101
|
Kuo CJ, Shie JJ, Fang JM, Yen GR, Hsu JTA, Liu HG, Tseng SN, Chang SC, Lee CY, Shih SR, Liang PH. Design, synthesis, and evaluation of 3C protease inhibitors as anti-enterovirus 71 agents. Bioorg Med Chem 2008; 16:7388-98. [PMID: 18583140 PMCID: PMC7125518 DOI: 10.1016/j.bmc.2008.06.015] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 06/07/2008] [Accepted: 06/10/2008] [Indexed: 10/31/2022]
Abstract
Human enterovirus (EV) belongs to the picornavirus family, which consists of over 200 medically relevant viruses. A peptidomimetic inhibitor AG7088 was developed to inhibit the 3C protease of rhinovirus (a member of the family), a chymotrypsin-like protease required for viral replication, by forming a covalent bond with the active site Cys residue. In this study, we have prepared the recombinant 3C protease from EV71 (TW/2231/98), a particular strain which causes severe outbreaks in Asia, and developed inhibitors against the protease and the viral replication. For inhibitor design, the P3 group of AG7088, which is not interacting with the rhinovirus protease, was replaced with a series of cinnamoyl derivatives directly linked to P2 group through an amide bond to simplify the synthesis. While the replacement caused decreased potency, the activity can be largely improved by substituting the alpha,beta-unsaturated ester with an aldehyde at the P1' position. The best inhibitor 10b showed EC(50) of 18 nM without apparent toxicity (CC(50)>25 microM). Our study provides potent inhibitors of the EV71 3C protease as anti-EV71 agents and facilitates the combinatorial synthesis of derivatives for further improving the inhibitory activity.
Collapse
Key Words
- ev, enterovirus
- rv, rhinovirus
- sars-cov, severe acute respiratory syndrome-coronavirus
- ninta, nickel nitrilo-tri-acetic acid
- dabcyl, 4-(4-dimethylaminophenylazo)benzoic acid
- edans, 5-[(2-aminoethyl)amino]naphthalene-1-sulfonic acid
- boc, tert-butyloxycarbonyl
- cbz, benzyloxycarbonyl
- mes, 2-n-morpholono-ethanesulfonic acid
- dmem, dulbecco’s modified eagle’s medium
- fbs, fetal bovine serum
- protease
- picornaviridae
- inhibitor
- enterovirus
- computer modeling
- fluorogenic substrate
Collapse
Affiliation(s)
- Chih-Jung Kuo
- Institute of Biological Chemistry, Academia Sinica, 128 Academia Road, Taipei 11529, Taiwan
- Taiwan International Graduate Program, Academia Sinica, Taipei 11529, Taiwan
| | - Jiun-Jie Shie
- The Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Jim-Min Fang
- The Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan
| | - Guei-Rung Yen
- Division of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Chu-Nan, Taiwan
| | - John T.-A. Hsu
- Division of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Chu-Nan, Taiwan
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Hun-Ge Liu
- Institute of Biological Chemistry, Academia Sinica, 128 Academia Road, Taipei 11529, Taiwan
| | - Sung-Nain Tseng
- Department of Medical Biotechnology & Laboratory Science, Chang Gung University, Tao-Yuan, Taiwan
| | - Shih-Cheng Chang
- Department of Medical Biotechnology & Laboratory Science, Chang Gung University, Tao-Yuan, Taiwan
- Clinical Virology Laboratory, Department of Clinical Pathology, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Ching-Yin Lee
- Department of Medical Biotechnology & Laboratory Science, Chang Gung University, Tao-Yuan, Taiwan
- Clinical Virology Laboratory, Department of Clinical Pathology, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Shin-Ru Shih
- Department of Medical Biotechnology & Laboratory Science, Chang Gung University, Tao-Yuan, Taiwan
- Clinical Virology Laboratory, Department of Clinical Pathology, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Po-Huang Liang
- Institute of Biological Chemistry, Academia Sinica, 128 Academia Road, Taipei 11529, Taiwan
- Taiwan International Graduate Program, Academia Sinica, Taipei 11529, Taiwan
- The Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
102
|
Bacha U, Barrila J, Gabelli SB, Kiso Y, Mario Amzel L, Freire E. Development of broad-spectrum halomethyl ketone inhibitors against coronavirus main protease 3CL(pro). Chem Biol Drug Des 2008; 72:34-49. [PMID: 18611220 PMCID: PMC2597651 DOI: 10.1111/j.1747-0285.2008.00679.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2008] [Accepted: 05/27/2008] [Indexed: 11/28/2022]
Abstract
Coronaviruses comprise a large group of RNA viruses with diverse host specificity. The emergence of highly pathogenic strains like the SARS coronavirus (SARS-CoV), and the discovery of two new coronaviruses, NL-63 and HKU1, corroborates the high rate of mutation and recombination that have enabled them to cross species barriers and infect novel hosts. For that reason, the development of broad-spectrum antivirals that are effective against several members of this family is highly desirable. This goal can be accomplished by designing inhibitors against a target, such as the main protease 3CL(pro) (M(pro)), which is highly conserved among all coronaviruses. Here 3CL(pro) derived from the SARS-CoV was used as the primary target to identify a new class of inhibitors containing a halomethyl ketone warhead. The compounds are highly potent against SARS 3CL(pro) with K(i)'s as low as 300 nM. The crystal structure of the complex of one of the compounds with 3CL(pro) indicates that this inhibitor forms a thioether linkage between the halomethyl carbon of the warhead and the catalytic Cys 145. Furthermore, Structure Activity Relationship (SAR) studies of these compounds have led to the identification of a pharmacophore that accurately defines the essential molecular features required for the high affinity.
Collapse
Affiliation(s)
- Usman Bacha
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jennifer Barrila
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sandra B. Gabelli
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yoshiaki Kiso
- Department of Medicinal Chemistry, Center for Frontier Research in Medicinal Science, Kyoto Pharmaceutical University, Yamashina‐ku, Kyoto 607‐8412, Japan
| | - L. Mario Amzel
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ernesto Freire
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
103
|
Lau KM, Lee KM, Koon CM, Cheung CSF, Lau CP, Ho HM, Lee MYH, Au SWN, Cheng CHK, Lau CBS, Tsui SKW, Wan DCC, Waye MMY, Wong KB, Wong CK, Lam CWK, Leung PC, Fung KP. Immunomodulatory and anti-SARS activities of Houttuynia cordata. JOURNAL OF ETHNOPHARMACOLOGY 2008; 118:79-85. [PMID: 18479853 PMCID: PMC7126383 DOI: 10.1016/j.jep.2008.03.018] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2007] [Revised: 03/05/2008] [Accepted: 03/10/2008] [Indexed: 05/20/2023]
Abstract
BACKGROUND Severe acute respiratory syndrome (SARS) is a life-threatening form of pneumonia caused by SARS coronavirus (SARS-CoV). From late 2002 to mid 2003, it infected more than 8000 people worldwide, of which a majority of cases were found in China. Owing to the absence of definitive therapeutic Western medicines, Houttuynia cordata Thunb. (Saururaceae)(HC) was shortlisted by Chinese scientists to tackle SARS problem as it is conventionally used to treat pneumonia. AIM OF THE STUDY The present study aimed to explore the SARS-preventing mechanisms of HC in the immunological and anti-viral aspects. RESULTS Results showed that HC water extract could stimulate the proliferation of mouse splenic lymphocytes significantly and dose-dependently. By flow cytometry, it was revealed that HC increased the proportion of CD4(+) and CD8(+) T cells. Moreover, it caused a significant increase in the secretion of IL-2 and IL-10 by mouse splenic lymphocytes. In the anti-viral aspect, HC exhibited significant inhibitory effects on SARS-CoV 3C-like protease (3CL(pro)) and RNA-dependent RNA polymerase (RdRp). On the other hand, oral acute toxicity test demonstrated that HC was non-toxic to laboratory animals following oral administration at 16 g/kg. CONCLUSION The results of this study provided scientific data to support the efficient and safe use of HC to combat SARS.
