Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Antimicrob Agents Chemother ; 68(4): e0137323, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38380945

ABSTRACT

Protease inhibitors (PIs) remain an important component of antiretroviral therapy for the treatment of HIV-1 infection due to their high genetic barrier to resistance development. Nevertheless, the two most commonly prescribed HIV PIs, atazanavir and darunavir, still require co-administration with a pharmacokinetic boosting agent to maintain sufficient drug plasma levels which can lead to undesirable drug-drug interactions. Herein, we describe GS-9770, a novel investigational non-peptidomimetic HIV PI with unboosted once-daily oral dosing potential due to improvements in its metabolic stability and its pharmacokinetic properties in preclinical animal species. This compound demonstrates potent inhibitory activity and high on-target selectivity for recombinant HIV-1 protease versus other aspartic proteases tested. In cell culture, GS-9770 inhibits Gag polyprotein cleavage and shows nanomolar anti-HIV-1 potency in primary human cells permissive to HIV-1 infection and against a broad range of HIV subtypes. GS-9770 demonstrates an improved resistance profile against a panel of patient-derived HIV-1 isolates with resistance to atazanavir and darunavir. In resistance selection experiments, GS-9770 prevented the emergence of breakthrough HIV-1 variants at all fixed drug concentrations tested and required multiple protease substitutions to enable outgrowth of virus exposed to escalating concentrations of GS-9770. This compound also remained fully active against viruses resistant to drugs from other antiviral classes and showed no in vitro antagonism when combined pairwise with drugs from other antiretroviral classes. Collectively, these preclinical data identify GS-9770 as a potent, non-peptidomimetic once-daily oral HIV PI with potential to overcome the persistent requirement for pharmacological boosting with this class of antiretroviral agents.


Subject(s)
HIV Infections , HIV Protease Inhibitors , HIV-1 , Humans , HIV Protease Inhibitors/pharmacology , HIV Protease Inhibitors/therapeutic use , Darunavir/pharmacology , Darunavir/therapeutic use , Atazanavir Sulfate/pharmacology , Atazanavir Sulfate/therapeutic use , Drug Resistance, Viral , HIV-1/genetics , Anti-Retroviral Agents/therapeutic use , HIV Infections/drug therapy , HIV Protease/genetics , HIV Protease/metabolism
2.
J Med Chem ; 66(3): 1761-1777, 2023 02 09.
Article in English | MEDLINE | ID: mdl-36652602

ABSTRACT

Non-nucleoside reverse transcriptase inhibitors (NNRTIs) represent cornerstones of current regimens for treatment of human immunodeficiency virus type 1 (HIV-1) infections. However, NNRTIs usually suffer from low aqueous solubility and the emergence of resistant viral strains. In the present work, novel bicyclic NNRTIs derived from etravirine (ETV) and rilpivirine (RPV), bearing modified purine, tetrahydropteridine, and pyrimidodiazepine cores, were designed and prepared. Compounds 2, 4, and 6 carrying the acrylonitrile moiety displayed single-digit nanomolar activities against the wild-type (WT) virus (EC50 = 2.5, 2.7, and 3.0 nM, respectively), where the low nanomolar activity was retained against HXB2 (EC50 = 2.2-2.8 nM) and the K103N and Y181C mutated strains (fold change, 1.2-6.7×). Most importantly, compound 2 exhibited significantly improved phosphate-buffered saline solubility (10.4 µM) compared to ETV and RPV (≪1 µM). Additionally, the binding modes of compounds 2, 4, and 6 to the reverse transcriptase were studied by X-ray crystallography.


Subject(s)
Anti-HIV Agents , HIV Infections , HIV-1 , Humans , Anti-HIV Agents/chemistry , HIV-1/metabolism , Reverse Transcriptase Inhibitors , HIV Reverse Transcriptase/metabolism , HIV Infections/drug therapy , Rilpivirine/therapeutic use , Drug Design
3.
J Biol Chem ; 293(51): 19659-19671, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30355736

