1
|
Sarkar A, Bhaskara Rao KV. Unraveling anticancer potential of a novel serine protease inhibitor from marine yeast Candida parapsilosis ABS1 against colorectal and breast cancer cells. World J Microbiol Biotechnol 2023; 39:225. [PMID: 37296286 DOI: 10.1007/s11274-023-03670-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023]
Abstract
The study was planned to isolate a serine protease inhibitor compound with anticancer potential against colorectal and breast cancer cells from marine yeast. Protease enzymes play a crucial role in the mechanism of life-threatening diseases like cancer, malaria and AIDS. Hence, blocking these enzymes with potential inhibitors can be an efficient approach in drug therapy for these diseases. A total of 12 marine yeast isolates, recovered from mangrove swamps of Sundarbans, India, showed inhibition activity against trypsin. The yeast isolate ABS1 showed highest inhibition activity (89%). The optimum conditions for protease inhibitor production were found to be glucose, ammonium phosphate, pH 7.0, 30 °C and 2 M NaCl. The PI protein from yeast isolate ABS1 was purified using ethyl acetate extraction and anion exchange chromatography. The purified protein was characterized using denaturing SDS-PAGE, Liquid Chromatography Electrospray Ionization Mass Spectrometry (LC-ESI-MS), Reverse Phase High Pressure Liquid Chromatography (RP-HPLC) and Fourier Transform Infra-red Spectroscopy (FTIR) analysis. The intact molecular weight of the PI protein was determined to be 25.584 kDa. The PI protein was further studied for in vitro anticancer activities. The IC50 value for MTT cell proliferation assay was found to be 43 µg/ml against colorectal cancer HCT15 cells and 48 µg/ml against breast cancer MCF7 cells. Hoechst staining, DAPI staining and DNA fragmentation assay were performed to check the apoptotic cells. The marine yeast was identified as Candida parapsilosis ABS1 (Accession No. MH782231) using 18s rRNA sequencing.
Collapse
Affiliation(s)
- Anwesha Sarkar
- Marine Biotechnology Laboratory, Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - K V Bhaskara Rao
- Marine Biotechnology Laboratory, Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| |
Collapse
|
2
|
Agbowuro AA, Huston WM, Gamble AB, Tyndall JDA. Proteases and protease inhibitors in infectious diseases. Med Res Rev 2017; 38:1295-1331. [PMID: 29149530 DOI: 10.1002/med.21475] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 09/10/2017] [Accepted: 10/17/2017] [Indexed: 12/31/2022]
Abstract
There are numerous proteases of pathogenic organisms that are currently targeted for therapeutic intervention along with many that are seen as potential drug targets. This review discusses the chemical and biological makeup of some key druggable proteases expressed by the five major classes of disease causing agents, namely bacteria, viruses, fungi, eukaryotes, and prions. While a few of these enzymes including HIV protease and HCV NS3-4A protease have been targeted to a clinically useful level, a number are yet to yield any clinical outcomes in terms of antimicrobial therapy. A significant aspect of this review discusses the chemical and pharmacological characteristics of inhibitors of the various proteases discussed. A total of 25 inhibitors have been considered potent and safe enough to be trialed in humans and are at different levels of clinical application. We assess the mechanism of action and clinical performance of the protease inhibitors against infectious agents with their developmental strategies and look to the next frontiers in the use of protease inhibitors as anti-infective agents.
Collapse
Affiliation(s)
| | - Wilhelmina M Huston
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, Australia
| | - Allan B Gamble
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
3
|
Abstract
Viruses are major pathogenic agents that can cause a variety of diseases, such as AIDS, hepatitis, respiratory diseases, and many more, in humans, plants, and animals. The most prominent of them have been adenoviruses, alphaviruses, flaviviruses, hepatitis C virus, herpesviruses, human immunodeficiency virus of type 1, and picornaviruses. This chapter presents an introductory remark on such viruses, mechanisms of their invasion, and diseases related to them. The inhibition of these viruses is of great concern to human beings. Each of these viruses encodes one or more proteases that play crucial roles in their replication, and thus they are important targets for the design and development of potent antiviral agents. The chapter, therefore, also introduces the readers to such proteases and their structures and functions. This chapter is thus a prelude to the remaining chapters in the book, which present in detail about the different viruses and their proteases.
Collapse
Affiliation(s)
- Anjana Sharma
- Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh, India
| | - Satya P. Gupta
- National Institute of Technical Teachers’ Training and Research, Bhopal, Madhya Pradesh, India
| |
Collapse
|
4
|
Humber DC, Bamford MJ, Bethell RC, Cammack N, Orr DC, Storer R, Weingarten GG, Wyatt PG. Synthesis and Structure Activity Relationships of a Series of Penicillin-Derived Pseudosymmetric Inhibitors of HIV-1 Proteinase. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/095632029400500311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The synthesis from penicillin G of a series of potent pseudosymmetric inhibitors 11 a-k of HIV-1 proteinase is described. The 2-pyridyl substituted compounds 11a and 11j showed improved antiviral activity compared to their C2-symmetric counterparts 3 and 4.
Collapse
Affiliation(s)
- D. C. Humber
- Departments of Medicinal Chemistry II, Glaxo Group Research Limited, Greenford, Middlesex, UB6 0HE, UK
| | - M. J. Bamford
- Departments of Medicinal Chemistry II, Glaxo Group Research Limited, Greenford, Middlesex, UB6 0HE, UK
| | - R. C. Bethell
- Departments of Virology, Glaxo Group Research Limited, Greenford, Middlesex, UB6 0HE, UK
| | - N. Cammack
- Departments of Virology, Glaxo Group Research Limited, Greenford, Middlesex, UB6 0HE, UK
| | - D. C. Orr
- Departments of Virology, Glaxo Group Research Limited, Greenford, Middlesex, UB6 0HE, UK
| | - R. Storer
- Departments of Medicinal Chemistry II, Glaxo Group Research Limited, Greenford, Middlesex, UB6 0HE, UK
| | - G. G. Weingarten
- Departments of Medicinal Chemistry II, Glaxo Group Research Limited, Greenford, Middlesex, UB6 0HE, UK
| | - P. G. Wyatt
- Departments of Medicinal Chemistry II, Glaxo Group Research Limited, Greenford, Middlesex, UB6 0HE, UK
| |
Collapse
|
5
|
Affiliation(s)
- B. M. Dunn
- Department of Biochemistry and Molecular Biology, J. Hillis Miller Health Center, University of Florida, Gainesville, Florida 32610, USA
| | - J. Kay
- Department of Biochemistry, University of Wales, College of Cardiff, PO Box 903, Cardiff CF1 1ST, UK
| |
Collapse
|
6
|
Billich A, Billich S, Rosenwirth B. Assay Systems for HIV-1 Proteinase and Their Use for Evaluation of Inhibitors. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/095632029100200201] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- A. Billich
- Sandoz Forschungsinstitut GmbH, Department of AntiRetroviral Therapy, Brunnerstr. 59, A-1235 Vienna, Austria
| | - S. Billich
- Sandoz Forschungsinstitut GmbH, Department of AntiRetroviral Therapy, Brunnerstr. 59, A-1235 Vienna, Austria
| | - B. Rosenwirth
- Sandoz Forschungsinstitut GmbH, Department of AntiRetroviral Therapy, Brunnerstr. 59, A-1235 Vienna, Austria
| |
Collapse
|
7
|
Abstract
Human rhinoviruses (HRV) are the major etiological agents of the common cold and asthma exacerbations, with significant worldwide health and economic impact. Although large-scale population vaccination has proved successful in limiting or even eradicating many viruses, the more than 100 distinct serotypes mean that conventional vaccination is not a feasible strategy to combat HRV. An alternative strategy is to target conserved viral proteins such as the HRV proteases, 2A(pro) and 3C(pro), the focus of this review. Necessary for host cell shutoff, virus replication, and pathogenesis, 2A(pro) and 3C(pro) are clearly viable drug targets, and indeed, 3C(pro) has been successfully targeted for treating the common cold in experimental infection. 2A(pro) and 3C(pro) are crucial for virus replication due to their role in polyprotein processing as well as cleavage of key cellular proteins to inhibit cellular transcription and translation. Intriguingly, the action of the HRV proteases also disrupts nucleocytoplasmic trafficking, contributing to HRV cytopathic effects. Improved understanding of the protease-cell interactions should enable new therapeutic approaches to be identified for drug development.