Collapse
Affiliation(s)
- Kit-Man Lau
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Shi J, Sivaraman J, Song J. Mechanism for controlling the dimer-monomer switch and coupling dimerization to catalysis of the severe acute respiratory syndrome coronavirus 3C-like protease. J Virol 2008; 82:4620-9. [PMID: 18305031 PMCID: PMC2293028 DOI: 10.1128/jvi.02680-07] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2007] [Accepted: 02/20/2008] [Indexed: 01/07/2023] Open
Abstract
Unlike 3C protease, the severe acute respiratory syndrome coronavirus (SARS-CoV) 3C-like protease (3CLpro) is only enzymatically active as a homodimer and its catalysis is under extensive regulation by the unique extra domain. Despite intense studies, two puzzles still remain: (i) how the dimer-monomer switch is controlled and (ii) why dimerization is absolutely required for catalysis. Here we report the monomeric crystal structure of the SARS-CoV 3CLpro mutant R298A at a resolution of 1.75 A. Detailed analysis reveals that Arg298 serves as a key component for maintaining dimerization, and consequently, its mutation will trigger a cooperative switch from a dimer to a monomer. The monomeric enzyme is irreversibly inactivated because its catalytic machinery is frozen in the collapsed state, characteristic of the formation of a short 3(10)-helix from an active-site loop. Remarkably, dimerization appears to be coupled to catalysis in 3CLpro through the use of overlapped residues for two networks, one for dimerization and another for the catalysis.
Collapse
Affiliation(s)
- Jiahai Shi
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Republic of Singapore.
| | | | | |
Collapse
|
105
|
Shao YM, Yang WB, Kuo TH, Tsai KC, Lin CH, Yang AS, Liang PH, Wong CH. Design, synthesis, and evaluation of trifluoromethyl ketones as inhibitors of SARS-CoV 3CL protease. Bioorg Med Chem 2008; 16:4652-60. [PMID: 18329272 PMCID: PMC7127754 DOI: 10.1016/j.bmc.2008.02.040] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2007] [Revised: 02/10/2008] [Accepted: 02/11/2008] [Indexed: 11/18/2022]
Abstract
A series of trifluoromethyl ketones as SARS-CoV 3CL protease inhibitors was developed. The inhibitors were synthesized in four steps from commercially available compounds. Three different amino acids were explored in the P1-position and in the P2–P4 positions varying amino acids and long alkyl chain were incorporated. All inhibitors were evaluated in an in vitro assay using purified enzyme and fluorogenic substrate peptide. One of the inhibitors showed a time-dependent inhibition, with a Ki value of 0.3 μM after 4 h incubation.
Collapse
Affiliation(s)
- Yi-Ming Shao
- Genomics Research Center, Academia Sinica, No. 128, Section 2, Academia Road, Taipei 11529, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
106
|
Chen S, Zhang J, Hu T, Chen K, Jiang H, Shen X. Residues on the dimer interface of SARS coronavirus 3C-like protease: dimer stability characterization and enzyme catalytic activity analysis. J Biochem 2008; 143:525-36. [PMID: 18182387 PMCID: PMC7109808 DOI: 10.1093/jb/mvm246] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
3C-like protease (3CLpro) plays pivotal roles in the life cycle of severe acute respiratory syndrome coronavirus (SARS-CoV) and only the dimeric protease is proposed as the functional form. Guided by the crystal structure and molecular dynamics simulations, we performed systematic mutation analyses to identify residues critical for 3CLpro dimerization and activity in this study. Seven residues on the dimer interface were selected for evaluating their contributions to dimer stability and catalytic activity by biophysical and biochemical methods. These residues are involved in dimerization through hydrogen bonding and broadly located in the N-terminal finger, the α-helix A′ of domain I, and the oxyanion loop near the S1 substrate-binding subsite in domain II. We revealed that all seven single mutated proteases still have the dimeric species but the monomer–dimer equilibria of these mutants vary from each other, implying that these residues might contribute differently to the dimer stability. Such a conclusion could be further verified by the results that the proteolytic activities of these mutants also decrease to varying degrees. The present study would help us better understand the dimerization-activity relationship of SARS-CoV 3CLpro and afford potential information for designing anti-viral compounds targeting the dimer interface of the protease.
Collapse
Affiliation(s)
- Shuai Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, China
| | | | | | | | | | | |
Collapse
|
107
|
Lee CC, Kuo CJ, Hsu MF, Liang PH, Fang JM, Shie JJ, Wang AHJ. Structural basis of mercury- and zinc-conjugated complexes as SARS-CoV 3C-like protease inhibitors. FEBS Lett 2007; 581:5454-8. [PMID: 17981158 PMCID: PMC7094272 DOI: 10.1016/j.febslet.2007.10.048] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2007] [Revised: 10/25/2007] [Accepted: 10/25/2007] [Indexed: 01/07/2023]
Abstract
Five active metal-conjugated inhibitors (PMA, TDT, EPDTC, JMF1586 and JMF1600) bound with the 3C-like protease of severe acute respiratory syndrome (SARS)-associated coronavirus were analyzed crystallographically. The complex structures reveal two major inhibition modes: Hg(2+)-PMA is coordinated to C(44), M(49) and Y(54) with a square planar geometry at the S3 pocket, whereas each Zn(2+) of the four zinc-inhibitors is tetrahedrally coordinated to the H(41)-C(145) catalytic dyad. For anti-SARS drug design, this Zn(2+)-centered coordination pattern would serve as a starting platform for inhibitor optimization.
Collapse
Affiliation(s)
- Cheng-Chung Lee
- Structural Biology Program, Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 11221, Taiwan,Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan,National Core Facility of High-Throughput Protein Crystallography, Academia Sinica, Taipei 11529, Taiwan
| | - Chih-Jung Kuo
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan,Taiwan International Graduate Program, Academia Sinica, Taipei 11529, Taiwan
| | - Min-Feng Hsu
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan,National Core Facility of High-Throughput Protein Crystallography, Academia Sinica, Taipei 11529, Taiwan
| | - Po-Huang Liang
- National Core Facility of High-Throughput Protein Crystallography, Academia Sinica, Taipei 11529, Taiwan,The Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan,Taiwan International Graduate Program, Academia Sinica, Taipei 11529, Taiwan,Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Jim-Min Fang
- The Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan,Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Jiun-Jie Shie
- The Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Andrew H.-J. Wang
- Structural Biology Program, Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 11221, Taiwan,Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan,National Core Facility of High-Throughput Protein Crystallography, Academia Sinica, Taipei 11529, Taiwan,The Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan,Taiwan International Graduate Program, Academia Sinica, Taipei 11529, Taiwan,Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
108
|
Yang CW, Yang YN, Liang PH, Chen CM, Chen WL, Chang HY, Chao YS, Lee SJ. Novel small-molecule inhibitors of transmissible gastroenteritis virus. Antimicrob Agents Chemother 2007; 51:3924-31. [PMID: 17709464 PMCID: PMC2151441 DOI: 10.1128/aac.00408-07] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We used swine testicle (ST) cells infected with transmissible gastroenteritis virus (TGEV) and an indirect immunofluorescent assay with antibodies against TGEV spike and nucleocapsid proteins to screen small-molecule compounds that inhibit TGEV replication. Analogues of initial hits were collected and subjected to a 3CL protease (3CL(pro)) inhibition assay with recombinant 3CL(pro) and a fluorogenic peptide substrate. A series of benzothiazolium compounds were found to have inhibitory activity against TGEV 3CL(pro) and to exert anti-TGEV activities in terms of viral protein and RNA replication in TGEV-infected ST cells, with consequent protection of TGEV-infected ST cells from cytopathic effect by blocking the activation of caspase-3.
Collapse
Affiliation(s)
- Cheng-Wei Yang
- Division of Biotechnology and Pharmaceutical Research, National Health Research Institute, 35, Keyan Road, Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
109
|
Elston C, Wallach J, Saulnier J. New continuous and specific fluorometric assays for Pseudomonas aeruginosa elastase and LasA protease. Anal Biochem 2007; 368:87-94. [PMID: 17553454 DOI: 10.1016/j.ab.2007.04.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2007] [Revised: 04/23/2007] [Accepted: 04/23/2007] [Indexed: 11/19/2022]
Abstract
A highly sensitive assay based on new internally quenched fluorogenic peptide substrates has been developed for monitoring protease activities. These novel substrates comprise an Edans (5-(2-aminoethylamino)-1-naphthalenesulfonic acid) group at the C terminus and a Dabsyl (4-(dimethylamino)azobenzene-4'-sulfonyl chloride) fluorophore at the N terminus of the peptide chains. The Edans fluorescence increases upon peptide hydrolysis by Pseudomonas aeruginosa proteases, and this increase is directly proportional to the amount of substrate cleaved, i.e., protease activity. The substrates Dabsyl-Ala-Ala-Phe-Ala-Edans and Dabsyl-Leu-Gly-Gly-Gly-Ala-Edans were used for testing the peptidasic activities of P. aeruginosa elastase and LasA protease, respectively. Elastase and LasA kinetic parameters were calculated and a sensitive assay was designed for the detection of P. aeruginosa proteases in bacterial supernatants. The sensitivity and the small sample requirements make the assay suitable for high-throughput screening of biological samples. Furthermore, this P. aeruginosa protease assay improves upon existing assays because it is simple, it requires only one step, and even more significantly it is enzyme specific.
