. Nucleic Acids Res. 1990;18:1825–1832.
    1. Brian D.A., Baric R.S. Coronavirus genome structure and replication. Curr. Top. Microbiol. Immunol. 2005;287:1–30.
    1. Calistri A., Munegato D., Carli I., Parolin C., Palu G. The ubiquitin-conjugating system: multiple roles in viral replication and infection. Cells. 2014;3:386–417.
    1. Chatterjee A., Johnson M.A., Serrano P., Pedrini B., Joseph J.S., Neuman B.W., Saikatendu K., Buchmeier M.J., Kuhn P., Wuthrich K. Nuclear magnetic resonance structure shows that the severe acute respiratory syndrome coronavirus-unique domain contains a macrodomain fold. J. Virol. 2009;83:1823–1836.
    1. Chen X., Chou C.Y., Chang G.G. Thiopurine analogue inhibitors of severe acute respiratory syndrome-coronavirus papain-like protease, a deubiquitinating and deISGylating enzyme. Antivir. Chem. Chemother. 2009;19:151–156.
    1. Cho J.K., Curtis-Long M.J., Lee K.H., Kim D.W., Ryu H.W., Yuk H.J., Park K.H. Geranylated flavonoids displaying SARS-CoV papain-like protease inhibition from the fruits of Paulownia tomentosa. Bioorg. Med. Chem. 2013;21:3051–3057.
    1. Chou C.Y., Chien C.H., Han Y.S., Prebanda M.T., Hsieh H.P., Turk B., Chang G.G., Chen X. Thiopurine analogues inhibit papain-like protease of severe acute respiratory syndrome coronavirus. Biochem. Pharmacol. 2008;75:1601–1609.
    1. Chou C.Y., Lai H.Y., Chen H.Y., Cheng S.C., Cheng K.W., Chou Y.W. Structural basis for catalysis and ubiquitin recognition by the severe acute respiratory syndrome coronavirus papain-like protease. Acta Crystallogr. D Biol. Crystallogr. 2014;70:572–581.
    1. Chouljenko V.N., Lin X.Q., Storz J., Kousoulas K.G., Gorbalenya A.E. Comparison of genomic and predicted amino acid sequences of respiratory and enteric bovine coronaviruses isolated from the same animal with fatal shipping pneumonia. J. Gen. Virol. 2001;82:2927–2933.
    1. Clementz M.A., Chen Z., Banach B.S., Wang Y., Sun L., Ratia K., Baez-Santos Y.M., Wang J., Takayama J., Ghosh A.K., Li K., Mesecar A.D., Baker S.C. Deubiquitinating and interferon antagonism activities of coronavirus papain-like proteases. J. Virol. 2010;84:4619–4629.
    1. Devaraj S.G., Wang N., Chen Z., Tseng M., Barretto N., Lin R., Peters C.J., Tseng C.T., Baker S.C., Li K. Regulation of IRF-3-dependent innate immunity by the papain-like protease domain of the severe acute respiratory syndrome coronavirus. J. Biol. Chem. 2007;282:32208–32221.
    1. Egloff M.P., Malet H., Putics A., Heinonen M., Dutartre H., Frangeul A., Gruez A., Campanacci V., Cambillau C., Ziebuhr J., Ahola T., Canard B. Structural and functional basis for ADP-ribose and poly(ADP-ribose) binding by viral macro domains. J. Virol. 2006;80:8493–8502.
    1. Fraile J.M., Quesada V., Rodriguez D., Freije J.M., Lopez-Otin C. Deubiquitinases in cancer: new functions and therapeutic options. Oncogene. 2012;31:2373–2388.
    1. Frieman M., Basu D., Matthews K., Taylor J., Jones G., Pickles R., Baric R., Engel D.A. Yeast based small molecule screen for inhibitors of SARS-CoV. PLoS One. 2011;6:e28479.
    1. Frieman M., Ratia K., Johnston R.E., Mesecar A.D., Baric R.S. Severe acute respiratory syndrome coronavirus papain-like protease ubiquitin-like domain and catalytic domain regulate antagonism of IRF3 and NF-kappaB signaling. J. Virol. 2009;83:6689–6705.
