Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Protein Sci ; 4(10): 1966-76, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8535233

ABSTRACT

The proteolytic enzyme stromelysin-1 is a member of the family of matrix metalloproteinases and is believed to play a role in pathological conditions such as arthritis and tumor invasion. Stromelysin-1 is synthesized as a pro-enzyme that is activated by removal of an N-terminal prodomain. The active enzyme contains a catalytic domain and a C-terminal hemopexin domain believed to participate in macromolecular substrate recognition. We have determined the three-dimensional structures of both a C-truncated form of the proenzyme and an inhibited complex of the catalytic domain by X-ray diffraction analysis. The catalytic core is very similar in the two forms and is similar to the homologous domain in fibroblast and neutrophil collagenases, as well as to the stromelysin structure determined by NMR. The prodomain is a separate folding unit containing three alpha-helices and an extended peptide that lies in the active site of the enzyme. Surprisingly, the amino-to-carboxyl direction of this peptide chain is opposite to that adopted by the inhibitor and by previously reported inhibitors of collagenase. Comparison of the active site of stromelysin with that of thermolysin reveals that most of the residues proposed to play significant roles in the enzymatic mechanism of thermolysin have equivalents in stromelysin, but that three residues implicated in the catalytic mechanism of thermolysin are not represented in stromelysin.


Subject(s)
Enzyme Precursors/chemistry , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/chemistry , Protein Structure, Secondary , Amino Acid Sequence , Binding Sites , Collagenases/chemistry , Crystallography, X-Ray , Fibroblasts/enzymology , Humans , Hydrogen Bonding , Matrix Metalloproteinase 3 , Models, Molecular , Neutrophils/enzymology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry
2.
J Med Chem ; 33(2): 861-7, 1990 Feb.
Article in English | MEDLINE | ID: mdl-2299649

ABSTRACT

6-(5-Cholesten-3 beta-yloxy)hexyl 1-thio-beta-D-mannopyranoside (L-644,257) enhances natural host resistance in cyclophosphamide-treated mice against Pseudomonas aeruginosa in a dose-dependent manner. It is active sc, im, and ip but not orally. L-644,257 is substantially more protective against P. aeruginosa than its alpha anomer. The beta-L-fucose glycolipid is more effective when given im and ip than sc. The lactose and beta-D-glucose glycolipids were only marginally effective to nonprotective. The 17 beta-steroidal side chain of L-644,257 can be modified without substantial loss of protective activity.


Subject(s)
Adjuvants, Immunologic , Cholesterol/analogs & derivatives , Glycolipids/pharmacology , Opportunistic Infections/therapy , Animals , Chemical Phenomena , Chemistry , Cholesterol/chemical synthesis , Cholesterol/pharmacology , Dose-Response Relationship, Drug , Female , Glycolipids/adverse effects , Immunotherapy , Mice , Pseudomonas Infections/therapy , Structure-Activity Relationship
3.
J Med Chem ; 29(8): 1436-41, 1986 Aug.
Article in English | MEDLINE | ID: mdl-3488405

ABSTRACT

A new class of tricyclic arylacetic acids was synthesized and evaluated as antiinflammatory/analgesic agents as well as inhibitors of prostaglandin synthetase. 11H-Dibenzo[b,e][1,4]dioxepin-2-, -3, -7, and -8-acetic and alpha-methylacetic acids and their derivatives were prepared by cyclization of diaryl ether precursors or by condensation of catechol and an aryl dihalide. The most potent compound in the carrageenan foot edema assay was alpha-methyl-11H-dibenzo[b,e][1,4]dioxepin-8-acetic acid (1 mg/kg = 43% inhibition). The most potent enzyme inhibitors were the 2-acetic acid and the alpha-methyl-7-acetic acid (IC50 = 0.1 microM). Some of these compounds were also found to be highly ulcerogenic.


Subject(s)
Analgesics/chemical synthesis , Anti-Inflammatory Agents/chemical synthesis , Benzoxepins/chemical synthesis , Analgesics/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Benzoxepins/pharmacology , Edema/drug therapy , Gastrointestinal Hemorrhage/drug therapy , Male , Mice , Pain/drug therapy , Rats , Rats, Inbred Strains , Structure-Activity Relationship
4.
J Med Chem ; 35(2): 252-8, 1992 Jan 24.
Article in English | MEDLINE | ID: mdl-1310118

ABSTRACT

The anaphylatoxin C5a is implicated in a number of inflammatory diseases. It is a highly cationic protein with 13 of 74 amino acids being either arginine or lysine. A search focusing on positively charged molecules, particularly amine-containing functionalities, led to the discovery of substituted 4,6-diaminoquinolines 1 [N,N'-bis(4-amino-2-methyl-6-quinolyl)urea] and 7 [6-N-(2-chlorocinnamoyl)-4,6-diamino-2-methylquinoline] as inhibitors of C5a receptor binding. These two compounds inhibited the binding of radiolabeled C5a to its receptor isolated from human neutrophils with IC50's = 3.3 and 12 micrograms/mL, respectively. Our efforts to enhance their potencies by chemical modification revealed a narrow profile of potency for effective C5a receptor binding inhibition.