Collapse
Affiliation(s)
- Lora M Jensen
- Faculty of Education, Science, Technology and Mathematics, Centre for Research in Therapeutic Solutions, University of Canberra, 1 Kirinari Street, Bruce, Canberra, ACT, 2601, Australia
| | | | | | | |
Collapse
|
8
|
Verdaguer N, Ferrero D, Murthy MRN. Viruses and viral proteins. IUCRJ 2014; 1:492-504. [PMID: 25485129 PMCID: PMC4224467 DOI: 10.1107/s205225251402003x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 09/04/2014] [Indexed: 05/30/2023]
Abstract
For more than 30 years X-ray crystallography has been by far the most powerful approach for determining the structures of viruses and viral proteins at atomic resolution. The information provided by these structures, which covers many important aspects of the viral life cycle such as cell-receptor recognition, viral entry, nucleic acid transfer and genome replication, has extensively enriched our vision of the virus world. Many of the structures available correspond to potential targets for antiviral drugs against important human pathogens. This article provides an overview of the current knowledge of different structural aspects of the above-mentioned processes.
Collapse
Affiliation(s)
- Nuria Verdaguer
- Institut de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, Baldiri i Reixac 15, 08028-Barcelona, Spain
| | - Diego Ferrero
- Institut de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, Baldiri i Reixac 15, 08028-Barcelona, Spain
| | - Mathur R. N. Murthy
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560 012, India
| |
Collapse
|
9
|
First generation inhibitors of the adenovirus proteinase. FEBS Lett 2013; 587:2332-9. [PMID: 23711373 DOI: 10.1016/j.febslet.2013.05.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 05/07/2013] [Accepted: 05/10/2013] [Indexed: 11/22/2022]
Abstract
As there are more than 50 adenovirus serotypes, the likelihood of developing an effective vaccine is low. Here we describe inhibitors of the adenovirus proteinase (AVP) with the ultimate objective of developing anti-adenovirus agents. Inhibitors were identified via structure-based drug design using as druggable sites the active site and a conserved cofactor pocket in the crystal structures of AVP. A lead compound was identified that had an IC50 of 18 μM. One of eight structural derivatives of the lead compound had an IC50 of 140 nM against AVP and an IC50 of 490 nM against the AVP with its cofactor bound.
Collapse
|
10
|
Rout MK, Reddy JG, Phillips M, Hosur RV. Single point mutation induced alterations in the equilibrium structural transitions on the folding landscape of HIV-1 protease. J Biomol Struct Dyn 2012; 31:684-93. [PMID: 22909351 DOI: 10.1080/07391102.2012.707459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Equilibrium folding-unfolding transitions are hard to study in HIV-1 protease (PR) because of its autolytic properties. Further, the protease exhibits many tolerant point mutations some of which also impart drug resistance to the protein. It is conceivable that the mutations affect protein's function by altering its folding characteristics; these would clearly depend on the nature of the mutations themselves. In this background, we report here NMR studies on the effects of D25 N mutation, which removes one negative charge from the protein at the active site, on the equilibrium folding behaviour of PR starting from its acetic acid denatured state. It is observed that in PRD25N two slowly exchanging conformations are present at the N-terminal. One of them is similar to that of PR. Though the conformational and dynamics preferences of PR and PRD25N are fairly similar in 9 M acetic acid, they seem to undergo different folding transitions when acetic acid concentration is reduced. The differences are seen in the active site, in the flap, and in the hinge of the flap regions. The present study suggests that such differences, though different in detail, would occur for other mutations as well, and also for different initial denatured states. These would have significant regulatory implications for the efficacy of protease function.
Collapse
Affiliation(s)
- Manoj Kumar Rout
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai, 400005, India
| | | | | | | |
Collapse
|
11
|
Kay J, Dunn BM. Substrate specificity and inhibitors of aspartic proteinases. Scandinavian Journal of Clinical and Laboratory Investigation 2011. [DOI: 10.1080/00365519209104651] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
12
|
Pinho RT, Beltramini LM, Alves CR, De-Simone SG. Trypanosoma cruzi: Isolation and characterization of aspartyl proteases. Exp Parasitol 2009; 122:128-33. [DOI: 10.1016/j.exppara.2009.02.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Revised: 08/18/2008] [Accepted: 02/06/2009] [Indexed: 11/28/2022]
|
13
|
Fluctuating partially native-like topologies in the acid denatured ensemble of autolysis resistant HIV-1 protease. Arch Biochem Biophys 2009; 482:33-41. [DOI: 10.1016/j.abb.2008.11.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Revised: 11/20/2008] [Accepted: 11/24/2008] [Indexed: 11/18/2022]
|
14
|
Deszcz L, Cencic R, Sousa C, Kuechler E, Skern T. An antiviral peptide inhibitor that is active against picornavirus 2A proteinases but not cellular caspases. J Virol 2006; 80:9619-27. [PMID: 16973565 PMCID: PMC1617246 DOI: 10.1128/jvi.00612-06] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The replication of many viruses is absolutely dependent on proteolytic cleavage. Infected cells also use this biological mechanism to induce programmed cell death in response to viral infection. Specific inhibitors for both viral and cellular proteases are therefore of vital importance. We have recently shown that the general caspase inhibitor zVAD.fmk inhibits not only caspases, but also the 2Apro of human rhinoviruses (HRVs) (L. Deszcz, J. Seipelt, E. Vassilieva, A. Roetzer, and E. Kuechler, FEBS Lett. 560:51-55, 2004). Here, we describe a derivative of zVAD.fmk that inhibits HRV2 2Apro but that has no effect on caspase 9. This gain in specificity was achieved by replacing the aspartic acid of zVAD.fmk with methionine to generate zVAM.fmk. Methionine was chosen because an oligopeptide with methionine at the P1 position was a much better substrate than an oligopeptide with an alanine residue, which is found at the P1 position of the wild-type HRV2 2Apro cleavage site. zVAM.fmk inhibits the replication of HRV type 2 (HRV2), HRV14, and HRV16. In contrast to zVAD.fmk, however, zVAM.fmk did not inhibit apoptosis induced by puromycin in HeLa cells. zVAM.fmk inhibited in vitro the intermolecular cleavage of eukaryotic initiation factor 4GI (eIF4GI) by HRV2 2Apro at nanomolar concentrations. However, much higher concentrations of zVAM.fmk were required to inhibit HRV14 2Apro cleavage of eIF4GI. In contrast, intramolecular self-processing of HRV14 2Apro was much more susceptible to inhibition by zVAM.fmk than that of HRV2 2Apro, suggesting that zVAM.fmk inhibits HRV2 and HRV14 replication by targeting different reactions of the same proteinase.
Collapse
Affiliation(s)
- Luiza Deszcz
- Max F. Perutz Laboratories, Medical University of Vienna, Dr. Bohr-Gasse 9/3, A-1030 Vienna, Austria
| | | | | | | | | |
Collapse
|
15
|
Byrd CM, Hruby DE. Vaccinia virus proteolysis--a review. Rev Med Virol 2006; 16:187-202. [PMID: 16710840 PMCID: PMC7169229 DOI: 10.1002/rmv.499] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2005] [Revised: 01/24/2006] [Accepted: 01/24/2006] [Indexed: 01/15/2023]
Abstract
It is well known that viruses, as obligate intracellular parasites, must use their hosts' metabolic machinery in order to replicate their genomes and form infectious progeny virions. What is less well known are the details of how viruses make sure that once all the necessary proteins are made, that they assume the correct configuration at the proper time in order to catalyse the efficient assembly of infectious virions. One of the methods employed by viruses to regulate this process is the proteolytic cleavage of viral proteins. Over the past several decades, studies in numerous laboratories have demonstrated that morphogenic proteolysis plays a major and essential role during the assembly and maturation of infectious poxvirus virions. In this review we describe the history of vaccinia virus proteolysis as a prototypic viral system.
