Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Haematologica ; 108(2): 409-419, 2023 02 01.
Article in English | MEDLINE | ID: mdl-35979719

ABSTRACT

Asparagine is a non-essential amino acid since it can either be taken up via the diet or synthesized by asparagine synthetase. Acute lymphoblastic leukemia (ALL) cells do not express asparagine synthetase or express it only minimally, which makes them completely dependent on extracellular asparagine for their growth and survival. This dependency makes ALL cells vulnerable to treatment with L-asparaginase, an enzyme that hydrolyzes asparagine. To date, all clinically approved L-asparaginases have significant L-glutaminase co-activity, associated with non-immune related toxic side effects observed during therapy. Therefore, reduction of L-glutaminase co-activity with concomitant maintenance of its anticancer L-asparaginase effect may effectively improve the tolerability of this unique drug. Previously, we designed a new alternative variant of Erwinia chrysanthemi (ErA; Erwinaze) with decreased L-glutaminase co-activity, while maintaining its L-asparaginase activity, by the introduction of three key mutations around the active site (ErA-TM). However, Erwinaze and our ErA-TM variant have very short half-lives in vivo. Here, we show that the fusion of ErA-TM with an albumin binding domain (ABD)-tag significantly increases its in vivo persistence. In addition, we evaluated the in vivo therapeutic efficacy of ABD-ErA-TM in a B-ALL xenograft model of SUP-B15. Our results show a comparable long-lasting durable antileukemic effect between the standard-of-care pegylated-asparaginase and ABD-ErA-TM L-asparaginase, but with fewer co-glutaminase-related acute side effects. Since the toxic side effects of current L-asparaginases often result in treatment discontinuation in ALL patients, this novel ErA-TM variant with ultra-low L-glutaminase co-activity and long in vivo persistence may have great clinical potential.


Subject(s)
Aspartate-Ammonia Ligase , Leukemia, Myeloid, Acute , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Asparaginase/pharmacology , Asparaginase/therapeutic use , Glutaminase/chemistry , Glutaminase/genetics , Glutaminase/metabolism , Asparagine , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Leukemia, Myeloid, Acute/drug therapy
2.
J Struct Biol ; 209(1): 107412, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31689502

ABSTRACT

Hemagglutnin (HA) mediates entry of influenza virus through a series of conformational changes triggered by the low pH of the endosome. The residue or combination of residues acting as pH sensors has not yet been fully elucidated. In this work, we assay pH effects on the structure of H5 HA by soaking HA crystallized at pH 6.5 in a series of buffers with lower pH, mimicking the conditions of the endosome. We find that HA1-H38, which is conserved in Group 1 HA, undergoes a striking change in side chain conformation, which we attribute to its protonation and cation-cation repulsion with conserved HA1-H18. This work suggests that x-ray crystallography can be applied for studying small-scale pH-induced conformational changes providing valuable information on the location of pH sensors in HA. Importantly, the observed change in HA1-H38 conformation is further evidence that the pH-induced conformational changes of HA are the result of a series of protonation events to conserved and non-conserved pH sensors.


Subject(s)
Hemagglutinins/ultrastructure , Influenza, Human/genetics , Orthomyxoviridae/ultrastructure , Virus Internalization , Crystallography, X-Ray , Endosomes/genetics , Endosomes/ultrastructure , Hemagglutinins/chemistry , Hemagglutinins/genetics , Humans , Hydrogen-Ion Concentration , Influenza, Human/pathology , Influenza, Human/virology , Models, Molecular , Orthomyxoviridae/genetics , Protein Conformation
3.
J Biol Chem ; 293(35): 13553-13565, 2018 08 31.
Article in English | MEDLINE | ID: mdl-29980598

