Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Biochemistry ; 53(30): 4970-8, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25003821

ABSTRACT

Canavan disease (CD) is a fatal, childhood neurological disorder caused by mutations in the ASPA gene, leading to catalytic deficiencies in the aspartoacylase (ASPA) enzyme and impaired N-acetyl-l-aspartic acid metabolism in the brain. To study the possible structural defects triggered by these mutations, four ASPA missense mutations associated with different disease severities have been structurally characterized. These mutant enzymes each have overall structures similar to that of the native ASPA enzyme, but with varying degrees of alterations that offer explanations for the respective loss of catalytic activity. The K213E mutant, a nonconservative mutant associated with a mild disease phenotype, has minimal structural differences compared to the native enzyme. In contrast, the loss of van der Waals contacts in the F295S mutant and the loss of hydrophobic and hydrogen bonding interactions in the Y231C mutant lead to a local collapse of the hydrophobic core structure in the carboxyl-terminal domain, contributing to a decrease in protein stability. The structure of the E285A mutant, the most common clinical mutant, reveals that the loss of hydrogen bonding interactions with the carboxylate side chain of Glu285 disturbs the active site architecture, leading to altered substrate binding and lower catalytic activity. Our improved understanding of the nature of these structural defects provides a basis for the development of treatment therapies for CD.


Subject(s)
Amidohydrolases/chemistry , Amidohydrolases/deficiency , Canavan Disease/enzymology , Catalytic Domain/genetics , Mutation, Missense/genetics , Amidohydrolases/genetics , Canavan Disease/genetics , Cell Line , Crystallography, X-Ray , Humans , Structure-Activity Relationship
2.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 2): 442-50, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24531478

ABSTRACT

S-Adenosylmethionine (AdoMet) participates in a wide range of methylation and other group-transfer reactions and also serves as the precursor for two groups of quorum-sensing molecules that function as regulators of the production of virulence factors in Gram-negative bacteria. The synthesis of AdoMet is catalyzed by AdoMet synthetases (MATs), a ubiquitous family of enzymes found in species ranging from microorganisms to mammals. The AdoMet synthetase from the bacterium Campylobacter jejuni (cjMAT) is an outlier among this homologous enzyme family, with lower sequence identity, numerous insertions and substitutions, and higher catalytic activity compared with other bacterial MATs. Alterations in the structure of this enzyme provide an explanation for its unusual dimeric quaternary structure relative to the other MATs. Taken together with several active-site substitutions, this new structure provides insights into its improved kinetic properties with alternative substrates.


Subject(s)
Bacterial Proteins/chemistry , Campylobacter jejuni/chemistry , Methionine Adenosyltransferase/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Campylobacter jejuni/enzymology , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Methionine Adenosyltransferase/genetics , Models, Molecular , Molecular Sequence Data , Mutation , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Alignment , Sequence Homology, Amino Acid
3.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 12): 3244-52, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25478842

ABSTRACT

The aspartate pathway is essential for the production of the amino acids required for protein synthesis and of the metabolites needed in bacterial development. This pathway also leads to the production of several classes of quorum-sensing molecules that can trigger virulence in certain microorganisms. The second enzyme in this pathway, aspartate ß-semialdehyde dehydrogenase (ASADH), is absolutely required for bacterial survival and has been targeted for the design of selective inhibitors. Fragment-library screening has identified a new set of inhibitors that, while they do not resemble the substrates for this reaction, have been shown to bind at the active site of ASADH. Structure-guided development of these lead compounds has produced moderate inhibitors of the target enzyme, with some selectivity observed between the Gram-negative and Gram-positive orthologs of ASADH. However, many of these inhibitor analogs and derivatives have not yet achieved the expected enhanced affinity. Structural characterization of these enzyme-inhibitor complexes has provided detailed explanations for the barriers that interfere with optimal binding. Despite binding in the same active-site region, significant changes are observed in the orientation of these bound inhibitors that are caused by relatively modest structural alterations. Taken together, these studies present a cautionary tale for issues that can arise in the systematic approach to the modification of lead compounds that are being used to develop potent inhibitors.