Collapse
Affiliation(s)
| | - Dennis E. Hruby
- SIGA Technologies, Inc., Corvallis, Oregon 97333, USA
- Department of Microbiology, Oregon State University, Corvallis, Oregon 97331, USA
| |
Collapse
|
16
|
Abstract
During antiviral drug development, any essential stage of the viral life cycle can serve as a potential drug target. Since most viruses encode specific proteases whose cleavage activity is required for viral replication, and whose structure and activity are unique to the virus and not the host cell, these enzymes make excellent targets for drug development. Success using this approach has been demonstrated with the plethora of protease inhibitors approved for use against HIV. This discussion is designed to review the field of antiviral drug development, focusing on the search for protease inhibitors, while highlighting some of the challenges encountered along the way. Protease inhibitor drug discovery efforts highlighting progress made with HIV, HCV, HRV, and vaccinia virus as a model system are included. Drug Dev. Res. 67:501–510, 2006. © 2006 Wiley‐Liss, Inc.
Collapse
Affiliation(s)
| | - Dennis E. Hruby
- SIGA Technologies, Inc., Corvallis, Oregon
- Department of Microbiology, Oregon State University, Corvallis, Oregon
| |
Collapse
|
17
|
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: 4.3] [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
|
18
|
Bernard D, Méhul B, Thomas-Collignon A, Delattre C, Donovan M, Schmidt R. Identification and Characterization of a Novel Retroviral-Like Aspartic Protease Specifically Expressed in Human Epidermis. J Invest Dermatol 2005; 125:278-87. [PMID: 16098038 DOI: 10.1111/j.0022-202x.2005.23816.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Proteases play a pivotal role in epidermal differentiation and desquamation. Separation of a total protein extract from human reconstructed epidermis by two-dimensional gel electrophoresis and subsequent peptide analysis of a specific protein spot identified a new protein exhibiting similarities with the retroviral aspartic protease family. Cloning of the corresponding full-length cDNA revealed an open reading frame encoding for a new protease of 343 amino acids, containing a putative aspartic protease catalytic domain. We named this protein Skin ASpartic Protease (SASPase). RT-PCR and northern blot analysis of various human tissues revealed that SASPase was specifically expressed within the epidermis. Immunohistochemical analysis showed a particularly intense expression restricted to the granular layers, whereas in diseased skin, its expression was changed. Western blot analysis, using a monoclonal antibody, revealed the expression of two forms of the enzyme: a 28 kDa putative proform and the active 14 kDa form. Recombinant truncated SASPase (SASP28) was generated from a prokaryotic expression system in Escherichia coli as a fusion protein with GST. SASP28 degraded insulin and to a lesser extent casein with a pH optimum of 5. As seen for retroviral proteases, an auto-activation processing was evidenced, generating a 14 kDa protein (SASP14). Site-directed mutagenesis inhibited auto-activation of the enzyme. Indinavir, a potent HIV protease inhibitor used in AIDS therapy, had a significant inhibitory effect on rSASPase auto-activation, which could explain its side effects on skin.
Collapse
|
19
|
Düzgüneş N, Simões S, Slepushkin V, Pretzer E, Flasher D, Salem II, Steffan G, Konopka K, Pedroso de Lima MC. Delivery of antiviral agents in liposomes. Methods Enzymol 2005; 391:351-73. [PMID: 15721391 DOI: 10.1016/s0076-6879(05)91020-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The intracellular activity of certain antiviral agents, including antisense oligonucleotides, acyclic nucleoside phosphonates, and protease inhibitors, is enhanced when they are delivered in liposome-encapsulated form. In this chapter we describe the preparation of pH-sensitive liposomes encapsulating antisense oligonucleotides, ribozymes, and acyclic nucleoside phosphonate analogues and their effects on HIV replication in macrophages. We outline the use of liposomal HIV protease inhibitors in infected macrophages. We present two methods for the covalent coupling of soluble CD4 to liposomes and show the association of these liposomes with HIV-infected cells. We also describe the synthesis of a novel antiviral agent based on cyclodextrin and its incorporation into liposomes.
Collapse
Affiliation(s)
- Nejat Düzgüneş
- Department of Microbiology, Arthur A. Dugoni School of Dentistry, University of the Pacific, San Francisco, CA 94115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Lee KW, Hwang SY, Kim CR, Nam DH, Chang JH, Choi SC, Choi BS, Choi HW, Lee KK, So B, Cho SW, Shin H. The Chemical Development of LB71350. Org Process Res Dev 2003. [DOI: 10.1021/op034062w] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Kyu Woong Lee
- Chemical Development Division, LG Life Sciences, Ltd./R&D, 104-1, Moonji-dong, Yusong-gu, Daejeon 305-380, Korea
| | - Sang Yeul Hwang
- Chemical Development Division, LG Life Sciences, Ltd./R&D, 104-1, Moonji-dong, Yusong-gu, Daejeon 305-380, Korea
| | - Chung Ryeol Kim
- Chemical Development Division, LG Life Sciences, Ltd./R&D, 104-1, Moonji-dong, Yusong-gu, Daejeon 305-380, Korea
| | - Do Hyun Nam
- Chemical Development Division, LG Life Sciences, Ltd./R&D, 104-1, Moonji-dong, Yusong-gu, Daejeon 305-380, Korea
| | - Jay Hyok Chang
- Chemical Development Division, LG Life Sciences, Ltd./R&D, 104-1, Moonji-dong, Yusong-gu, Daejeon 305-380, Korea
| | - Sang Chul Choi
- Chemical Development Division, LG Life Sciences, Ltd./R&D, 104-1, Moonji-dong, Yusong-gu, Daejeon 305-380, Korea
| | - Bo Seung Choi
- Chemical Development Division, LG Life Sciences, Ltd./R&D, 104-1, Moonji-dong, Yusong-gu, Daejeon 305-380, Korea
| | - Hyeong-wook Choi
- Chemical Development Division, LG Life Sciences, Ltd./R&D, 104-1, Moonji-dong, Yusong-gu, Daejeon 305-380, Korea
| | - Ki Kon Lee
- Chemical Development Division, LG Life Sciences, Ltd./R&D, 104-1, Moonji-dong, Yusong-gu, Daejeon 305-380, Korea
| | - Byungran So
- Chemical Development Division, LG Life Sciences, Ltd./R&D, 104-1, Moonji-dong, Yusong-gu, Daejeon 305-380, Korea
| | - Sung Wook Cho
- Chemical Development Division, LG Life Sciences, Ltd./R&D, 104-1, Moonji-dong, Yusong-gu, Daejeon 305-380, Korea
| | - Hyunik Shin
- Chemical Development Division, LG Life Sciences, Ltd./R&D, 104-1, Moonji-dong, Yusong-gu, Daejeon 305-380, Korea
| |
Collapse
|
21
|
Affiliation(s)
- Liang Tong
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA.
| |
Collapse
|
22
|
Deo SK, Lewis JC, Daunert S. Bioluminescence detection of proteolytic bond cleavage by using recombinant aequorin. Anal Biochem 2000; 281:87-94. [PMID: 10847614 DOI: 10.1006/abio.2000.4539] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Detection of proteolytic bond cleavage was achieved by taking advantage of the bioluminescence emission generated by the photoprotein aequorin. A genetically engineered HIV-1 protease substrate was coupled with a cysteine-free mutant of aequorin by employing the polymerase chain reaction to produce a fusion protein that incorporates an optimum natural protease cleavage site. The fusion protein was immobilized on a solid phase and employed as the substrate for the HIV-1 protease. Proteolytic bond cleavage was detected by a decrease in the bioluminescence generated by the aequorin fusion protein on the solid phase. A dose-response curve for HIV-1 protease was constructed by relating the decrease in bioluminescence signal with varying amounts of the protease. The system was also used to evaluate two competitive and one noncompetitive inhibitor of the HIV-1 protease. Among the advantages of this assay is that by using recombinant methods a complete bioluminescently labeled protease recognition site can be designed and produced. The assay yields very sensitive detection limits, which are inherent to bioluminescence-based methods. An application of this system may be in the high-throughput screening of biopharmaceutical drugs that are potential inhibitors of a target protease.
