Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Chem Sci ; 10(45): 10595-10600, 2019 Dec 07.
Article in English | MEDLINE | ID: mdl-32110345

ABSTRACT

Protein-protein interactions involve hotspots as small as 4 sequential amino acids. Corresponding tetrapeptides have no structure in water. Here we report linking side chains of amino acids X and Z to form 24 cyclic tetrapeptides, cyclo-[XAAZ]-NH2, and stabilise 14-18 membered rings that mimic different kinds of non-regular secondary structures found in protein hotspots. 2D NMR spectra allowed determination of 3D structures for 14 cyclic tetrapeptides in water. Five formed two (i, i + 3) hydrogen bonds and a beta/gamma (6, 7) or beta (9, 19, 20) turn; eight formed one (i, i + 4) hydrogen bond and twisted into a non-helical (13, 18, 21, 22, 24) or helical (5, 17, 23) alpha turn; one was less structured (15). A beta or gamma turn was favoured for Z = Dab, Orn or Glu due to a χ1 gauche (+) rotamer, while an alpha turn was favoured for Z = Dap (but not X = Dap) due to a gauche (-) rotamer. Surprisingly, an unstructured peptide ARLARLARL could be twisted into a helix when either a helical or non-helical alpha turn (5, 13, 17, 18, 21-24) with Z = Dap was attached to the N-terminus. These structural models provide insights into stability for different turns and twists corresponding to non-regular folds in protein hotspots.

2.
Org Biomol Chem ; 14(48): 11525, 2016 Dec 28.
Article in English | MEDLINE | ID: mdl-27879976

ABSTRACT

Correction for 'Downsizing the BAD BH3 peptide to small constrained α-helices with improved ligand efficiency' by Nicholas E. Shepherd et al., Org. Biomol. Chem., 2016, DOI: 10.1039/c6ob02185a.

3.
Org Biomol Chem ; 14(46): 10939-10945, 2016 Nov 22.
Article in English | MEDLINE | ID: mdl-27819377

ABSTRACT

Bcl2 Homology (BH) proteins can either trigger or prevent programmed cell death or apoptosis. Deregulation of the BH protein family network leads to evasion of apoptosis, uncontrolled proliferation and is a hallmark of cancer. Inhibition of pro-survival BH proteins is a promising chemotherapeutic strategy for certain cancers. We have examined whether helix-constrained peptides based on the BAD BH3 domain (residues 103-127) can be downsized to much smaller more drug-like peptides. We report the preparation, structural characterisation, in vitro Bcl-xL inhibition and leukemic T-cell killing ability of 45 linear, mono-, bi- and tricyclic helical peptidomimetics between 8- and 19-residues in length. We show that the BAD BH3 can be downsized to 8-14 residues and still maintain appreciable affinity for Bcl-xL. In addition, the binding efficiency indices (BEI) of the downsized mimetics are significantly higher than the BAD BH3 and similar stapled BH3 mimetics, approaching drug-like molecules. This suggests that bicyclic and monocyclic mimetics based on BH3 domains are much more efficient binding ligands than the longer peptides which they mimic.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , bcl-Associated Death Protein/chemistry , Amino Acid Sequence , Cell Line, Tumor , Humans , Jurkat Cells , Ligands , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Domains , bcl-X Protein/antagonists & inhibitors
4.
Hum Vaccin Immunother ; 11(5): 1251-7, 2015.
Article in English | MEDLINE | ID: mdl-26018444