ABSTRACT

Mixed-lineage kinase 3 (MLK3; also known as MAP3K11) is a Ser/Thr protein kinase widely expressed in normal and cancerous tissues, including brain, lung, liver, heart, and skeletal muscle tissues. Its Src homology 3 (SH3) domain has been implicated in MLK3 autoinhibition and interactions with other proteins, including those from viruses. The MLK3 SH3 domain contains a six-amino-acid insert corresponding to the n-Src insert, suggesting that MLK3 may bind additional peptides. Here, affinity selection of a phage-displayed combinatorial peptide library for MLK3's SH3 domain yielded a 13-mer peptide, designated "MLK3 SH3-interacting peptide" (MIP). Unlike most SH3 domain peptide ligands, MIP contained a single proline. The 1.2-Å crystal structure of the MIP-bound SH3 domain revealed that the peptide adopts a ß-hairpin shape, and comparison with a 1.5-Å apo SH3 domain structure disclosed that the n-Src loop in SH3 undergoes an MIP-induced conformational change. A 1.5-Å structure of the MLK3 SH3 domain bound to a canonical proline-rich peptide from hepatitis C virus nonstructural 5A (NS5A) protein revealed that it and MIP bind the SH3 domain at two distinct sites, but biophysical analyses suggested that the two peptides compete with each other for SH3 binding. Moreover, SH3 domains of MLK1 and MLK4, but not MLK2, also bound MIP, suggesting that the MLK1-4 family may be differentially regulated through their SH3 domains. In summary, we have identified two distinct peptide-binding sites in the SH3 domain of MLK3, providing critical insights into mechanisms of ligand binding by the MLK family of kinases.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Peptides/metabolism , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Humans , MAP Kinase Kinase Kinases/chemistry , Molecular Docking Simulation , Peptide Library , Peptides/chemistry , Protein Binding , src Homology Domains , Mitogen-Activated Protein Kinase Kinase Kinase 11
4.
Proc Natl Acad Sci U S A ; 113(15): 4027-32, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27035974

ABSTRACT

Deoxycytidine kinase (dCK), a rate-limiting enzyme in the cytosolic deoxyribonucleoside (dN) salvage pathway, is an important therapeutic and positron emission tomography (PET) imaging target in cancer. PET probes for dCK have been developed and are effective in mice but have suboptimal specificity and sensitivity in humans. To identify a more suitable probe for clinical dCK PET imaging, we compared the selectivity of two candidate compounds-[(18)F]Clofarabine; 2-chloro-2'-deoxy-2'-[(18)F]fluoro-9-ß-d-arabinofuranosyl-adenine ([(18)F]CFA) and 2'-deoxy-2'-[(18)F]fluoro-9-ß-d-arabinofuranosyl-guanine ([(18)F]F-AraG)-for dCK and deoxyguanosine kinase (dGK), a dCK-related mitochondrial enzyme. We demonstrate that, in the tracer concentration range used for PET imaging, [(18)F]CFA is primarily a substrate for dCK, with minimal cross-reactivity. In contrast, [(18)F]F-AraG is a better substrate for dGK than for dCK. [(18)F]CFA accumulation in leukemia cells correlated with dCK expression and was abrogated by treatment with a dCK inhibitor. Although [(18)F]CFA uptake was reduced by deoxycytidine (dC) competition, this inhibition required high dC concentrations present in murine, but not human, plasma. Expression of cytidine deaminase, a dC-catabolizing enzyme, in leukemia cells both in cell culture and in mice reduced the competition between dC and [(18)F]CFA, leading to increased dCK-dependent probe accumulation. First-in-human, to our knowledge, [(18)F]CFA PET/CT studies showed probe accumulation in tissues with high dCK expression: e.g., hematopoietic bone marrow and secondary lymphoid organs. The selectivity of [(18)F]CFA for dCK and its favorable biodistribution in humans justify further studies to validate [(18)F]CFA PET as a new cancer biomarker for treatment stratification and monitoring.


Subject(s)
Adenine Nucleotides/chemistry , Arabinonucleosides/chemistry , Biomarkers, Tumor/chemistry , Deoxycytidine Kinase/analysis , Deoxycytidine Kinase/metabolism , Positron-Emission Tomography/methods , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Clofarabine , Contrast Media/chemistry , Deoxycytidine Kinase/antagonists & inhibitors , Humans , Leukemia/enzymology , Mice , Neoplasms/drug therapy , Prodrugs/chemistry , Rats
5.
Biochemistry ; 57(8): 1316-1325, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29389115

ABSTRACT

Choline kinase α (ChoKα) is an enzyme that is upregulated in many types of cancer and has been shown to be tumorigenic. As such, it makes a promising target for inhibiting tumor growth. Though there have been several inhibitors synthesized for ChoKα, not all of them demonstrate the same efficacy in vivo, though the reasons behind this difference in potency are not clear. One particular inhibitor, designated TCD-717, has recently completed phase I clinical trials. Cell culture and in vitro studies support the powerful inhibitory effect TCD-717 has on ChoKα, but an examination of the inhibitor's interaction with the ChoKα enzyme has been missing prior to this work. Here we detail the 2.35 Å structure of ChoKα in complex with TCD-717. Examination of this structure in conjunction with kinetic assays reveals that TCD-717 does not bind directly in the choline pocket as do previously characterized ChoKα inhibitors, but rather in a proximal but novel location near the surface of the enzyme. The unique binding site identified for TCD-717 lends insight for the future design of more potent in vivo inhibitors for ChoKα.