    1. Ghosh A.K., Takayama J., Aubin Y., Ratia K., Chaudhuri R., Baez Y., Sleeman K., Coughlin M., Nichols D.B., Mulhearn D.C., Prabhakar B.S., Baker S.C., Johnson M.E., Mesecar A.D. Structure-based design, synthesis, and biological evaluation of a series of novel and reversible inhibitors for the severe acute respiratory syndrome-coronavirus papain-like protease. J. Med. Chem. 2009;52:5228–5240.
    1. Ghosh A.K., Takayama J., Rao K.V., Ratia K., Chaudhuri R., Mulhearn D.C., Lee H., Nichols D.B., Baliji S., Baker S.C., Johnson M.E., Mesecar A.D. Severe acute respiratory syndrome coronavirus papain-like novel protease inhibitors: design, synthesis, protein-ligand X-ray structure and biological evaluation. J. Med. Chem. 2010;53:4968–4979.
    1. Ghosh A.K., Xi K., Johnson M.E., Baker S.C., Mesecar A.D. Progress in anti-SARS coronavirus chemistry, biology and chemotherapy. Annu. Rep. Med. Chem. 2006;41:183–196.
    1. Ghosh A.K., Xi K., Ratia K., Santarsiero B.D., Fu W.T., Harcourt B.H., Rota P.A., Baker S.C., Johnson M.E., Mesecar A.D. Design and synthesis of peptidomimetic severe acute respiratory syndrome chymotrypsin-like protease inhibitors. J. Med. Chem. 2005;48:6767–6771.
    1. Gosert R., Kanjanahaluethai A., Egger D., Bienz K., Baker S.C. RNA replication of mouse hepatitis virus takes place at double-membrane vesicles. J. Virol. 2002;76:3697–3708.
    1. Hagemeijer M.C., Verheije M.H., Ulasli M., Shaltiel I.A., de Vries L.A., Reggiori F., Rottier P.J., de Haan C.A. Dynamics of coronavirus replication-transcription complexes. J. Virol. 2010;84:2134–2149.
    1. Han Y.S., Chang G.G., Juo C.G., Lee H.J., Yeh S.H., Hsu J.T., Chen X. Papain-like protease 2 (PLP2) from severe acute respiratory syndrome coronavirus (SARS-CoV): expression, purification, characterization, and inhibition. Biochemistry. 2005;44:10349–10359.
    1. Harcourt B.H., Jukneliene D., Kanjanahaluethai A., Bechill J., Severson K.M., Smith C.M., Rota P.A., Baker S.C. Identification of severe acute respiratory syndrome coronavirus replicase products and characterization of papain-like protease activity. J. Virol. 2004;78:13600–13612.
    1. Hu M., Li P., Li M., Li W., Yao T., Wu J.W., Gu W., Cohen R.E., Shi Y. Crystal structure of a UBP-family deubiquitinating enzyme in isolation and in complex with ubiquitin aldehyde. Cell. 2002;111:1041–1054.
    1. Imbert I., Snijder E.J., Dimitrova M., Guillemot J.C., Lecine P., Canard B. The SARS-Coronavirus PLnc domain of nsp3 as a replication/transcription scaffolding protein. Virus Res. 2008;133:136–148.
    1. Jacobs J., Grum-Tokars V., Zhou Y., Turlington M., Saldanha S.A., Chase P., Eggler A., Dawson E.S., Baez-Santos Y.M., Tomar S., Mielech A.M., Baker S.C., Lindsley C.W., Hodder P., Mesecar A., Stauffer S.R. Discovery, synthesis, and structure-based optimization of a series of n-(tert-butyl)-2-(n-arylamido)-2-(pyridin-3-yl) acetamides (ML188) as potent noncovalent small molecule inhibitors of the severe acute respiratory syndrome coronavirus (SARS-CoV) 3CL protease. J. Med. Chem. 2013;56:534–546.
