1
|
Serrano-Aparicio N, Moliner V, Świderek K. On the Origin of the Different Reversible Characters of Salinosporamide A and Homosalinosporamide A in the Covalent Inhibition of the Human 20S Proteasome. ACS Catal 2021. [DOI: 10.1021/acscatal.1c02614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Natalia Serrano-Aparicio
- Biocomp Group, Institute of Advanced Materials (INAM), Universitat Jaume I, 12071 Castellón, Spain
| | - Vicent Moliner
- Biocomp Group, Institute of Advanced Materials (INAM), Universitat Jaume I, 12071 Castellón, Spain
| | - Katarzyna Świderek
- Biocomp Group, Institute of Advanced Materials (INAM), Universitat Jaume I, 12071 Castellón, Spain
| |
Collapse
|
2
|
Friday SN, Cheng DW, Zagler SG, Zanella BS, Dietz JD, Calbat CN, Roach LT, Bagnal C, Faile IS, Halkides CJ, Viola RE. Design and testing of selective inactivators against an antifungal enzyme target. Drug Dev Res 2021; 83:447-460. [PMID: 34469014 DOI: 10.1002/ddr.21875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/03/2021] [Accepted: 08/19/2021] [Indexed: 11/10/2022]
Abstract
Systemic infections from fungal organisms are becoming increasingly difficult to treat as drug resistance continues to emerge. To substantially expand the antifungal drug landscape new compounds must be identified and developed with novel modes of action against previously untested drug targets. Most drugs block the activity of their targets through reversible, noncovalent interactions. However, a significant number of drugs form irreversible, covalent bonds with their selected targets. While more challenging to develop, these irreversible inactivators offer some significant advantages as novel antifungal agents. Vinyl sulfones contain a potentially reactive functional group that could function as a selective enzyme inactivator, and members of this class of compounds are now being developed as inactivators against an antifungal drug target. The enzyme aspartate semialdehyde dehydrogenase (ASADH) catalyzes a key step in an essential microbial pathway and is essential for the survival of every microorganism examined. A series of vinyl sulfones have been designed, guided by molecular modeling and docking studies to enhance their affinity for fungal ASADHs. These newly synthesized compounds have been examined against this target enzyme from the pathogenic fungal organism Candida albicans. Vinyl sulfones containing complementary structural elements inhibit this enzyme with inhibition constants in the low-micromolar range. These inhibitors have also led to the rapid and irreversible inactivation of this enzyme, and show some initial selectivity when compared to the inactivation of a bacterial ASADH. The best inactivators will serve as lead compounds for the development of potent and selective antifungal agents.
Collapse
Affiliation(s)
- Samantha N Friday
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio, USA
| | - Daniel W Cheng
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina, USA
| | - Sebastian G Zagler
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina, USA
| | - Brady S Zanella
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina, USA
| | - Jordan D Dietz
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina, USA
| | - Christopher N Calbat
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina, USA
| | - Logan T Roach
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina, USA
| | - Cindy Bagnal
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina, USA
| | - Ian S Faile
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina, USA
| | - Christopher J Halkides
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina, USA
| | - Ronald E Viola
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio, USA
| |
Collapse
|
3
|
Abstract
Covalent enzyme inhibitors are widely applied as biochemical tools and therapeutic agents. As a complement to categorization of these inhibitors by reactive group or modification site, we present a categorization by mechanism, which highlights common advantages and disadvantages inherent to each approach. Established categories for reversible and irreversible covalent inhibition are reviewed with representative examples given for each class, including covalent reversible inhibitors, slow substrates, residue-specific reagents, affinity labels (classical, quiescent, and photoaffinity), and mechanism-based inactivators. The relationships of these categories to proteomic profiling probes (activity-based and reactivity-based) as well as complementary approaches such as prodrug and soft drug design are also discussed. A wide variety of strategies are used to balance reactivity and selectivity in the design of covalent enzyme inhibitors. Use of a shared terminology is encouraged to clearly convey these mechanisms, to relate them to prior use of covalent inhibitors in enzymology, and to facilitate the development of more effective covalent inhibitors.