ABSTRACT

Na-APR-1(M74) is an aspartic protease that is rendered enzymatically inactive by site-directed mutagenesis and is a candidate antigen component in the Human Hookworm Vaccine. The mutant protease exerts vaccine efficacy by inducing antibodies that neutralize the enzymatic activity of wild type enzyme (Na-APR-1wt) in the gut of the hookworm, thereby depriving the worm of its ability to digest its blood meal. Previously, canines immunized with Na-APR-1(M74) and challenged with Ancylostoma caninum were partially protected against hookworm challenge infection, especially from the loss in hemoglobin observed in control canines and canine immunoglobulin (Ig) G raised against Na-APR-1 was shown to inhibit the enzymatic activity of Na-APR-1 wt in vitro, thereby providing proof of concept of Na-APR-1(M74) as a vaccine antigen. The mutated version, Na-APR-1(M74), was then expressed at the cGMP level using a Nicotiana benthamiana expression system (Fraunhofer, CMB, Delaware, MD), formulated with Alhydrogel®, and used to immunize mice in a dose-ranging study to explore the enzyme-neutralizing capacity of the resulting anti- Na-APR-1(M74) IgG. As little as 0.99 µg of recombinant Na-APR-1(M74) could induce anti Na-APR-1(M74) IgG in mice that were capable of inhibiting Na-APR-1w t-mediated digestion of a peptide substrate by 89%. In the absence of enzymatic activity of Na-APR-1(M74) as a surrogate marker of protein functionality, we developed an assay based on the binding of a quenched fluorescence-labeled inhibitor of aspartic proteases, BODIPY-FL pepstatin A (BDP). Binding of BDP in the active site of Na-APR-1 wt was demonstrated by inhibition of enzymatic activity, and competitive binding with unlabelled pepstatin A. BDP also bound to Na-APR-1(M74) which was assessed by fluorescence polarization, but with an ∼ 50-fold reduction in the dissociation constant. Taken together, these assays comprise a "toolbox" that could be useful for the analyses of Na-APR-1(M74) as it proceeds through the clinical development as part of the Human Hookworm Vaccine pipeline.


Subject(s)
Ancylostoma/enzymology , Ancylostomiasis/prevention & control , Antigens, Helminth/immunology , Aspartic Acid Proteases/immunology , Hookworm Infections/prevention & control , Recombinant Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Ancylostoma/immunology , Animals , Antibodies, Helminth/blood , Antibodies, Neutralizing/blood , Antigens, Helminth/genetics , Aspartic Acid Proteases/genetics , Drug Discovery/methods , Female , Mice, Inbred BALB C , Mutant Proteins/genetics , Mutant Proteins/immunology , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , Quality Control , Recombinant Proteins/genetics , Nicotiana/genetics , Nicotiana/metabolism , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
6.
J Med Chem ; 53(23): 8400-8, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-21067234

ABSTRACT

The nociceptin opioid peptide receptor (NOP, NOR, ORL-1) is a GPCR that recognizes nociceptin, a 17-residue peptide hormone. Nociceptin regulates pain transmission, learning, memory, anxiety, locomotion, cardiovascular and respiratory stress, food intake, and immunity. Nociceptin was constrained using an optimized helix-inducing cyclization strategy to produce the most potent NOP agonist (EC50 = 40 pM) and antagonist (IC50 = 7.5 nM) known. Alpha helical structures were measured in water by CD and 2D (1)H NMR spectroscopy. Agonist and antagonist potencies, evaluated by ERK phosphorylation in mouse neuroblastoma cells natively expressing NOR, increased 20-fold and 5-fold, respectively, over nociceptin. Helix-constrained peptides with key amino acid substitutions had much higher in vitro activity, serum stability, and thermal analgesic activity in mice, without cytotoxicity. The most potent agonist increased hot plate contact time from seconds up to 60 min; the antagonist prevented this effect. Such helix-constrained peptides may be valuable physiological probes and therapeutics for treating some forms of pain.


Subject(s)
Analgesics/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Opioid Peptides/pharmacology , Phosphorylation/drug effects , Temperature , Amino Acid Sequence , Analgesics/chemistry , Animals , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Molecular Sequence Data , Opioid Peptides/chemistry , Nociceptin
7.
J Med Chem ; 53(15): 5576-86, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20684600

ABSTRACT

Success in discovering bioactive peptide mimetics is often limited by the difficulties in correctly transposing known binding elements of the active peptide onto a small and metabolically more stable scaffold while maintaining bioactivity. Here we describe a scanning approach using a library of pyranose-based peptidomimetics that is structurally diverse in a systematic manner, designed to cover all possible conformations of tripeptide motifs containing two aromatic groups and one positive charge. Structural diversity was achieved by efficient selection of various chemoforms, characterized by a choice of pyranose scaffold of defined chirality and substitution pattern. A systematic scanning library of 490 compounds was thus designed, produced, and screened in vitro for activity at the somatostatin (sst(1-5)) and melanin-concentrating hormone (MCH(1)) receptors. Bioactive compounds were found for each target, with specific chemoform preferences identified in each case, which can be used to guide follow-on drug discovery projects without the need for scaffold hopping.