Subject(s)
Aminoquinolines/pharmacology , Complement C5a/metabolism , Receptors, Complement/antagonists & inhibitors , Aminoquinolines/chemical synthesis , Cell Degranulation/drug effects , Cell Degranulation/immunology , Complement C5a/antagonists & inhibitors , Humans , In Vitro Techniques , Leukotriene B4/metabolism , N-Formylmethionine Leucyl-Phenylalanine/metabolism , Neutrophils/drug effects , Receptor, Anaphylatoxin C5a , Structure-Activity Relationship
5.
J Med Chem ; 35(21): 3745-54, 1992 Oct 16.
Article in English | MEDLINE | ID: mdl-1433189

ABSTRACT

A thorough analysis of the mechanism of inhibition of human leukocyte elastase (HLE) by a monocyclic beta-lactam and the mechanism of beta-lactam hydrolysis led to the preparation of potent and highly stable inhibitors of HLE. This work led to the identification of 4-[(4-carboxyphenyl)-oxy]-3,3-diethyl-1- [[(phenylmethyl)amino]carbonyl]-2-azetidinone (2) as the first orally active inhibitor of human leukocyte elastase (HLE). Analogs of 2 with different substituents on the urea N were synthesized and evaluated for their activity in vitro against HLE as well as in vivo in a hamster lung hemorrhage model. Compounds with a methyl or a methoxy group in the para position of the benzene ring were very potent in both assays. The results are discussed on the basis of the proposed model for the binding of this class of inhibitors to HLE and a possible mechanism of inhibition is presented.


Subject(s)
Azetidines/pharmacology , Pancreatic Elastase/antagonists & inhibitors , beta-Lactams/pharmacology , Administration, Oral , Amino Acid Sequence , Animals , Azetidines/administration & dosage , Azetidines/chemistry , Cricetinae , Humans , Hydrolysis , Leukocyte Elastase , Magnetic Resonance Spectroscopy , Molecular Sequence Data , X-Ray Diffraction , beta-Lactams/administration & dosage
6.
J Med Chem ; 36(6): 771-7, 1993 Mar 19.
Article in English | MEDLINE | ID: mdl-8459404

ABSTRACT

The effect of changing the C-4 substituent of 3,3-diethyl-1-[(benzylamino)carbonyl]-2-azetidinone on inhibition of HLE and in a model of HLE-induced lung damage in hamsters was explored. Substituents at this position do not appear to interact strongly with HLE with the most potent compounds having k(obs)/[I] = 6900 M-1 s-1. However, substituents at this position had a marked effect on in vivo activity. The greatest oral activity in the lung hemorrhage assay was achieved with C-4 aryl carboxylic acid ethers (60-85% inhibition at 30 mg/kg po). Based upon the established mechanism of inhibition by these compounds, the C-4 substituent would be released, and therefore, the pharmacological potential of these C-4 substituents was of considerable concern. Fortunately, compounds containing 4-hydroxybenzoic acid and 4-hydroxyphenylacetic acid ethers at C-4 were among the most active analogs. These phenolic acids are also found as urinary metabolites in healthy humans. Other heteroaryls at C-4 were also orally active in this model despite relatively modest enzyme activity.