    1. Jain R.P., Pettersson H.I., Zhang J.M., Aull K.D., Fortin P.D., Huitema C., Eltis L.D., Parrish J.C., James M.N.G., Wishart D.S., Vederas J.C. Synthesis and evaluation of keto-glutamine analogues as potent inhibitors of severe acute respiratory syndrome 3CL(pro) J. Med. Chem. 2004;47:6113–6116.
    1. Johnson M.A., Chatterjee A., Neuman B.W., Wuthrich K. SARS coronavirus unique domain: three-domain molecular architecture in solution and RNA binding. J. Mol. Biol. 2010;400:724–742.
    1. Kanjanahaluethai A., Chen Z., Jukneliene D., Baker S.C. Membrane topology of murine coronavirus replicase nonstructural protein 3. Virology. 2007;361:391–401.
    1. Knoops K., Kikkert M., Worm S.H., Zevenhoven-Dobbe J.C., van der Meer Y., Koster A.J., Mommaas A.M., Snijder E.J. SARS-coronavirus replication is supported by a reticulovesicular network of modified endoplasmic reticulum. PLoS Biol. 2008;6:e226.
    1. Lei J., Mesters J.R., Drosten C., Anemuller S., Ma Q., Hilgenfeld R. Crystal structure of the papain-like protease of MERS coronavirus reveals unusual, potentially druggable active-site features. Antiviral Res. 2014;109:72–82.
    1. Lindner H.A., Fotouhi-Ardakani N., Lytvyn V., Lachance P., Sulea T., Menard R. The papain-like protease from the severe acute respiratory syndrome coronavirus is a deubiquitinating enzyme. J. Virol. 2005;79:15199–15208.
    1. Lindner H.A., Lytvyn V., Qi H., Lachance P., Ziomek E., Menard R. Selectivity in ISG15 and ubiquitin recognition by the SARS coronavirus papain-like protease. Arch. Biochem. Biophys. 2007;466:8–14.
    1. Love K.R., Catic A., Schlieker C., Ploegh H.L. Mechanisms, biology and inhibitors of deubiquitinating enzymes. Nat. Chem. Biol. 2007;3:697–705.
    1. McNaught A.D., Wilkinson A. second ed. IUPAC; 1997. Compendium of Chemical Terminology. (the “Gold Book”)
    1. Melandri F., Grenier L., Plamondon L., Huskey W.P., Stein R.L. Kinetic studies on the inhibition of isopeptidase T by ubiquitin aldehyde. Biochemistry. 1996;35:12893–12900.
    1. Mielech A.M., Chen Y., Mesecar A.D., Baker S.C. Nidovirus papain-like proteases: Multifunctional enzymes with protease, deubiquitinating and deISGylating activities. Virus Res. 2014
    1. Mielech A.M., Kilianski A., Baez-Santos Y.M., Mesecar A.D., Baker S.C. MERS-CoV papain-like protease has deISGylating and deubiquitinating activities. Virology. 2014;450–451:64–70.
    1. Molland K., Zhou Q., Mesecar A.D. A 2.2 Å resolution structure of the USP7 catalytic domain in a new space group elaborates upon structural rearrangements resulting from ubiquitin binding. Acta Crystallogr. F Struct. Biol. Commun. 2014;70:283–287.
    1. Neuman B.W., Joseph J.S., Saikatendu K.S., Serrano P., Chatterjee A., Johnson M.A., Liao L., Klaus J.P., Yates J.R., 3rd, Wuthrich K., Stevens R.C., Buchmeier M.J., Kuhn P. Proteomics analysis unravels the functional repertoire of coronavirus nonstructural protein 3. J. Virol. 2008;82:5279–5294.
    1. Nicholson B., Leach C.A., Goldenberg S.J., Francis D.M., Kodrasov M.P., Tian X., Shanks J., Sterner D.E., Bernal A., Mattern M.R., Wilkinson K.D., Butt T.R. Characterization of ubiquitin and ubiquitin-like-protein isopeptidase activities. Protein Sci. 2008;17:1035–1043.