Subject(s)
Monosaccharides/chemistry , Oligopeptides/chemistry , Amino Acids/chemistry , Animals , Binding, Competitive , CHO Cells , Cricetinae , Cricetulus , Databases, Factual , Humans , Models, Molecular , Molecular Conformation , Molecular Mimicry , Monosaccharides/pharmacology , Oligopeptides/pharmacology , Radioligand Assay , Receptors, Somatostatin/antagonists & inhibitors , Stereoisomerism , Structure-Activity Relationship
8.
Proc Natl Acad Sci U S A ; 107(26): 11686-91, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20543141

ABSTRACT

Recombinant proteins are important therapeutics due to potent, highly specific, and nontoxic actions in vivo. However, they are expensive medicines to manufacture, chemically unstable, and difficult to administer with low patient uptake and compliance. Small molecule drugs are cheaper and more bioavailable, but less target-specific in vivo and often have associated side effects. Here we combine some advantages of proteins and small molecules by taking short amino acid sequences that confer potency and selectivity to proteins, and fixing them as small constrained molecules that are chemically and structurally stable and easy to make. Proteins often use short alpha-helices of just 1-4 helical turns (4-15 amino acids) to interact with biological targets, but peptides this short usually have negligible alpha-helicity in water. Here we show that short peptides, corresponding to helical epitopes from viral, bacterial, or human proteins, can be strategically fixed in highly alpha-helical structures in water. These helix-constrained compounds have similar biological potencies as proteins that bear the same helical sequences. Examples are (i) a picomolar inhibitor of Respiratory Syncytial Virus F protein mediated fusion with host cells, (ii) a nanomolar inhibitor of RNA binding to the transporter protein HIV-Rev, (iii) a submicromolar inhibitor of Streptococcus pneumoniae growth induced by quorum sensing pheromone Competence Stimulating Peptide, and (iv) a picomolar agonist of the GPCR pain receptor opioid receptor like receptor ORL-1. This approach can be generally applicable to downsizing helical regions of proteins with broad applications to biology and medicine.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Viral Proteins/chemistry , Viral Proteins/pharmacology , Amino Acid Sequence , Cell Line , Circular Dichroism , Humans , Models, Molecular , Molecular Mimicry , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Oligopeptides/chemistry , Oligopeptides/pharmacology , Protein Stability , Protein Structure, Secondary , Water
9.
J Med Chem ; 53(13): 4938-48, 2010 Jul 08.
Article in English | MEDLINE | ID: mdl-20527893

ABSTRACT

Human anaphylatoxin C3a, formed through cleavage of complement protein C3, is a potent effector of innate immunity via activation of its G protein coupled receptor, human C3aR. Previously reported short peptide ligands for this receptor either have low potency or lack receptor selectivity. Here we report the first small peptide agonists that are both potent and selective for human C3aR, derived from structure-activity relationships of peptides based on the C-terminus of C3a. Affinity for C3aR was examined by competitive binding with (125)I-labeled C3a to human PBMCs [corrected], agonist versus antagonist activity measured using fluorescence detection of intracellular calcium, and general selectivity monitored by C3a-induced receptor desensitization. An NMR structure for an agonist in DMSO showed a beta-turn motif that may be important for C3aR binding and activation. Derivatization produced a noncompetitive and insurmountable antagonist of C3aR. Small molecule C3a agonists and antagonists may be valuable probes of immunity and inflammatory diseases.


Subject(s)
Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Receptors, Complement/agonists , Receptors, Complement/antagonists & inhibitors , Binding, Competitive , Calcium/metabolism , Complement C3a/metabolism , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Oligopeptides/chemistry , Receptors, Complement/metabolism , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship , U937 Cells
11.
J Med Chem ; 53(6): 2651-5, 2010 Mar 25.
Article in English | MEDLINE | ID: mdl-20170165

ABSTRACT

Potent and noncovalent inhibitors of caspase-1 were produced by incorporating a secondary amine (reduced amide) isostere in place of the conventional electrophile (e.g., aldehyde) that normally confers high potency to cysteine protease inhibitors. Benzyl- or cyclohexylamines produced potent, reversible, and competitive inhibitors that were selective for caspase-1 (e.g., K(i) = 47 nM) over caspases 3 and 8 with minimal cytotoxicity. Unlike most cysteine protease inhibitors, these compounds do not react covalently and indiscriminately with thiols.