Subject(s)
Monobactams/chemical synthesis , Pancreatic Elastase/antagonists & inhibitors , Administration, Oral , Animals , Cricetinae , Hemorrhage/prevention & control , Leukocyte Elastase , Lung/drug effects , Lung/enzymology , Monobactams/pharmacology , Pancreatic Elastase/toxicity , Structure-Activity Relationship
7.
J Med Chem ; 40(6): 1026-40, 1997 Mar 14.
Article in English | MEDLINE | ID: mdl-9083493

ABSTRACT

Carboxyalkyl peptides containing a biphenylylethyl group at the P1' position were found to be potent inhibitors of stromelysin-1 (MMP-3) and gelatinase A (MMP-2), in the range of 10-50 nM, but poor inhibitors of collagenase (MMP-1). Combination of a biphenylylethyl moiety at P1', a tert-butyl group at P2', and a methyl group at P3' produced orally bioavailable inhibitors as measured by an in vivo model of MMP-3 degradation of radiolabeled transferrin in the mouse pleural cavity. The X-ray structure of a complex of a P1-biphenyl inhibitor and the catalytic domain of MMP-3 is described. Inhibitors that contained halogenated biphenylylethyl residues at P1' proved to be superior in terms of enzyme potency and oral activity with 2(R)-[2-(4'-fluoro-4-biphenylyl)ethyl]-4(S)-n-butyl-1,5-pentane dioic acid 1-(alpha(S)-tert-butylglycine methylamide) amide (L-758,354, 26) having a Ki of 10 nM against MMP-3 and an ED50 of 11 mg/kg po in the mouse pleural cavity assay. This compound was evaluated in acute (MMP-3 and IL-1 beta injection in the rabbit) and chronic (rat adjuvant-induced arthritis and mouse collagen-induced arthritis) models of cartilage destruction but showed activity only in the MMP-3 injection model (ED50 = 6 mg/kg iv).


Subject(s)
Dipeptides/pharmacology , Matrix Metalloproteinase Inhibitors , Protease Inhibitors/pharmacology , Animals , Arthritis/drug therapy , Binding Sites , Cartilage/drug effects , Crystallography, X-Ray , Dipeptides/chemical synthesis , Dipeptides/chemistry , Dipeptides/metabolism , Disease Models, Animal , Gelatinases/antagonists & inhibitors , Interleukin-1/administration & dosage , Interleukin-1/pharmacology , Magnetic Resonance Spectroscopy , Matrix Metalloproteinase 1 , Matrix Metalloproteinase 2 , Metalloendopeptidases/antagonists & inhibitors , Mice , Models, Molecular , Molecular Structure , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Rabbits , Rats , Recombinant Proteins/antagonists & inhibitors , Structure-Activity Relationship , Transferrin/metabolism , Zinc/chemistry , Zinc/metabolism
9.
Xenobiotica ; 38(2): 223-37, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18197560

ABSTRACT

N-(1-(3,5-dichlorobenzenesulfonyl)-2S-methyl-azetidine-2-carbonyl)-L-4-(2',6'-dimethoxyphenyl)phenylalanine (1) is a potent antagonist of the very late activating (VLA) antigen-4. During initial screening, 1 exhibited an apparent plasma clearance (CL) of 227 ml min(-1) kg(-1) in Sprague-Dawley rats following an intravenous bolus dose formulated in an aqueous solution containing 40% polyethylene glycol. Such a high CL value led to speculation that the elimination of compound 1 involved extra-hepatic tissues. However, the apparent plasma CL was reduced to 97 ml in(-1) kg(-1) when a 2-min time point was added to sample collections, and further decreased to 48 ml min(-1) kg(-1) after the dose was formulated in rat plasma. The lung extraction of 1 in rats was negligible whereas the hepatic extraction was > or =90%, based on comparison of area under the curve (AUC) values derived from intra-artery, intravenous, and portal vein administration. In rats dosed intravenously with [(14)C]-1, approximately 91% of the radioactivity was recovered in bile over 48 h, with 85% accounted for in the first 4-h samples. The biliary radioactivity profile consisted of approximately 30% intact parent compound, 20% 1-glucuronide, and 50% oxidation products resulting from O-demethylation or hydroxylation reactions. When incubated with rat liver microsomes, oxidative metabolism of 1 was inhibited completely by 1-aminobenzotriazole (ABT), whereas the oxidation and glucuronidation reactions were little affected in the presence of cyclosporin A (CsA). In contrast, the hepatic extraction of 1 in rats was unperturbed in animals pre-dosed with ABT, but was reduced approximately 60% following treatment with CsA. In vitro, 1 was a substrate of the rat organic anion transporter Oatp1b2, and its cellular uptake was inhibited by CsA. In addition, the hepatic extraction of 1 was approximately 30% lower in Eisai hyperbilirubinaemic rats which lack functional multidrug resistant protein-2 (MRP2). Collectively, these data suggest that the clearance of 1 in rats likely is a result of the combined processes of hepatic oxidation, glucuronidation and biliary excretion, all of which are facilitated by active hepatic uptake of parent compound and subsequent active efflux of both unchanged parent and its metabolites into bile. It was concluded, therefore, that multiple mechanisms contribute to the clearance of 1 in rats, and suggest that appropriate pharmacokinetic properties might be difficult to achieve for this class of compounds. This case study demonstrates that an integrated strategy, incorporating both rapid screening and mechanistic investigations, is of particular value in supporting drug discovery efforts and decision-making processes.