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Aspartate-Semialdehyde Dehydrogenase/chemistry , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Streptococcus pneumoniae/enzymology , Vibrio cholerae/enzymology , Aspartate-Semialdehyde Dehydrogenase/metabolism , Aspartic Acid/metabolism , Biosynthetic Pathways , Catalytic Domain , Cholera/microbiology , Crystallography, X-Ray , Humans , Models, Molecular , Pneumococcal Infections/microbiology , Protein Conformation , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Streptococcus pneumoniae/chemistry , Streptococcus pneumoniae/metabolism , Vibrio cholerae/chemistry , Vibrio cholerae/metabolism
4.
Bioorg Med Chem ; 20(9): 2950-6, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22464683

ABSTRACT

Microbes that have gained resistance against antibiotics pose a major emerging threat to human health. New targets must be identified that will guide the development of new classes of antibiotics. The selective inhibition of key microbial enzymes that are responsible for the biosynthesis of essential metabolites can be an effective way to counter this growing threat. Aspartate semialdehyde dehydrogenases (ASADHs) produce an early branch point metabolite in a microbial biosynthetic pathway for essential amino acids and for quorum sensing molecules. In this study, molecular modeling and docking studies were performed to achieve two key objectives that are important for the identification of new selective inhibitors of ASADH. First, virtual screening of a small library of compounds was used to identify new core structures that could serve as potential inhibitors of the ASADHs. Compounds have been identified from diverse chemical classes that are predicted to bind to ASADH with high affinity. Next, molecular docking studies were used to prioritize analogs within each class for synthesis and testing against representative bacterial forms of ASADH from Streptococcus pneumoniae and Vibrio cholerae. These studies have led to new micromolar inhibitors of ASADH, demonstrating the utility of this molecular modeling and docking approach for the identification of new classes of potential enzyme inhibitors.


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Aspartate-Semialdehyde Dehydrogenase/metabolism , Binding Sites , Enzyme Inhibitors/chemical synthesis , Kinetics , Molecular Dynamics Simulation , Protein Structure, Tertiary , Streptococcus pneumoniae/enzymology , Vibrio cholerae/enzymology
5.
Acta Crystallogr D Biol Crystallogr ; 66(Pt 2): 205-12, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20124701

ABSTRACT

The enzyme aspartate semialdehyde dehydrogenase (ASADH) catalyzes a critical transformation that produces the first branch-point intermediate in an essential microbial amino-acid biosynthetic pathway. The first structure of an ASADH isolated from a fungal species (Candida albicans) has been determined as a complex with its pyridine nucleotide cofactor. This enzyme is a functional dimer, with a similar overall fold and domain organization to the structurally characterized bacterial ASADHs. However, there are differences in the secondary-structural elements and in cofactor binding that are likely to cause the lower catalytic efficiency of this fungal enzyme. Alterations in the dimer interface, through deletion of a helical subdomain and replacement of amino acids that participate in a hydrogen-bonding network, interrupt the intersubunit-communication channels required to support an alternating-site catalytic mechanism. The detailed functional information derived from this new structure will allow an assessment of ASADH as a possible target for antifungal drug development.


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/chemistry , Candida albicans/enzymology , Amino Acid Sequence , Aspartate-Semialdehyde Dehydrogenase/metabolism , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Protein Folding , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment
6.
Bioorg Med Chem Lett ; 20(2): 576-80, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20005097

ABSTRACT

Potent, highly selective and orally-bioavailable MMP-13 inhibitors have been identified based upon a (pyridin-4-yl)-2H-tetrazole scaffold. Co-crystal structure analysis revealed that the inhibitors bind at the S(1)(') active site pocket and are not ligands for the catalytic zinc atom. Compound 29b demonstrated reduction of cartilage degradation biomarker (TIINE) levels associated with cartilage protection in a preclinical rat osteoarthritis model.


Subject(s)
Matrix Metalloproteinase Inhibitors , Osteoarthritis/drug therapy , Picolinic Acids/chemistry , Protease Inhibitors/chemistry , Tetrazoles/chemistry , Administration, Oral , Animals , Binding Sites , Cartilage/drug effects , Cartilage/metabolism , Catalytic Domain , Crystallography, X-Ray , Disease Models, Animal , Drug Discovery , Matrix Metalloproteinase 13/metabolism , Picolinic Acids/chemical synthesis , Picolinic Acids/pharmacology , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacology , Rats , Tetrazoles/chemical synthesis , Tetrazoles/pharmacology , Zinc/chemistry
7.
Nat Struct Mol Biol ; 11(12): 1192-7, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15543157