Subject(s)
Amines/pharmacology , Caspase Inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Amines/chemical synthesis , Amines/chemistry , Benzene/chemistry , Caspase 1/chemistry , Caspase 1/metabolism , Cell Survival/drug effects , Cyclohexylamines/chemical synthesis , Cyclohexylamines/chemistry , Cyclohexylamines/pharmacology , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/chemistry , HT29 Cells , Humans , Kinetics , Models, Chemical , Models, Molecular , Molecular Structure , Protein Binding , Protein Structure, Tertiary
12.
J Org Chem ; 75(1): 197-203, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19961235

ABSTRACT

The pyranose scaffold is unique in its ability to position pharmacophore substituents in various ways in 3D space, and unique pharmacophore scanning libraries could be envisaged that focus on scanning topography rather than diversity in the type of substituents. Approaches have been described that make use of amine and acid functionalities on the pyranose scaffolds to append substituents, and this has enabled the generation of libraries of significant structural diversity. Our general aim was to generate libraries of pyranose-based drug-like mimetics, where the substituents are held close to the scaffold, in order to obtain molecules with better defined positions for the pharmacophore substituents. Here we describe the development of a versatile synthetic route toward peptide mimetics build on 2-amino pyranose scaffolds. The method allows introduction of a wide range of substituent types, it is regio- and stereospecific, and the later diversity steps are performed on solid phase. Further, the same process was applied on glucose and allose scaffolds, in the exemplified cases, and is likely adaptable to other pyranose building blocks. The methods developed in this work give access to molecules that position the three selected binding elements in various 3D orientations on a pyranose scaffold and have been applied for the production of a systematically diverse library of several hundred monosaccharide-based mimetics.


Subject(s)
Amines/chemistry , Monosaccharides/chemistry , Monosaccharides/chemical synthesis , Peptides/chemistry , Peptides/chemical synthesis , Sugar Acids/chemistry , Sugar Acids/chemical synthesis , Combinatorial Chemistry Techniques , Glycosylation , Magnetic Resonance Spectroscopy , Molecular Structure
13.
J Am Chem Soc ; 131(43): 15877-86, 2009 Nov 04.
Article in English | MEDLINE | ID: mdl-19807085

ABSTRACT

Proteins typically consist of right-handed alpha helices, whereas left-handed alpha helices are rare in nature. Peptides of 20 amino acids or less corresponding to protein helices do not form thermodynamically stable alpha helices in water away from protein environments. The smallest known water-stable right- (alpha(R)) and left- (alpha(L)) handed alpha helices are reported, each stabilized in cyclic pentapeptide units containing all L- or all D-amino acids. Homochiral decapeptides comprising two identical cyclic pentapeptides (alpha(R)alpha(R) or alpha(L)alpha(L)) are continuous alpha-helical structures that are extremely stable to denaturants, degradative proteases, serum, and additives like TFE, acid, and base. Heterochiral decapeptides comprising two different cyclic pentapeptides (alpha(L)alpha(R) or alpha(R)alpha(L)) maintain the respective helical handedness of each monocyclic helical turn component but adopt extended or bent helical structures depending on the solvent environment. Adding TFE to their aqueous solutions caused a change to bent helical structures with slightly distorted N-terminal alpha(R) or alpha(L)-helical turns terminated by a Schellman-like motif adjacent to the C-terminal alpha(L) or alpha(R)-turn. This hinge-like switching between structures in response to an external cue suggests possible uses in larger structures to generate smart materials. The library of left- and right-handed 1-3 turn alpha-helical compounds reported herein project their amino acid side chains into very different regions of 3D space, constituting a unique and potentially valuable class of novel scaffolds.


Subject(s)
Oligopeptides/chemistry , Circular Dichroism , Glycine/chemistry , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Stereoisomerism
14.
J Med Chem ; 52(19): 6042-52, 2009 Oct 08.
Article in English | MEDLINE | ID: mdl-19743866

ABSTRACT

Human complement is a cascading network of plasma proteins important in immune defense, cooperatively effecting recognition, opsonization, destruction, and removal of pathogens and infected/damaged cells. Overstimulated or unregulated complement activation can result in immunoinflammatory diseases. Key serine proteases in this cascade are difficult to study due to their multiprotein composition, short lifetimes, formation on membranes, or serum circulation as inactive zymogens. Factor B is inactive at pH 7, but a catalytically active serine protease under alkaline conditions, enabling structure-activity relationship studies for 63 substrate-based peptide inhibitors with 4-7 residues and a C-terminal aldehyde. A potent factor B inhibitor was hexpeptide Ac-RLTbaLAR-H (IC(50) 250 nM, pH 9.5), which at pH 7 also blocked formation of membrane attack complex via the "alternative pathway" of complement activation and inhibited human complement mediated lysis of rabbit erythrocytes. Inhibitors of factor B may be valuable probes and drug leads for complement mediated immunity and disease.