Subject(s)
Choline Kinase/antagonists & inhibitors , Choline Kinase/chemistry , Protein Kinase Inhibitors/pharmacology , Binding Sites , Choline Kinase/metabolism , Crystallography, X-Ray , Drug Design , Humans , Molecular Docking Simulation , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/chemistry
6.
J Biol Chem ; 292(52): 21590-21597, 2017 12 29.
Article in English | MEDLINE | ID: mdl-29127198

ABSTRACT

Viral entry into host cells is mediated by membrane proteins in a metastable state that transition to a more stable state upon a stimulus. For example, in the influenza envelope protein hemagglutinin (HA), the low pH in the endosome triggers a transition from the metastable prefusion conformation to the stable fusion conformation. To identify probes that interfere with HA function, here we screened a library of H7 HA peptides for inhibition of H7 HA-mediated entry. We discovered a peptide, PEP87 (WSYNAELLVAMENQHTI), that inhibited H7 and H5 HA-mediated entry. PEP87 corresponds to a highly conserved helical region of the HA2 subunit of HA that self-interacts in the neutral pH conformation. Mutagenesis experiments indicated that PEP87 binds to its native region in the HA trimer. We also found that PEP87 is unstructured in isolation but tends to form a helix as evidenced by CD and NMR studies. Fluorescence, chemical cross-linking, and saturation transfer difference NMR data suggested that PEP87 binds to the neutral pH conformation of HA and disrupts the HA structure without affecting its oligomerization state. Together, this work provides support for a model in which PEP87 disrupts HA function by displacing native interactions of the neutral pH conformation. Moreover, our observations indicate that the HA prefusion structure (and perhaps the metastable states of other viral entry proteins) is more dynamic with transient motions being larger than generally appreciated. These findings also suggest that the ensemble of prefusion structures presents many potential sites for targeting in therapeutic interventions.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Amino Acid Sequence , Crystallography, X-Ray/methods , Hemagglutinins/chemistry , Humans , Hydrogen-Ion Concentration , Influenza, Human/metabolism , Membrane Fusion/physiology , Models, Molecular , Mutagenesis , Peptides/metabolism , Protein Conformation , Virus Internalization
7.
J Biol Chem ; 291(34): 17664-76, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27354283

ABSTRACT

Current FDA-approved l-asparaginases also possess significant l-glutaminase activity, which correlates with many of the toxic side effects of these drugs. Therefore, l-asparaginases with reduced l-glutaminase activity are predicted to be safer. We exploited our recently described structures of the Erwinia chrysanthemi l-asparaginase (ErA) to inform the design of mutants with diminished ability to hydrolyze l-glutamine. Structural analysis of these variants provides insight into the molecular basis for the increased l-asparagine specificity. A primary role is attributed to the E63Q mutation that acts to hinder the correct positioning of l-glutamine but not l-asparagine. The substitution of Ser-254 with either an asparagine or a glutamine increases the l-asparagine specificity but only when combined with the E63Q mutation. The A31I mutation reduces the substrate Km value; this is a key property to allow the required therapeutic l-asparagine depletion. Significantly, an ultra-low l-glutaminase ErA variant maintained its cell killing ability. By diminishing the l-glutaminase activity of these highly active l-asparaginases, our engineered ErA variants hold promise as l-asparaginases with fewer side effects.


Subject(s)
Asparaginase/chemistry , Bacterial Proteins/chemistry , Dickeya chrysanthemi/enzymology , Glutaminase , Mutation, Missense , Amino Acid Substitution , Asparaginase/genetics , Asparaginase/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/pharmacology , Cell Line, Tumor , Dickeya chrysanthemi/genetics , Humans
8.
J Biol Chem ; 291(10): 5088-100, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26733195