Collapse
Affiliation(s)
- Alfred Tuley
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy , University of Texas , Austin , Texas 78712 , United States
| | - Walter Fast
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy , University of Texas , Austin , Texas 78712 , United States
| |
Collapse
|
4
|
Soualmia F, El Amri C. Serine protease inhibitors to treat inflammation: a patent review (2011-2016). Expert Opin Ther Pat 2017; 28:93-110. [PMID: 29171765 DOI: 10.1080/13543776.2018.1406478] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Inflammation is a physiological part of the complex biological response of tissues to counteract various harmful signals. This process involves diverse actors such as immune cells, blood vessels, and nerves as sources of mediators for inflammation control. Among them serine proteases are key elements in both physiological and pathological inflammation. AREAS COVERED Serine protease inhibitors to treat inflammatory diseases are being actively investigated by various industrial and academic institutions. The present review covers patent literature on serine protease inhibitors for the therapy of inflammatory diseases patented between 2011 and 2016. EXPERT OPINION Serine proteases regulating inflammation are versatile enzymes, usually involved in proinflammatory cytokine production and activation of immune cells. Their dysregulation during inflammation can have devastating consequences, promoting various diseases including skin and lung inflammation, neuroinflammation, and inflammatory arthritis. Several serine proteases were selected for their contribution to inflammatory diseases and significant efforts that are spread to develop inhibitors. Strategies developed for inhibitor identification consist on either peptide-based inhibitor derived from endogenous protein inhibitors or small-organic molecules. It is also worth noting that among the recent patents on serine protease inhibitors related to inflammation a significant number are related to retinal vascular dysfunction and skin diseases.
Collapse
Affiliation(s)
- Feryel Soualmia
- a B2A, Biological Adaptation and Ageing, Integrated Cellular Ageing and Inflammation, Molecular & Functional Enzymology , Sorbonne Universités , UPMC Univ Paris 06, UMR 8256 , Paris , France
| | - Chahrazade El Amri
- a B2A, Biological Adaptation and Ageing, Integrated Cellular Ageing and Inflammation, Molecular & Functional Enzymology , Sorbonne Universités , UPMC Univ Paris 06, UMR 8256 , Paris , France
| |
Collapse
|
5
|
Schardon CL, Tuley A, Er JAV, Swartzel JC, Fast W. Selective Covalent Protein Modification by 4-Halopyridines through Catalysis. Chembiochem 2017; 18:1551-1556. [PMID: 28470883 DOI: 10.1002/cbic.201700104] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Indexed: 12/12/2022]
Abstract
We have investigated 4-halopyridines as selective, tunable, and switchable covalent protein modifiers for use in the development of chemical probes. Nonenzymatic reactivity of 4-chloropyridine with amino acids and thiols was ranked with respect to common covalent protein-modifying reagents and found to have reactivity similar to that of acrylamide, but could be switched to a reactivity similar to that of iodoacetamide upon stabilization of the positively charged pyridinium. Diverse, fragment-sized 4-halopyridines inactivated human dimethylarginine dimethylaminohydrolase-1 (DDAH1) through covalent modification of the active site cysteine, acting as quiescent affinity labels that required off-pathway catalysis through stabilization of the protonated pyridinium by a neighboring aspartate residue. A series of 2-fluoromethyl-substituted 4-chloropyridines demonstrated that the pKa and kinact /KI values could be predictably varied over several orders of magnitude. Covalent labeling of proteins in an Escherichia coli lysate was shown to require folded proteins, indicating that alternative proteins can be targeted, and modification is likely to be catalysisdependent. 4-Halopyridines, and quiescent affinity labels in general, represent an attractive strategy to develop reagents with switchable electrophilicity as selective covalent protein modifiers.
