Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Genome Announc ; 5(45)2017 Nov 09.
Article in English | MEDLINE | ID: mdl-29122874

ABSTRACT

Here, we report the draft genome sequences of three laboratory variants of Bacillus anthracis Sterne and their double (Δlef Δcya) and triple (Δpag Δlef Δcya) toxin gene deletion derivatives.

2.
Genome Announc ; 4(3)2016 May 12.
Article in English | MEDLINE | ID: mdl-27174274

ABSTRACT

Here, we present the complete genome sequences of two Zika virus (ZIKV) strains, Zika virus/Homo sapiens-tc/THA/2014/SV0127-14 and Zika virus/H. sapiens-tc/PHL/2012/CPC-0740, isolated from the blood of patients collected in Thailand, 2014, and the Philippines, 2012, respectively. Sequencing and phylogenetic analysis showed that both strains belong to the Asian lineage.

3.
Osteoarthritis Cartilage ; 18(9): 1159-66, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20633676

ABSTRACT

OBJECTIVE: To develop a short-term in vivo model in rats, with an enzyme-linked immunosorbent assay (ELISA) readout for specific aggrecanase-cleaved aggrecan fragments, to facilitate testing of aggrecanase inhibitors. METHODS: Monosodium iodoacetate (MIA), a metabolic inhibitor, was injected into the right knee joint of male Lewis rats and the release of aggrecanase-cleaved fragments of aggrecan containing the NITEGE or ARGN neoepitope was measured in the synovial fluid at 7 days post MIA injection using novel ELISAs. The ELISAs utilize a commercial antibody directed against the hyaluronic-acid binding region (HABR) of aggrecan, in combination with either an alpha-NITEGE antibody (NITEGE ELISA) or an alpha-ARGS/BC3 antibody (ARGS ELISA), to detect aggrecanase-cleavage of aggrecan within the interglobular domain (IGD). Aggrecan fragments present in in vitro digests, in cytokine-treated cartilage explant culture supernatants and in rat synovial fluid lavage samples were detected and quantified using the two ELISAs. Small molecule inhibitors of aggrecanase activity were dosed orally on days 3-7 to determine their ability to inhibit MIA-induced generation of the NITEGE and ARGN neoepitopes measured in the rat synovial fluid. RESULTS: The NITEGE assay was shown to specifically detect the N-terminal fragment of aggrecan comprising the G1 domain and the NITEGE neoepitope sequence. This assay can readily measure aggrecanase-cleaved bovine, human and rat aggrecan without the need for deglycosylation. The ARGS assay specifically detects C-terminal fragments of aggrecan comprising the ARGS/ARGN neoepitope and the G2 domain. Keratan sulfate (KS) residues of aggrecan interfere with this ELISA, and hence this assay works well with native rat articular cartilage aggrecan (that lacks KS residues) and with deglycosylated bovine and human aggrecan. Injection of MIA into the rat knee joints resulted in a time-dependent increase in the release of aggrecanase-cleaved aggrecan fragments into the synovial fluid and treatment with an aggrecanase inhibitor resulted in a dose-dependent inhibition of the generation of these neoepitopes. CONCLUSIONS: We have established a short-term in vivo model in rats that involves measurement of synovial fluid biomarkers that are dependent on aggrecanase activity in the joint. The short duration of the model combined with the mechanistic biomarker readout makes it very useful for the initial in vivo screening of aggrecanase inhibitors prior to testing them in time and resource-intensive disease models of osteoarthritis (OA).


Subject(s)
Aggrecans/metabolism , Endopeptidases/pharmacokinetics , Iodoacetates/pharmacology , Synovial Fluid/metabolism , Animals , Biomarkers/metabolism , Cattle , Endopeptidases/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Epitopes/analysis , Humans , Knee Joint/metabolism , Male , Rats , Rats, Inbred Lew
4.
Osteoarthritis Cartilage ; 18(9): 1150-8, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20633682

ABSTRACT

OBJECTIVE: Proteolytic degradation of aggrecan in articular cartilage is a hallmark feature of osteoarthritis (OA). The present study was aimed at developing a sensitive enzyme linked immunosorbent assay (ELISA) for the detection of aggrecanase-cleaved fragments of aggrecan in human serum and urine to facilitate the clinical development of aggrecanase inhibitors for OA. METHODS: The BC3 monoclonal antibody that detects the ARGS neoepitope sequence in aggrecanase-cleaved aggrecan was engineered and optimized using complementarity determining region (CDR)-saturation mutagenesis to improve its binding affinity to the neoepitope. A sandwich ELISA (BC3-C2 ELISA) was developed using the optimized alpha-ARGS antibody (BC3-C2) as capture antibody and a commercially available antibody directed against the hyaluronic-acid binding region (HABR) of aggrecan as detection antibody. Aggrecanase-cleaved fragments of aggrecan present in in vitro digests, human cartilage explant culture supernatants and in human synovial fluid, serum and urine were detected and quantified using this ELISA. RESULTS: The optimized antibody had a 4-log improvement in affinity for the ARGS containing peptide compared to the parental BC3 antibody, while maintaining the ability to not cross-react with a spanning peptide. The BC3-C2 ELISA demonstrated the ability to detect aggrecanase-cleaved aggrecan fragments in the native state, without the need for deglycosylation. This ELISA was able to measure aggrecanase-generated ARGS containing aggrecan fragments in human articular cartilage (HAC) explant cultures in the basal state (without cytokine stimulation). Treatment with an aggrecanase inhibitor resulted in a dose-dependent inhibition of ARGS neoepitope released into the culture supernatant. The ELISA assay also enabled the detection of ARGS containing fragments in human synovial fluid, serum and urine, suggesting its potential utility as a biomarker of aggrecanase activity. CONCLUSIONS: We have developed a novel ELISA using an optimized ARGS antibody and have demonstrated for the first time, an ELISA-based measurement of aggrecan degradation products in human serum and urine. This assay has the potential to serve as a mechanistic drug activity biomarker in the clinic and is expected to significantly impact/accelerate the clinical development of aggrecanase inhibitors and other disease modifying drugs for OA.