ABSTRACT

Bacterial L-asparaginases play an important role in the treatment of certain types of blood cancers. We are exploring the guinea pig L-asparaginase (gpASNase1) as a potential replacement of the immunogenic bacterial enzymes. The exact mechanism used by L-asparaginases to catalyze the hydrolysis of asparagine into aspartic acid and ammonia has been recently put into question. Earlier experimental data suggested that the reaction proceeds via a covalent intermediate using a ping-pong mechanism, whereas recent computational work advocates the direct displacement of the amine by an activated water. To shed light on this controversy, we generated gpASNase1 mutants of conserved active site residues (T19A, T116A, T19A/T116A, K188M, and Y308F) suspected to play a role in hydrolysis. Using x-ray crystallography, we determined the crystal structures of the T19A, T116A, and K188M mutants soaked in asparagine. We also characterized their steady-state kinetic properties and analyzed the conversion of asparagine to aspartate using NMR. Our structures reveal bound asparagine in the active site that has unambiguously not formed a covalent intermediate. Kinetic and NMR assays detect significant residual activity for all of the mutants. Furthermore, no burst of ammonia production was observed that would indicate covalent intermediate formation and the presence of a ping-pong mechanism. Hence, despite using a variety of techniques, we were unable to obtain experimental evidence that would support the formation of a covalent intermediate. Consequently, our observations support a direct displacement rather than a ping-pong mechanism for l-asparaginases.


Subject(s)
Asparaginase/chemistry , Amino Acid Sequence , Ammonia/metabolism , Animals , Asparaginase/genetics , Asparaginase/metabolism , Asparagine/metabolism , Aspartic Acid/metabolism , Catalytic Domain , Guinea Pigs , Molecular Sequence Data , Mutation, Missense , Protein Binding
9.
Biochemistry ; 55(8): 1246-53, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26855287

ABSTRACT

l-Asparaginases of bacterial origin are a mainstay of acute lymphoblastic leukemia treatment. The mechanism of action of these enzyme drugs is associated with their capacity to deplete the amino acid l-asparagine from the blood. However, clinical use of bacterial l-asparaginases is complicated by their dual l-asparaginase and l-glutaminase activities. The latter, even though representing only ∼10% of the overall activity, is partially responsible for the observed toxic side effects. Hence, l-asparaginases devoid of l-glutaminase activity hold potential as safer drugs. Understanding the key determinants of l-asparaginase substrate specificity is a prerequisite step toward the development of enzyme variants with reduced toxicity. Here we present crystal structures of the Erwinia chrysanthemi l-asparaginase in complex with l-aspartic acid and with l-glutamic acid. These structures reveal two enzyme conformations-open and closed-corresponding to the inactive and active states, respectively. The binding of ligands induces the positioning of the catalytic Thr15 into its active conformation, which in turn allows for the ordering and closure of the flexible N-terminal loop. Notably, l-aspartic acid is more efficient than l-glutamic acid in inducing the active positioning of Thr15. Structural elements explaining the preference of the enzyme for l-asparagine over l-glutamine are discussed with guidance to the future development of more specific l-asparaginases.


Subject(s)
Asparaginase/metabolism , Dickeya chrysanthemi/enzymology , Asparaginase/chemistry , Aspartic Acid/metabolism , Crystallography, X-Ray , Dickeya chrysanthemi/chemistry , Dickeya chrysanthemi/metabolism , Glutamic Acid/metabolism , Humans , Models, Molecular , Protein Conformation , Substrate Specificity
10.
J Biol Chem ; 290(27): 16595-606, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-26023235

ABSTRACT

The molecular seal between epithelial cells, called the tight junction (TJ), is built by several membrane proteins, with claudins playing the most prominent role. The scaffold proteins of the zonula occludens family are required for the correct localization of claudins and hence formation of the TJ. The intracellular C terminus of claudins binds to the N-terminal PDZ domain of zonula occludens proteins (PDZ1). Of the 23 identified human claudin proteins, nine possess a tyrosine at the -6 position. Here we show that the claudin affinity for PDZ1 is dependent on the presence or absence of this tyrosine and that the affinity is reduced if the tyrosine is modified by phosphorylation. The PDZ1 ß2-ß3 loop undergoes a significant conformational change to accommodate this tyrosine. Cell culture experiments support a regulatory role for this tyrosine. Plasticity has been recognized as a critical property of TJs that allow cell remodeling and migration. Our work provides a molecular framework for how TJ plasticity may be regulated.