Subject(s)
Integrin alpha4beta1/antagonists & inhibitors , Phenylalanine/analogs & derivatives , Animals , Cells, Cultured , Cyclosporine/metabolism , Dogs , Inactivation, Metabolic , Microsomes, Liver/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Phenylalanine/metabolism , Phenylalanine/physiology , Rats , Rats, Sprague-Dawley , Solute Carrier Organic Anion Transporter Family Member 1B3 , Time Factors , Triazoles/pharmacology
10.
Agents Actions ; 29(3-4): 266-76, 1990 Mar.
Article in English | MEDLINE | ID: mdl-2160187

ABSTRACT

Control of myeloperoxidase (MPO) may be an important consideration in disorders where excessive PMN elastase activity is a significant factor. There are, however, two mechanisms for the apparent regulation of MPO: 1) inhibit the enzyme directly, and ii) prevent the ensuing HOC1 induced oxidation by using a surrogate reducing agent. Appropriate methodology has been devised to distinguish true MPO inhibitors. With the exception of NaN3, many MPO inhibitors fall into the latter category and do not actually regulate the enzyme. Several potent organic inhibitors have been discovered, which, because of their structural selectivity, appear to associate specifically with a binding site on the enzyme, rather than attaching indiscriminately to a hydrophobic domain. By controlling the enzyme, these compounds protect alpha-1-PI from MPO induced damage, and could serve better than antioxidants to define the role of MPO in elastase induced injury.


Subject(s)
Naphthalenes/pharmacology , Peroxidase/antagonists & inhibitors , Animals , Binding Sites , Dianisidine/metabolism , Dogs , Hydrogen Peroxide/metabolism , Hypochlorous Acid , Leukocyte Elastase , Methionine/pharmacology , Molecular Structure , Oxidation-Reduction , Pancreatic Elastase/metabolism , Peroxidase/metabolism , Structure-Activity Relationship , alpha 1-Antitrypsin/metabolism
11.
Biochemistry ; 33(6): 1356-65, 1994 Feb 15.
Article in English | MEDLINE | ID: mdl-8312254

ABSTRACT

We have investigated the inhibition of the human matrix metalloproteinase stromelysin (SLN) by the N-(carboxyalkyl)dipeptide Ala[N]hPhe-Leu-anilide and find that it is a competitive, slow-binding inhibitor of this enzyme with Ki = 3 x 10(-8) M (pH 6.0, 25 degrees C). The dependence of k(obs), the observed first-order rate constant for the approach to steady state, on Ala[N]hPhe-Leu-anilide concentrations less than 10(-5) M is linear and suggests a simple, one-step mechanism with kon = 3.4 x 10(4) M-1 s-1 and k(off) = 1.2 x 10(-3) s-1 (pH 6.0, 25 degrees C). Using rapid kinetic techniques, we extended the concentration range of Ala[N]hPhe-Leu-anilide to 2 x 10(-3) M and found that the [Ala[N]hPhe-Leu-anilide] dependence of K(obs) suggests saturation kinetics with a Ki' near 5 x 10(-4) M. Detailed analysis of these data reveal that the dependence of k(obs) on [Ala[N]hPhe-Leu-anilide] is, in fact, sigmoidal. To probe the chemical mechanism of inhibition, we determined pH and temperature dependencies and solvent deuterium isotope effects. For k(on), delta H not equal to = 12.4 kcal/mol and -T delta S not equal to = 6.2 kcal/mol (T = 298 K; [I]steady-state = 10(-6) M), while for k(off), delta H not equal to = 12.5 kcal/mol and -T delta S not equal to = 8.9 kcal/mol (T = 298 K). pH dependencies of the kinetic parameters for inhibition are complex but reflect greater potency at lower pH and suggest a mechanism involving the same active-site groups that are involved in catalysis.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Dipeptides/pharmacology , Metalloendopeptidases/antagonists & inhibitors , Amino Acid Sequence , Binding, Competitive , Deuterium , Dipeptides/chemistry , Humans , Hydrogen-Ion Concentration , Kinetics , Matrix Metalloproteinase 3 , Molecular Sequence Data , Temperature , Thermodynamics
12.
Int J Immunopharmacol ; 12(2): 241-6, 1990.
Article in English | MEDLINE | ID: mdl-2329016