ABSTRACT

MEK1 and MEK2 are closely related, dual-specificity tyrosine/threonine protein kinases found in the Ras/Raf/MEK/ERK mitogen-activated protein kinase (MAPK) signaling pathway. Approximately 30% of all human cancers have a constitutively activated MAPK pathway, and constitutive activation of MEK1 results in cellular transformation. Here we present the X-ray structures of human MEK1 and MEK2, each determined as a ternary complex with MgATP and an inhibitor to a resolution of 2.4 A and 3.2 A, respectively. The structures reveal that MEK1 and MEK2 each have a unique inhibitor-binding pocket adjacent to the MgATP-binding site. The presence of the potent inhibitor induces several conformational changes in the unphosphorylated MEK1 and MEK2 enzymes that lock them into a closed but catalytically inactive species. Thus, the structures reported here reveal a novel, noncompetitive mechanism for protein kinase inhibition.


Subject(s)
Enzyme Inhibitors/pharmacology , MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/chemistry , MAP Kinase Kinase 2/metabolism , Binding Sites , Conserved Sequence , Dimerization , Enzyme Inhibitors/chemistry , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Models, Molecular , Molecular Structure , Protein Structure, Quaternary , Structural Homology, Protein
8.
J Med Chem ; 51(1): 31-45, 2008 Jan 10.
Article in English | MEDLINE | ID: mdl-18072721

ABSTRACT

In light of accumulating evidence that aggressive LDL-lowering therapy may offer increased protection against coronary heart disease, we undertook the design and synthesis of a novel series of HMG-CoA reductase inhibitors based upon a substituted pyrazole template. Optimizing this series using both structure-based design and molecular property considerations afforded a class of highly efficacious and hepatoselective inhibitors resulting in the identification of (3 R,5 R)-7-[2-(4-fluoro-phenyl)-4-isopropyl-5-(4-methyl-benzylcarbamoyl)-2 H-pyrazol-3-yl]-3,5-dihydroxy-heptanoic (PF-3052334) as a candidate for the treatment of hypercholesterolemia.


Subject(s)
Heptanoic Acids/chemical synthesis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemical synthesis , Hypercholesterolemia/drug therapy , Liver/drug effects , Pyrazoles/chemical synthesis , Animals , Cholesterol, LDL/biosynthesis , Cholesterol, LDL/blood , Cricetinae , Guinea Pigs , Hepatocytes/drug effects , Hepatocytes/metabolism , Heptanoic Acids/chemistry , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , In Vitro Techniques , Liver/metabolism , Male , Mesocricetus , Muscle Cells/drug effects , Muscle Cells/metabolism , Pyrazoles/chemistry , Pyrazoles/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship
9.
Bioorg Med Chem Lett ; 18(23): 6171-4, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18951019

ABSTRACT

This paper reports a second generation MEK inhibitor. The previously reported potent and efficacious MEK inhibitor, PD-184352 (CI-1040), contains an integral hydroxamate moiety. This compound suffered from less than ideal solubility and metabolic stability. An oxadiazole moiety behaves as a bioisostere for the hydroxamate group, leading to a more metabolically stable and efficacious MEK inhibitor.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Oxadiazoles/chemical synthesis , Oxadiazoles/pharmacology , Antineoplastic Agents/chemistry , Benzamides/chemistry , Colonic Neoplasms/chemically induced , Colonic Neoplasms/drug therapy , Combinatorial Chemistry Techniques , Drug Screening Assays, Antitumor , Esters , Humans , Hydroxamic Acids/chemistry , Microsomes, Liver/drug effects , Molecular Structure , Oxadiazoles/chemistry , Structure-Activity Relationship
10.
J Med Chem ; 50(21): 5090-102, 2007 Oct 18.
Article in English | MEDLINE | ID: mdl-17880056

ABSTRACT

A new series of MEK1 inhibitors, the 4-anilino-5-carboxamido-2-pyridones, were designed and synthesized using a combination of medicinal chemistry, computational chemistry, and structural elucidation. The effect of variation in the carboxamide side chain, substitution on the pyridone nitrogen, and replacement of the 4'-iodide were all investigated. This study afforded several compounds which were either equipotent or more potent than the clinical candidate CI-1040 (1) in an isolated enzyme assay, as well as murine colon carcinoma (C26) cells, as measured by suppression of phosphorylated ERK substrate. Most notably, pyridone 27 was found to be more potent than 1 in vitro and produced a 100% response rate at a lower dose than 1, when tested for in vivo efficacy in animals bearing C26 tumors.