Collapse
Affiliation(s)
- Caroline Elston
- Laboratoire de Biochimie Analytique & Synthèse Bioorganique, UFR Chimie-Biochimie, Université Claude Bernard Lyon 1, 43 boulevard du 11 Novembre 1918, 69622 Villeurbanne Cedex, France
| | | | | |
Collapse
|
110
|
Wang HM, Liang PH. Pharmacophores and biological activities of severe acute respiratory syndrome viral protease inhibitors. Expert Opin Ther Pat 2007. [DOI: 10.1517/13543776.17.5.533] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
111
|
Grum-Tokars V, Ratia K, Begaye A, Baker SC, Mesecar AD. Evaluating the 3C-like protease activity of SARS-Coronavirus: recommendations for standardized assays for drug discovery. Virus Res 2007; 133:63-73. [PMID: 17397958 PMCID: PMC4036818 DOI: 10.1016/j.virusres.2007.02.015] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2006] [Revised: 01/24/2007] [Accepted: 02/19/2007] [Indexed: 01/28/2023]
Abstract
Although the initial outbreaks of the deadly coronavirus that causes severe acute respiratory syndrome (SARS-CoV) were controlled by public health measures, the development of vaccines and antiviral agents for SARS-CoV is essential for improving control and treatment of future outbreaks. One potential target for SARS-CoV antiviral drug development is the 3C-like protease (3CLpro). This enzyme is an attractive target since it is essential for viral replication, and since there are now a number of high resolution X-ray structures of SARS-CoV 3CLpro available making structure-based drug-design possible. As a result, SARS-CoV 3CLpro has become the focus of numerous drug discovery efforts worldwide, but as a consequence, a variety of different 3CLpro expression constructs and kinetic assays have been independently developed making evaluation and comparison between potential inhibitors problematic. Here, we review the literature focusing on different SARS-CoV 3CLpro expression constructs and assays used to measure enzymatic activity. Moreover, we provide experimental evidence showing that the activity of 3CLpro enzymatic is significantly reduced when non-native sequences or affinity-tags are added to the N- or C-termini of the enzyme, or when the enzyme used in assays is at concentrations below the equilibrium dissociation constant of the 3CLpro dimer. We demonstrate for the first time the utility of a highly sensitive and novel Alexa488-QSY7 FRET-based peptide substrate designed for routine analysis and high-throughput screening, and show that kinetic constants determined from FRET-based assays that are uncorrected for inner-filter effects can lead to artifacts. Finally, we evaluated the effects of common assay components including DTT, NaCl, EDTA and DMSO on enzymatic activity, and we recommend standardized assay conditions and constructs for routine SARS-CoV 3CLpro assays to facilitate direct comparisons between SARS-CoV 3CLpro inhibitors under development worldwide.
Collapse
Affiliation(s)
- Valerie Grum-Tokars
- Center for Pharmaceutical Biotechnology and Department of Medicinal Chemistry and Pharmacognosy, University of Illinois, Chicago, IL 60607, United States
| | | | | | | | | |
Collapse
|
112
|
Barrila J, Bacha U, Freire E. Long-range cooperative interactions modulate dimerization in SARS 3CLpro. Biochemistry 2007; 45:14908-16. [PMID: 17154528 PMCID: PMC2570436 DOI: 10.1021/bi0616302] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Severe acute respiratory syndrome (SARS) is an infectious disease caused by the human coronavirus, SARS-CoV. The main viral protease, SARS 3CLpro, is a validated target for the development of antiviral therapies. Since the enzyme is a homodimer and the individual monomers are inactive, two approaches are being used to develop inhibitors: enzyme activity inhibitors that target the active site and dimerization inhibitors. Dimerization inhibitors are usually targeted to the dimerization interface and need to compete with the attractive forces between subunits to be effective. In this paper, we show that the dimerization of SARS 3CLpro is also under allosteric control and that additional and energetically more favorable target sites away from the dimerization interface may also lead to subunit dissociation. We previously identified a cluster of conserved serine residues (Ser139, Ser144, and Ser147) located adjacent to the active site of 3CLpro that could effectively be targeted to inactivate the protease [Bacha, U et al. (2004) Biochemistry 43, 4906-4912]. Mutation of any of these serine residues to alanine had a debilitating effect on the catalytic activity of 3CLpro. In particular, the mutation of Ser147, which does not make any contact with the opposing subunit and is located approximately 9 A away from the dimer interface, totally inhibited dimerization and resulted in a complete loss of enzymatic activity. The finding that residues away from the dimer interface are able to control dimerization defines alternative targets for the design of dimerization inhibitors.
Collapse
Affiliation(s)
| | | | - Ernesto Freire
- All correspondence should be addressed to E. Freire, Department of Biology, The Johns Hopkins University, Baltimore, MD 21218; Phone (410) 516-7743; Fax (410) 516-6469; e-mail
| |
Collapse
|
113
|
Ghosh AK, Xi K, Johnson ME, Baker SC, Mesecar AD. Progress in Anti-SARS Coronavirus Chemistry, Biology and Chemotherapy. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2007; 41:183-196. [PMID: 19649165 PMCID: PMC2718771 DOI: 10.1016/s0065-7743(06)41011-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Proteolytic processing of the coronavirus replicase polyproteins is essential for ongoing viral ribonucleic acid (RNA) synthesis. Therefore, the severe acute respiratory syndrome (SARS)-coronaviruses (SARS-CoV) proteases are attractive targets for the development of antiviral drugs to reduce viral replication and pathogenicity. The structure and activity of the coronavirus 3C-like protease (3CLpro) has already been elucidated, and the design of inhibitors to 3CLpro as therapeutics has been proposed. The chapter discusses SARS-CoV 3CLpro inhibitors that include covalent inhibitors, noncovalent inhibitors, and inhibitors from screening. SARS-CoV papain-like protease (PLpro) is considered an equally viable target to 3CLpro for drug design because both are essential for viral replication. However, PLpro has likely not been pursued because of the paucity of structural information. Several compounds have been identified that have shown inhibitory activity against SARS-CoV. However, no information regarding their mechanism of action or the corresponding target is known. Glycyrrhizin showed inhibitory activity for SARS-CoV replication with EC50 = 300 mg/L after virus absorption in Vero cells. Some glycyrrhizin acid derivatives were found to inhibit SARS-CoV replication in vitro with EC50 values ranging from 5 to 50 μ M. Unfortunately, these compounds show high cytotoxity.
Collapse
Affiliation(s)
- Arun K Ghosh
- Departments of Chemistry and Medicinal Chemistry, Purdue University, West Lafayette, IN 47907
| | | | | | | | | |
Collapse
|
114
|
Chen L, Li J, Luo C, Liu H, Xu W, Chen G, Liew OW, Zhu W, Puah CM, Shen X, Jiang H. Binding interaction of quercetin-3-beta-galactoside and its synthetic derivatives with SARS-CoV 3CL(pro): structure-activity relationship studies reveal salient pharmacophore features. Bioorg Med Chem 2006; 14:8295-306. [PMID: 17046271 PMCID: PMC7125754 DOI: 10.1016/j.bmc.2006.09.014] [Citation(s) in RCA: 194] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Revised: 09/02/2006] [Accepted: 09/08/2006] [Indexed: 02/06/2023]
Abstract
The 3C-like protease (3CL(pro)) of severe acute respiratory syndrome-associated coronavirus (SARS-CoV) is one of the most promising targets for discovery of drugs against SARS, because of its critical role in the viral life cycle. In this study, a natural compound called quercetin-3-beta-galactoside was identified as an inhibitor of the protease by molecular docking, SPR/FRET-based bioassays, and mutagenesis studies. Both molecular modeling and Q189A mutation revealed that Gln189 plays a key role in the binding. Furthermore, experimental evidence showed that the secondary structure and enzymatic activity of SARS-CoV 3CL(pro) were not affected by the Q189A mutation. With the help of molecular modeling, eight new derivatives of the natural product were designed and synthesized. Bioassay results reveal salient features of the structure-activity relationship of the new compounds: (1) removal of the 7-hydroxy group of the quercetin moiety decreases the bioactivity of the derivatives; (2) acetoxylation of the sugar moiety abolishes inhibitor action; (3) introduction of a large sugar substituent on 7-hydroxy of quercetin can be tolerated; (4) replacement of the galactose moiety with other sugars does not affect inhibitor potency. This study not only reveals a new class of compounds as potential drug leads against the SARS virus, but also provides a solid understanding of the mechanism of inhibition against the target enzyme.