Subject(s)
ADAM Proteins/analysis , Aggrecans/analysis , Antibodies, Monoclonal , Cartilage, Articular/enzymology , Enzyme-Linked Immunosorbent Assay/methods , Peptide Fragments/analysis , Procollagen N-Endopeptidase/analysis , ADAMTS4 Protein , Aggrecans/immunology , Biomarkers , Cartilage, Articular/immunology , Creatinine/urine , Humans , Osteoarthritis, Knee/enzymology , Peptide Fragments/immunology , Synovial Fluid/enzymology
6.
J Org Chem ; 65(10): 2924-32, 2000 May 19.
Article in English | MEDLINE | ID: mdl-10814179

ABSTRACT

Further exploration of the scope of our solid-phase method for the synthesis of 3-aminobenzisoxazoles (using the Kaiser oxime resin 1) is described. The effects of base, leaving group, and solvent on the nucleophilic aromatic substitution based resin-loading reaction are discussed. Representative aryloxime intermediates were subjected to a variety of acidic conditions commonly used in protecting group removal to establish the acid stability profile of this linker. Regioselectivity was evaluated with various di- and trifluorobenzonitriles, which gave single benzisoxazole products after loading and cyclorelease reactions. Substituent effects observed in the course of the acid stability and regioselectivity studies suggest that the nitrile plays a critical role in the oxime hydrolysis mechanism. Finally, to establish the compatibility of the aryloxime linker with a variety of useful on-resin synthetic transformations, functionalized substrates were loaded onto resin 1, and carbon-nitrogen, carbon-oxygen, and carbon-carbon bond-forming reactions were successfully executed.


Subject(s)
Isoxazoles/chemical synthesis , Amides/chemistry , Chromatography, High Pressure Liquid , Hydrogen-Ion Concentration , Indicators and Reagents , Magnetic Resonance Spectroscopy , Oximes/chemical synthesis , Solvents , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared
7.
J Med Chem ; 43(5): 859-72, 2000 Mar 09.
Article in English | MEDLINE | ID: mdl-10715153

ABSTRACT

High-throughput screening of a combinatorial library of diamidophenols yielded lead compounds with the ability to inhibit human factor Xa (fXa) at micromolar concentrations (e.g. compound 4, fXa apparent K(ass) = 0.64 x 10(6) L/mol). SAR studies in this novel structural series of fXa inhibitors showed that the phenolic hydroxyl group was not essential for activity. The best activity was found in substituted 1,2-dibenzamidobenzenes in which the phenyl group of one benzoyl group (A-ring) was substituted in the 4-position with relatively small lipophilic or polarizable groups such as methoxy, vinyl, or chloro and the phenyl group of the other benzoyl group (B-ring) was substituted in the 4-position with larger lipophilic groups such as tert-butyl or dimethylamino. The central phenyl ring (C-ring) tolerated a wide variety of substituents, but methoxy, methanesulfonamido, hydroxyl, and carboxyl substitution produced slightly higher levels of activity than other substituents when present in combination with favorable B-ring substitution. Methylation of the amide nitrogen atoms was found to greatly decrease activity. Compound 12 is the highest affinity fXa inhibitor in this group of compounds, having fXa apparent K(ass) = 25.5 x 10(6) L/mol, about 40x more active than the original lead. This lead series does not show potent inhibition of human thrombin. A model for the binding of these ligands to the fXa active site is proposed. The model is consistent with the observed SAR and can serve to guide future SAR studies.