Subject(s)
Amides/chemical synthesis , Aniline Compounds/chemical synthesis , Antineoplastic Agents/chemical synthesis , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Pyridones/chemical synthesis , Amides/chemistry , Amides/pharmacology , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 2/chemistry , Male , Mice , Models, Molecular , Neoplasm Transplantation , Phosphorylation , Pyridones/chemistry , Pyridones/pharmacology , Rats , Structure-Activity Relationship
11.
Nat Commun ; 8: 15559, 2017 05 30.
Article in English | MEDLINE | ID: mdl-28555620

ABSTRACT

Inflammation and thrombosis occur together in many diseases. The leukocyte integrin Mac-1 (also known as integrin αMß2, or CD11b/CD18) is crucial for leukocyte recruitment to the endothelium, and Mac-1 engagement of platelet GPIbα is required for injury responses in diverse disease models. However, the role of Mac-1 in thrombosis is undefined. Here we report that mice with Mac-1 deficiency (Mac-1-/-) or mutation of the Mac-1-binding site for GPIbα have delayed thrombosis after carotid artery and cremaster microvascular injury without affecting parameters of haemostasis. Adoptive wild-type leukocyte transfer rescues the thrombosis defect in Mac-1-/- mice, and Mac-1-dependent regulation of the transcription factor Foxp1 contributes to thrombosis as evidenced by delayed thrombosis in mice with monocyte-/macrophage-specific overexpression of Foxp1. Antibody and small-molecule targeting of Mac-1:GPIbα inhibits thrombosis. Our data identify a new pathway of thrombosis involving leukocyte Mac-1 and platelet GPIbα, and suggest that targeting this interaction has anti-thrombotic therapeutic potential with reduced bleeding risk.


Subject(s)
Blood Platelets/immunology , Leukocytes/metabolism , Macrophage-1 Antigen/genetics , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombosis/immunology , Animals , Binding Sites , Bleeding Time , Blood Coagulation , Blood Platelets/cytology , Blood Platelets/metabolism , Carotid Arteries/pathology , Glucosamine/chemistry , Hemostasis , Inflammation , Leukocytes/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microcirculation , NIH 3T3 Cells , Partial Thromboplastin Time , Phagocytosis , Platelet Activation , Platelet Count , Protein Binding , Protein Domains , Signal Transduction , Thrombin/metabolism
13.
Article in English | MEDLINE | ID: mdl-17012784

ABSTRACT

The activation of the beta-carboxyl group of aspartate catalyzed by aspartokinase is the commitment step to amino-acid biosynthesis in the aspartate pathway. The first structure of a microbial aspartokinase, that from Methanococcus jannaschii, has been determined in the presence of the amino-acid substrate L-aspartic acid and the nucleotide product MgADP. The enzyme assembles into a dimer of dimers, with the interfaces mediated by both the N- and C-terminal domains. The active-site functional groups responsible for substrate binding and specificity have been identified and roles have been proposed for putative catalytic functional groups.


Subject(s)
Archaeal Proteins/chemistry , Aspartate Kinase/chemistry , Aspartic Acid/biosynthesis , Archaeal Proteins/metabolism , Aspartate Kinase/metabolism , Binding Sites , Catalysis , Feedback, Physiological , Methanococcus/chemistry , Methanococcus/enzymology , Models, Molecular , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/metabolism , Structure-Activity Relationship
14.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 10): 1340-5, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25286936

ABSTRACT

Homoserine O-acetyltransferase (HTA) catalyzes the formation of L-O-acetyl-homoserine from L-homoserine through the transfer of an acetyl group from acetyl-CoA. This is the first committed step required for the biosynthesis of methionine in many fungi, Gram-positive bacteria and some Gram-negative bacteria. The structure of HTA from Staphylococcus aureus (SaHTA) has been determined to a resolution of 2.45 Å. The structure belongs to the α/ß-hydrolase superfamily, consisting of two distinct domains: a core α/ß-domain containing the catalytic site and a lid domain assembled into a helical bundle. The active site consists of a classical catalytic triad located at the end of a deep tunnel. Structure analysis revealed some important differences for SaHTA compared with the few known structures of HTA.