Collapse
Affiliation(s)
- Lili Chen
- Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Hamill P, Hudson D, Kao RY, Chow P, Raj M, Xu H, Richer MJ, Jean F. Development of a red-shifted fluorescence-based assay for SARS-coronavirus 3CL protease: identification of a novel class of anti-SARS agents from the tropical marine sponge Axinella corrugata. Biol Chem 2006; 387:1063-74. [PMID: 16895476 DOI: 10.1515/bc.2006.131] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
SARS-coronavirus (SARS-CoV) encodes a main protease, 3CLpro, which plays an essential role in the viral life cycle and is currently the prime target for discovering new anti-coronavirus agents. In this article, we report our success in developing a novel red-shifted (RS) fluorescence-based assay for 3CLpro and its application for identifying small-molecule anti-SARS agents from marine organisms. We have synthesised and characterised the first generation of a red-shifted internally quenched fluorogenic substrate (RS-IQFS) for 3CLpro based on resonance energy transfer between the donor and acceptor pair CAL Fluor Red 610 and Black Hole Quencher-1 (Km and kcat values of 14 microM and 0.65 min-1). The RS-IQFS primary sequence was selected based on the results of our screening analysis of 3CLpro performed using a series of blue-shifted (BS)-IQFSs corresponding to the 3CLpro-mediated cleavage junctions of the SARS-CoV polyproteins. In contrast to BS-IQFSs, the RS-IQFS was not susceptible to fluorescence interference from coloured samples and allowed for successful screening of marine natural products and identification of a coumarin derivative, esculetin-4-carboxylic acid ethyl ester, a novel 3CLpro inhibitor (IC50=46 microM) and anti-SARS agent (EC50=112 microM; median toxic concentration>800 microM) from the tropical marine sponge Axinella corrugata.
Collapse
Affiliation(s)
- Pamela Hamill
- Department of Microbiology and Immunology, Life Sciences Centre, University of British Columbia, 3559-2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | | | | | | | | | | | | | | |
Collapse
|
116
|
Lee TW, Cherney MM, Liu J, James KE, Powers JC, Eltis LD, James MNG. Crystal structures reveal an induced-fit binding of a substrate-like Aza-peptide epoxide to SARS coronavirus main peptidase. J Mol Biol 2006; 366:916-32. [PMID: 17196984 PMCID: PMC7094323 DOI: 10.1016/j.jmb.2006.11.078] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2006] [Revised: 11/18/2006] [Accepted: 11/28/2006] [Indexed: 01/06/2023]
Abstract
The SARS coronavirus main peptidase (SARS-CoV M(pro)) plays an essential role in the life-cycle of the virus and is a primary target for the development of anti-SARS agents. Here, we report the crystal structure of M(pro) at a resolution of 1.82 Angstroms, in space group P2(1) at pH 6.0. In contrast to the previously reported structure of M(pro) in the same space group at the same pH, the active sites and the S1 specificity pockets of both protomers in the structure of M(pro) reported here are in the catalytically competent conformation, suggesting their conformational flexibility. We report two crystal structures of M(pro) having an additional Ala at the N terminus of each protomer (M(+A(-1))(pro)), both at a resolution of 2.00 Angstroms, in space group P4(3)2(1)2: one unbound and one bound by a substrate-like aza-peptide epoxide (APE). In the unbound form, the active sites and the S1 specificity pockets of both protomers of M(+A(-1))(pro) are observed in a collapsed (catalytically incompetent) conformation; whereas they are in an open (catalytically competent) conformation in the APE-bound form. The observed conformational flexibility of the active sites and the S1 specificity pockets suggests that these parts of M(pro) exist in dynamic equilibrium. The structural data further suggest that the binding of APE to M(pro) follows an induced-fit model. The substrate likely also binds in an induced-fit manner in a process that may help drive the catalytic cycle.
Collapse
Affiliation(s)
- Ting-Wai Lee
- Group in Protein Structure and Function, Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | |
Collapse
|
117
|
Wu CY, King KY, Kuo CJ, Fang JM, Wu YT, Ho MY, Liao CL, Shie JJ, Liang PH, Wong CH. Stable benzotriazole esters as mechanism-based inactivators of the severe acute respiratory syndrome 3CL protease. ACTA ACUST UNITED AC 2006; 13:261-8. [PMID: 16638531 PMCID: PMC7111201 DOI: 10.1016/j.chembiol.2005.12.008] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Revised: 12/16/2005] [Accepted: 12/27/2005] [Indexed: 11/25/2022]
Abstract
Severe acute respiratory syndrome (SARS) is caused by a newly emerged coronavirus that infected more than 8000 individuals and resulted in more than 800 fatalities in 2003. Currently, there is no effective treatment for this epidemic. SARS-3CLpro has been shown to be essential for replication and is thus a target for drug discovery. Here, a class of stable benzotriazole esters was reported as mechanism-based inactivators of 3CLpro, and the most potent inactivator exhibited a kinact of 0.0011 s−1 and a Ki of 7.5 nM. Mechanistic investigation with kinetic and mass spectrometry analyses indicates that the active site Cys145 is acylated, and that no irreversible inactivation was observed with the use of the C145A mutant. In addition, a noncovalent, competitive inhibition became apparent by using benzotriazole ester surrogates in which the bridged ester-oxygen group is replaced with carbon.
Collapse
Affiliation(s)
- Chung-Yi Wu
- The Genomics Research Center and Institute of Biological Chemistry, Academia Sinica No. 128, Academia Road Section 2, Nan-Kang, Taipei, 115, Taiwan
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Ke-Yung King
- The Genomics Research Center and Institute of Biological Chemistry, Academia Sinica No. 128, Academia Road Section 2, Nan-Kang, Taipei, 115, Taiwan
| | - Chih-Jung Kuo
- The Genomics Research Center and Institute of Biological Chemistry, Academia Sinica No. 128, Academia Road Section 2, Nan-Kang, Taipei, 115, Taiwan
- Taiwan International Graduate Program, Academia Sinica, Nan-Kang, Taipei, 115, Taiwan
| | - Jim-Min Fang
- The Genomics Research Center and Institute of Biological Chemistry, Academia Sinica No. 128, Academia Road Section 2, Nan-Kang, Taipei, 115, Taiwan
- Department of Chemistry, National Taiwan University, Taipei, 106, Taiwan
| | - Ying-Ta Wu
- The Genomics Research Center and Institute of Biological Chemistry, Academia Sinica No. 128, Academia Road Section 2, Nan-Kang, Taipei, 115, Taiwan
| | - Ming-Yi Ho
- The Genomics Research Center and Institute of Biological Chemistry, Academia Sinica No. 128, Academia Road Section 2, Nan-Kang, Taipei, 115, Taiwan
| | - Chung-Lin Liao
- The Genomics Research Center and Institute of Biological Chemistry, Academia Sinica No. 128, Academia Road Section 2, Nan-Kang, Taipei, 115, Taiwan
| | - Jiun-Jie Shie
- The Genomics Research Center and Institute of Biological Chemistry, Academia Sinica No. 128, Academia Road Section 2, Nan-Kang, Taipei, 115, Taiwan
| | - Po-Huang Liang
- The Genomics Research Center and Institute of Biological Chemistry, Academia Sinica No. 128, Academia Road Section 2, Nan-Kang, Taipei, 115, Taiwan
- Ph: 886-2-27855696, ext. 6070; Fax: 886-2-27889759
| | - Chi-Huey Wong
- The Genomics Research Center and Institute of Biological Chemistry, Academia Sinica No. 128, Academia Road Section 2, Nan-Kang, Taipei, 115, Taiwan
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
- Department of Chemistry, National Taiwan University, Taipei, 106, Taiwan
- Ph: 858-784-2487; Fax: 858-784-2409
| |
Collapse
|
118
|
Sydnes MO, Hayashi Y, Sharma VK, Hamada T, Bacha U, Barrila J, Freire E, Kiso Y. Synthesis of glutamic acid and glutamine peptides possessing a trifluoromethyl ketone group as SARS-CoV 3CL protease inhibitors. Tetrahedron 2006; 62:8601-8609. [PMID: 32287416 PMCID: PMC7111793 DOI: 10.1016/j.tet.2006.06.052] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2006] [Revised: 06/13/2006] [Accepted: 06/14/2006] [Indexed: 12/21/2022]
Abstract
Trifluoromethyl-β-amino alcohol 11 [(4S)-tert-butyl 4-amino-6,6,6-trifluoro-5-hydroxyhexanoate] was synthesized in five steps starting from Cbz-l-Glu-OH 5 where the key step involved the introduction of the trifluoromethyl (CF3) group to oxazolidinone 7, resulting in the formation of silyl ether 8 [(4S,5S)-benzyl 4-(2-(tert-butoxycarbonyl)ethyl)-5-(trifluoromethyl)-5-(trimethylsilyloxy)oxazolidine-3-carboxylate]. Compound 11 was then converted into four tri- and tetra-glutamic acid and glutamine peptides (1-4) possessing a CF3-ketone group that exhibited inhibitory activity against severe acute respiratory syndrome coronavirus protease (SARS-CoV 3CLpro).