Collapse
Affiliation(s)
| | - Alfred Tuley
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, The University of Texas, Austin, TX, 78712, USA
| | - Joyce A V Er
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, The University of Texas, Austin, TX, 78712, USA
| | - Jake C Swartzel
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, The University of Texas, Austin, TX, 78712, USA
| | - Walter Fast
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, The University of Texas, Austin, TX, 78712, USA.,LaMontagne Center for Infectious Disease, The University of Texas, Austin, TX, 78712, USA
| |
Collapse
|
6
|
Tago N, Katolik A, Clark NE, Montemayor EJ, Seio K, Sekine M, Hart PJ, Damha MJ. Design, Synthesis, and Properties of Phosphoramidate 2',5'-Linked Branched RNA: Toward the Rational Design of Inhibitors of the RNA Lariat Debranching Enzyme. J Org Chem 2015; 80:10108-18. [PMID: 26378468 PMCID: PMC4749351 DOI: 10.1021/acs.joc.5b01719] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Two RNA fragments linked by means of a 2',5' phosphodiester bridge (2' hydroxyl of one fragment connected to the 5' hydroxyl of the other) constitute a class of nucleic acids known as 2'-5' branched RNAs (bRNAs). In this report we show that bRNA analogues containing 2'-5' phosphoramidate linkages (bN-RNAs) inhibit the lariat debranching enzyme, a 2',5'-phosphodiesterase that has recently been implicated in neurodegenerative diseases associated with aging. bN-RNAs were efficiently generated using automated solid-phase synthesis and suitably protected branchpoint building blocks. Two orthogonally removable groups, namely the 4-monomethoxytrityl (MMTr) group and the fluorenylmethyl-oxycarbonyl (Fmoc) groups, were evaluated as protecting groups of the 2' amino functionality. The 2'-N-Fmoc methodology was found to successfully produce bN-RNAs on solid-phase oligonucleotide synthesis. The synthesized bN-RNAs resisted hydrolysis by the lariat debranching enzyme (Dbr1) and, in addition, were shown to attenuate the Dbr1-mediated hydrolysis of native bRNA.
Collapse
Affiliation(s)
- Nobuhiro Tago
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec H3A 0B8, Canada
- Department of Life Science, Tokyo Institute of Technology, 4259 Nagatsuta, Midoriku, Yokohama, Kanagawa, 226-8501, Japan
| | - Adam Katolik
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec H3A 0B8, Canada
| | - Nathaniel E. Clark
- Department of Veterans Affairs, South Texas Veterans Health Care System, San Antonio, TX 78229, United States
| | - Eric J. Montemayor
- Department of Veterans Affairs, South Texas Veterans Health Care System, San Antonio, TX 78229, United States
- Departments of Biochemistry and Biomolecular Chemistry, University of Wisconsin-Madison, 433 Babcock Dr., Madison, WI 53706, United States
| | - Kohji Seio
- Department of Life Science, Tokyo Institute of Technology, 4259 Nagatsuta, Midoriku, Yokohama, Kanagawa, 226-8501, Japan
| | - Mitsuo Sekine
- Department of Life Science, Tokyo Institute of Technology, 4259 Nagatsuta, Midoriku, Yokohama, Kanagawa, 226-8501, Japan
| | - P. John Hart
- Department of Veterans Affairs, South Texas Veterans Health Care System, San Antonio, TX 78229, United States
- Department of Biochemistry, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Masad J. Damha
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec H3A 0B8, Canada
| |
Collapse
|
7
|
Johnson CM, Monzingo AF, Ke Z, Yoon DW, Linsky TW, Guo H, Robertus JD, Fast W. On the mechanism of dimethylarginine dimethylaminohydrolase inactivation by 4-halopyridines. J Am Chem Soc 2011; 133:10951-9. [PMID: 21630706 PMCID: PMC3135753 DOI: 10.1021/ja2033684] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Small molecules capable of selective covalent protein modification are of significant interest for the development of biological probes and therapeutics. We recently reported that 2-methyl-4-bromopyridine is a quiescent affinity label for the nitric oxide controlling enzyme dimethylarginine dimethylaminohydrolase (DDAH) (Johnson, C. M.; Linsky, T. W.; Yoon, D. W.; Person, M. D.; Fast, W. J. Am. Chem. Soc. 2011, 133, 1553-1562). Discovery of this novel protein modifier raised the possibility that the 4-halopyridine motif may be suitable for wider application. Therefore, the inactivation mechanism of the related compound 2-hydroxymethyl-4-chloropyridine is probed here in more detail. Solution studies support an inactivation mechanism in which the active site Asp66 residue stabilizes the pyridinium form of the inactivator, which has enhanced reactivity toward the active site Cys, resulting in covalent bond formation, loss of the halide, and irreversible inactivation. A 2.18 Å resolution X-ray crystal structure of the inactivated complex elucidates the orientation of the inactivator and its covalent attachment to the active site Cys, but the structural model does not show an interaction between the inactivator and Asp66. Molecular modeling is used to investigate inactivator binding, reaction, and also a final pyridinium deprotonation step that accounts for the apparent differences between the solution-based and structural studies with respect to the role of Asp66. This work integrates multiple approaches to elucidate the inactivation mechanism of a novel 4-halopyridine "warhead," emphasizing the strategy of using pyridinium formation as a "switch" to enhance reactivity when bound to the target protein.