Subject(s)
Complement Activation/drug effects , Complement Factor B/antagonists & inhibitors , Oligopeptides/pharmacology , Animals , Complement Pathway, Alternative , Erythrocytes , Hemolysis/drug effects , Humans , Molecular Mimicry , Oligopeptides/chemistry , Rabbits , Structure-Activity Relationship
15.
Biochemistry ; 48(35): 8466-72, 2009 Sep 08.
Article in English | MEDLINE | ID: mdl-19642650

ABSTRACT

The innate immune response to infection or injury involves an antigen-antibody triggered classical pathway (CP) of complement activation, in which soluble and cell surface plasma proteins cooperatively effect elimination of foreign organisms and damaged host cells. However, protracted or dysfunctional complement activation can lead to inflammatory diseases. Complement component 2 bound to C4b is cleaved by classical (C1s) or lectin (MASP2) proteases to produce C4bC2a, a very short-lived C3 convertase (t(1/2) 2 min) that in turn cleaves C3 to C3a and C3b, leading ultimately to formation of Membrane Attack Complex (MAC) and lysis of bacteria and damaged cells. C2 has the same serine protease domain as C4bC2a but in an inactive zymogen-like conformation, requiring cofactor-induced conformational change for activity. Here, we show that C2 has catalytic protease activity in its own right above pH 7, in the absence of cofactor, processing C3 and C3-derived chromogenic peptide fragments. In contrast to the instability of C3 convertase (t(1/2) 2 min, pH 7), the C2 enzyme is indefinitely stable under alkaline conditions, facilitating studies of its catalytic properties and development of small molecule inhibitors. We characterize the catalytic activity of C2 against C3 and short paranitroanilide peptide substrates, and identify potent small molecule inhibitors of C2 that also inhibit classical pathway C3 convertase, MAC formation, and hemolysis of sensitized sheep erythrocytes. These results provide a new avenue and valuable new insights to inhibiting CP complement activation relevant to inflammatory diseases.


Subject(s)
Complement C2/pharmacology , Complement C3-C5 Convertases/antagonists & inhibitors , Complement Pathway, Classical , Serine Endopeptidases/drug effects , Animals , Complement Activation , Complement C2/chemistry , Complement C2b/metabolism , Complement C3/metabolism , Complement C3-C5 Convertases/metabolism , Complement C3b/metabolism , Complement C4b/metabolism , Complement Membrane Attack Complex , Erythrocytes , Humans , Hydrogen-Ion Concentration , Protein Conformation , Protein Folding , Serine Endopeptidases/metabolism , Sheep
16.
Org Lett ; 11(14): 3092-5, 2009 Jul 16.
Article in English | MEDLINE | ID: mdl-19534544

ABSTRACT

A protein alpha-helix is defined by 3.6 amino acids per turn. Cyclization of the tripeptide Alanine-Leucine-Glutamate through a side chain to the N-terminus lactam bond produces cyclo-(1,3)-[ALE]-NH(2) which displays a circular dichroism spectrum typical of an alpha-helix backbone. However, proton NMR spectra show a novel cyclic peptide featuring two non-hydrogen-bonded antiparallel beta-strands connected by an Ala-Leu cis-amide bond. This example highlights that the common practice of characterizing alpha-helices by CD spectra alone can be misleading.


Subject(s)
Models, Molecular , Peptides, Cyclic/chemistry , Amino Acid Sequence , Circular Dichroism , Cyclization , Molecular Structure , Protein Structure, Secondary , Stereoisomerism
17.
Chem Commun (Camb) ; (43): 4501-3, 2007 Nov 21.
Article in English | MEDLINE | ID: mdl-17971969

ABSTRACT

Alkyl azides have been found to undergo an unexpectedly facile photodecomposition under mild conditions (laboratory light) and the reaction gives rise to small amounts of aldehydes and monoacyl aminal rearrangement products that can dramatically impact on the uses of azides in chemistry, biology and medicine.