Subject(s)
Acetyltransferases/chemistry , Bacterial Proteins/chemistry , Staphylococcus aureus/enzymology , Amino Acid Sequence , Apoenzymes/chemistry , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary
15.
Chem Biol Drug Des ; 79(1): 128-36, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22039970

ABSTRACT

The aspartate biosynthetic pathway provides essential metabolites for many important biological functions, including the production of four essential amino acids. As this critical pathway is only present in plants and microbes, any disruptions will be fatal to these organisms. An early pathway enzyme, l-aspartate-ß-semialdehyde dehydrogenase, produces a key intermediate at the first branch point of this pathway. Developing potent and selective inhibitors against several orthologs in the l-aspartate-ß-semialdehyde dehydrogenase family can serve as lead compounds for antibiotic development. Kinetic studies of two small molecule fragment libraries have identified inhibitors that show good selectivity against l-aspartate-ß-semialdehyde dehydrogenases from two different bacterial species, Streptococcus pneumoniae and Vibrio cholerae, despite the presence of an identical constellation of active site amino acids in this homologous enzyme family. Structural characterization of enzyme-inhibitor complexes have elucidated different modes of binding between these structurally related enzymes. This information provides the basis for a structure-guided approach to the development of more potent and more selective inhibitors.


Subject(s)
Anti-Bacterial Agents/chemistry , Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Aspartate-Semialdehyde Dehydrogenase/metabolism , Binding Sites , Catalytic Domain , Computer Simulation , Crystallography, X-Ray , Drug Design , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Propionates/chemistry , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/enzymology , Substrate Specificity , Vibrio cholerae/drug effects , Vibrio cholerae/enzymology
16.
J Amino Acids ; 2011: 352538, 2011.
Article in English | MEDLINE | ID: mdl-22332000

ABSTRACT

The aspartate pathway of amino acid biosynthesis is essential for all microbial life but is absent in mammals. Characterizing the enzyme-catalyzed reactions in this pathway can identify new protein targets for the development of antibiotics with unique modes of action. The enzyme aspartate ß-semialdehyde dehydrogenase (ASADH) catalyzes an early branch point reaction in the aspartate pathway. Kinetic, mutagenic, and structural studies of ASADH from various microbial species have been used to elucidate mechanistic details and to identify essential amino acids involved in substrate binding, catalysis, and enzyme regulation. Important structural and functional differences have been found between ASADHs isolated from these bacterial and fungal organisms, opening the possibility for developing species-specific antimicrobial agents that target this family of enzymes.

17.
J Biomol Screen ; 15(9): 1042-50, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20855558

ABSTRACT

The microbial threat to human health is growing due to the dramatic increase in the number of multidrug-resistant organisms. The decline in effective antibiotics available to treat these growing threats has provided greater urgency to the search for new antibiotics. Clearly, new approaches must be developed against novel targets to control these resistant infectious organisms. The screening of low molecular weight compounds against new protein targets provides an opportunity to identify novel inhibitors as starting points for the development of new antibiotics. Custom fragment libraries have been assembled and screened against 3 representative forms of a key enzyme in an essential microbial biosynthetic pathway. Although each of these aspartate semialdehyde dehydrogenases (ASADHs) catalyzes the same reaction and each shares identical active site functional groups, subtle differences in enzyme structures have led to different binding selectivity among the initial hits from these fragment libraries. Amino acid analogues have been identified that show selectivity for either the gram-negative or gram-positive bacterial enzyme forms. A series of benzophenone analogues selectively inhibit the gram-negative ASADH, whereas some haloacids and substituted aromatic acids have been found to inhibit only the fungal form of ASADH. Each of these low molecular weight compounds possesses high ligand binding efficiency for their target enzyme forms. These results support the goal of designing lead compounds that will selectively target ASADHs from different microbial species.


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/analysis , Enzyme Inhibitors/pharmacology , Small Molecule Libraries/analysis , Small Molecule Libraries/pharmacology , Amino Acids/analysis , Amino Acids/pharmacology , Benzophenones/analysis , Benzophenones/pharmacology , Biocatalysis/drug effects , Candida albicans/drug effects , Microbial Sensitivity Tests , Streptococcus pneumoniae/drug effects , Substrate Specificity , Vibrio cholerae/drug effects
18.
Eur J Cancer ; 45(9): 1589-96, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19188061