Collapse
Affiliation(s)
| | | | | | | | | | - Hua Guo
- To whom correspondence should be addressed. W.F.: College of Pharmacy, PHAR-MED CHEM, 1 University Station; C0850, Austin, Texas 78712; Phone: (512) 232-4000; Fax: (512) 232-2606; ; J.D.R.: Department of Chemistry and Biochemistry, University of Texas, Austin, TX 78712. Phone: (512) 471-3175. Fax: (512) 471-6135. , and H.G.: Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM 87131;
| | - Jon D. Robertus
- To whom correspondence should be addressed. W.F.: College of Pharmacy, PHAR-MED CHEM, 1 University Station; C0850, Austin, Texas 78712; Phone: (512) 232-4000; Fax: (512) 232-2606; ; J.D.R.: Department of Chemistry and Biochemistry, University of Texas, Austin, TX 78712. Phone: (512) 471-3175. Fax: (512) 471-6135. , and H.G.: Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM 87131;
| | - Walter Fast
- To whom correspondence should be addressed. W.F.: College of Pharmacy, PHAR-MED CHEM, 1 University Station; C0850, Austin, Texas 78712; Phone: (512) 232-4000; Fax: (512) 232-2606; ; J.D.R.: Department of Chemistry and Biochemistry, University of Texas, Austin, TX 78712. Phone: (512) 471-3175. Fax: (512) 471-6135. , and H.G.: Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM 87131;
| |
Collapse
|
8
|
Johnson CM, Linsky TW, Yoon DW, Person MD, Fast W. Discovery of halopyridines as quiescent affinity labels: inactivation of dimethylarginine dimethylaminohydrolase. J Am Chem Soc 2011; 133:1553-62. [PMID: 21222447 PMCID: PMC3038607 DOI: 10.1021/ja109207m] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In an effort to develop novel covalent modifiers of dimethylarginine dimethylaminohydrolase (DDAH) that are useful for biological applications, a set of "fragment"-sized inhibitors that were identified using a high-throughput screen are tested for time-dependent inhibition. One structural class of inactivators, 4-halopyridines, show time- and concentration-dependent inactivation of DDAH, and the inactivation mechanism of one example, 4-bromo-2-methylpyridine (1), is characterized in detail. The neutral form of halopyridines is not very reactive with excess glutathione. However, 1 readily reacts, with loss of its halide, in a selective, covalent, and irreversible manner with the active-site Cys249 of DDAH. This active-site Cys is not particularly reactive (pK(a) ca. 8.8), and 1 does not inactivate papain (Cys pK(a) ca. ≤4), suggesting that, unlike many reagents, Cys nucleophilicity is not a predominating factor in selectivity. Rather, binding and stabilization of the more reactive pyridinium form of the inactivator by a second moiety, Asp66, is required for facile reaction. This constraint imparts a unique selectivity profile to these inactivators. To our knowledge, halopyridines have not previously been reported as protein modifiers, and therefore they represent a first-in-class example of a novel type of quiescent affinity label.