ABSTRACT

Systemic light-chain amyloidosis (AL) is a human disease caused by overexpression of monoclonal immunoglobulin light chains that form pathogenic amyloid fibrils. These amyloid fibrils deposit in tissues and cause organ failure. Proteins form amyloid fibrils when they partly or fully unfold and expose segments capable of stacking into ß-sheets that pair and thereby form a tight, dehydrated interface. These structures, termed steric zippers, constitute the spines of amyloid fibrils. Here, using a combination of computational (with ZipperDB and Boston University ALBase), mutational, biochemical, and protein structural analyses, we identified segments within the variable domains of Ig light chains that drive the assembly of amyloid fibrils in AL. We demonstrate that there are at least two such segments and that each one can drive amyloid fibril assembly independently of the other. Our analysis revealed that peptides derived from these segments form steric zippers featuring a typical dry interface with high-surface complementarity and occupy the same spatial location of the Greek-key immunoglobulin fold in both λ and κ variable domains. Of note, some predicted steric-zipper segments did not form amyloid fibrils or assembled into fibrils only when removed from the whole protein. We conclude that steric-zipper propensity must be experimentally validated and that the two segments identified here may represent therapeutic targets. In addition to elucidating the molecular pathogenesis of AL, these findings also provide an experimental approach for identifying segments that drive fibril formation in other amyloid diseases.


Subject(s)
Amyloid/metabolism , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/metabolism , Immunoglobulin Light-chain Amyloidosis/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Amino Acid Sequence , Immunoglobulin Light-chain Amyloidosis/drug therapy , Models, Molecular , Molecular Targeted Therapy , Protein Domains
4.
Elife ; 4: e10935, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26576950

ABSTRACT

Overproduction of immunoglobulin light chains leads to systemic amyloidosis, a lethal disease characterized by the formation of amyloid fibrils in patients' tissues. Excess light chains are in equilibrium between dimers and less stable monomers which can undergo irreversible aggregation to the amyloid state. The dimers therefore must disassociate into monomers prior to forming amyloid fibrils. Here we identify ligands that inhibit amyloid formation by stabilizing the Mcg light chain variable domain dimer and shifting the equilibrium away from the amyloid-prone monomer.


Subject(s)
Amyloid/antagonists & inhibitors , Immunoglobulin Light Chains/metabolism , Ligands
5.
J Biol Chem ; 289(40): 27513-25, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25138218

ABSTRACT

Systemic light chain amyloidosis is a lethal disease characterized by excess immunoglobulin light chains and light chain fragments composed of variable domains, which aggregate into amyloid fibers. These fibers accumulate and damage organs. Some light chains induce formation of amyloid fibers, whereas others do not, making it unclear what distinguishes amyloid formers from non-formers. One mechanism by which sequence variation may reduce propensity to form amyloid fibers is by shifting the equilibrium toward an amyloid-resistant quaternary structure. Here we identify the monomeric form of the Mcg immunoglobulin light chain variable domain as the quaternary unit required for amyloid fiber assembly. Dimers of Mcg variable domains remain stable and soluble, yet become prone to assemble into amyloid fibers upon disassociation into monomers.


Subject(s)
Amyloid/chemistry , Amyloidosis/metabolism , Immunoglobulin Variable Region/chemistry , Amino Acid Sequence , Amyloid/genetics , Amyloid/metabolism , Amyloidosis/genetics , Crystallography, X-Ray , Humans , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Immunoglobulin lambda-Chains/genetics , Immunoglobulin lambda-Chains/metabolism , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary
6.
Org Biomol Chem ; 9(11): 4160-7, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21483943

ABSTRACT

Cyclodextrin-based host-guest chemistry has been exploited to facilitate co-crystallization of recombinant human acid ß-glucosidase (ß-glucocerebrosidase, GlcCerase) with amphiphilic bicyclic nojirimycin analogues of the sp(2)-iminosugar type. Attempts to co-crystallize GlcCerase with 5-N,6-O-[N'-(n-octyl)iminomethylidene]nojirimycin (NOI-NJ) or with 5-N,6-S-[N'-(n-octyl)iminomethylidene]-6-thionojirimycin (6S-NOI-NJ), two potent inhibitors of the enzyme with promising pharmacological chaperone activity for several Gaucher disease-associated mutations, were unsuccessful probably due to the formation of aggregates that increase the heterogeneity of the sample and affect nucleation and growth of crystals. Cyclomaltoheptaose (ß-cyclodextrin, ßCD) efficiently captures NOI-NJ and 6S-NOI-NJ in aqueous media to form inclusion complexes in which the lipophilic tail is accommodated in the hydrophobic cavity of the cyclooligosaccharide. The dissociation constant of the complex of the amphiphilic sp(2)-iminosugars with ßCD is two orders of magnitude higher than that of the corresponding complex with GlcCerase, allowing the efficient transfer of the inhibitor from the ßCD cavity to the GlcCerase active site. Enzyme-inhibitor complexes suitable for X-ray analysis were thus grown in the presence of ßCD. In contrast to what was previously observed for the complex of GlcCerase with the more basic derivative, 6-amino-6-deoxy-5-N,6-N-[N'-(n-octyl)iminomethylidene]nojirimycin (6N-NOI-NJ), the ß-anomers of both NOI-NJ and 6S-NOI-NJ were seen in the active site, even though the α-anomer was exclusively detected both in aqueous solution and in the corresponding ßCD:sp(2)-iminosugar complexes. Our results further suggest that cyclodextrin derivatives might serve as suitable delivery systems of amphiphilic glycosidase inhibitors in a biomedical context.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Cyclodextrins/chemistry , Glucosylceramidase/chemistry , 1-Deoxynojirimycin/chemistry , Crystallography, X-Ray , Glucosylceramidase/metabolism , Hydrogen Bonding , Models, Molecular , Molecular Structure , Stereoisomerism
7.
Glycobiology ; 20(1): 24-32, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19741058