ABSTRACT

A steroidal glycolipid that enhances the nonspecific cellular response to opportunistic infection in an immunocompromised host has been discovered. A dose dependent response with 6-(5-cholesten-3 beta-yloxy)hexyl 1-thio-beta-D-mannopyranoside, L-644,257, was observed against several infective agents including bacterial, fungal, and viral pathogens in cyclophosphamide-treated mice. A mechanism for this protective action is proposed.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cholesterol/analogs & derivatives , Immunity, Innate/drug effects , Adjuvants, Immunologic/administration & dosage , Administration, Oral , Animals , Candidiasis/immunology , Cholesterol/administration & dosage , Cholesterol/pharmacology , Female , Injections, Intramuscular , Injections, Intraperitoneal , Injections, Subcutaneous , Mice , Pseudomonas Infections/immunology , Staphylococcal Infections/immunology , Time Factors
13.
Biochemistry ; 31(19): 4535-40, 1992 May 19.
Article in English | MEDLINE | ID: mdl-1581308

ABSTRACT

A determination of the zinc stoichiometry of the catalytic domain of the human matrix metalloproteinase stromelysin-1 has been carried out using enzyme purified from recombinant Escherichia coli that express C-terminally truncated protein. Atomic absorption spectrometry revealed that both the proenzyme (prostrom255) and the mature active form (strom255) contained nearly 2 mol of Zn/mol of protein. Full-length prostromelysin purified from a mammalian cell culture line also contained zinc in excess of 1 equiv. While zinc in prostrom255 could not be removed by dialysis against o-phenanthroline, similar treatment of mature strom255 resulted in the loss of one-half of the original zinc content. The peptidase activity of the zinc-depleted protein was reduced by greater than 85% but could be restored upon addition of Zn2+ or Co2+. Addition of a thiol-containing inhibitor to a CoZn hybrid enzyme resulted in marked spectral changes in both the visible and ultraviolet regions characteristic of sulfur ligation to Co2+. This direct evidence for an integral role in catalysis and inhibitor binding confirms the location of the exchangeable metal at the active site. To examine the environment of zinc in the proenzyme, a fully cobalt-substituted proenzyme was prepared by in vivo metal replacement. The absorbance features of dicobalt prostrom255 were consistent with metal coordination by the single cysteine present in the propeptide, although the data do not allow assignment to a particular zinc site.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Carrier Proteins/chemistry , Cysteine/chemistry , Enzyme Precursors/chemistry , Metalloendopeptidases/chemistry , Zinc/chemistry , Amino Acid Sequence , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Catalysis , Cobalt/chemistry , Enzyme Precursors/genetics , Humans , Matrix Metalloproteinase 3 , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Protein Binding , Recombinant Proteins/chemistry , Substrate Specificity
14.
Biochemistry ; 32(48): 13098-108, 1993 Dec 07.
Article in English | MEDLINE | ID: mdl-8241164

ABSTRACT

Stromelysin-1, a member of the matrix metalloendoprotease family, is a zinc protease involved in the degradation of connective tissue in the extracellular matrix. As a step toward determining the structure of this protein, multidimensional heteronuclear NMR experiments have been applied to an inhibited truncated form of human stromelysin-1. Extensive 1H, 13C, and 15N sequential assignments have been obtained with a combination of three- and four-dimensional experiments. On the basis of sequential and short-range NOEs and 13C alpha chemical shifts, two helices have been delineated, spanning residues Asp-111 to Val-127 and Leu-195 to Ser-206. A third helix spanning residues Asp-238 to Gly-247 is characterized by sequential NOEs and 13C alpha chemical shifts, but not short-range NOEs. The lack of the latter NOEs suggests that this helix is either distorted or mobile. Similarly, sequential and interstrand NOEs and 13C alpha chemical shifts characterize a four-stranded beta-sheet with three parallel strands (Arg-100 to Ile-101, Ile-142 to Ala-147, Asp-177 to Asp-181) and one antiparallel strand (Ala-165 to Tyr-168). Two zinc sites have been identified in stromelysin [Salowe et al. (1992) Biochemistry 31, 4535-4540]. The NMR spectral properties, including chemical shift, pH dependence, and proton coupling of the imidazole nitrogens of six histidine residues (151, 166, 179, 201, 205, and 211), invariant in the matrix metalloendoprotease family, suggest that these residues are zinc ligands. NOE data indicate that these histidines form two clusters: one ligates the catalytic zinc (His-201, -205, and -211), and the other ligates a structural zinc (His-151, -166, and -179). Heteronuclear multiple quantum correlated spectra and specific labeling experiments indicate His-151, -179, -201, -205, and -211 are in the N delta 1H tautomer and His-166 is in the N epsilon 2H tautomer.