Collapse
Affiliation(s)
- Corey M. Johnson
- Division of Medicinal Chemistry, University of Texas, Austin, Texas 78712
| | - Thomas W. Linsky
- Graduate Program in Biochemistry, University of Texas, Austin, Texas 78712
| | - Dae-Wi Yoon
- Division of Medicinal Chemistry, University of Texas, Austin, Texas 78712
| | - Maria D. Person
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas, Austin, Texas 78712
| | - Walter Fast
- Division of Medicinal Chemistry, University of Texas, Austin, Texas 78712
- Graduate Program in Biochemistry, University of Texas, Austin, Texas 78712
| |
Collapse
|
9
|
John S, Thangapandian S, Sakkiah S, Lee KW. Discovery of potential pancreatic cholesterol esterase inhibitors using pharmacophore modelling, virtual screening, and optimization studies. J Enzyme Inhib Med Chem 2010; 26:535-45. [PMID: 21143043 DOI: 10.3109/14756366.2010.535795] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Shalini John
- Department of Biochemistry and Division of Applied Life Science (BK21 Program), Environmental Biotechnology National Core Research Center (EB-NCRC), Gyeongsang National University (GNU), Jinju, Republic of Korea
| | - Sundarapandian Thangapandian
- Department of Biochemistry and Division of Applied Life Science (BK21 Program), Environmental Biotechnology National Core Research Center (EB-NCRC), Gyeongsang National University (GNU), Jinju, Republic of Korea
| | - Sugunadevi Sakkiah
- Department of Biochemistry and Division of Applied Life Science (BK21 Program), Environmental Biotechnology National Core Research Center (EB-NCRC), Gyeongsang National University (GNU), Jinju, Republic of Korea
| | - Keun Woo Lee
- Department of Biochemistry and Division of Applied Life Science (BK21 Program), Environmental Biotechnology National Core Research Center (EB-NCRC), Gyeongsang National University (GNU), Jinju, Republic of Korea
| |
Collapse
|
10
|
Krämer S, Testa B. The Biochemistry of Drug Metabolism - An Introduction. Chem Biodivers 2009; 6:1477-660, table of contents. [DOI: 10.1002/cbdv.200900233] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
11
|
Schepetkin IA, Khlebnikov AI, Kirpotina LN, Quinn MT. Novel small-molecule inhibitors of anthrax lethal factor identified by high-throughput screening. J Med Chem 2006; 49:5232-44. [PMID: 16913712 DOI: 10.1021/jm0605132] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Anthrax lethal factor (LF) is a key virulence factor of anthrax lethal toxin. We screened a chemolibrary of 10,000 drug-like molecules for their ability to inhibit LF and identified 18 novel small molecules with potent LF inhibitory activity. Three additional LF inhibitors were identified through further structure-activity relationship (SAR) analysis. All 21 compounds inhibited LF with an IC50 range of 0.8 to 11 muM, utilizing mixed-mode competitive inhibition. An evaluation of inhibitory activity against a range of unrelated proteases showed relatively high specificity for LF. Furthermore, pharmacophore modeling of these compounds showed a high degree of similarity to the model published by Panchal et al. (Nat. Struct. Mol. Biol. 2004, 11, 67-72), indicating that the conformational features of these inhibitors are structurally compatible with the steric constraints of the substrate-binding pocket. These novel LF inhibitors and the structural scaffolds identified as important for inhibitory activity represent promising leads to pursue for further LF inhibitor development.
Collapse
Affiliation(s)
- Igor A Schepetkin
- Department of Veterinary Molecular Biology, Montana State University, Bozeman, Montana 59717, USA
| | | | | | | |
Collapse
|
12
|
Gautier A, Pitrat D, Hasserodt J. An unusual functional group interaction and its potential to reproduce steric and electrostatic features of the transition states of peptidolysis. Bioorg Med Chem 2006; 14:3835-47. [PMID: 16464600 DOI: 10.1016/j.bmc.2006.01.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2005] [Revised: 01/13/2006] [Accepted: 01/17/2006] [Indexed: 10/25/2022]
Abstract
The donor-acceptor interaction between a tertiary amine and an aldehyde, first observed among a select class of alkaloids, was deliberately established in a peptidomimetic (1a-c) to mimic features of the two principal transition states of peptide hydrolysis. Compounds 1a-c show preferential adoption in methanol and water of a 'folded' conformation displaying the interaction. Proportions of the folded form in MeOH range from 45% to 70% and can reach 84% in buffer. Significantly, three tendencies for the folded/unfolded equilibrium are observed: increasing solubility and polarity of the medium and decreasing temperature results in a higher extent of folding. In the absence of any parameter set available for this weak bond, no modeling studies were conducted to aid in the design of 1a-c. The successful straightforward synthesis of 1 and its folding and inhibition results with HIV-1 peptidase using FRET technology encourage studies to further pre-organize candidate molecules and to screen the structure space by modeling and parallel combinatorial chemistry.