    1. Oostra M., Hagemeijer M.C., van Gent M., Bekker C.P., te Lintelo E.G., Rottier P.J., de Haan C.A. Topology and membrane anchoring of the coronavirus replication complex: not all hydrophobic domains of nsp3 and nsp6 are membrane spanning. J. Virol. 2008;82:12392–12405.
    1. Oostra M., te Lintelo E.G., Deijs M., Verheije M.H., Rottier P.J., de Haan C.A. Localization and membrane topology of coronavirus nonstructural protein 4: involvement of the early secretory pathway in replication. J. Virol. 2007;81:12323–12336.
    1. Park J.Y., Jeong H.J., Kim J.H., Kim Y.M., Park S.J., Kim D., Park K.H., Lee W.S., Ryu Y.B. Diarylheptanoids from Alnus japonica inhibit papain-like protease of severe acute respiratory syndrome coronavirus. Biol. Pharm. Bull. 2012;35:2036–2042.
    1. Park J.Y., Kim J.H., Kim Y.M., Jeong H.J., Kim D.W., Park K.H., Kwon H.J., Park S.J., Lee W.S., Ryu Y.B. Tanshinones as selective and slow-binding inhibitors for SARS-CoV cysteine proteases. Bioorg. Med. Chem. 2012;20:5928–5935.
    1. Ratia K., Kilianski A., Baez-Santos Y.M., Baker S.C., Mesecar A.D. Structural basis for the ubiquitin-linkage specificity and deISGylating activity of SARS-CoV papain-like protease. PLoS Pathog. 2014;10:e1004113.
    1. Ratia K., Pegan S., Takayama J., Sleeman K., Coughlin M., Baliji S., Chaudhuri R., Fu W., Prabhakar B.S., Johnson M.E., Baker S.C., Ghosh A.K., Mesecar A.D. A noncovalent class of papain-like protease/deubiquitinase inhibitors blocks SARS virus replication. Proc. Natl. Acad. Sci. U.S.A. 2008;105:16119–16124.
    1. Ratia K., Saikatendu K.S., Santarsiero B.D., Barretto N., Baker S.C., Stevens R.C., Mesecar A.D. Severe acute respiratory syndrome coronavirus papain-like protease: structure of a viral deubiquitinating enzyme. Proc. Natl. Acad. Sci. U.S.A. 2006;103:5717–5722.
    1. Saikatendu K.S., Joseph J.S., Subramanian V., Clayton T., Griffith M., Moy K., Velasquez J., Neuman B.W., Buchmeier M.J., Stevens R.C., Kuhn P. Structural basis of severe acute respiratory syndrome coronavirus ADP-ribose-1″-phosphate dephosphorylation by a conserved domain of nsP3. Structure (Camb) 2005;13:1665–1675.
    1. Serrano P., Johnson M.A., Almeida M.S., Horst R., Herrmann T., Joseph J.S., Neuman B.W., Subramanian V., Saikatendu K.S., Buchmeier M.J., Stevens R.C., Kuhn P., Wuthrich K. Nuclear magnetic resonance structure of the N-terminal domain of nonstructural protein 3 from the severe acute respiratory syndrome coronavirus. J. Virol. 2007;81:12049–12060.
    1. Serrano P., Johnson M.A., Chatterjee A., Neuman B.W., Joseph J.S., Buchmeier M.J., Kuhn P., Wuthrich K. Nuclear magnetic resonance structure of the nucleic acid-binding domain of severe acute respiratory syndrome coronavirus nonstructural protein 3. J. Virol. 2009;83:12998–13008.
    1. Snijder E.J., Bredenbeek P.J., Dobbe J.C., Thiel V., Ziebuhr J., Poon L.L., Guan Y., Rozanov M., Spaan W.J., Gorbalenya A.E. Unique and conserved features of genome and proteome of SARS-coronavirus, an early split-off from the coronavirus group 2 lineage. J. Mol. Biol. 2003;331:991–1004.