Subject(s)
Claudin-1/metabolism , Claudin-2/metabolism , Zonula Occludens-1 Protein/chemistry , Amino Acid Motifs , Amino Acid Sequence , Claudin-1/chemistry , Claudin-1/genetics , Claudin-2/chemistry , Claudin-2/genetics , Humans , Molecular Sequence Data , PDZ Domains , Phosphorylation , Protein Binding , Protein Structure, Secondary , Sequence Alignment , Tight Junctions/chemistry , Tight Junctions/genetics , Tight Junctions/metabolism , Tyrosine/chemistry , Tyrosine/genetics , Tyrosine/metabolism , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
11.
J Biol Chem ; 289(48): 33175-86, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25320094

ABSTRACT

The initial observation that guinea pig serum kills lymphoma cells marks the serendipitous discovery of a new class of anti-cancer agents. The serum cell killing factor was shown to be an enzyme with L-asparaginase (ASNase) activity. As a direct result of this observation, several bacterial L-asparaginases were developed and are currently approved by the Food and Drug Administration for the treatment of the subset of hematological malignancies that are dependent on the extracellular pool of the amino acid asparagine. As drugs, these enzymes act to hydrolyze asparagine to aspartate, thereby starving the cancer cells of this amino acid. Prior to the work presented here, the precise identity of this guinea pig enzyme has not been reported in the peer-reviewed literature. We discovered that the guinea pig enzyme annotated as H0W0T5_CAVPO, which we refer to as gpASNase1, has the required low Km property consistent with that possessed by the cell-killing guinea pig serum enzyme. Elucidation of the ligand-free and aspartate complex gpASNase1 crystal structures allows a direct comparison with the bacterial enzymes and serves to explain the lack of L-glutaminase activity in the guinea pig enzyme. The structures were also used to generate a homology model for the human homolog hASNase1 and to help explain its vastly different kinetic properties compared with gpASNase1, despite a 70% sequence identity. Given that the bacterial enzymes frequently present immunogenic and other toxic side effects, this work suggests that gpASNase1 could be a promising alternative to these bacterial enzymes.


Subject(s)
Antineoplastic Agents/chemistry , Asparaginase/chemistry , Animals , Antineoplastic Agents/therapeutic use , Asparaginase/genetics , Asparaginase/therapeutic use , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/therapeutic use , Crystallography, X-Ray , Guinea Pigs , Humans , Recombinant Proteins , Structural Homology, Protein
12.
Biochemistry ; 53(14): 2318-28, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24669941

ABSTRACT

We investigated whether an uncharacterized protein from guinea pig could be the enzyme behind Kidd's serendipitous discovery, made over 60 years ago, that guinea pig serum has cell killing ability. It has been long known that an enzyme with l-asparaginase activity is responsible for cell killing, although astonishingly, its identity remains unclear. Bacterial asparaginases with similar cell killing properties have since become a mainstay therapy of certain cancers such as acute lymphoblastic leukemia. By hydrolyzing asparagine to aspartate and ammonia, these drugs deplete the asparagine present in the blood, killing cancer cells that rely on extracellular asparagine uptake for survival. However, bacterial asparaginases can elicit an adverse immune response. We propose that replacement of bacterial enzymes with the guinea pig asparaginase responsible for serum activity, by its virtue of being more closely related to human enzymes, will be less immunogenic. To this goal, we investigated whether an uncharacterized protein from guinea pig with putative asparaginase activity, which we call gpASNase3, could be that enzyme. We examined its self-activation process (gpASNase3 requires autocleavage to become active), kinetically characterized it for asparaginase and ß-aspartyl dipeptidase activity, and elucidated its crystal structure in both the uncleaved and cleaved states. This work reveals that gpASNase3 is not the enzyme responsible for the antitumor effects of guinea pig serum. It exhibits a low affinity for asparagine as measured by a high Michaelis constant, KM, in the millimolar range, in contrast to the low KM (micromolar range) required for asparaginase to be effective as an anticancer agent.


Subject(s)
Asparaginase/metabolism , Amino Acid Sequence , Animals , Asparaginase/chemistry , Asparaginase/genetics , Catalytic Domain , Cloning, Molecular , Guinea Pigs , Kinetics , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid
13.
Biochemistry ; 53(5): 872-80, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24437575

ABSTRACT

A component of the shikimate biosynthetic pathway, dehydroquinate dehydratase (DHQD) catalyzes the dehydration of 3-dehydroquniate (DHQ) to 3-dehydroshikimate. In the type I DHQD reaction mechanism a lysine forms a Schiff base intermediate with DHQ. The Schiff base acts as an electron sink to facilitate the catalytic dehydration. To address the mechanism of Schiff base formation, we determined structures of the Salmonella enterica wild-type DHQD in complex with the substrate analogue quinate and the product analogue shikimate. In addition, we determined the structure of the K170M mutant (Lys170 being the Schiff base forming residue) in complex with quinate. Combined with nuclear magnetic resonance and isothermal titration calorimetry data that revealed altered binding of the analogue to the K170M mutant, these structures suggest a model of Schiff base formation characterized by the dynamic interplay of opposing forces acting on either side of the substrate. On the side distant from the substrate 3-carbonyl group, closure of the enzyme's ß8-α8 loop is proposed to guide DHQ into the proximity of the Schiff base-forming Lys170. On the 3-carbonyl side of the substrate, Lys170 sterically alters the position of DHQ's reactive ketone, aligning it at an angle conducive for nucleophilic attack. This study of a type I DHQD reveals the interplay between the enzyme and substrate required for the correct orientation of a functional group constrained within a cyclic substrate.