Collapse
Affiliation(s)
- Arnaud Gautier
- Laboratoire de Chimie, UMR 5182 ENS/CNRS, Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364 Lyon cedex 07, France
| | | | | |
Collapse
|
13
|
Taguchi H, Burr G, Karle S, Planque S, Zhou YX, Paul S, Nishiyama Y. A mechanism-based probe for gp120-Hydrolyzing antibodies. Bioorg Med Chem Lett 2002; 12:3167-70. [PMID: 12372525 DOI: 10.1016/s0960-894x(02)00640-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
An antigenic peptide analogue consisting of HIV gp120 residues 421-431 (an antigen recognition site probe) with diphenyl amino(4-amidinophenyl)methanephosphonate located at the C-terminus (a catalytic site probe) was synthesized and its trypsin and antibody reactivity characteristics were studied. Antibodies to the peptide determinant recognized the peptidyl phosphonate probe. Trypsin was inhibited equipotently by the peptidyl phosphonate and its simple phosphonate counterpart devoid of the peptide determinant. The peptidyl phosphonate inhibited the gp120-hydrolyzing activity of a catalytic antibody light chain. It was bound covalently by the light chain and the binding was inhibited by the classical active-site directed inhibitor of serine proteinase, diisopropyl fluorophosphate. These results reveal that the peptidyl phosphonate ester can serve as a probe for the antigen recognition and catalytic subsites of proteolytic antibodies.
Collapse
Affiliation(s)
- Hiroaki Taguchi
- Chemical Immunology and Therapeutics Research Center, Department of Pathology and Laboratory Medicine, University of Texas-Houston Medical School, 6431 Fannin, Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Pietsch M, Gutschow M. Alternate substrate inhibition of cholesterol esterase by thieno[2,3-d][1,3]oxazin-4-ones. J Biol Chem 2002; 277:24006-13. [PMID: 11959857 DOI: 10.1074/jbc.m112252200] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In a kinetic study, the interaction of bovine pancreatic cholesterol esterase (CEase) with fused 1,3-oxazin-4-ones and 1,3-thiazin-4-ones was investigated, and the compounds were characterized as alternate substrate inhibitors. Inhibition assays were performed in the presence of sodium taurocholate with p-nitrophenyl butyrate as chromogenic substrate. Strong active site-directed inhibition was detected for 2-diethylaminothieno[2,3-d][1,3]oxazin-4-ones with a cycloaliphatic chain at positions 5,6. The most potent inhibitors, compounds 3 and 4, exhibited K(i) values of 0.58 and 1.86 microm, respectively. An exchange of the ring oxygen by sulfur and the removal of the cycloaliphatic moiety as well as the replacement of the thiophene ring by benzene led to a loss of inhibitory potency. CEase has the capability to catalyze the hydrolysis of representatives of fused 1,3-oxazin-4-ones as well as the highly stable 1,3-thiazin-4-ones by using an acylation-deacylation mechanism. Hydrolyses were performed in the presence of a high enzyme concentration, and products were identified spectrophotometrically and by means of high performance liquid chromatography. The kinetic parameters V(max)I and V(max)I/K(m)(I) for the CEase-catalyzed turnover were determined. The compounds whose enzyme-catalyzed hydrolysis followed first-order kinetics (K(m)(I) > 25 microm) failed to inhibit CEase. On the other hand, inhibitors 3 (initial concentration of 25 microm) and 4 (20 microm) were hydrolyzed by CEase under steady-state conditions in the first phase of the reaction. Rate-limiting deacylation was demonstrated in nucleophilic competition experiments with ethanol as acyl acceptor wherein the conversion of compound 3 was accelerated up to an ethanol concentration of 1.5 m. The characterization of these compounds (i.e. 3 and 4) as alternate substrate inhibitors is not only based on the verification of the CEase-catalyzed hydrolysis. It also rests upon the concurrence of corresponding K(i) values obtained in the inhibition assay compared with separately determined K(m)(I) values of their enzyme-catalyzed consumption, as could be predicted from the kinetic model used in this study.