ABSTRACT

PURPOSE: There are currently no approved therapies for patients with metastatic pancreatic adenocarcinoma previously treated with gemcitabine. This Phase III trial evaluated the efficacy and safety of glufosfamide as compared with best supportive care (BSC) in this patient population. METHODS: Patients were randomised to glufosfamide plus BSC or to BSC alone with baseline performance status as a stratification factor. The primary end-point was overall survival. RESULTS: Three hundred and three patients were randomised: 148 to glufosfamide plus BSC and 155 to BSC alone. There was an 18% increase in overall survival for glufosfamide that was not statistically significant: hazard ratio (HR) 0.85 (95% confidence interval (CI) 0.66-1.08, p=0.19). Median survival was 105 (range 5-875) days for glufosfamide and 84 (range 2+ to 761) days for BSC. Grade 3/4 creatinine increase occurred in 6 patients on glufosfamide, including 4 with dosing errors. CONCLUSION: These results suggest low activity of glufosfamide in this very refractory patient population.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/secondary , Antineoplastic Agents, Alkylating/therapeutic use , Pancreatic Neoplasms/drug therapy , Phosphoramide Mustards/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/adverse effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Female , Glucose/analogs & derivatives , Humans , Ifosfamide/analogs & derivatives , Male , Middle Aged , Phosphoramide Mustards/adverse effects , Survival Analysis , Treatment Outcome , Gemcitabine
19.
J Biol Chem ; 283(23): 16216-25, 2008 Jun 06.
Article in English | MEDLINE | ID: mdl-18334478

ABSTRACT

The commitment step to the aspartate pathway of amino acid biosynthesis is the phosphorylation of aspartic acid catalyzed by aspartokinase (AK). Most microorganisms and plants have multiple forms of this enzyme, and many of these isofunctional enzymes are subject to feedback regulation by the end products of the pathway. However, the archeal species Methanococcus jannaschii has only a single, monofunctional form of AK. The substrate l-aspartate binds to this recombinant enzyme in two different orientations, providing the first structural evidence supporting the relaxed regiospecificity previously observed with several alternative substrates of Escherichia coli AK ( Angeles, T. S., Hunsley, J. R., and Viola, R. E. (1992) Biochemistry 31, 799-805 ). Binding of the nucleotide substrate triggers significant domain movements that result in a more compact quaternary structure. In contrast, the highly cooperative binding of the allosteric regulator l-threonine to multiple sites on this dimer of dimers leads to an open enzyme structure. A comparison of these structures supports a mechanism for allosteric regulation in which the domain movements induced by threonine binding causes displacement of the substrates from the enzyme, resulting in a relaxed, inactive conformation.


Subject(s)
Archaeal Proteins/chemistry , Aspartate Kinase/chemistry , Methanococcus/enzymology , Threonine/chemistry , Allosteric Regulation/physiology , Archaeal Proteins/genetics , Aspartate Kinase/genetics , Dimerization , Methanococcus/genetics , Protein Structure, Quaternary/physiology , Protein Structure, Tertiary/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Structure-Activity Relationship , Substrate Specificity/physiology
20.
Biochemistry ; 47(11): 3484-92, 2008 Mar 18.
Article in English | MEDLINE | ID: mdl-18293939

ABSTRACT

Canavan disease is a fatal neurological disorder caused by the malfunctioning of a single metabolic enzyme, aspartoacylase, that catalyzes the deacetylation of N-acetyl-L-aspartate to produce L-aspartate and acetate. The structure of human brain aspartoacylase has been determined in complex with a stable tetrahedral intermediate analogue, N-phosphonomethyl-L-aspartate. This potent inhibitor forms multiple interactions between each of its heteroatoms and the substrate binding groups arrayed within the active site. The binding of the catalytic intermediate analogue induces the conformational ordering of several substrate binding groups, thereby setting up the active site for catalysis. The highly ordered binding of this inhibitor has allowed assignments to be made for substrate binding groups and provides strong support for a carboxypeptidase-type mechanism for the hydrolysis of the amide bond of the substrate, N-acetyl- l-aspartate.


Subject(s)
Amidohydrolases/chemistry , Aspartic Acid/analogs & derivatives , Brain/enzymology , Organophosphorus Compounds/metabolism , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/genetics , Amidohydrolases/metabolism , Apoenzymes/antagonists & inhibitors , Apoenzymes/chemistry , Apoenzymes/genetics , Apoenzymes/metabolism , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Binding Sites/genetics , Carboxypeptidases/chemistry , Carboxypeptidases/metabolism , Catalysis , Crystallography, X-Ray , Enzyme Inhibitors/metabolism , Glycosylation , Humans , Mutagenesis, Site-Directed , Organophosphorus Compounds/chemistry , Protein Binding/genetics
SELECTION OF CITATIONS
SEARCH DETAIL