Collapse
Affiliation(s)
- S K Deo
- Department of Chemistry, University of Kentucky, Lexington 40506, USA
| | | | | |
Collapse
|
23
|
Patick AK, Binford SL, Brothers MA, Jackson RL, Ford CE, Diem MD, Maldonado F, Dragovich PS, Zhou R, Prins TJ, Fuhrman SA, Meador JW, Zalman LS, Matthews DA, Worland ST. In vitro antiviral activity of AG7088, a potent inhibitor of human rhinovirus 3C protease. Antimicrob Agents Chemother 1999; 43:2444-50. [PMID: 10508022 PMCID: PMC89498 DOI: 10.1128/aac.43.10.2444] [Citation(s) in RCA: 167] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/1999] [Accepted: 07/15/1999] [Indexed: 11/20/2022] Open
Abstract
AG7088 is a potent, irreversible inhibitor of human rhinovirus (HRV) 3C protease (inactivation rate constant (k(obs)/[I]) = 1,470,000 +/- 440,000 M(-1) s(-1) for HRV 14) that was discovered by protein structure-based drug design methodologies. In H1-HeLa and MRC-5 cell protection assays, AG7088 inhibited the replication of all HRV serotypes (48 of 48) tested with a mean 50% effective concentration (EC(50)) of 0.023 microM (range, 0.003 to 0.081 microM) and a mean EC(90) of 0.082 microM (range, 0.018 to 0.261 microM) as well as that of related picornaviruses including coxsackieviruses A21 and B3, enterovirus 70, and echovirus 11. No significant reductions in the antiviral activity of AG7088 were observed when assays were performed in the presence of alpha(1)-acid glycoprotein or mucin, proteins present in nasal secretions. The 50% cytotoxic concentration of AG7088 was >1,000 microM, yielding a therapeutic index of >12,346 to >333,333. In a single-cycle, time-of-addition assay, AG7088 demonstrated antiviral activity when added up to 6 h after infection. In contrast, a compound targeting viral attachment and/or uncoating was effective only when added at the initiation of virus infection. Direct inhibition of 3C proteolytic activity in infected cells treated with AG7088 was demonstrated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis of radiolabeled proteins, which showed a dose-dependent accumulation of viral precursor polyproteins and reduction of processed protein products. The broad spectrum of antiviral activity of AG7088, combined with its efficacy even when added late in the virus life cycle, highlights the advantages of 3C protease as a target and suggests that AG7088 will be a promising clinical candidate.
Collapse
Affiliation(s)
- A K Patick
- Agouron Pharmaceuticals, Inc., San Diego, California 92121, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Matthews DA, Dragovich PS, Webber SE, Fuhrman SA, Patick AK, Zalman LS, Hendrickson TF, Love RA, Prins TJ, Marakovits JT, Zhou R, Tikhe J, Ford CE, Meador JW, Ferre RA, Brown EL, Binford SL, Brothers MA, DeLisle DM, Worland ST. Structure-assisted design of mechanism-based irreversible inhibitors of human rhinovirus 3C protease with potent antiviral activity against multiple rhinovirus serotypes. Proc Natl Acad Sci U S A 1999; 96:11000-7. [PMID: 10500114 PMCID: PMC34232 DOI: 10.1073/pnas.96.20.11000] [Citation(s) in RCA: 245] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human rhinoviruses, the most important etiologic agents of the common cold, are messenger-active single-stranded monocistronic RNA viruses that have evolved a highly complex cascade of proteolytic processing events to control viral gene expression and replication. Most maturation cleavages within the precursor polyprotein are mediated by rhinovirus 3C protease (or its immediate precursor, 3CD), a cysteine protease with a trypsin-like polypeptide fold. High-resolution crystal structures of the enzyme from three viral serotypes have been used for the design and elaboration of 3C protease inhibitors representing different structural and chemical classes. Inhibitors having alpha,beta-unsaturated carbonyl groups combined with peptidyl-binding elements specific for 3C protease undergo a Michael reaction mediated by nucleophilic addition of the enzyme's catalytic Cys-147, resulting in covalent-bond formation and irreversible inactivation of the viral protease. Direct inhibition of 3C proteolytic activity in virally infected cells treated with these compounds can be inferred from dose-dependent accumulations of viral precursor polyproteins as determined by SDS/PAGE analysis of radiolabeled proteins. Cocrystal-structure-assisted optimization of 3C-protease-directed Michael acceptors has yielded molecules having extremely rapid in vitro inactivation of the viral protease, potent antiviral activity against multiple rhinovirus serotypes and low cellular toxicity. Recently, one compound in this series, AG7088, has entered clinical trials.
Collapse
Affiliation(s)
- D A Matthews
- Agouron Pharmaceuticals, Inc., 3565 General Atomics Court, San Diego, CA 92121, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Seipelt J, Guarné A, Bergmann E, James M, Sommergruber W, Fita I, Skern T. The structures of picornaviral proteinases. Virus Res 1999; 62:159-68. [PMID: 10507325 DOI: 10.1016/s0168-1702(99)00043-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Picornaviruses are a family of positive-strand RNA viruses the members of which include poliovirus, hepatitis A virus, rhinovirus, foot-and-mouth disease virus and encephalomyocarditis virus. The genetic information contained in the single-stranded, positive sense RNA genome is expressed as a single protein of around 2000 amino acids. This primary product of protein synthesis, designated the polyprotein, is subsequently cleaved into the mature viral proteins by proteinases present within it. The properties of the three defined proteolytic activities present in the picornaviruses are reviewed and the three-dimensional structures of the hepatitis A 3C proteinase and the leader proteinase of foot-and-mouth disease virus as well as a model of the structure of the HRV2 2A proteinase are compared with those of chymotrypsin, papain and streptomyces griseus A proteinase, respectively.
Collapse
Affiliation(s)
- J Seipelt
- Institute of Biochemistry, Medical Faculty, University of Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
26
|
Sprietsma JE. Cysteine, glutathione (GSH) and zinc and copper ions together are effective, natural, intracellular inhibitors of (AIDS) viruses. Med Hypotheses 1999; 52:529-38. [PMID: 10459834 DOI: 10.1054/mehy.1997.0689] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Sufficient essential nutrients such as methionine, cysteine, copper, selenium, zinc and vitamins C and E are indispensable for the maintenance of optimal (immune) cell functions. Parasitic organisms such as protozoa, fungi, bacteria and viruses also depend on these essential nutrients for their multiplication and functioning. An evolutionarily developed optimal distribution of available nutrients between host (cells) and parasitic organisms normally prevents diseases, the nature of which will depend on genetic and environmental factors. The way in which the right amount of cysteine, glutathione (GSH), and copper and zinc ions made available in the right place at the right time and in the right form can prevent an unchecked multiplication of (AIDS) viruses in a more passive or active way forms the basis for the AIDS zinc-deficiency hypothesis (A-Z hypothesis) presented in this article. Zinc and copper ions stimulate/inhibit/block in a concentration-dependent way the (intracellular) activation of essential protein-splitting enzymes such as HIV proteases. Zinc and copper ions as 'passive' virus inhibitors. Apart from this, zinc ions directly or indirectly regulate, via zinc finger protein molecular structures, the activities of virus-combating Th-1 cells such as cytotoxic T-cells (CTLs). Zinc ions as regulators of the active, virus-combating Th-1 cells. Zinc and copper ions that remain available in sufficient amounts via cysteine/GSH are effective natural inhibitors/combaters of (AIDS) viruses and thereby prevent the development of chronic virus diseases that can lead to AIDS, autoimmune diseases, (food) allergies and/or cancer. A safe, relatively inexpensive and extensively tested medicine such as N-acetylcysteine (NAC) can help in supplying extra cysteine. The anti-HIV peptide T22, synthesized on the basis of two natural peptides from the Tachypleus tridentatus and Limnus polyphemus crabs, appears to be able to serve as supplier/carrier molecule of cysteine and zinc and/or to hinder the entry of HIVs into cells by way of the CD4 receptor.