Collapse
Affiliation(s)
- Markus Pietsch
- Pharmaceutical Institute, Poppelsdorf, University of Bonn, D-53115 Bonn, Germany
| | | |
Collapse
|
15
|
Abstract
Mammalian cysteine proteases of the papain superfamily are interesting targets for the development of new drugs against diseases connected to abnormal degradation of muscle or bone proteins. The high nucleophilicity of the active site of these proteases as well as the characteristics of the well-known epoxysuccinic acid derived cysteine protease inhibitors provided a basis for the design of new types of selective and irreversible inhibitors for these enzymes. We designed and synthesized a novel class of peptidic cysteine protease inhibitors containing aziridine-2,3-dicarboxylic acid as electrophilic amino acid. Three types of aziridinyl peptides that differ in the position of the aziridine building block within the peptide chain have been synthesized and tested as inhibitors of several cysteine proteases. Remarkable differences could be observed between the three types of inhibitors concerning their activity, stereospecificity, pH dependency of inhibition, and selectivity between different cysteine proteases, respectively, indicating that different binding modes of the three types of inhibitors in respect to their orientation in the S and S' binding sites of the enzymes may be present.
Collapse
Affiliation(s)
- T Schirmeister
- Department of Pharmaceutical Chemistry, Albert-Ludwigs University of Freiburg, Germany
| |
Collapse
|
16
|
Gütschow M, Kuerschner L, Neumann U, Pietsch M, Löser R, Koglin N, Eger K. 2-(diethylamino)thieno1,3ŏxazin-4-ones as stable inhibitors of human leukocyte elastase. J Med Chem 1999; 42:5437-47. [PMID: 10639285 DOI: 10.1021/jm991108w] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A series of 2-(diethylamino)thieno1,3ŏxazin-4-ones was synthesized and evaluated in vitro for inhibitory activity toward human leukocyte elastase (HLE). The Gewald thiophene synthesis was utilized to obtain several ethyl 2-aminothiophene-3-carboxylates. These precursors were subjected to a five-step route to obtain thieno2,3-d1,3ŏxazin-4-ones bearing various substituents at positions 5 and 6. Both thieno2,3-d and thieno3,2-d fused oxazin-4-ones possess extraordinary chemical stability, which was expressed as rate constants of the alkaline hydrolysis. The kinetic parameters of the HLE inhibition were determined. The most potent compound, 2-(diethylamino)-4H-1benzothieno2,3-d1,3ŏxazin-4-one, exhibited a K(i) value of 5.8 nM. 2-(Diethylamino)thieno1, 3ŏxazin-4-ones act as acyl-enzyme inhibitors of HLE, similar to the inhibition of serine proteases by 4H-3,1-benzoxazin-4-ones. The isosteric benzene-thiophene replacement accounts for an enhanced stability of the acyl-enzyme intermediates.
Collapse
Affiliation(s)
- M Gütschow
- Institute of Pharmacy, University of Leipzig, D-04103 Leipzig, Germany
| | | | | | | | | | | | | |
Collapse
|
17
|
Chen JM, Xu SL, Wawrzak Z, Basarab GS, Jordan DB. Structure-based design of potent inhibitors of scytalone dehydratase: displacement of a water molecule from the active site. Biochemistry 1998; 37:17735-44. [PMID: 9922139 DOI: 10.1021/bi981848r] [Citation(s) in RCA: 103] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Scytalone dehydratase (SD) is a molecular target of inhibitor design efforts aimed at protecting rice plants from the fungal disease caused by Magnaporthe grisea. As determined from X-ray diffraction data of an SD-inhibitor complex [Lundqvist et al. (1994) Structure (London) 2, 937-944], there is an extended hydrogen-bonding network between protein side chains, the inhibitor, and two bound water molecules. From models of SD complexed to quinazoline and benztriazine inhibitors, a new class of potent SD inhibitors involving the displacement of an active-site water molecule were designed. We were able to increase inhibitory potency by synthesizing compounds with a nitrile functionality displayed into the space occupied by one of the crystallographic water molecules. Sixteen inhibitors are compared. The net conversion of potent quinazoline and benztriazine inhibitors to cyanoquinolines and cyanocinnolines increased binding potency 2-20-fold. Replacement of the nitrile with a hydrogen atom lowered binding affinity 100-30,000-fold. X-ray crystallographic data at 1.65 A resolution on a SD-inhibitor complex confirmed that the nitrile functionality displaced the water molecule as intended and that a favorable orientation was created with tyrosines 30 and 50 which had been part of the hydrogen-bonding network with the water molecule. Additional data on inhibitors presented herein reveals the importance of two hydrogen-bonding networks toward inhibitory potency: one between Asn131 and an appropriately positioned inhibitor heteroatom and one between a bound water molecule and a second inhibitor heteroatom.