Subject(s)
Metalloendopeptidases/ultrastructure , Amino Acid Sequence , Histidine/chemistry , Humans , Hydrogen Bonding , Imidazoles/chemistry , Ligands , Magnetic Resonance Spectroscopy , Matrix Metalloproteinase 3 , Metalloendopeptidases/metabolism , Molecular Sequence Data , Protein Structure, Secondary , Recombinant Proteins , Zinc/metabolism
15.
J Biomol NMR ; 7(1): 8-28, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8720828

ABSTRACT

Stromelysin-1 is a matrix metalloprotease that has been implicated in a number of degenerative diseases. Here we present the refined NMR solution structure of the catalytic domain of stromelysin-1 complexed with a small inhibitor and compare it to the X-ray crystal structure of the same complex. The structures are similar in global fold and show an unusual bottomless S1' subsite. There are differences, however, in the least well defined regions, Phe83-Ile89, His224-Phe232 and Pro249- Pro250, reflecting the lack of NOE data and large B-factors. The region His224-Phe232 contains residues of the S1' subsite and, consequently, small differences are observed in this subsite. Hydrogen-bond data show that, in contrast to the crystal structure, the solution structure lacks a hydrogen bond between the amide of Tyr223 and the carbonyl of the P3' residue. Analysis of bound water shows two tightly bound water molecules both in the solution and the crystal structure; neither of these waters are in the inhibitor binding site.


Subject(s)
Metalloendopeptidases/chemistry , Protein Structure, Secondary , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Humans , Hydrogen Bonding , Magnetic Resonance Spectroscopy , Matrix Metalloproteinase 3 , Metalloendopeptidases/metabolism , Models, Molecular , Molecular Sequence Data , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
16.
Biochemistry ; 31(21): 4980-6, 1992 Jun 02.
Article in English | MEDLINE | ID: mdl-1599924

ABSTRACT

The cephalosporin derivatives L 658758 [1-[[3-(acetoxymethyl)-7 alpha-methoxy-8-oxo-5-thia-1-azabicyclo [4.2.0]oct-2-en-2-yl]carbonyl]proline S,S-dioxide] and L 659286 [1-[[7 alpha-methoxy-8-oxo-3-[[(1,2,5,6-tetrahydro-2-methyl-5,6-dioxo- 1,2,4-triazin-3-yl)thio]methyl]-5-thia-1-aza-(6R)-bicyclo[4.2.0]-o ct-2-en-2-yl]carbonyl]pyrrolidine S,S-dioxide] are mechanism based inhibitors of human leukocyte elastase (HLE). The mechanism involves initial formation of a Michaelis complex followed by acylation of the active site serine. The group on the 3'-methylene is liberated during the course of these reactions, followed by partitioning of an intermediate between hydrolysis to regenerate active enzyme and further modification to produce a stable HLE-inhibitor complex. The partition ratio of 2.0 obtained for the reaction with L 658758 approaches that of an optimal inhibitor. These compounds are functionally irreversible inhibitors as the recovery of activity after inactivation is slow. The half-lives at 37 degrees C of the L 658758 and L 659286 derived HLE-I complexes were 9 and 6.5 h, respectively. The complexes produced by both inhibitors are similar chemically since the thermodynamic parameters for activation to regenerate active enzyme are essentially identical. The free energy of activation for this process is dominated primarily by the enthalpy term. The stability of the final complexes likely arises from Michael addition on the active site histidine to the 3'-methylene.


Subject(s)
Cephalosporins/pharmacology , Pancreatic Elastase/antagonists & inhibitors , Pyrrolidines/pharmacology , Catalysis , Chromatography, High Pressure Liquid , Humans , Kinetics , Leukocyte Elastase , Spectrophotometry, Ultraviolet
17.
Biochemistry ; 30(26): 6476-83, 1991 Jul 02.
Article in English | MEDLINE | ID: mdl-1647201