Subject(s)
Azides/chemistry , Aldehydes/chemistry , Cysteine Proteinase Inhibitors/chemistry , Photolysis
18.
J Biol Chem ; 282(48): 34809-16, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17921140

ABSTRACT

Human complement factor B is the crucial catalytic component of the C3 convertase enzyme that activates the alternative pathway of complement-mediated immunity. Although a serine protease in its own right, factor B circulates in human serum as an inactive zymogen and there is a crystal structure only for the inactive state of factor B and various fragments. To provide greater insight to the catalytic function and properties of factor B, we have used short para-nitroanilide derivatives of 4- to 15-residue peptides as substrates to profile the catalytic properties of factor B. Among factors found to influence catalytic activity of factor B was an unusual dependence on pH. Non-physiological alkaline conditions strongly promoted substrate cleavage by factor B, consistent with a pH-accessible conformation of the enzyme that may be critical for catalytic function. Small N-terminal extensions to conventional hexapeptide para-nitroanilide substrates significantly increased catalytic activity of factor B, which was more selective for its cleavage site than trypsin. The new chromogenic assay enabled optimization of catalysis conditions, the profiling of different substrate sequences, and the development of the first reversible and competitive substrate-based inhibitor of factor B. The inhibitor was also shown to prevent in vitro formation of C3a from C3 by factor B, by synthetic and by natural C3 convertase of the alternative complement activation pathway, and to block formation of membrane attack complex. The availability of a reversible substrate-based inhibitor that could stabilize the active conformation of factor B, in conjunction with a pH-promoted higher processing activity, may offer a new avenue to obtain crystal structures of factor B and C3 convertase in an active conformation.


Subject(s)
Complement Factor B/antagonists & inhibitors , Complement Factor B/chemistry , Animals , Brain/metabolism , Catalysis , Complement System Proteins , Crystallography, X-Ray , Elapid Venoms/metabolism , Enzyme Inhibitors/pharmacology , Humans , Hydrogen-Ion Concentration , Hydrolysis , Ischemia , Kinetics , Models, Chemical , Substrate Specificity , Trypsin/chemistry
19.
J Am Chem Soc ; 128(38): 12396-7, 2006 Sep 27.
Article in English | MEDLINE | ID: mdl-16984172

ABSTRACT

Cysteine proteases are crucial regulatory enzymes in human physiology and disease. Inhibitors are usually designed with reactive electrophiles to covalently bond to the catalytic cysteinyl sulfur, and consequently they also indiscriminately interact with biological thiolates and other nucleophiles, leading to toxic side effects in vivo. Here we describe an alternative to using reactive electrophiles, demonstrating the use of a much less reactive azidomethylene substituent (-CH2-N3) that confers potent inhibition of cysteine proteases. This new approach resulted in potent, reversible, competitive inhibitors of caspase-1 (IC50 < 10 nM), with significant advantages over aldehydes such as high stability in vitro to thiols (10 mM dithiothreitol (pH 7.2), 20 mM glutathione (pH 7.2, 9, 11)) and aqueous media, as well as some highly desirable druglike features. It was also demonstrated that azides can be incorporated into inhibitors of other caspases (e.g. 3, 8) and cathepsins (e.g. K, S, B), indicating the versatility of this valuable new approach to cysteine protease inhibition.


Subject(s)
Azides/chemistry , Azides/pharmacology , Caspase Inhibitors , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Aldehydes/chemistry , Aldehydes/pharmacology , Binding Sites , Caspase 1/metabolism , Ketones/chemistry , Ketones/pharmacology , Magnetic Resonance Spectroscopy , Models, Molecular , Structure-Activity Relationship
20.
Curr Med Chem ; 12(25): 2963-77, 2005.
Article in English | MEDLINE | ID: mdl-16378499

ABSTRACT

TNF-alpha neutralising agents such as Infliximab (Remicade), Etanercept (Enbrel) and the IL-1 receptor antagonist Anakinra (Kineret), are currently used clinically for the treatment of many inflammatory diseases such as Crohn's disease, rheumatoid arthritis, ankylosing spondylitis, juvenile rheumatoid arthritis, psoriatic arthritis and psoriasis. These protein preparations are expensive to manufacture and administer, need to be injected and can cause allergic reactions. An alternative approach to lowering the levels of TNF-alpha and IL-1beta in inflammatory disease, is to inhibit the enzymes that generate these cytokines using cheaper small molecules. This paper is a broad overview of the progress that has been achieved so far, with respect to small molecule inhibitor design and pharmacological studies (in animals and humans), for the metalloprotease Tumour Necrosis Factor-alpha Converting Enzyme (TACE) and the cysteine protease Caspase-1 (Interleukin-1beta Converting Enzyme, ICE). Inhibitors of these two enzymes are currently considered to be good therapeutic targets that have the potential to provide relatively inexpensive and orally bioavailable anti-inflammatory agents in the future.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Caspase Inhibitors , Enzyme Inhibitors/pharmacology , ADAM17 Protein , Animals , Anti-Inflammatory Agents/chemistry , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...