Collapse
Affiliation(s)
- J M Chen
- E.I. DuPont de Nemours Agricultural Products, Stine-Haskell Research Center, Newark, Delaware 19714, USA
| | | | | | | | | |
Collapse
|
18
|
Amour A, Reboud-Ravaux M, de Rosny E, Abouabdellah A, Bégue JP, Bonnet-Delpon D, Le Gall M. Stereoselective synthesis of peptidyl trifluoromethyl alcohols and ketones: inhibitory potency against human leucocyte elastase, cathepsin G, porcine pancreatic elastase and HIV-1 protease. J Pharm Pharmacol 1998; 50:593-600. [PMID: 9680068 DOI: 10.1111/j.2042-7158.1998.tb06892.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
New fluorinated inhibitors have been designed to target two major proteases-human leucocyte elastase and HIV-1 protease. Two series of beta-peptidyl trifluoromethyl alcohols (TFMAs) Z-L-Val-NH-*CH(Y)*CH(OH)-CF3, where *C is the chiral centre, varied in the nature of the substituent Y, a phenylethyl [-(CH2)2-C6H5] or an isopropyl [-CH(CH3)2] group. These TFMAs were first synthesized as two pairs of the syn and anti diastereoisomers. The inhibitory effects of these mixtures were then assessed on three serine proteases chosen on the basis of the aromatic and aliphatic nature of the substituents-human leucocyte elastase (HLE), human cathepsin G (HCG) and porcine pancreatic elastase (PPE). In the presence of detectable inhibition, each epimer at C2 was separated to determine its inhibition constant (Ki) towards HLE, HCG and PPE. The stereoisomerically pure TFMAs were then oxidized into peptidyl trifluoromethyl ketones (TFMKs) for similar inhibition assays. The absolute configuration of the compounds remained unknown. One epimer at C2 of each syn and anti TFMA with the phenylethyl substituent behaved as a competitive inhibitor towards HLE and HCG with inhibition constants below the millimolar range, whereas their TFMK counterparts were non-inhibitors. In the second series, the two ketones inhibited both elastases with Ki values in the micromolar range, whereas only the syn TFMA was active towards HLE (Ki = 5.65 x 10(-4)M). The tested compounds also had structural properties compatible with recognition by HIV-1 protease. The inhibition of the enzyme was observed with TFMK only (IC50 = 15-200 microM). The phenylethyl substituent promoted inhibition by a factor of 10 (IC50 = 15 microM) compared with the isopropyl substituent (IC50 = 200 microM) leading to selective inhibition of HIV-1 protease. Isomerically pure TFMKs were more potent towards HLE than the alcohols from which they were obtained. However, an enantiomerically pure TFMA selectively inhibited HLE unlike its TFMK analogue which also inhibited PPE. This last result together with the selective inhibition of HIV-1 protease by TFMK with a phenylethyl substituent might be relevant to the design of specific HLE and HIV-1 inhibitors as therapeutic agents.
Collapse
Affiliation(s)
- A Amour
- Département de Biologie Cellulaire et Supramoléculaire, Institut Jacques Monod, Université Paris VII, France
| | | | | | | | | | | | | |
Collapse
|
19
|
Babine RE, Bender SL. Molecular Recognition of Proteinminus signLigand Complexes: Applications to Drug Design. Chem Rev 1997; 97:1359-1472. [PMID: 11851455 DOI: 10.1021/cr960370z] [Citation(s) in RCA: 714] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Robert E. Babine
- Agouron Pharmaceuticals, Inc., 3565 General Atomics Court, San Diego, California 92121-1122
| | | |
Collapse
|
20
|
Affiliation(s)
- Hans-Hartwig Otto
- Department of Pharmaceutical Chemistry, University of Freiburg, Hermann-Herder-Str. 9, D-79104 Freiburg i.Br., Germany
| | | |
Collapse
|
21
|
Affiliation(s)
- R B Silverman
- Department of Pharmacology, University of Florida, Gainesville 32610, USA
| |
Collapse
|
22
|
Luisi G, Calcagni A, Pinnen F. ψ(SO2-NH) transition state isosteres of peptides. Synthesis of the glutathione disulfide analogue. Tetrahedron Lett 1993. [DOI: 10.1016/s0040-4039(00)77622-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|