Collapse
|
27
|
Abstract
Although our knowledge of HIV-1 growth, from a molecular mechanistic perspective, has rapidly increased, we do not yet know how the overall growth rate of HIV-1 depends on its constituent biochemical reactions. Such an understanding would be of fundamental importance and potentially useful for designing and evaluating anti-HIV strategies. As a first step toward addressing this need we formulate and implement here a global computer simulation for the intracellular growth of HIV-1 on a CD4+ T lymphocyte. Our simulation accounts for the kinetics of reverse transcription, integration of proviral DNA into the host genome, transcription, mRNA splicing and transport from the nucleus, translation, feedback of regulatory proteins to the nucleus, transport of viral proteins to the cell membrane, particle assembly, budding, and maturation. The simulation quantitatively captures the experimentally observed intracellular dynamics of viral DNA, mRNA, and proteins while employing no "fudge factors." Moreover, it provides an estimate of the intracellular growth rate of HIV-1 and enables evaluation of mono- and combined anti-HIV strategies.
Collapse
Affiliation(s)
- B Reddy
- Department of Chemical Engineering, University of Wisconsin-Madison, 53706-1691, USA
| | | |
Collapse
|
28
|
Düzgüneş N, Pretzer E, Simões S, Slepushkin V, Konopka K, Flasher D, de Lima MC. Liposome-mediated delivery of antiviral agents to human immunodeficiency virus-infected cells. Mol Membr Biol 1999; 16:111-8. [PMID: 10332745 DOI: 10.1080/096876899294832] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Intracellular delivery of novel macromolecular drugs against human immunodeficiency virus type-1 (HIV-1), including antisense oligodeoxynucleotides, ribozymes and therapeutic genes, may be achieved by encapsulation in or association with certain types of liposomes. Liposomes may also protect these drugs against nucleases. Low-molecular-weight, charged antiviral drugs may also be delivered more efficiently via liposomes. Liposomes were targeted to HIV-1-infected cells via covalently coupled soluble CD4. An HIV-1 protease inhibitor encapsulated in conventional negatively charged multilamellar liposomes was about 10-fold more effective and had a lower EC90 than the free drug in inhibiting HIV-1 production in human monocyte-derived macrophages. The drug encapsulated in sterically stabilized liposomes was as effective as the free drug. The EC50 of the reverse transcriptase inhibitor 9-(2-phosphonylmethoxyethyl)adenine (PMEA) was reduced by an order of magnitude when delivered to HIV-1-infected macrophages in pH-sensitive liposomes. A 15-mer antisense oligodeoxynucleotide against the Rev response element was ineffective in free form against HIV-1 replication in macrophages, while delivery of the oligonucleotide in pH-sensitive liposomes inhibited virus replication. The oligodeoxynucleotide encapsulated in sterically stabilized pH-sensitive liposomes with prolonged circulation in vivo, which were recently developed in the laboratories of the authors, was also highly effective. A ribozyme complementary to HIV-1 5'-LTR delivered in pH-sensitive liposomes inhibited virus production by 90%, while the free ribozyme caused only a slight inhibition. Cationic liposome-mediated co-transfection of the HIV-regulated diphtheria toxin A fragment gene and a proviral HIV clone into HeLa cells completely inhibited virus production, while the frame-shifted mutant gene was ineffective. Co-transfection of the proviral genome and a gene encoding a Rev-binding aptamer into HeLa cells via transferrin-associated cationic liposomes inhibited virus production. These studies indicate that liposomes can be used to facilitate the intracellular delivery of certain anti-HIV agents and to enhance their therapeutic effects. These properties may be particularly advantageous in the development of novel macromolecular drugs, which may be necessary because of the emergence of virus strains resistant to the currently available drugs.
Collapse
Affiliation(s)
- N Düzgüneş
- Department of Microbiology, School of Dentistry, University of the Pacific, San Francisco, CA 94115, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Proteolytic Enzymes of the Viruses of the Family Picornaviridae. PROTEASES OF INFECTIOUS AGENTS 1999. [PMCID: PMC7155532 DOI: 10.1016/b978-012420510-9/50032-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
This chapter deals with proteolytic enzymes of the viruses of the family picornaviridae. The picornaviral 3C proteinases constitute an ideal target for the rational design of antiviral drugs. The chapter discusses the chymotrypsin-like cysteine proteinases, which constitute a unique class of enzymes with a distinct substrate specificity, and are so far only found in +RNA viruses. Within these viruses the 3C proteinases perform a central and indispensable role during the viral life cycle and 3C proteinase inhibitors have the potential to limit the spread of viral infections. The chapter concludes that there is a wealth of experimental information available for the best-studied examples of the viruses of the Picornaviridae. This information provides an opportunity to design inhibitors against the viral 3C proteinase. Effective inhibitors of the picornaviral 3C proteinase have the potential to become effective antiviral drugs against human diseases such as the common cold, HAV, enteroviral infections, and diseases caused by related + RNA viruses.
Collapse
|
30
|
Abstract
Currently, there are a number of approved antiviral agents for use in the treatment of viral infections. However, many instances exist in which the use of a second antiviral agent would be beneficial because it would allow the option of either an alternative or a combination therapeutic approach. Accordingly, virus-encoded proteases have emerged as new targets for antiviral intervention. Molecular studies have indicated that viral proteases play a critical role in the life cycle of many viruses by effecting the cleavage of high-molecular-weight viral polyprotein precursors to yield functional products or by catalyzing the processing of the structural proteins necessary for assembly and morphogenesis of virus particles. This review summarizes some of the important general features of virus-encoded proteases and highlights new advances and/or specific challenges that are associated with the research and development of viral protease inhibitors. Specifically, the viral proteases encoded by the herpesvirus, retrovirus, hepatitis C virus, and human rhinovirus families are discussed.
Collapse
Affiliation(s)
- A K Patick
- Agouron Pharmaceuticals, Inc., San Diego, California 92121, USA.
| | | |
Collapse
|
31
|
Persichini T, Colasanti M, Lauro GM, Ascenzi P. Cysteine nitrosylation inactivates the HIV-1 protease. Biochem Biophys Res Commun 1998; 250:575-6. [PMID: 9784385 DOI: 10.1006/bbrc.1998.9350] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nitric oxide (NO) may modulate the catalytic activity of cysteine-containing enzymes. HIV-1 protease action is modulated by the redox equilibrium of Cys67 and Cys95 regulatory residues. In the present study, the inhibitory effect of NO, released by the NO-donor (+/-)-(E)-4-ethyl-2-[(E)-hydroxyimino]-5-nitro-3-hexenamide (NOR-3), on the aspartyl HIV-1 protease action is reported. HIV-1 protease inactivation via NO-mediated nitrosylation of Cys regulatory residue(s) may represent a possible mechanism for inhibition of HIV-1 replication.
Collapse
Affiliation(s)
- T Persichini
- Department of Biology, University of Rome 'Roma Tre,&rsquo, Viale Marconi 446, I-00146, Rome, Italy
| | | | | | | |
Collapse
|
32
|
Wu JL, Hong JR, Chang CY, Hui CF, Liao CF, Hsu YL. Involvement of serine proteinase in infectious pancreatic necrosis virus capsid protein maturation and NS proteinase cleavage in CHSE-214 cells. JOURNAL OF FISH DISEASES 1998; 21:215-220. [PMID: 21361976 DOI: 10.1046/j.1365-2761.1998.00097.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
An investigation of virus-specific protein maturation in infectious pancreatic necrosis virus (IPNV) infected Chinook salmon embryo cells (CHSE-214) was undertaken. The precursor protein (pVP2-1) of the major mature capsid protein (VP2) was processed sequentially from pVP2-1 to pVP2-2 and VP2. Experiments using serine proteinase inhibitors showed that the maturation of the VP2 was blocked in the pVP2-1 post-translational cleavage steps. A protinin, a potent proteinase inhibitor, at 800 μg ml(-1) blocked pVP2-2 to VP2 and the cleavage of VP4 (28 kDa) to VP4-1 (25 kDa). Therefore, our data showed that the maturation of the capsid protein (VP2) and cleavage of VP4 (NS proteinase) can be blocked by serine proteinase inhibitors.