Collapse
Affiliation(s)
- Magne O Sydnes
- Department of Medicinal Chemistry, Center for Frontier Research in Medicinal Science, 21st Century COE Program, Kyoto Pharmaceutical University, Kyoto 607-8412, Japan
| | - Yoshio Hayashi
- Department of Medicinal Chemistry, Center for Frontier Research in Medicinal Science, 21st Century COE Program, Kyoto Pharmaceutical University, Kyoto 607-8412, Japan
| | - Vinay K Sharma
- Department of Medicinal Chemistry, Center for Frontier Research in Medicinal Science, 21st Century COE Program, Kyoto Pharmaceutical University, Kyoto 607-8412, Japan
| | - Takashi Hamada
- Department of Medicinal Chemistry, Center for Frontier Research in Medicinal Science, 21st Century COE Program, Kyoto Pharmaceutical University, Kyoto 607-8412, Japan
| | - Usman Bacha
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Jennifer Barrila
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Ernesto Freire
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Yoshiaki Kiso
- Department of Medicinal Chemistry, Center for Frontier Research in Medicinal Science, 21st Century COE Program, Kyoto Pharmaceutical University, Kyoto 607-8412, Japan
| |
Collapse
|
119
|
Graziano V, McGrath WJ, DeGruccio AM, Dunn JJ, Mangel WF. Enzymatic activity of the SARS coronavirus main proteinase dimer. FEBS Lett 2006; 580:2577-83. [PMID: 16647061 PMCID: PMC7094300 DOI: 10.1016/j.febslet.2006.04.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2005] [Revised: 03/23/2006] [Accepted: 04/03/2006] [Indexed: 02/01/2023]
Abstract
The enzymatic activity of the SARS coronavirus main proteinase dimer was characterized by a sensitive, quantitative assay. The new, fluorogenic substrate, (Ala‐Arg‐Leu‐Gln‐NH)2‐Rhodamine, contained a severe acute respiratory syndrome coronavirus (SARS CoV) main proteinase consensus cleavage sequence and Rhodamine 110, one of the most detectable compounds known, as the reporter group. The gene for the enzyme was cloned in the absence of purification tags, expressed in Escherichia coli and the enzyme purified. Enzyme activity from the SARS CoV main proteinase dimer could readily be detected at low pM concentrations. The enzyme exhibited a high Km, and is unusually sensitive to ionic strength and reducing agents.
Collapse
Affiliation(s)
- Vito Graziano
- Biology Department, Brookhaven National Laboratory, Upton, NY 11973, USA
| | - William J. McGrath
- Biology Department, Brookhaven National Laboratory, Upton, NY 11973, USA
| | | | - John J. Dunn
- Biology Department, Brookhaven National Laboratory, Upton, NY 11973, USA
| | - Walter F. Mangel
- Biology Department, Brookhaven National Laboratory, Upton, NY 11973, USA
| |
Collapse
|
120
|
Abstract
The 3C‐like protease of the severe acute respiratory syndrome (SARS) coronavirus has a C‐terminal extra domain in addition to the chymotrypsin‐fold adopted by piconavirus 3C proteases hosting the complete catalytic machinery. Previously we identified the extra domain to be involved in enzyme dimerization which has been considered essential for the catalytic activity. In an initial attempt to map out the extra‐domain residues critical for dimerization, we have systematically generated 15 point mutations, five deletions and one triple mutation and subsequently characterized them by enzymatic assay, dynamic light scattering, CD and NMR spectroscopy. The results led to identification of four regions critical for enzyme dimerization. Interestingly, Asn214Ala mutant with a significant tendency to form a monomer still retained ≈ 30% activity, indicating that the relationship between the activity and dimerization might be very complex. Very surprisingly, two regions (one over Ser284–Thr285–Ile286 and another around Phe291) were discovered on which Ala‐mutations significantly increased the enzymatic activities. Based on this, a super‐active triple‐mutant STI/A with a 3.7‐fold activity enhancement was thus engineered by mutating residues Ser284, Thr285 and Ile286 to Ala. The dynamic light scattering, CD and NMR characterizations indicate that the wild‐type (WT) and STI/A mutant share similar structural and dimerization properties, thus implying that in addition to dimerization, the extra domain might have other mechanisms to regulate the catalytic machinery. We rationalized these results based on the enzyme structure and consequently observed an interesting picture: the majority of the dimerization‐critical residues plus Ser284–Thr285–Ile286 and Phe291 are clustered together to form a nano‐scale channel passing through the central region of the enzyme. We therefore speculate that this channel might play a role in relaying regulatory effects from the extra domain to the catalytic machinery.
Collapse
Affiliation(s)
- Jiahai Shi
- Department of Biochemistry, The Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | |
Collapse
|
121
|
Weimer S, Oertel K, Fuchsbauer HL. A quenched fluorescent dipeptide for assaying dispase- and thermolysin-like proteases. Anal Biochem 2006; 352:110-9. [PMID: 16564490 DOI: 10.1016/j.ab.2006.02.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Revised: 02/22/2006] [Accepted: 02/26/2006] [Indexed: 11/23/2022]
Abstract
Metalloproteases such as dispase and thermolysin play a crucial role in the life cycle of bacteria. Commonly, they prefer hydrophobic amino acids at P1' of substrate proteins, thereby cleaving the peptide bond at the alpha amino group. Activity of such proteases has been measured by the use of tailor-made oligopeptides provided with fluorescence resonance energy transfer dyes. We can now show that the short dipeptide Dabcyl-Ser-Phe-EDANS is an appropriate substrate of dispase and thermolysin. It was cleaved by both enzymes at the single peptide bond accompanied by a steep increase in fluorescence. Substantial quenching effects of the formed products were observed only when more than 80microM substrate was hydrolyzed. High affinity of the proteases for the dipeptide resulted in low K(m) values of 91+/-9 and 104+/-18microM, which are comparable to those measured for longer peptides. Dabcyl-Ser-Phe-EDANS was also used to determine the pH and optimal temperature of dispase, which were found at pH 7.0 and 50 degrees C. Buffer substances such as acetate, citrate, and tris(hydroxymethyl)aminomethane had no significant effect on enzyme activity. Measurements up to 100 degrees C revealed that hydrolysis of the quenched fluorescent dipeptide took place only in the presence of active dispase.
Collapse
Affiliation(s)
- Stefanie Weimer
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, D-64287 Darmstadt, Germany
| | | | | |
Collapse
|
122
|
Chen LR, Wang YC, Lin YW, Chou SY, Chen SF, Liu LT, Wu YT, Kuo CJ, Chen TSS, Juang SH. Synthesis and evaluation of isatin derivatives as effective SARS coronavirus 3CL protease inhibitors. Bioorg Med Chem Lett 2005; 15:3058-62. [PMID: 15896959 PMCID: PMC7119080 DOI: 10.1016/j.bmcl.2005.04.027] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2005] [Revised: 04/08/2005] [Accepted: 04/14/2005] [Indexed: 01/13/2023]
Abstract
N-Substituted isatin derivatives were prepared from the reaction of isatin and various bromides via two steps. Bioactivity assay results (in vitro tests) demonstrated that some of these compounds are potent and selective inhibitors against SARS coronavirus 3CL protease with IC50 values ranging from 0.95 to 17.50 microM. Additionally, isatin 4o exhibited more potent inhibition for SARS coronavirus protease than for other proteases including papain, chymotrypsin, and trypsin.