ABSTRACT

Gaucher disease, the most common lysosomal storage disease, can be treated with enzyme replacement therapy (ERT), in which defective acid-beta-glucosidase (GlcCerase) is supplemented by a recombinant, active enzyme. The X-ray structures of recombinant GlcCerase produced in Chinese hamster ovary cells (imiglucerase, Cerezyme) and in transgenic carrot cells (prGCD) have been previously solved. We now describe the structure and characteristics of a novel form of GlcCerase under investigation for the treatment of Gaucher disease, Gene-Activated human GlcCerase (velaglucerase alfa). In contrast to imiglucerase and prGCD, velaglucerase alfa contains the native human enzyme sequence. All three GlcCerases consist of three domains, with the active site located in domain III. The distances between the carboxylic oxygens of the catalytic residues, E340 and E235, are consistent with distances proposed for acid-base hydrolysis. Kinetic parameters (K(m) and V(max)) of velaglucerase alfa and imiglucerase, as well as their specific activities, are similar. However, analysis of glycosylation patterns shows that velaglucerase alfa displays distinctly different structures from imiglucerase and prGCD. The predominant glycan on velaglucerase alfa is a high-mannose type, with nine mannose units, while imiglucerase contains a chitobiose tri-mannosyl core glycan with fucosylation. These differences in glycosylation affect cellular internalization; the rate of velaglucerase alfa internalization into human macrophages is at least 2-fold greater than that of imiglucerase.


Subject(s)
Glucosylceramidase/genetics , Macrophages/metabolism , Animals , CHO Cells , Catalytic Domain , Cricetinae , Cricetulus , Crystallography, X-Ray/methods , Daucus carota/genetics , Glucosylceramidase/chemistry , Glycosylation , Humans , Kinetics , Molecular Conformation , Plants, Genetically Modified
9.
Biol Chem ; 389(11): 1361-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18783340

ABSTRACT

In mammalian cells, glucosylceramide (GlcCer), the simplest glycosphingolipid, is hydrolyzed by the lysosomal enzyme acid beta-glucosidase (GlcCerase). In the human metabolic disorder Gaucher disease, GlcCerase activity is significantly decreased owing to one of approximately 200 mutations in the GlcCerase gene. The most common therapy for Gaucher disease is enzyme replacement therapy (ERT), in which patients are given intravenous injections of recombinant human GlcCerase; the Genzyme product Cerezyme has been used clinically for more than 15 years and is administered to approximately 4000 patients worldwide. Here we review the crystal structure of Cerezyme and other recombinant forms of GlcCerase, as well as of their complexes with covalent and non-covalent inhibitors. We also discuss the stability of Cerezyme, which can be altered by modification of its N-glycan chains with possible implications for improved ERT in Gaucher disease.


Subject(s)
Gaucher Disease/drug therapy , Gaucher Disease/enzymology , Glucosylceramidase/chemistry , Glucosylceramidase/therapeutic use , Amino Acid Sequence , Animals , Catalytic Domain , Glucosylceramidase/antagonists & inhibitors , Glucosylceramidase/metabolism , Humans , Molecular Sequence Data , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/pharmacology , Protein Stability/drug effects
10.
J Appl Crystallogr ; 41(Pt 5): 969-971, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-19461852

ABSTRACT

Microbatch crystallization under oil is a powerful procedure for obtaining protein crystals. Using this method, aqueous protein solutions are dispensed under liquid oil, and water evaporates through the layer of oil, with a concomitant increase in the concentrations of both protein and precipitant until the nucleation point is reached. A technique is presented for regulating the rate of water evaporation, which permits fine tuning of the crystallization conditions as well as preventing complete desiccation of the drops in the microbatch crystallization trays.