    1. Sun L., Xing Y., Chen X., Zheng Y., Yang Y., Nichols D.B., Clementz M.A., Banach B.S., Li K., Baker S.C., Chen Z. Coronavirus papain-like proteases negatively regulate antiviral innate immune response through disruption of STING-mediated signaling. PLoS One. 2012;7:e30802.
    1. Tan J., Kusov Y., Mutschall D., Tech S., Nagarajan K., Hilgenfeld R., Schmidt C.L. The “SARS-unique domain” (SUD) of SARS coronavirus is an oligo(G)-binding protein. Biochem. Biophys. Res. Commun. 2007;364:877–882.
    1. Tan J., Vonrhein C., Smart O.S., Bricogne G., Bollati M., Kusov Y., Hansen G., Mesters J.R., Schmidt C.L., Hilgenfeld R. The SARS-unique domain (SUD) of SARS coronavirus contains two macrodomains that bind G-quadruplexes. PLoS Pathog. 2009;5:e1000428.
    1. Thiel V., Ivanov K.A., Putics A., Hertzig T., Schelle B., Bayer S., Weissbrich B., Snijder E.J., Rabenau H., Doerr H.W., Gorbalenya A.E., Ziebuhr J. Mechanisms and enzymes involved in SARS coronavirus genome expression. J. Gen. Virol. 2003;84:2305–2315.
    1. Tong T.R. Drug targets in severe acute respiratory syndrome (SARS) virus and other coronavirus infections. Infect. Disord. Drug Targets. 2009;9:223–245.
    1. van Hemert M.J., van den Worm S.H., Knoops K., Mommaas A.M., Gorbalenya A.E., Snijder E.J. SARS-coronavirus replication/transcription complexes are membrane-protected and need a host factor for activity in vitro. PLoS Pathog. 2008;4:e1000054.
    1. Wilkinson K.D., Gan-Erdene T., Kolli N. Derivitization of the C-terminus of ubiquitin and ubiquitin-like proteins using intein chemistry: methods and uses. Methods Enzymol. 2005;399:37–51.
    1. Xue X.Y., Yu H.W., Yang H.T., Xue F., Wu Z.X., Shen W., Li J., Zhou Z., Ding Y., Zhao Q., Zhang X.J.C., Liao M., Bartlam M., Rao Z. Structures of two coronavirus main proteases: Implications for substrate binding and antiviral drug design. J. Virol. 2008;82:2515–2527.
    1. Yang S., Chen S.J., Hsu M.F., Wu J.D., Tseng C.T.K., Liu Y.F., Chen H.C., Kuo C.W., Wu C.S., Chang L.W., Chen W.C., Liao S.Y., Chang T.Y., Hung H.H., Shr H.L., Liu C.Y., Huang Y.A., Chang L.Y., Hsu J.C., Peters C.J., Wang A.H.J., Hsu M.C. Synthesis, crystal structure, structure-activity relationships, and antiviral activity of a potent SARS coronavirus 3CL protease inhibitor. J. Med. Chem. 2006;49:4971–4980.
    1. Zhang J.M., Pettersson H.I., Huitema C., Niu C.Y., Yin J., James M.N.G., Eltis L.D., Vederas J.C. Design, synthesis, and evaluation of inhibitors for severe acute respiratory syndrome 3C-like protease based on phthalhydrazide ketones or heteroaromatic esters. J. Med. Chem. 2007;50:1850–1864.
    1. Ziebuhr J. Molecular biology of severe acute respiratory syndrome coronavirus. Curr. Opin. Microbiol. 2004;7:412–419.
    1. Ziebuhr J., Snijder E.J., Gorbalenya A.E. Virus-encoded proteinases and proteolytic processing in the Nidovirales. J. Gen. Virol. 2000;81:853–879.
    1. Ziebuhr J., Thiel V., Gorbalenya A.E. The autocatalytic release of a putative RNA virus transcription factor from its polyprotein precursor involves two paralogous papain-like proteases that cleave the same peptide bond. J. Biol. Chem. 2001;276:33220–33232.

Source: PubMed

3
Sottoscrivi