Subject(s)
Anticoagulants/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Factor Xa Inhibitors , Phenylenediamines/chemical synthesis , Sulfonamides/chemical synthesis , Thrombin/antagonists & inhibitors , Anticoagulants/chemistry , Anticoagulants/metabolism , Binding Sites , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Factor Xa/chemistry , Factor Xa/metabolism , Humans , Models, Molecular , Phenylenediamines/chemistry , Phenylenediamines/metabolism , Phenylenediamines/pharmacology , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/metabolism , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/metabolism , Sulfonamides/pharmacology , Thrombin/metabolism
8.
J Med Chem ; 43(5): 873-82, 2000 Mar 09.
Article in English | MEDLINE | ID: mdl-10715154

ABSTRACT

Reversal of the A-ring amide link in 1,2-dibenzamidobenzene 1 (fXa K(ass) = 0.81 x 10(6) L/mol) led to a series of human factor Xa (hfXa) inhibitors based on N(2)-aroylanthranilamide 4. Expansion of the SAR around 4 showed that only small planar substituents could be accommodated in the A-ring for binding to the S1 site of hfXa. Bulky groups such as 4-isopropyl, 4-tert-butyl, and 4-dimethylamino were favored in the B-ring to interact with the S4 site of hfXa. The central (C) ring containing a 5-methanesulfonamido group yielded greater activity than carbamoyl groups. Combining the beneficial features from the B- and C-ring SAR, compound 55 represents the most potent hfXa inhibitor in the N(2)-aroylanthranilamide 4 series with hfXa K(ass) = 58 x 10(6) L/mol (K(i) = 11.5 nM).


Subject(s)
Anticoagulants/chemical synthesis , Benzamides/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Factor Xa Inhibitors , Sulfonamides/chemical synthesis , Anticoagulants/chemistry , Anticoagulants/metabolism , Benzamides/chemistry , Benzamides/metabolism , Binding Sites , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Factor Xa/chemistry , Factor Xa/metabolism , Humans , Models, Molecular , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/metabolism
9.
J Med Chem ; 43(5): 883-99, 2000 Mar 09.
Article in English | MEDLINE | ID: mdl-10715155

ABSTRACT

To enhance the potency of 1,2-dibenzamidobenzene-derived inhibitors of factor Xa (fXa), an amidine substituent was incorporated on one of the benzoyl side chains to interact with Asp189 in the S1 specificity pocket. Lead molecule 1 was docked into the active site of fXa to facilitate inhibitor design. Subsequently, iterative SAR studies and molecular modeling led to a 1000-fold increase in fXa affinity and a refined model of the new inhibitors in the fXa active site. Strong support for the computational model was achieved through the acquisition of an X-ray crystal structure using thrombin as a surrogate protein. The amidines in this series show high levels of selectivity for the inhibition of fXa relative to other trypsin-like serine proteases. Furthermore, the fXa affinity of compounds in this series (K(ass) = 50-500 x 10(6) L/mol) translates effectively into both anticoagulant activity in vitro and antithrombotic activity in vivo.


Subject(s)
Amidines/chemical synthesis , Anticoagulants/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Factor Xa Inhibitors , Fibrinolytic Agents/chemical synthesis , Amidines/chemistry , Amidines/metabolism , Amidines/pharmacology , Animals , Anticoagulants/chemistry , Anticoagulants/metabolism , Anticoagulants/pharmacology , Binding Sites , Crystallography, X-Ray , Dogs , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Factor Xa/chemistry , Factor Xa/metabolism , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/pharmacology , Humans , In Vitro Techniques , Male , Models, Molecular , Prothrombin Time , Rabbits , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Thrombin/chemistry , Thrombin/metabolism , Thrombosis/drug therapy
10.
Bioorg Med Chem Lett ; 9(18): 2767-72, 1999 Sep 20.
Article in English | MEDLINE | ID: mdl-10509932

ABSTRACT

Modeling of neutral or mildly basic functional groups in the S1 site of thrombin led to the targeting of imidazole as a S1 binding element and correctly predicted the optimal chain length for connecting this group with the S2 and S3 binding elements. Derivatives of 4-(3-aminopropyl)-imidazole can be selective inhibitors of thrombin demonstrating potent anticoagulant activity.


Subject(s)
Antithrombins/chemical synthesis , Imidazoles/metabolism , Peptides/chemical synthesis , Antithrombins/chemistry , Antithrombins/pharmacology , Binding Sites , Crystallography, X-Ray , Drug Design , Humans , Models, Molecular , Peptides/chemistry , Peptides/pharmacology
11.
Science ; 273(5280): 1389-91, 1996 Sep 06.
Article in English | MEDLINE | ID: mdl-8703074

ABSTRACT

A modulator of the enzymatic activity of human thrombin, designated LY254603, was identified that enhances the thrombin-catalyzed generation of the anticoagulant factor activated protein C, yet inhibits thrombin-dependent fibrinogen clotting. By means of mutant substrates, it was shown that LY254603 mediates the change in enzymatic substrate specificity through an alteration in thrombin's S3 substrate recognition site, a mechanism that appeared to be independent of allosteric changes induced by either sodium ions or by thrombomodulin. This compound may represent the prototype of a class of agents that specifically modulates the balance between thrombin's procoagulant and anticoagulant functions.


Subject(s)
Fibrinogen/metabolism , Naphthalenes/pharmacology , Phenyl Ethers/pharmacology , Protein C/metabolism , Thrombin/pharmacology , Blood Coagulation/drug effects , Calcium/pharmacology , Choline/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Humans , Naphthalenes/chemistry , Partial Thromboplastin Time , Phenyl Ethers/chemistry , Protein C/chemistry , Protein Conformation , Recombinant Proteins/metabolism , Sodium Chloride/pharmacology , Substrate Specificity/drug effects , Thrombomodulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...