Collapse
Affiliation(s)
- J L Wu
- 1 Molecular Biology Laboratory, Institute of Zoology, Academia Sinica, Nankang, Taipei, Taiwan, 2 Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
33
|
Konvalinka J, Litera J, Weber J, Vondrásek J, Hradílek M, Soucek M, Pichová I, Majer P, Strop P, Sedlácek J, Heuser AM, Kottler H, Kräusslich HG. Configurations of diastereomeric hydroxyethylene isosteres strongly affect biological activities of a series of specific inhibitors of human-immunodeficiency-virus proteinase. EUROPEAN JOURNAL OF BIOCHEMISTRY 1997; 250:559-66. [PMID: 9428710 DOI: 10.1111/j.1432-1033.1997.0559a.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Human immunodeficiency virus (HIV) proteinase (PR) represents an important target for antiviral chemotherapy. We present an analysis of inhibitory activities of a series of pseudopeptide inhibitors of HIV-1 PR. All inhibitors were N-protected tetrapeptides with the scissile bond replaced by a nonhydrolysable hydroxyethylene or hydroxyethylamine isostere. To elucidate subtle structural requirements of the PR binding cleft, we synthesised inhibitors with four combinations of configurations at the asymmetric carbons of the isostere. Compounds were tested in vitro using purified recombinant enzyme and a chromogenic peptide substrate. The differences in inhibition constants between individual diastereoisomers reached three orders of magnitude. The most active hydroxyethylene-containing inhibitor possessed the 2R,4S,5S configuration at the isostere. Inhibitor activity was also tested in mammalian cell culture by analysing reduction of viral polyprotein processing and virus infectivity. The results obtained in tissue culture were generally in agreement with the in vitro data, giving a similar order of potency for the individual diastereoisomers. The most active compounds completely blocked production of infectious virus. A simulation method for interaction was employed to build a model of the inhibitors in the PR active site, to identify the interactions responsible for the differences in activities of individual stereoisomers, and to estimate the relative contribution of individual structural features to the overall inhibitory activity.
Collapse
Affiliation(s)
- J Konvalinka
- Institute of Organic Chemistry and Biochemistry, Academy of Science of the Czech Republic, Praha.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ro YT, Scheffter SM, Patterson JL. Specific in vitro cleavage of a Leishmania virus capsid-RNA-dependent RNA polymerase polyprotein by a host cysteine-like protease. J Virol 1997; 71:8983-90. [PMID: 9371554 PMCID: PMC230198 DOI: 10.1128/jvi.71.12.8983-8990.1997] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Antibodies raised against baculovirus-expressed RNA-dependent RNA polymerase (RDRP) recognized a 95-kDa antigen and two smaller proteins in sucrose-purified Leishmania virus particles isolated from infected parasites. The 95-kDa antigen is similar in size to one predicted by translation of the RDRP open reading frame (ORF) alone. In an effort to reconcile in vitro observations of translational frameshifting on Leishmania RNA virus 1-4 transcripts, we have developed an in vitro cleavage assay system to explore the possibility that the fusion polyprotein is proteolytically processed. We show that coincubation a synthetic Cap-Pol fusion protein with lysates from Leishmania parasites yields major cleavage products similar in size to those encoded by the individual capsid and RDRP genes as well as the antigens detected in vivo. The major 82- and 95-kDa major cleavage products are specifically immunoprecipitated by capsid- or polymerase-specific antibodies, respectively, showing that cleavage occurs at or near the junction of the two functional domains. Protease inhibitor studies suggest that a cysteine-like protease is responsible for cleavage in the in vitro assay system developed here. From these results, we suggest that failure to detect a capsid-polymerase fusion protein produced by translational frameshifting in vivo may be due to specific proteolytic processing.
Collapse
Affiliation(s)
- Y T Ro
- Department of Virology and Immunology, Southwest Foundation for Biomedical Research, San Antonio, Texas 78245-0549, USA
| | | | | |
Collapse
|
35
|
Choy N, Choi HI, Jung WH, Kim CR, Yoon H, Kim SC, Lee TG, Koh JS. Synthesis of irreversible HIV-1 protease inhibitors containing sulfonamide and sulfone as amide bond isosteres. Bioorg Med Chem Lett 1997. [DOI: 10.1016/s0960-894x(97)10054-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
Pretzer E, Flasher D, Düzgüneş N. Inhibition of human immunodeficiency virus type-1 replication in macrophages and H9 cells by free or liposome-encapsulated L-689,502, an inhibitor of the viral protease. Antiviral Res 1997; 34:1-15. [PMID: 9107381 DOI: 10.1016/s0166-3542(96)01017-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Macrophages are recognized as a major reservoir of HIV-1 in infected individuals. We examined the effect of an inhibitor of the viral protease, L-689,502, on virus production by monocyte-derived macrophages infected with HIV-1BaL. Continuous treatment with L-689,502 drastically inhibited virus production in a dose-dependent manner in the range of 10-200 nM, in some cases by more than 1000-fold, compared to untreated cells. Since liposomes can be targeted to macrophages in vivo, we examined whether the inhibitor was effective following delivery in liposomes. The inhibitor encapsulated in multilamellar liposomes was more effective than the free drug in inhibiting virus production in macrophages, throughout the concentration range studied. The EC90 of the liposomal inhibitor was 2.9- to 4.5-fold lower than that of the free compound. L-689,502 encapsulated in sterically stabilized liposomes with prolonged circulation time inhibited virus production at a level comparable to the free inhibitor. When macrophages were infected and treated for only a limited time, L-689,502 in multilamellar liposomes was the most effective of the three treatments. In chronically infected H9 cells treated continuously, the free inhibitor was more effective than the liposome-encapsulated drug, but virus production was reduced only to 40-60% of controls. In contrast, treatment of acutely infected H9 cells with either free or encapsulated L-689,502 inhibited virus production by up to three orders of magnitude. Our results indicate that liposomes may be useful for the delivery of HIV protease inhibitors with low aqueous solubility and low oral bio-availability, and for the targeting of these drugs to lymph nodes.
Collapse
Affiliation(s)
- E Pretzer
- Department of Microbiology, School of Dentistry, University of the Pacific, San Francisco, CA 94115, USA
| | | | | |
Collapse
|
37
|
Mamoun RZ, Dye D, Rebeyrotte N, Bouamr F, Cerutti M, Desgranges C. Mouse monoclonal antibodies directed against the HTLV-I protease recognize epitopes internal to the dimer. JOURNAL OF ACQUIRED IMMUNE DEFICIENCY SYNDROMES AND HUMAN RETROVIROLOGY : OFFICIAL PUBLICATION OF THE INTERNATIONAL RETROVIROLOGY ASSOCIATION 1997; 14:184-8. [PMID: 9052730 DOI: 10.1097/00042560-199702010-00014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The proteases (PR) of retroviruses are expressed as gag-PR fused polyprotein. The active PR is a dimer obtained after the aggregation of the gag and gag-pro precursors, which leads to the formation and the release of the viral particle. Subsequently, in the cell, the PR is present essentially as a monomeric polyprotein. To mimic the antigenic properties of such an intracellular form of the PR, we produced a monomeric form of the HTLV-I (human T-cell leukemia virus, type-I) PR fused to the maltose binding protein (MBP-PR). Monoclonal antibodies (mabs) directed against MBP-PR were developed. Three mabs were obtained that recognized different epitopes. Two were directed against the NH2-terminus, a region that contributes to the dimerization interface. The other was specific to a peptide that lines the substrate binding pocket. This latter epitope is located just downstream of the D-T-G peptide of the catalytic site. The two identified regions contained the amino acids Asp6, Arg10 and Asp36, which were previously shown to be important in the stabilization of the dimer. In view of the localization of the recognized epitopes, these mabs will be useful for inhibition studies of the HTLV-I PR by intracellular immunization.