ABSTRACT

Stromelysin-1 is a member of a tissue metalloproteinase family whose members are all capable of degrading extracellular matrix components. A truncated form of human fibroblast prostromelysin 1 lacking the C-terminal, hemopexin-like domain has been expressed in Escherichia coli and purified to homogeneity. Treatment of this short form of prostromelysin with (aminophenyl)mercuric acetate resulted in activation and loss of the propeptide in a manner identical with the wild-type, full-length protein. Kinetic comparisons using Nle11-substance P as a substrate showed that the wild-type stromelysin and the truncated form of the enzyme had similar kcat and Km values. Likewise, both enzymes displayed similar Ki values for a hydroxamate-containing peptide inhibitor. Taken together, these results indicate that the C-terminal portion of stromelysin is not required for proper folding of the catalytic domain, maintenance of the enzyme in a latent form, activation with an organomercurial, cleavage of a peptide substrate, or interaction with an inhibitor. Moreover, the active short form of stromelysin displayed a reduction in the C-terminal heterogeneity, a characteristic degradation of the full-length stromelysin, and thereby provides a more suitable protein for future structural studies.


Subject(s)
Metalloendopeptidases/genetics , Amino Acid Sequence , Base Sequence , Binding Sites , Chromatography, Ion Exchange , Electrophoresis, Polyacrylamide Gel , Enzyme Precursors/isolation & purification , Enzyme Precursors/metabolism , Escherichia coli/genetics , Fibroblasts/enzymology , Humans , Hydroxamic Acids/pharmacology , Kinetics , Macromolecular Substances , Matrix Metalloproteinase 3 , Metalloendopeptidases/isolation & purification , Metalloendopeptidases/metabolism , Microbial Collagenase/genetics , Models, Structural , Molecular Sequence Data , Molecular Weight , Oligonucleotide Probes , Plasmids , Polymerase Chain Reaction/methods , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Restriction Mapping , Sequence Homology, Nucleic Acid
18.
J Biol Chem ; 274(13): 8694-7, 1999 Mar 26.
Article in English | MEDLINE | ID: mdl-10085108

ABSTRACT

We have identified a series of potent, orally bioavailable, non-peptidyl, triarylimidazole and triarylpyrrole glucagon receptor antagonists. 2-(4-Pyridyl)-5-(4-chlorophenyl)-3-(5-bromo-2-propyloxyphenyl)p yrr ole (L-168,049), a prototypical member of this series, inhibits binding of labeled glucagon to the human glucagon receptor with an IC50 = 3. 7 +/- 3.4 nM (n = 7) but does not inhibit binding of labeled glucagon-like peptide to the highly homologous human glucagon-like peptide receptor at concentrations up to 10 microM. The binding affinity of L-168,049 for the human glucagon receptor is decreased 24-fold by the inclusion of divalent cations (5 mM). L-168,049 increases the apparent EC50 for glucagon stimulation of adenylyl cyclase in Chinese hamster ovary cells expressing the human glucagon receptor and decreases the maximal glucagon stimulation observed, with a Kb (concentration of antagonist that shifts the agonist dose-response 2-fold) of 25 nM. These data suggest that L-168,049 is a noncompetitive antagonist of glucagon action. Inclusion of L-168, 049 increases the rate of dissociation of labeled glucagon from the receptor 4-fold, confirming that the compound is a noncompetitive glucagon antagonist. In addition, we have identified two putative transmembrane domain residues, phenylalanine 184 in transmembrane domain 2 and tyrosine 239 in transmembrane domain 3, for which substitution by alanine reduces the affinity of L-168,049 46- and 4. 5-fold, respectively. These mutations do not alter the binding of labeled glucagon, suggesting that the binding sites for glucagon and L-168,049 are distinct.


Subject(s)
Peptides/metabolism , Pyridines/pharmacology , Pyrroles/pharmacology , Receptors, Glucagon/antagonists & inhibitors , Receptors, Glucagon/metabolism , Adenylyl Cyclases/metabolism , Animals , CHO Cells , Cations, Divalent/pharmacology , Cricetinae , Enzyme Activation/drug effects , Glucagon-Like Peptide-1 Receptor , Glucagon-Like Peptides , Hormone Antagonists/chemistry , Hormone Antagonists/pharmacology , Humans , Molecular Structure , Mutation , Protein Binding/drug effects , Receptors, Glucagon/genetics
19.
Connect Tissue Res ; 33(4): 291-9, 1996.
Article in English | MEDLINE | ID: mdl-8834447