11.
Arh Hig Rada Toksikol ; 58(3): 347-53, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17913690

ABSTRACT

Serum paraoxonases (PONs) exhibit a wide range of physiologically important hydrolytic activities, including drug metabolism and detoxification of nerve gases. PON1 and PON3 reside on high-density lipoprotein (HDL) (the "good cholesterol"), and are involved in the alleviation of atherosclerosis. Members of the PON family have been identified not only in mammals and other vertebrates, but also in invertebrates. We earlier described the first crystal structure of a PON family member, a directly-evolved variant of PON1, at 2.2 A resolution. PON1 is a 6-bladed beta-propeller with a unique active-site lid which is also involved in binding to HDL. The 3-D structure, taken together with directed evolution studies, permitted analysis of mutations which enhanced the stability, solubility and crystallizability of this PON1 variant. The structure permits a detailed description of PON1's active site and suggests possible mechanisms for its catalytic activity on certain substrates.


Subject(s)
Aryldialkylphosphatase/chemistry , Animals , Aryldialkylphosphatase/blood , Catalysis , Chemical Phenomena , Chemistry, Physical , Crystallization , Humans , Molecular Conformation , Thromboxane A2
12.
J Biol Chem ; 282(39): 29052-29058, 2007 Sep 28.
Article in English | MEDLINE | ID: mdl-17666401

ABSTRACT

Gaucher disease is caused by mutations in the gene encoding acid beta-glucosidase (GlcCerase), resulting in glucosylceramide (GlcCer) accumulation. The only currently available orally administered treatment for Gaucher disease is N-butyl-deoxynojirimycin (Zavesca, NB-DNJ), which partially inhibits GlcCer synthesis, thus reducing levels of GlcCer accumulation. NB-DNJ also acts as a chemical chaperone for GlcCerase, although at a different concentration than that required to completely inhibit GlcCer synthesis. We now report the crystal structures, at 2A resolution, of complexes of NB-DNJ and N-nonyl-deoxynojirimycin (NN-DNJ) with recombinant human GlcCerase, expressed in cultured plant cells. Both inhibitors bind at the active site of GlcCerase, with the imino sugar moiety making hydrogen bonds to side chains of active site residues. The alkyl chains of NB-DNJ and NN-DNJ are oriented toward the entrance of the active site where they undergo hydrophobic interactions. Based on these structures, we make a number of predictions concerning (i) involvement of loops adjacent to the active site in the catalytic process, (ii) the nature of nucleophilic attack by Glu-340, and (iii) the role of a conserved water molecule located in a solvent cavity adjacent to the active site. Together, these results have significance for understanding the mechanism of action of GlcCerase and the mode of GlcCerase chaperoning by imino sugars.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Enzyme Inhibitors/chemistry , Gaucher Disease/enzymology , Glucosylceramidase/chemistry , Molecular Chaperones/chemistry , 1-Deoxynojirimycin/administration & dosage , 1-Deoxynojirimycin/chemistry , Administration, Oral , Binding Sites/physiology , Crystallography, X-Ray , Enzyme Inhibitors/administration & dosage , Gaucher Disease/drug therapy , Glucosylceramides/biosynthesis , Glucosylceramides/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Recombinant Proteins/chemistry , Structure-Activity Relationship
13.
Acta Crystallogr D Biol Crystallogr ; 62(Pt 12): 1458-65, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17139081

ABSTRACT

Gaucher disease is caused by mutations in the gene encoding acid-beta-glucosidase. A recombinant form of this enzyme, Cerezyme, is used to treat Gaucher disease patients by ;enzyme-replacement therapy'. Crystals of Cerezyme after its partial deglycosylation were obtained earlier and the structure was solved to 2.0 A resolution [Dvir et al. (2003), EMBO Rep. 4, 704-709]. The crystal structure of unmodified Cerezyme is now reported, in which a substantial number of sugar residues bound to three asparagines via N-glycosylation could be visualized. The structure of intact fully glycosylated Cerezyme is virtually identical to that of the partially deglycosylated enzyme. However, the three loops at the entrance to the active site, which were previously observed in alternative conformations, display additional variability in their structures. Comparison of the structure of acid-beta-glucosidase with that of xylanase, a bacterial enzyme from a closely related protein family, demonstrates a close correspondence between the active-site residues of the two enzymes.