Collapse
Affiliation(s)
- R Z Mamoun
- Lab. Rétrovirus et thérapie, Université Victor Segalen Bordeaux II, France
| | | | | | | | | | | |
Collapse
|
38
|
β-Methanesulfonyl-L-valine as a novel, unnatural amino acid surrogate for P2 in the design of HIV protease inhibitors. Bioorg Med Chem Lett 1996. [DOI: 10.1016/0960-894x(96)00086-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
39
|
Wondrak EM, Nashed NT, Haber MT, Jerina DM, Louis JM. A transient precursor of the HIV-1 protease. Isolation, characterization, and kinetics of maturation. J Biol Chem 1996; 271:4477-81. [PMID: 8626801 DOI: 10.1074/jbc.271.8.4477] [Citation(s) in RCA: 55] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Recently, the mechanism of autoprocessing of the protease (PR) of the human immunodeficiency virus type 1 from the model polyprotein, MBP-DeltaTF-PR-DeltaPol, which contains the protease linked to short native flanking sequences (DeltaTF and DeltaPol) fused to the maltose binding protein (MBP) of Escherichia coli, was reported (Louis, J. M., Nashed, N. T., Parris, K. D., Kimmel, A. R., and Jerina, D. M. (1994) Proc. Natl. Acad. Sci. U. S. A. 91, 7970-7974). According to this mechanism, intramolecular cleavage of the N-terminal strands of the dimeric MBP-DeltaTF-PR-DeltaPol protein leads to the formation of the PR-DeltaPol intermediate, which is subsequently converted to the mature protease by cleavage of the C-terminal strands. We now report the purification and characterization of the PR-DeltaPol intermediate and the kinetics of its processing to the mature protease. Unlike the MBP-DeltaTF-PR-DeltaPol precursor, PR-DeltaPol has proteolytic activity similar to that of the mature enzyme at pH 5.0. The pH rate profile for kcat/Km is similar to that of the mature protease above pH 4.0. Although the PR-DeltaPol is more sensitive than the mature protease toward denaturing reagents, both the enzymatic activity and the intrinsic fluorescence of PR-DeltaPol are linearly dependent on the protein concentration, indicating that the protein is largely in its dimeric form above 10 nM. In contrast to the first-order kinetics observed for the proteolytic reaction at the N terminus of the protease, the proteolytic reaction at the C terminus of the protease is second order in protein concentration. These results are discussed in terms of a mechanism in which the C-terminally located DeltaPol peptide chains are cleaved intermolecularly to release the mature protease.
Collapse
Affiliation(s)
- E M Wondrak
- Molecular Mechanisms of Development Section, Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
40
|
Henskens YM, Veerman EC, Nieuw Amerongen AV. Cystatins in health and disease. BIOLOGICAL CHEMISTRY HOPPE-SEYLER 1996; 377:71-86. [PMID: 8868064 DOI: 10.1515/bchm3.1996.377.2.71] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Proteolytic enzymes have many physiological functions in plants, bacteria, viruses, protozoa and mammals. They play a role in processes such as food digestion, complement activation or blood coagulation. The action of proteolytic enzymes is biologically controlled by proteinase inhibitors and increasing attention is being paid to the physiological significance of these natural inhibitors in pathological processes. The reason for this growing interest is that uncontrolled proteolysis can lead to irreversible damage e.g. in chronic inflammation or tumor metastasis. This review focusses on the possible role of the cystatins, natural and specific inhibitors of the cysteine proteinases, in pathological processes.
Collapse
Affiliation(s)
- Y M Henskens
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), Netherlands
| | | | | |
Collapse
|
41
|
Hall DJ, Palmenberg AC. Mengo virus 3C proteinase: recombinant expression, intergenus substrate cleavage and localization in vivo. Virus Genes 1996; 13:99-110. [PMID: 8972564 DOI: 10.1007/bf00568903] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Mengo virus 3C proteinase was cloned and expressed to high levels in a bacterial vector system. The protein was solubilized from inclusion bodies then purified to homogeneity (> 95%) by ion exchange chromatography. The recombinant enzyme was proteolytically active in cell-free processing assays with a Mengo capsid precursor substrate, L-P1-2A, correctly and proficiently cleaving it into L, 1AB, 1C, 1D and 2A protein products. Further analyses with synthetic peptide substrates encompassing the Mengo or rhinovirus-14 2C/3A cleavage sequences, showed the Mengo 3C could recognize and process specific glutamine-glycine sites within these peptides. The reactivity with the rhinovirus peptide was unexpected, because cross-reactivity between a picornavirus 3C enzyme and a protein substrate from different genus of this family has otherwise never been observed. In reciprocal reactions, a rhinovirus-14 3C preparation was unable to cleave the Mengo-derived synthetic peptide substrate. The recombinant Mengo 3C reactions were also characterized with regard to substrate Km, optimum pH and temperature. The protein was additionally used to raise monoclonal antibodies (mAbs) in mice, which in turn localized natural 3C, 3ABC, 3CD and P3 in immunoblots, immunoprecipitations and indirect immunofluorescence assays of Mengo-infected HeLa cells. The monoclonals showed cross-reactivity with 3C and 3C-containing precursors from encephalomyocarditis virus (EMCV), but did not react with 3C proteins from rhinovirus-14 or poliovirus-1M.
Collapse
Affiliation(s)
- D J Hall
- Institute for Molecular Virology, University of Wisconsin-Madison 53706, USA
| | | |
Collapse
|
42
|
Affiliation(s)
- J Saunders
- Department of Medicinal Chemistry II, Glaxo Research and Development Limited, Stevenage, Herts, UK
| | | |
Collapse
|
43
|
|
44
|
Kräusslich HG, Fäcke M, Heuser AM, Konvalinka J, Zentgraf H. The spacer peptide between human immunodeficiency virus capsid and nucleocapsid proteins is essential for ordered assembly and viral infectivity. J Virol 1995; 69:3407-19. [PMID: 7745687 PMCID: PMC189053 DOI: 10.1128/jvi.69.6.3407-3419.1995] [Citation(s) in RCA: 191] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Morphogenesis of retroviruses involves ordered assembly of the structural Gag- and Gag-Pol polyproteins, with subsequent budding from the plasma membrane and proteolytic cleavage by the viral proteinase (PR). Two cleavage sites exist between the capsid (CA) and nucleocapsid (NC) domains of the human immunodeficiency virus (HIV) type 1 Gag polyprotein which are separated by a 14-amino-acid spacer peptide of unknown function. To analyze the role of the two cleavage sites and the spacer peptide, both sites were individually mutated and a deletion mutation that precisely removes the spacer peptide was constructed. Following transfection of proviral DNA carrying the point mutations, mutant polyproteins were synthesized and assembled like wild-type polyprotein, and release of particles was not significantly altered. Both mutations abolished cleavage at the respective site and reduced or abolished viral infectivity. Deletion of the spacer peptide severely affected ordered assembly and reduced particle release. The extracellular particles that were released exhibited normal density but were heterogeneous in size. Electron micrographs revealed large electron-dense plaques underneath the plasma membrane of transfected cells which appeared like confluent ribonucleoprotein complexes arrested early in the budding process. Extracellular particles exhibited very aberrant and heterogeneous morphology and were incapable of inducing viral spread. These particles may correspond to membrane vesicles sequestered by the rigid structures underneath the cell membrane and not released by a regular budding process.