ABSTRACT

The objective of this study was to compare the specificity and potency of recombinant human SLN-1 (rhSLN) and human leukocyte elastase (HLE) as proteoglycan (PG)-degrading enzymes after intraarticular injection into rabbits. Another objective was to evaluate the elicitation of a rhSLN-induced hyaluronan-binding region (HABR) fragment from rabbit aggrecan in joints using a polyclonal antiserum (anti-FVDIPEN) against the synthetic peptide, Phe-Val-Asp-Ile-Pro-Glu-Asn (FVDIPEN). The intraarticular injection of either activated rhSLN or HLE resulted in enzyme-specific quantitative release of PG fragments into synovial fluid. Based on the criteria used herein, HLE appears to be a more potent PG-degrading enzyme than SLN. Intraarticular injection of rhSLN also resulted in time- and dose-dependent release of a new HABR fragment of aggrecan (HABR-FMDIPEN) into both articular cartilage and synovial fluid. HABR-FVDIPEN is likely to be a good marker of matrix metalloproteinase (MMP)-induced degradation of aggrecan.


Subject(s)
Extracellular Matrix Proteins , Joints/metabolism , Leukocyte Elastase/metabolism , Matrix Metalloproteinase 3/metabolism , Proteoglycans/metabolism , Synovial Fluid/metabolism , Aggrecans , Amino Acid Sequence , Animals , Cartilage, Articular/metabolism , Dose-Response Relationship, Drug , Female , Humans , Hyaluronic Acid/metabolism , Lectins, C-Type , Leukocyte Elastase/antagonists & inhibitors , Matrix Metalloproteinase Inhibitors , Molecular Sequence Data , Proteoglycans/drug effects , Rabbits , Time Factors
20.
Biochemistry ; 32(34): 8970-80, 1993 Aug 31.
Article in English | MEDLINE | ID: mdl-8364042

ABSTRACT

The kinetic and catalytic mechanisms of time-dependent inhibition of human polymorphonuclear leukocyte elastase (HLE) by the monocyclic beta-lactams described by Knight et al. [Knight, W.B., et al. (1992) Biochemistry 31, 8160] are investigated in this work. The dependence of the pseudo-first-order rate constant (k(obs)) on inhibitor concentration was saturable. The individual kinetic constants for the inhibition by L-680,833, [S-(R*,S*)]-4-[(1-(((1-(4- methylphenyl)butyl)amino)carbonyl)-3,3-diethyl-4-oxo-2- azetidinyl)oxy]benzeneacetic acid, and L-683,845, [S-(R*,S*)]-4-[(1-(((1-(5-benzofuranyl)butyl)amino)carbonyl)- 3,3-diethyl-4-oxo-2-azetidinyl)oxy]benzeneacetic acid, at pH 7.5 were k(inact) = 0.08 and 0.06 s-1 and Ki = 0.14 and 0.06 microM, respectively. The relative potency of this class of compounds as measured by k(inact)/Ki is primarily controlled by the Ki, term which ranged from 6 nM to 8 mM, while K(inact) was relatively insensitive to structural changes and varied by only an order of magnitude. Inactivation by the beta-lactams was efficient, requiring only 1.3 and 1.7 equiv of L-680,833 and L-683,845 to inactivate HLE. These values are indicative of some partitioning between turnover of inhibitor and inactivation. The partition ratio ranged as high as 3.5:1 depending upon the structure of the inhibitors, but this ratio was essentially independent of the availability and identity of a leaving group at C-4 of the lactam ring. Inactivation and partitioning liberate the leaving group when present at C-4. p-Hydroxy-m-nitrophenylacetic acid is liberated from this position at a rate similar to that for enzyme inactivation, suggesting kinetic competence of this process. Other products observed during the interaction of L-680,833 with HLE include a substituted urea, a species previously observed during the base-catalyzed decomposition of this class of compounds, and small amounts of products observed during reactivation of beta-lactam-derived HLE-I complexes. Both the pH dependence of k(inact)/Ki for the inactivation of HLE by [S-(R*,S*)]-4-[(1-(((1-(4-methylphenyl)butyl)amino)carbonyl)-3,3-diethyl - 4-oxo-2-azetidinyl)oxyl]benzoic acid and V/K for HLE-catalyzed substrate hydrolysis indicate that a single ionizable group with a pK of approximately 7 must be deprotonated for both processes. This group is likely the active site histidine. The data are consistent with initial formation of a Michaelis complex, acylation of the catalytic serine, and loss of the leaving group at C-4 of the original beta-lactam ring followed by partitioning between regeneration of active enzyme and production of a stable enzyme-inhibitor complex.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Pancreatic Elastase/antagonists & inhibitors , beta-Lactams/pharmacology , Catalysis , Humans , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Leukocyte Elastase
SELECTION OF CITATIONS
SEARCH DETAIL