Subject(s)
Gaucher Disease/enzymology , Glucosylceramidase/chemistry , Glucosylceramidase/metabolism , Amino Acid Sequence , Binding Sites , Catalysis , Crystallography, X-Ray , Endo-1,4-beta Xylanases/chemistry , Endo-1,4-beta Xylanases/metabolism , Glycosylation , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Molecular Sequence Data , Protein Structure, Secondary
14.
J Med Chem ; 49(18): 5491-500, 2006 Sep 07.
Article in English | MEDLINE | ID: mdl-16942022

ABSTRACT

The X-ray crystal structures were solved for complexes with Torpedo californica acetylcholinesterase of two bivalent tacrine derivative compounds in which the two tacrine rings were separated by 5- and 7-carbon spacers. The derivative with the 7-carbon spacer spans the length of the active-site gorge, making sandwich interactions with aromatic residues both in the catalytic anionic site (Trp84 and Phe330) at the bottom of the gorge and at the peripheral anionic site near its mouth (Tyr70 and Trp279). The derivative with the 5-carbon spacer interacts in a similar manner at the bottom of the gorge, but the shorter tether precludes a sandwich interaction at the peripheral anionic site. Although the upper tacrine group does interact with Trp279, it displaces the phenyl residue of Phe331, thus causing a major rearrangement in the Trp279-Ser291 loop. The ability of this inhibitor to induce large-scale structural changes in the active-site gorge of acetylcholinesterase has significant implications for structure-based drug design because such conformational changes in the target enzyme are difficult to predict and to model.


Subject(s)
Acetylcholinesterase/chemistry , Alkenes/chemistry , Cholinesterase Inhibitors/chemistry , Models, Molecular , Tacrine/chemistry , Animals , Binding Sites , Crystallization , Crystallography, X-Ray , Dimerization , Molecular Structure , Protein Conformation , Torpedo
15.
Chem Biol Interact ; 157-158: 153-7, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16289500

ABSTRACT

The anticancer prodrug CPT-11 is a highly effective camptothecin analog that has been approved for the treatment of colon cancer. The 2.6 angstroms resolution crystal structure of its complex with Torpedo californica acetylcholinesterase (TcAChE) demonstrates that CPT-11 binds to TcAChE and spans its gorge similarly to the Alzheimer drug, Aricept. The crystal structure clearly reveals the interactions, which contribute to the inhibitory action of CPT-11. Modeling of the complexes of CPT-11 with mammalian butyrylcholinesterase and carboxylesterase, both of which are known to hydrolyze the drug, shows how binding to either of the two enzymes yields a productive substrate-enzyme complex.


Subject(s)
Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Antineoplastic Agents/chemistry , Camptothecin/analogs & derivatives , Carboxylesterase/metabolism , Cholinesterase Inhibitors/chemistry , Torpedo , Animals , Antineoplastic Agents/metabolism , Butyrylcholinesterase/metabolism , Camptothecin/chemistry , Cholinesterase Inhibitors/metabolism , Crystallography, X-Ray , Hydrolysis , Irinotecan , Liver/enzymology , Models, Molecular , Protein Structure, Tertiary , Rabbits
16.
Chem Biol Interact ; 157-158: 247-52, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16257398

ABSTRACT

CPT-11 (irinotecan, 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin) is an anticancer prodrug that has been approved for the treatment of colon cancer. It is a member of the camptothecin class of drugs and activation to the active metabolite SN-38, is mediated by carboxylesterases (CE). SN-38 is a potent topoisomerase I poison and is highly effective at killing human tumor cells, with IC50 values in the low nM range. However, upon high dose administration of CPT-11 to cancer patients, a cholinergic syndrome is observed, that can be rapidly ameliorated by atropine. This suggests a direct interaction of the drug or its metabolites with acetylcholinesterase (AChE). Kinetic studies indicated that CPT-11 was primarily responsible for AChE inhibition with the 4-piperidinopiperidine moiety, the major determinant in the loss of enzyme activity. Structural analogs of 4-piperidinopiperidine however, did not inhibit AChE, including a benzyl piperazine derivate of CPT-11. These results suggest that novel anticancer drugs could be synthesized that do not inhibit AChE, or alternatively, that novel AChE inhibitors could be designed based around the camptothecin scaffold.