Collapse
Affiliation(s)
- H G Kräusslich
- Angewandte Tumorvirologie, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
45
|
Priestle JP, Fässler A, Rösel J, Tintelnot-Blomley M, Strop P, Grütter MG. Comparative analysis of the X-ray structures of HIV-1 and HIV-2 proteases in complex with CGP 53820, a novel pseudosymmetric inhibitor. Structure 1995; 3:381-9. [PMID: 7613867 DOI: 10.1016/s0969-2126(01)00169-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND The human immunodeficiency virus (HIV) is the causative agent of acquired immunodeficiency syndrome (AIDS). Two subtypes of the virus, HIV-1 and HIV-2, have been characterized. The protease enzymes from these two subtypes, which are aspartic acid proteases and have been found to be essential for maturation of the infectious particle, share about 50% sequence identity. Differences in substrate and inhibitor binding between these enzymes have been previously reported. RESULTS We report the X-ray crystal structures of both HIV-1 and HIV-2 proteases each in complex with the pseudosymmetric inhibitor, CGP 53820, to 2.2 A and 2.3 A, respectively. In both structures, the entire enzyme and inhibitor could be located. The structures confirmed earlier modeling studies. Differences between the CGP 53820 inhibitory binding constants for the two enzymes could be correlated with structural differences. CONCLUSIONS Minor sequence changes in subsites at the active site can explain some of the observed differences in substrate and inhibitor binding between the two enzymes. The information gained from this investigation may help in the design of equipotent HIV-1/HIV-2 protease inhibitors.
Collapse
Affiliation(s)
- J P Priestle
- Department of Core Drug Discovery Technologies, Pharma Research, Ciba-Geigy Ltd., Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
46
|
Pillay D, Bryant M, Getman D, Richman DD. HIV-1 Protease inhibitors: Their development, mechanism of action and clinical potential. Rev Med Virol 1995. [DOI: 10.1002/rmv.1980050104] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
47
|
Donaghy G, Jupp R. Characterization of the Epstein-Barr virus proteinase and comparison with the human cytomegalovirus proteinase. J Virol 1995; 69:1265-70. [PMID: 7815503 PMCID: PMC188701 DOI: 10.1128/jvi.69.2.1265-1270.1995] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The BVRF2 gene of Epstein-Barr virus (EBV) shows homology to the UL26 and UL80 genes of herpes simplex virus type 1 (HSV-1) and cytomegalovirus (CMV), respectively. These genes are believed to provide a scaffold protein for the assembly of capsids leading to the formation of infectious viral particles. We have cloned the BVRF2 gene from the B95.8 strain of EBV and shown that the BVRF2 gene product is a polyprotein capable of autoproteolytic cleavage. Two Ala-Ser-containing recognition sequences were identified in the BVRF2 polyprotein at amino acid positions 568/569 and 570/571 where this cleavage was expected to occur. Here, we show that EBV proteinase is capable of cleaving at the first Ala-Ser bond but not the second. Comparison of the processing of the EBV and human CMV assembly domains in vitro by either EBV or human CMV proteinase revealed that, while both proteinases could cleave their native assembly domain, only EBV proteinase was able to cleave the assembly domain of the other virus.
Collapse
Affiliation(s)
- G Donaghy
- Department of Biology, Roche Research Centre, Welwyn Garden City, Hertfordshire, United Kingdom
| | | |
Collapse
|
48
|
Marczinovits I, Molnár J, Patthy A. Staphylococcal protein A is a novel heterologous substrate for the HIV-1 protease. J Biotechnol 1994; 37:79-83. [PMID: 7765414 DOI: 10.1016/0168-1656(94)90205-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Upon in vitro processing of the recombinant HIV-1/gag p24 protein, expressed in Escherichia coli as a fusion protein, by HIV-1 protease, a cleavage site within the staphylococcal protein A fusion partner was found. N-terminal sequencing of the protein A fragments showed that HIV-1 protease cleavage occurred between phenylalanine-235 and tyrosine-236 within the sequence Gln-Asn-Ala-Phe/Tyr-Glu-Ile-Leu (QNAF/YEIL) in the IgG-binding domain C of the protein A encoded by the pRIT2T fusion gene vector (Pharmacia). Results presented here have proven that the protease-sensitive site is viable in vitro on the protein A alone and other chimeric protein, protein A/beta-galactosidase. A possible significance of this phenomenon in biotechnology work is discussed.
Collapse
Affiliation(s)
- I Marczinovits
- Institute of Microbiology, Albert Szent-Györgyi Medical University, Szeged, Hungary
| | | | | |
Collapse
|
49
|
Louis JM, Nashed NT, Parris KD, Kimmel AR, Jerina DM. Kinetics and mechanism of autoprocessing of human immunodeficiency virus type 1 protease from an analog of the Gag-Pol polyprotein. Proc Natl Acad Sci U S A 1994; 91:7970-4. [PMID: 8058744 PMCID: PMC44526 DOI: 10.1073/pnas.91.17.7970] [Citation(s) in RCA: 78] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Upon renaturation, the polyprotein MBP-delta TF-Protease-delta Pol, consisting of HIV-1 protease and short native sequences from the trans-frame protein (delta TF) and the polymerase (delta Pol) fused to the maltose-binding protein (MBP) of Escherichia coli, undergoes autoprocessing to produce the mature protease in two steps. The initial step corresponds to cleavage of the N-terminal sequence to release the protein intermediate Protease-delta Pol, which has enzymatic activity comparable to that of the mature enzyme. Subsequently, the mature enzyme is formed by a slower cleavage at the C terminus. The rate of increase in enzymatic activity is identical to that of the appearance of MBP-delta TF and the disappearance of the MBP-delta TF-Protease-delta Pol. Initial rates are linearly dependent on the protein concentration, indicating that the N-terminal cleavage is first-order in protein concentration. The reaction is competitively inhibited by pepstatin A and has a pH rate profile similar to that of the mature enzyme. These results and molecular modeling studies are discussed in terms of a mechanism in which a dimeric full-length fusion protein must form prior to rate-limiting intramolecular cleavage of the N-terminal sequence that leads to an increase in enzymatic activity.
Collapse
Affiliation(s)
- J M Louis
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | | | | | | | | |
Collapse
|
50
|
Dame JB, Reddy GR, Yowell CA, Dunn BM, Kay J, Berry C. Sequence, expression and modeled structure of an aspartic proteinase from the human malaria parasite Plasmodium falciparum. Mol Biochem Parasitol 1994; 64:177-90. [PMID: 7935597 DOI: 10.1016/0166-6851(94)90024-8] [Citation(s) in RCA: 95] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
A clone encoding the aspartic proteinase (PFAPD) from Plasmodium falciparum strain HB3 was obtained during the course of a project designed to sequence and identify the protein coding regions of the parasite's genome. The protein encoded by the clone contains a sequence identical to the N-terminal sequence determined for an aspartic proteinase isolated from the digestive vacuole of P. falciparum and demonstrated to participate in the hemoglobin digestive pathway (D. Goldberg, personal communication). The translated polypeptide sequence encompasses a number of features characteristic of aspartic proteinases, having > 30% identity and > 50% similarity overall to human cathepsin D, cathepsin E and renin. A model of the three-dimensional structure of PFAPD was constructed using rule-based procedures. This confirms that the primary sequence may be folded as a single chain into a three dimensional structure closely resembling those of other known aspartic proteinases. It includes a lengthy prosegment, two typical-hydrophobic-hydrophobic-Asp-Thr/Ser-Gly motifs and a tyrosine residue positioned in a beta-hairpin loop. The distribution of hydrophobic residues throughout the active site cleft is indicative of a likely preference for hydrophobic polypeptide substrates. The recombinant form of this enzyme expressed using the pGEX2T vector in Escherichia coli is active in digesting hemoglobin at acidic pH and in hydrolyzing a synthetic peptide corresponding to the putative initial cleavage site in hemoglobin. Activity is inhibited completely by pepstatin, confirming the identity of PFAPD as a member of the aspartic proteinase family. Specific mRNA for PFAPD is expressed in the erythrocytic stages of the life cycle.
Collapse
Affiliation(s)
- J B Dame
- Department of Infectious Diseases, College of Veterinary Medicine, University of Florida, Gainesville 32611
| | | | | | | | | | | |
Collapse
|