Subject(s)
Bacterial Proteins/chemistry , Hydro-Lyases/chemistry , Quinic Acid/chemistry , Salmonella enterica/enzymology , Schiff Bases/chemistry , Shikimic Acid/chemistry , Bacterial Proteins/genetics , Crystallography, X-Ray , Hydro-Lyases/genetics , Mutation , Promoter Regions, Genetic , Protein Binding , Protein Conformation
14.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 1): 68-78, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24419380

ABSTRACT

Deoxycytidine kinase (dCK) is a key enzyme in the nucleoside salvage pathway that is also required for the activation of several anticancer and antiviral nucleoside analog prodrugs. Additionally, dCK has been implicated in immune disorders and has been found to be overexpressed in several cancers. To allow the probing and modulation of dCK activity, a new class of small-molecule inhibitors of the enzyme were developed. Here, the structural characterization of four of these inhibitors in complex with human dCK is presented. The structures reveal that the compounds occupy the nucleoside-binding site and bind to the open form of dCK. Surprisingly, a slight variation in the nature of the substituent at the 5-position of the thiazole ring governs whether the active site of the enzyme is occupied by one or two inhibitor molecules. Moreover, this substituent plays a critical role in determining the affinity, improving it from >700 to 1.5 nM in the best binder. These structures lay the groundwork for future modifications that would result in even tighter binding and the correct placement of moieties that confer favorable pharmacodynamics and pharmacokinetic properties.


Subject(s)
Deoxycytidine Kinase/antagonists & inhibitors , Deoxycytidine Kinase/chemistry , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Crystallography, X-Ray , Humans , Models, Molecular , Protein Binding , Protein Conformation/drug effects , Uridine Diphosphate/metabolism
15.
J Struct Funct Genomics ; 14(1): 25-30, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23539270

ABSTRACT

Dehydroquinate dehydratase (DHQD) catalyzes the third step in the biosynthetic shikimate pathway. Here we identify a Bifidobacterium longum protein with high sequence homology to type II DHQDs but no detectable DHQD activity under standard assay conditions. A crystal structure reveals that the B. longum protein adopts a DHQD-like tertiary structure but a distinct quaternary state. Apparently forming a dimer, the B. longum protein lacks the active site aspartic acid contributed from a neighboring protomer in the type II DHQD dodecamer. Relating to the absence of protein-protein interactions established in the type II DHQD dodecameric assembly, substantial conformational changes distinguish the would-be active site of the B. longum protein. As B. longum possess no other genes with homology to known DHQDs, these findings imply a unique DHQD activity within B. longum.


Subject(s)
Bacterial Proteins/chemistry , Bifidobacterium/chemistry , Hydro-Lyases/chemistry , Models, Molecular , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bifidobacterium/genetics , Bifidobacterium/metabolism , Crystallography, X-Ray , Escherichia coli/genetics , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Molecular Sequence Data , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Shikimic Acid/metabolism
16.
J Biol Chem ; 287(1): 446-454, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22074768

ABSTRACT

Positron emission tomography (PET) reporter gene imaging can be used to non-invasively monitor cell-based therapies. Therapeutic cells engineered to express a PET reporter gene (PRG) specifically accumulate a PET reporter probe (PRP) and can be detected by PET imaging. Expanding the utility of this technology requires the development of new non-immunogenic PRGs. Here we describe a new PRG-PRP system that employs, as the PRG, a mutated form of human thymidine kinase 2 (TK2) and 2'-deoxy-2'-18F-5-methyl-1-ß-L-arabinofuranosyluracil (L-18F-FMAU) as the PRP. We identified L-18F-FMAU as a candidate PRP and determined its biodistribution in mice and humans. Using structure-guided enzyme engineering, we generated a TK2 double mutant (TK2-N93D/L109F) that efficiently phosphorylates L-18F-FMAU. The N93D/L109F TK2 mutant has lower activity for the endogenous nucleosides thymidine and deoxycytidine than wild type TK2, and its ectopic expression in therapeutic cells is not expected to alter nucleotide metabolism. Imaging studies in mice indicate that the sensitivity of the new human TK2-N93D/L109F PRG is comparable with that of a widely used PRG based on the herpes simplex virus 1 thymidine kinase. These findings suggest that the TK2-N93D/L109F/L-18F-FMAU PRG-PRP system warrants further evaluation in preclinical and clinical applications of cell-based therapies.