Subject(s)
Acetylcholinesterase/metabolism , Antineoplastic Agents/pharmacology , Camptothecin/analogs & derivatives , Cholinesterase Inhibitors/pharmacology , Antineoplastic Agents/chemistry , Butyrylcholinesterase/metabolism , Camptothecin/chemistry , Camptothecin/pharmacology , Cholinesterase Inhibitors/chemistry , Enzyme Activation/drug effects , Humans , Irinotecan , Molecular Structure , Structure-Activity Relationship
17.
Mol Pharmacol ; 67(6): 1874-81, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15772291

ABSTRACT

The anticancer prodrug 7-ethyl-10-[4-(1-piperidino)-1-piperidino-]carbonyloxycamptothecin (CPT-11) is a highly effective camptothecin analog that has been approved for the treatment of colon cancer. It is hydrolyzed by carboxylesterases to yield 7-ethyl-10-hydroxycamptothecin (SN-38), a potent topoisomerase I poison. However, upon high-dose intravenous administration of CPT-11, a cholinergic syndrome is observed that can be ameliorated by atropine. Previous studies have indicated that CPT-11 can inhibit acetylcholinesterase (AChE), and here, we provide a detailed analysis of the inhibition of AChE by CPT-11 and by structural analogs. These studies demonstrate that the terminal dipiperidino moiety in CPT-11 plays a major role in enzyme inhibition, and this has been confirmed by X-ray crystallographic studies of a complex of the drug with Torpedo californica AChE. Our results indicate that CPT-11 binds within the active site gorge of the protein in a fashion similar to that observed with the Alzheimer drug donepezil. The 3D structure of the CPT-11/AChE complex also permits modeling of CPT-11 complexed with mammalian butyrylcholinesterase and carboxylesterase, both of which are known to hydrolyze the drug to the active metabolite. Overall, the results presented here clarify the mechanism of AChE inhibition by CPT-11 and detail the interaction of the drug with the protein. These studies may allow the design of both novel camptothecin analogs that would not inhibit AChE and new AChE inhibitors derived from the camptothecin scaffold.


Subject(s)
Acetylcholinesterase/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Camptothecin/analogs & derivatives , Camptothecin/chemistry , Cholinesterase Inhibitors/chemistry , Prodrugs/chemistry , Acetylcholinesterase/metabolism , Animals , Antineoplastic Agents, Phytogenic/metabolism , Camptothecin/metabolism , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/pharmacology , Crystallization , Crystallography, X-Ray , Humans , Irinotecan , Prodrugs/metabolism , Torpedo
18.
Nat Struct Mol Biol ; 11(5): 412-9, 2004 May.
Article in English | MEDLINE | ID: mdl-15098021

ABSTRACT

Members of the serum paraoxonase (PON) family have been identified in mammals and other vertebrates, and in invertebrates. PONs exhibit a wide range of physiologically important hydrolytic activities, including drug metabolism and detoxification of nerve agents. PON1 and PON3 reside on high-density lipoprotein (HDL, 'good cholesterol') and are involved in the prevention of atherosclerosis. We describe the first crystal structure of a PON family member, a variant of PON1 obtained by directed evolution, at a resolution of 2.2 A. PON1 is a six-bladed beta-propeller with a unique active site lid that is also involved in HDL binding. The three-dimensional structure and directed evolution studies permit a detailed description of PON1's active site and catalytic mechanism, which are reminiscent of secreted phospholipase A2, and of the routes by which PON family members diverged toward different substrate and reaction selectivities.


Subject(s)
Aryldialkylphosphatase/blood , Aryldialkylphosphatase/genetics , Evolution, Molecular , Amino Acid Sequence , Aryldialkylphosphatase/chemistry , Aryldialkylphosphatase/metabolism , Catalysis , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid , Substrate Specificity
19.
Acta Crystallogr D Biol Crystallogr ; 58(Pt 9): 1476-8, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12198307

ABSTRACT

Manganese is recruited in microorganisms by way of ABC-type transporter systems. Here, the expression, purification and preliminary crystallographic analysis of a soluble form of the MntC solute-binding protein component of the MntABC manganese-import system from the cyanobacterium Synechococystis sp. PCC 6803 is reported. The protein (321 amino-acid residues) was expressed exclusively in inclusion bodies, which required unfolding and refolding in the presence of manganese prior to purification. The purified protein was crystallized in the presence of PEG and zinc. The crystals belong to space group P6(2)22, with unit-cell parameters a = b = 128.1, c = 90.0 A and a single molecule in the asymmetric unit. The crystals diffract to 2.6 A under cryoconditions using synchrotron radiation.


Subject(s)
Cyanobacteria/chemistry , Manganese/metabolism , Periplasmic Binding Proteins/chemistry , Crystallography, X-Ray , Periplasmic Binding Proteins/metabolism , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...