Collapse
Affiliation(s)
- Li-Rung Chen
- Development Center for Biotechnology, 102, Lane 169, Kang Ning St., Xi Zhi 221, Taipei, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Hsu MF, Kuo CJ, Chang KT, Chang HC, Chou CC, Ko TP, Shr HL, Chang GG, Wang AHJ, Liang PH. Mechanism of the maturation process of SARS-CoV 3CL protease. J Biol Chem 2005; 280:31257-66. [PMID: 15788388 PMCID: PMC8062786 DOI: 10.1074/jbc.m502577200] [Citation(s) in RCA: 199] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2005] [Revised: 05/31/2005] [Indexed: 01/07/2023] Open
Abstract
Severe acute respiratory syndrome (SARS) is an emerging infectious disease caused by a novel human coronavirus. Viral maturation requires a main protease (3CL(pro)) to cleave the virus-encoded polyproteins. We report here that the 3CL(pro) containing additional N- and/or C-terminal segments of the polyprotein sequences undergoes autoprocessing and yields the mature protease in vitro. The dimeric three-dimensional structure of the C145A mutant protease shows that the active site of one protomer binds with the C-terminal six amino acids of the protomer from another asymmetric unit, mimicking the product-bound form and suggesting a possible mechanism for maturation. The P1 pocket of the active site binds the Gln side chain specifically, and the P2 and P4 sites are clustered together to accommodate large hydrophobic side chains. The tagged C145A mutant protein served as a substrate for the wild-type protease, and the N terminus was first digested (55-fold faster) at the Gln(-1)-Ser1 site followed by the C-terminal cleavage at the Gln306-Gly307 site. Analytical ultracentrifuge of the quaternary structures of the tagged and mature proteases reveals the remarkably tighter dimer formation for the mature enzyme (K(d) = 0.35 nm) than for the mutant (C145A) containing 10 extra N-terminal (K(d) = 17.2 nM) or C-terminal amino acids (K(d) = 5.6 nM). The data indicate that immature 3CL(pro) can form dimer enabling it to undergo autoprocessing to yield the mature enzyme, which further serves as a seed for facilitated maturation. Taken together, this study provides insights into the maturation process of the SARS 3CL(pro) from the polyprotein and design of new structure-based inhibitors.
Collapse
Affiliation(s)
- Min-Feng Hsu
- Institute of Biochemical Sciences, National Taiwan University, Taipei 106
| | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Shie JJ, Fang JM, Kuo TH, Kuo CJ, Liang PH, Huang HJ, Wu YT, Jan JT, Cheng YSE, Wong CH. Inhibition of the severe acute respiratory syndrome 3CL protease by peptidomimetic alpha,beta-unsaturated esters. Bioorg Med Chem 2005; 13:5240-52. [PMID: 15994085 PMCID: PMC7119063 DOI: 10.1016/j.bmc.2005.05.065] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2005] [Revised: 05/26/2005] [Accepted: 05/26/2005] [Indexed: 11/06/2022]
Abstract
The proteolytic processing of polyproteins by the 3CL protease of severe acute respiratory syndrome coronavirus is essential for the viral propagation. A series of tripeptide alpha,beta-unsaturated esters and ketomethylene isosteres, including AG7088, are synthesized and assayed to target the 3CL protease. Though AG7088 is inactive (IC50 > 100 microM), the ketomethylene isosteres and tripeptide alpha,beta-unsaturated esters containing both P1 and P2 phenylalanine residues show modest inhibitory activity (IC50 = 11-39 microM). The Phe-Phe dipeptide inhibitors 18a-e are designed on the basis of computer modeling of the enzyme-inhibitor complex. The most potent inhibitor 18c with an inhibition constant of 0.52 microM is obtained by condensation of the Phe-Phe dipeptide alpha,beta-unsaturated ester with 4-(dimethylamino)cinnamic acid. The cell-based assays also indicate that 18c is a nontoxic anti-SARS agent with an EC50 value of 0.18 microM.
Collapse
Affiliation(s)
- Jiun-Jie Shie
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan
| | - Jim-Min Fang
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan
- The Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Tun-Hsun Kuo
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Chih-Jung Kuo
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Po-Huang Liang
- The Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Hung-Jyun Huang
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan
| | - Yin-Ta Wu
- The Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Jia-Tsrong Jan
- Institute of Preventive Medicine, National Defense University, Taipei, Taiwan
| | | | - Chi-Huey Wong
- The Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
125
|
Liu YC, Huang V, Chao TC, Hsiao CD, Lin A, Chang MF, Chow LP. Screening of drugs by FRET analysis identifies inhibitors of SARS-CoV 3CL protease. Biochem Biophys Res Commun 2005; 333:194-9. [PMID: 15950190 PMCID: PMC7092914 DOI: 10.1016/j.bbrc.2005.05.095] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2005] [Accepted: 05/14/2005] [Indexed: 11/20/2022]
Abstract
SARS-CoV 3CL protease is essential for viral protein processing and is regarded as a good drug target to prevent SARS-CoV replication. In the present study, we established a high-throughput FRET technique for screening for anti-SARS-CoV 3CL protease drugs. Of a thousand existing drugs examined, hexachlorophene was identified as the most potent in inhibiting SARS-CoV 3CL protease. Further characterization showed that it was effective at micromolar concentrations (K(i) = 4 microM). The binding mode was competitive, and the inhibitory effect was dependent on preincubation time. Two other drugs, triclosan and nelfinavir, were about 10 times less potent. The structure-based search and biological evaluation of various hexachlorophene analogues were described. These analogues gave optimal inhibitory activity against SARS-CoV 3CL protease with IC(50) values ranging from 7.6 to 84.5 microM. Optimization of hexachlorophene analogues was shown to provide several active 3CL protease inhibitors that function as potential anti-SARS agents.
Collapse
Affiliation(s)
- Yu-Chih Liu
- Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 106, Taiwan
| | - Vicky Huang
- Department of Biochemical Pharmacology, MDS Pharma Services, Taipei 112, Taiwan
| | - Ti-Chun Chao
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Chwan-Deng Hsiao
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Atsui Lin
- Department of Biochemical Pharmacology, MDS Pharma Services, Taipei 112, Taiwan
| | - Ming-Fu Chang
- Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 106, Taiwan
| | - Lu-Ping Chow
- Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 106, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 106, Taiwan
- Corresponding author. Fax: +886 2 23958814.
| |
Collapse
|
126
|
Ding L, Zhang XX, Wei P, Fan K, Lai L. The interaction between severe acute respiratory syndrome coronavirus 3C-like proteinase and a dimeric inhibitor by capillary electrophoresis. Anal Biochem 2005; 343:159-65. [PMID: 15935325 PMCID: PMC7094366 DOI: 10.1016/j.ab.2005.04.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2005] [Revised: 04/12/2005] [Accepted: 04/18/2005] [Indexed: 01/28/2023]
Abstract
3C-like proteinase of severe acute respiratory syndrome (SARS) coronavirus has been demonstrated to be a key target for drug design against SARS. The interaction between SARS coronavirus 3C-like (3CL) proteinase and an octapeptide interface inhibitor was studied by affinity capillary electrophoresis (ACE). The binding constants were estimated by the change of migration time of the analytes in the buffer solution containing different concentrations of SARS 3CL proteinase. The results showed that SARS 3CL proteinase was able to complex with the octapeptide competitively, with binding constants of 2.44 x 10(4) M(-1) at 20 degrees C and 2.11 x 10(4)M(-1) at 37 degrees C. In addition, the thermodynamic parameters deduced reveal that hydrophobic interaction might play major roles, along with electrostatic force, in the binding process. The ACE method used here could be developed to be an effective and simple way of applying large-scale drug screening and evaluation.
Collapse
Affiliation(s)
- Li Ding
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | | | | | | | | |
Collapse
|
127
|
Chen L, Gui C, Luo X, Yang Q, Günther S, Scandella E, Drosten C, Bai D, He X, Ludewig B, Chen J, Luo H, Yang Y, Yang Y, Zou J, Thiel V, Chen K, Shen J, Shen X, Jiang H. Cinanserin is an inhibitor of the 3C-like proteinase of severe acute respiratory syndrome coronavirus and strongly reduces virus replication in vitro. J Virol 2005; 79:7095-103. [PMID: 15890949 PMCID: PMC1112131 DOI: 10.1128/jvi.79.11.7095-7103.2005] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The 3C-like proteinase (3CLpro) of severe acute respiratory syndrome-associated coronavirus (SARS-CoV) is one of the most promising targets for anti-SARS-CoV drugs due to its crucial role in the viral life cycle. In this study, a database containing structural information of more than 8,000 existing drugs was virtually screened by a docking approach to identify potential binding molecules of SARS-CoV 3CLpro. As a target for screening, both a homology model and the crystallographic structure of the binding pocket of the enzyme were used. Cinanserin (SQ 10,643), a well-characterized serotonin antagonist that has undergone preliminary clinical testing in humans in the 1960s, showed a high score in the screening and was chosen for further experimental evaluation. Binding of both cinanserin and its hydrochloride to bacterially expressed 3CLpro of SARS-CoV and the related human coronavirus 229E (HCoV-229E) was demonstrated by surface plasmon resonance technology. The catalytic activity of both enzymes was inhibited with 50% inhibitory concentration (IC50) values of 5 microM, as tested with a fluorogenic substrate. The antiviral activity of cinanserin was further evaluated in tissue culture assays, namely, a replicon system based on HCoV-229E and quantitative test assays with infectious SARS-CoV and HCoV-229E. All assays revealed a strong inhibition of coronavirus replication at nontoxic drug concentrations. The level of virus RNA and infectious particles was reduced by up to 4 log units, with IC50 values ranging from 19 to 34 microM. These findings demonstrate that the old drug cinanserin is an inhibitor of SARS-CoV replication, acting most likely via inhibition of the 3CL proteinase.
Collapse
Affiliation(s)
- Lili Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Fan K, Ma L, Han X, Liang H, Wei P, Liu Y, Lai L. The substrate specificity of SARS coronavirus 3C-like proteinase. Biochem Biophys Res Commun 2005; 329:934-40. [PMID: 15752746 PMCID: PMC7092912 DOI: 10.1016/j.bbrc.2005.02.061] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2005] [Indexed: 11/21/2022]
Abstract
The 3C-like proteinase of severe acute respiratory syndrome coronavirus (SARS) has been proposed to be a key target for structural based drug design against SARS. We have designed and synthesized 34 peptide substrates and determined their hydrolysis activities. The conserved core sequence of the native cleavage site is optimized for high hydrolysis activity. Residues at position P4, P3, and P3′ are critical for substrate recognition and binding, and increment of β-sheet conformation tendency is also helpful. A comparative molecular field analysis (CoMFA) model was constructed. Based on the mutation data and CoMFA model, a multiply mutated octapeptide S24 was designed for higher activity. The experimentally determined hydrolysis activity of S24 is the highest in all designed substrates and is close to that predicted by CoMFA. These results offer helpful information for the research on the mechanism of substrate recognition of coronavirus 3C-like proteinase.
Collapse
Affiliation(s)
- Keqiang Fan
- State Key Laboratory of Structural Chemistry of Stable and Unstable Species, College of Chemistry, Peking University, Beijing 100871, China
| | | | | | | | | | | | | |
Collapse
|
129
|
Abstract
Severe acute respiratory syndrome (SARS) is a disease that has newly emerged in the 21st century, and is both severe and highly contagious. SARS first surfaced in late 2002 and spread within a few months from its origin in Guandong province, China, to more than 30 countries (World Health Organization, 2003). In this review, several antiviral substances shown to be active in vitro will be introduced and summarized in the order of the virus' replication steps; that is, binding to cellular receptor, fusion and entry to the cells, viral RNA replication and transcription, protein processing and so on. The possible clinical use of several synthetic peptides, including those that mimic the S-binding domain, the HR2 fusion protein and SARS proteinase substrates, will be discussed. Monoclonal antibodies (Mabs) and established drugs, such as interferons and HIV proteinase inhibitors, are also discussed in relation to anti-SARS clinical use.
Collapse
Affiliation(s)
- Shiro Shigeta
- Department of Microbiology, Fukushima Medical University, School of Medicine, Fukushima, Japan.
| | | |
Collapse
|
130
|
Hsu WC, Chang HC, Chou CY, Tsai PJ, Lin PI, Chang GG. Critical assessment of important regions in the subunit association and catalytic action of the severe acute respiratory syndrome coronavirus main protease. J Biol Chem 2005; 280:22741-8. [PMID: 15831489 PMCID: PMC8060872 DOI: 10.1074/jbc.m502556200] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The severe acute respiratory syndrome (SARS) coronavirus (CoV) main protease represents an attractive target for the development of novel anti-SARS agents. The tertiary structure of the protease consists of two distinct folds. One is the N-terminal chymotrypsin-like fold that consists of two structural domains and constitutes the catalytic machinery; the other is the C-terminal helical domain, which has an unclear function and is not found in other RNA virus main proteases. To understand the functional roles of the two structural parts of the SARS-CoV main protease, we generated the full-length of this enzyme as well as several terminally truncated forms, different from each other only by the number of amino acid residues at the C- or N-terminal regions. The quaternary structure and K(d) value of the protease were analyzed by analytical ultracentrifugation. The results showed that the N-terminal 1-3 amino acid-truncated protease maintains 76% of enzyme activity and that the major form is a dimer, as in the wild type. However, the amino acids 1-4-truncated protease showed the major form to be a monomer and had little enzyme activity. As a result, the fourth amino acid seemed to have a powerful effect on the quaternary structure and activity of this protease. The last C-terminal helically truncated protease also exhibited a greater tendency to form monomer and showed little activity. We concluded that both the C- and the N-terminal regions influence the dimerization and enzyme activity of the SARS-CoV main protease.
Collapse
Affiliation(s)
- Wen-Chi Hsu
- Faculty of Life Sciences, Institute of Biochemistry, and Structural Biology Program, National Yang-Ming University, Taipei 112, Taiwan
| | | | | | | | | | | |
Collapse
|
131
|
Chen S, Chen LL, Luo HB, Sun T, Chen J, Ye F, Cai JH, Shen JK, Shen X, Jiang HL. Enzymatic activity characterization of SARS coronavirus 3C-like protease by fluorescence resonance energy transfer technique. Acta Pharmacol Sin 2005; 26:99-106. [PMID: 15659121 PMCID: PMC7091904 DOI: 10.1111/j.1745-7254.2005.00010.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Aim: To characterize enzymatic activity of severe acute respiratory syndrome (SARS) coronavirus (CoV) 3C-like protease (3CLpro) and its four site-directed mutants. Methods: Based on the fluorescence resonance energy transfer (FRET) principle using 5-[(2′-aminoethyl)-amino] naphthelenesulfonic acid (EDANS) and 4-[[4-(dimethylamino) phenyl] azo] benzoic acid (Dabcyl) as the energy transfer pair, one fluorogenic substrate was designed for the evaluation of SARS-CoV 3CLpro proteolytic activity. Results: The kinetic parameters of the fluorogenic substrate have been determined as Km=404 μmol·L−1, kcat=1.08 min−1, and kcat/Km=2.7 mmol−1·L·min−1. SARS-CoV 3CLpro showed substantial pH and temperature-triggered activity switches, and site-directed mutagenesis analysis of SARS-CoV 3CLpro revealed that substitutions of His41, Cys145, and His163 resulted in complete loss of enzymatic activity, while replacement of Met162 with Ala caused strongly increased activity. Conclusion: This present work has provided valuable information for understanding the catalytic mechanism of SARS-CoV 3CLpro. This FRET-based assay might supply an ideal approach for the exploration SARS-CoV 3CLpro putative inhibitors.
Collapse
Affiliation(s)
- Shuai Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, 201203 China
| | - Li-li Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, 201203 China
| | - Hai-bin Luo
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, 201203 China
| | - Tao Sun
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, 201203 China
| | - Jing Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, 201203 China
| | - Fei Ye
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, 201203 China
| | - Jian-hua Cai
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, 201203 China
| | - Jing-kang Shen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, 201203 China
| | - Xu Shen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, 201203 China
| | - Hua-liang Jiang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, 201203 China
| |
Collapse
|
132
|
Kao RY, To APC, Ng LWY, Tsui WHW, Lee TSW, Tsoi HW, Yuen KY. Characterization of SARS-CoV main protease and identification of biologically active small molecule inhibitors using a continuous fluorescence-based assay. FEBS Lett 2004; 576:325-30. [PMID: 15498556 PMCID: PMC7134596 DOI: 10.1016/j.febslet.2004.09.026] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2004] [Revised: 09/09/2004] [Accepted: 09/13/2004] [Indexed: 11/25/2022]
Abstract
Severe acute respiratory syndrome associated coronavirus main protease (SARS‐CoV Mpro) has been proposed as a prime target for anti‐SARS drug development. We have cloned and overexpressed the SARS‐CoV Mpro in Escherichia coli, and purified the recombinant Mpro to homogeneity. The kinetic parameters of the recombinant SARS‐CoV Mpro were characterized by high performance liquid chromatography‐based assay and continuous fluorescence‐based assay. Two novel small molecule inhibitors of the SARS‐CoV Mpro were identified by high‐throughput screening using an internally quenched fluorogenic substrate. The identified inhibitors have Ki values at low μM range with comparable anti‐SARS‐CoV activity in cell‐based assays.
Collapse
Affiliation(s)
- Richard Y Kao
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | | | | | | | | | | | | |
Collapse
|
133
|
Chen S, Chen L, Tan J, Chen J, Du L, Sun T, Shen J, Chen K, Jiang H, Shen X. Severe acute respiratory syndrome coronavirus 3C-like proteinase N terminus is indispensable for proteolytic activity but not for enzyme dimerization. Biochemical and thermodynamic investigation in conjunction with molecular dynamics simulations. J Biol Chem 2004; 280:164-73. [PMID: 15507456 PMCID: PMC7982548 DOI: 10.1074/jbc.m408211200] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Severe acute respiratory syndrome (SARS) coronavirus is a novel human coronavirus and is responsible for SARS infection. SARS coronavirus 3C-like proteinase (SARS 3CLpro) plays key roles in viral replication and transcription and is an attractive target for anti-SARS drug discovery. In this report, we quantitatively characterized the dimerization features of the full-length and N-terminal residues 1–7 deleted SARS 3CLpros by using glutaraldehyde cross-linking SDS-PAGE, size-exclusion chromatography, and isothermal titration calorimeter techniques. Glutaraldehyde cross-linking SDS-PAGE and size-exclusion chromatography results show that, similar to the full-length SARS 3CLpro, the N-terminal deleted SARS 3CLpro still remains a dimer/monomer mixture within a wide range of protein concentrations. Isothermal titration calorimeter determinations indicate that the equilibrium dissociation constant (Kd) of the N-terminal deleted proteinase dimer (262 μm) is very similar to that of the full-length proteinase dimer (227 μm). Enzymatic activity assay using the fluorescence resonance energy transfer method reveals that N-terminal deletion results in almost complete loss of enzymatic activity for SARS 3CLpro. Molecular dynamics and docking simulations demonstrate the N-terminal deleted proteinase dimer adopts a state different from that of the full-length proteinase dimer, which increases the angle between the two protomers and reduces the binding pocket that is not beneficial to the substrate binding. This conclusion is verified by the surface plasmon resonance biosensor determination, indicating that the model substrate cannot bind to the N-terminal deleted proteinase. These results suggest the N terminus is not indispensable for the proteinase dimerization but may fix the dimer at the active state and is therefore vital to enzymatic activity.
Collapse
Affiliation(s)
- Shuai Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Graduate School of Chinese Academy of Sciences, Shanghai 201203, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Hsu JTA, Kuo CJ, Hsieh HP, Wang YC, Huang KK, Lin CPC, Huang PF, Chen X, Liang PH. Evaluation of metal-conjugated compounds as inhibitors of 3CL protease of SARS-CoV. FEBS Lett 2004; 574:116-20. [PMID: 15358550 PMCID: PMC7134601 DOI: 10.1016/j.febslet.2004.08.015] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2004] [Revised: 08/06/2004] [Accepted: 08/10/2004] [Indexed: 11/21/2022]
Abstract
3C-like (3CL) protease is essential for the life cycle of severe acute respiratory syndrome-coronavirus (SARS-CoV) and therefore represents a key anti-viral target. A compound library consisting of 960 commercially available drugs and biologically active substances was screened for inhibition of SARS-CoV 3CL protease. Potent inhibition was achieved using the mercury-containing compounds thimerosal and phenylmercuric acetate, as well as hexachlorophene. As well, 1-10 microM of each compound inhibited viral replication in Vero E6 cell culture. Detailed mechanism studies using a fluorescence-based protease assay demonstrated that the three compounds acted as competitive inhibitors (Ki=0.7, 2.4, and 13.7 microM for phenylmercuric acetate, thimerosal, and hexachlorophene, respectively). A panel of metal ions including Zn2+ and its conjugates were then evaluated for their anti-3CL protease activities. Inhibition was more pronounced using a zinc-conjugated compound (1-hydroxypyridine-2-thione zinc; Ki=0.17 microM) than using the ion alone (Ki=1.1 microM).
Collapse
Affiliation(s)
- John T.-A. Hsu
- Division of Biotechnology and Pharmaceutical Research, National Health Research Institutes, National Defense Medical Center, Taipei 115, Taiwan, ROC
| | - Chih-Jung Kuo
- Institute of Biological Chemistry, Academia Sinica, National Defense Medical Center, Taipei 115, Taiwan, ROC
| | - Hsing-Pang Hsieh
- Division of Biotechnology and Pharmaceutical Research, National Health Research Institutes, National Defense Medical Center, Taipei 115, Taiwan, ROC
| | - Yeau-Ching Wang
- Institute of Preventive Medicine, National Defense Medical Center, Taipei 115, Taiwan, ROC
| | - Kuo-Kuei Huang
- Division of Biotechnology and Pharmaceutical Research, National Health Research Institutes, National Defense Medical Center, Taipei 115, Taiwan, ROC
| | - Coney P.-C. Lin
- Division of Biotechnology and Pharmaceutical Research, National Health Research Institutes, National Defense Medical Center, Taipei 115, Taiwan, ROC
| | - Ping-Fang Huang
- Institute of Biological Chemistry, Academia Sinica, National Defense Medical Center, Taipei 115, Taiwan, ROC
| | - Xin Chen
- Division of Biotechnology and Pharmaceutical Research, National Health Research Institutes, National Defense Medical Center, Taipei 115, Taiwan, ROC
| | - Po-Huang Liang
- Institute of Biological Chemistry, Academia Sinica, National Defense Medical Center, Taipei 115, Taiwan, ROC
| |
Collapse
|
135
|
Wu CY, Jan JT, Ma SH, Kuo CJ, Juan HF, Cheng YSE, Hsu HH, Huang HC, Wu D, Brik A, Liang FS, Liu RS, Fang JM, Chen ST, Liang PH, Wong CH. Small molecules targeting severe acute respiratory syndrome human coronavirus. Proc Natl Acad Sci U S A 2004; 101:10012-7. [PMID: 15226499 PMCID: PMC454157 DOI: 10.1073/pnas.0403596101] [Citation(s) in RCA: 366] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Severe acute respiratory syndrome (SARS) is an infectious disease caused by a novel human coronavirus. Currently, no effective antiviral agents exist against this type of virus. A cell-based assay, with SARS virus and Vero E6 cells, was developed to screen existing drugs, natural products, and synthetic compounds to identify effective anti-SARS agents. Of >10,000 agents tested, approximately 50 compounds were found active at 10 microM; among these compounds, two are existing drugs (Reserpine 13 and Aescin 5) and several are in clinical development. These 50 active compounds were tested again, and compounds 2-6, 10, and 13 showed active at 3 microM. The 50% inhibitory concentrations for the inhibition of viral replication (EC(50)) and host growth (CC(50)) were then measured and the selectivity index (SI = CC(50)/EC(50)) was determined. The EC(50), based on ELISA, and SI for Reserpine, Aescim, and Valinomycin are 3.4 microM (SI = 7.3), 6.0 microM (SI = 2.5), and 0.85 microM (SI = 80), respectively. Additional studies were carried out to further understand the mode of action of some active compounds, including ELISA, Western blot analysis, immunofluorescence and flow cytometry assays, and inhibition against the 3CL protease and viral entry. Of particular interest are the two anti-HIV agents, one as an entry blocker and the other as a 3CL protease inhibitor (K(i) = 0.6 microM).
Collapse
Affiliation(s)
- Chung-Yi Wu
- Genomics Research Center and Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|