Subject(s)
Genes, Reporter/genetics , Positron-Emission Tomography/methods , Protein Engineering/methods , Thymidine Kinase/chemistry , Thymidine Kinase/genetics , Thymidine/analogs & derivatives , Thymidine/metabolism , Adult , Animals , Arabinofuranosyluracil/analogs & derivatives , Arabinofuranosyluracil/chemistry , Arabinofuranosyluracil/metabolism , Arabinofuranosyluracil/pharmacokinetics , Female , Fluorine Radioisotopes , Guanine/analogs & derivatives , Guanine/chemistry , Guanine/metabolism , Guanine/pharmacokinetics , Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/genetics , Humans , Male , Mice , Middle Aged , Models, Molecular , Phosphorylation , Protein Conformation , Thymidine/pharmacokinetics , Thymidine Kinase/metabolism
17.
Mol Ther ; 20(5): 1002-13, 2012 May.
Article in English | MEDLINE | ID: mdl-22273576

ABSTRACT

The safety of cell therapy applications can be enhanced by the introduction of Cell Fate Control (CFC) elements, which encode pharmacologically controlled cellular suicide switches. CFC Gene Therapy (CFCGT) offers the possibility of establishing control over gene-modified cells (GMCs) with regards to their proliferation, differentiation, or function. However, enzymes commonly employed in these approaches often possess poor kinetics and high immunogenicity. We describe a novel CFCGT system based on engineered variants of human deoxyCytidine Kinase (dCK) that overcomes limitations of current modalities. Mutants of dCK with rationally designed active sites that make them thymidine-activating were stably introduced into cells by recombinant lentiviral vectors (LVs). Transduced cells maintained growth kinetics and function. These dCK mutants efficiently activate bromovinyl-deoxyuridine (BVdU), L-deoxythymidine (LdT), and L-deoxyuridine (LdU), which are otherwise not toxic to wild-type cells. We show that mutant dCK-expressing Jurkat, Molt-4, and U87mg cells could be efficiently eliminated in vitro and in xenogeneic leukemia and tumor models in vivo. We also describe a fusion construct of the thymidine-activating dCK to the cytoplasmic tail-truncated LNGFR molecule and applications to in vivo eradication of primary human T cells. This novel CFCGT system offers unique plasticity with respect to the wide range of prodrugs it can potentiate, and can be used as a reliable safety switch in cell and gene therapy.


Subject(s)
Deoxycytidine Kinase/genetics , Genetic Therapy/methods , Neoplasms/drug therapy , Animals , Bromodeoxyuridine/administration & dosage , Catalytic Domain , Cell Line, Tumor , Deoxycytidine Kinase/metabolism , Deoxyuridine/administration & dosage , Enzyme Activation/drug effects , Genetic Engineering , Genetic Vectors , Humans , Lentivirus/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Primary Cell Culture , Prodrugs/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Thymidine/administration & dosage , Thymidine/genetics , Transduction, Genetic , Xenograft Model Antitumor Assays
18.
Biochemistry ; 51(34): 6816-26, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22861376

ABSTRACT

Asparaginases catalyze the hydrolysis of the amino acid asparagine to aspartate and ammonia. Bacterial asparaginases are used in cancer chemotherapy to deplete asparagine from the blood, because several hematological malignancies depend on extracellular asparagine for growth. To avoid the immune response against the bacterial enzymes, it would be beneficial to replace them with human asparaginases. However, unlike the bacterial asparaginases, the human enzymes have a millimolar K(m) value for asparagine, making them inefficient in depleting the amino acid from blood. To facilitate the development of human variants suitable for therapeutic use, we determined the structure of human l-asparaginase (hASNase3). This asparaginase is an N-terminal nucleophile (Ntn) family member that requires autocleavage between Gly167 and Thr168 to become catalytically competent. For most Ntn hydrolases, this autoproteolytic activation occurs efficiently. In contrast, hASNas3 is relatively stable in its uncleaved state, and this allowed us to observe the structure of the enzyme prior to cleavage. To determine the structure of the cleaved state, we exploited our discovery that the free amino acid glycine promotes complete cleavage of hASNase3. Both enzyme states were elucidated in the absence and presence of the product aspartate. Together, these structures provide insight into the conformational changes required for cleavage and the precise enzyme-substrate interactions. The new understanding of hASNase3 will serve to guide the design of variants that possess a decreased K(m) value for asparagine, making the human enzyme a suitable replacement for the bacterial asparaginases in cancer therapy.


Subject(s)
Asparaginase/chemistry , Asparaginase/metabolism , Asparagine/metabolism , Amino Acid Sequence , Apoenzymes/chemistry , Apoenzymes/genetics , Apoenzymes/metabolism , Asparaginase/genetics , Binding Sites , Biocatalysis , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Sequence Alignment , Substrate Specificity
19.
J Biol Chem ; 286(50): 43352-60, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22030391

ABSTRACT

Tight junctions are cell-cell contacts that regulate the paracellular flux of solutes and prevent pathogen entry across cell layers. The assembly and permeability of this barrier are dependent on the zonula occludens (ZO) membrane-associated guanylate kinase (MAGUK) proteins ZO-1, -2, and -3. MAGUK proteins are characterized by a core motif of protein-binding domains that include a PDZ domain, a Src homology 3 (SH3) domain, and a region of homology to guanylate kinase (GUK); the structure of this core motif has never been determined for any MAGUK. To better understand how ZO proteins organize the assembly of protein complexes we have crystallized the entire PDZ3-SH3-GUK core motif of ZO-1. We have also crystallized this core motif in complex with the cytoplasmic tail of the ZO-1 PDZ3 ligand, junctional adhesion molecule A (JAM-A) to determine how the activity of different domains is coordinated. Our study shows a new feature for PDZ class II ligand binding that implicates the two highly conserved Phe(-2) and Ser(-3) residues of JAM. Our x-ray structures and NMR experiments also show for the first time a role for adjacent domains in the binding of ligands to PDZ domains in the MAGUK proteins family.


Subject(s)
Membrane Proteins/metabolism , PDZ Domains/physiology , Phosphoproteins/metabolism , src Homology Domains/physiology , Cell Adhesion Molecules/metabolism , Crystallography, X-Ray , Humans , Junctional Adhesion Molecules , Membrane Proteins/chemistry , Membrane Proteins/genetics , PDZ Domains/genetics , Phosphoproteins/chemistry , Phosphoproteins/genetics , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Tight Junctions/metabolism , Zonula Occludens-1 Protein , src Homology Domains/genetics
20.
J Biol Chem ; 286(5): 3531-9, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21087925

ABSTRACT

The biosynthetic shikimate pathway consists of seven enzymes that catalyze sequential reactions to generate chorismate, a critical branch point in the synthesis of the aromatic amino acids. The third enzyme in the pathway, dehydroquinate dehydratase (DHQD), catalyzes the dehydration of 3-dehydroquinate to 3-dehydroshikimate. We present three crystal structures of the type I DHQD from the intestinal pathogens Clostridium difficile and Salmonella enterica. Structures of the enzyme with substrate and covalent pre- and post-dehydration reaction intermediates provide snapshots of successive steps along the type I DHQD-catalyzed reaction coordinate. These structures reveal that the position of the substrate within the active site does not appreciably change upon Schiff base formation. The intermediate state structures reveal a reaction state-dependent behavior of His-143 in which the residue adopts a conformation proximal to the site of catalytic dehydration only when the leaving group is present. We speculate that His-143 is likely to assume differing catalytic roles in each of its observed conformations. One conformation of His-143 positions the residue for the formation/hydrolysis of the covalent Schiff base intermediates, whereas the other conformation positions the residue for a role in the catalytic dehydration event. The fact that the shikimate pathway is absent from humans makes the enzymes of the pathway potential targets for the development of non-toxic antimicrobials. The structures and mechanistic insight presented here may inform the design of type I DHQD enzyme inhibitors.


Subject(s)
Clostridioides difficile/enzymology , Hydro-Lyases/chemistry , Salmonella enterica/enzymology , Bacterial Proteins , Catalysis , Catalytic Domain , Crystallography, X-Ray , Hydro-Lyases/metabolism , Protein Binding , Protein Conformation , Quinic Acid/analogs & derivatives , Quinic Acid/chemistry , Quinic Acid/metabolism , Schiff Bases , Shikimic Acid/analogs & derivatives , Shikimic Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL