Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Int J Mol Sci ; 23(7)2022 Mar 31.
Article in English | MEDLINE | ID: mdl-35409221

ABSTRACT

Glycogen synthase kinase 3 beta (GSK-3ß) is an evolutionarily conserved serine-threonine kinase dysregulated in numerous pathologies, such as Alzheimer's disease and cancer. Even though GSK-3ß is a validated pharmacological target most of its inhibitors have two main limitations: the lack of selectivity due to the high homology that characterizes the ATP binding site of most kinases, and the toxicity that emerges from GSK-3ß complete inhibition which translates into the impairment of the plethora of pathways GSK-3ß is involved in. Starting from a 1D 19F NMR fragment screening, we set up several biophysical assays for the identification of GSK-3ß inhibitors capable of binding protein hotspots other than the ATP binding pocket or to the ATP binding pocket, but with an affinity able of competing with a reference binder. A phosphorylation activity assay on a panel of several kinases provided selectivity data that were further rationalized and corroborated by structural information on GSK-3ß in complex with the hit compounds. In this study, we identified promising fragments, inhibitors of GSK-3ß, while proposing an alternative screening workflow that allows facing the flaws that characterize the most common GSK-3ß inhibitors through the identification of selective inhibitors and/or inhibitors able to modulate GSK-3ß activity without leading to its complete inhibition.


Subject(s)
Alzheimer Disease , Adenosine Triphosphate/metabolism , Alzheimer Disease/metabolism , Binding Sites , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Phosphorylation
2.
Int J Mol Sci ; 22(21)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34769210

ABSTRACT

After almost two years from its first evidence, the COVID-19 pandemic continues to afflict people worldwide, highlighting the need for multiple antiviral strategies. SARS-CoV-2 main protease (Mpro/3CLpro) is a recognized promising target for the development of effective drugs. Because single target inhibition might not be sufficient to block SARS-CoV-2 infection and replication, multi enzymatic-based therapies may provide a better strategy. Here we present a structural and biochemical characterization of the binding mode of MG-132 to both the main protease of SARS-CoV-2, and to the human Cathepsin-L, suggesting thus an interesting scaffold for the development of double-inhibitors. X-ray diffraction data show that MG-132 well fits into the Mpro active site, forming a covalent bond with Cys145 independently from reducing agents and crystallization conditions. Docking of MG-132 into Cathepsin-L well-matches with a covalent binding to the catalytic cysteine. Accordingly, MG-132 inhibits Cathepsin-L with nanomolar potency and reversibly inhibits Mpro with micromolar potency, but with a prolonged residency time. We compared the apo and MG-132-inhibited structures of Mpro solved in different space groups and we identified a new apo structure that features several similarities with the inhibited ones, offering interesting perspectives for future drug design and in silico efforts.


Subject(s)
COVID-19 Drug Treatment , Cathepsin L/drug effects , Coronavirus 3C Proteases/drug effects , Leupeptins/chemistry , Leupeptins/pharmacology , SARS-CoV-2/chemistry , SARS-CoV-2/drug effects , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Catalytic Domain/drug effects , Cathepsin L/chemistry , Coronavirus 3C Proteases/chemistry , Drug Design , Drug Discovery , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptidomimetics , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Virus Replication/drug effects , X-Ray Diffraction
3.
Int J Mol Sci ; 22(3)2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33525510

ABSTRACT

Huntington disease (HD) is a devastating and presently untreatable neurodegenerative disease characterized by progressively disabling motor and mental manifestations. The sigma-1 receptor (σ1R) is a protein expressed in the central nervous system, whose 3D structure has been recently determined by X-ray crystallography and whose agonists have been shown to have neuroprotective activity in neurodegenerative diseases. To identify therapeutic agents against HD, we have implemented a drug repositioning strategy consisting of: (i) Prediction of the ability of the FDA-approved drugs publicly available through the ZINC database to interact with σ1R by virtual screening, followed by computational docking and visual examination of the 20 highest scoring drugs; and (ii) Assessment of the ability of the six drugs selected by computational analyses to directly bind purified σ1R in vitro by Surface Plasmon Resonance and improve the growth of fibroblasts obtained from HD patients, which is significantly impaired with respect to control cells. All six of the selected drugs proved able to directly bind purified σ1R in vitro and improve the growth of HD cells from both or one HD patient. These results support the validity of the drug repositioning procedure implemented herein for the identification of new therapeutic tools against HD.


Subject(s)
Fibroblasts/cytology , Huntington Disease/metabolism , Pharmaceutical Preparations/chemistry , Receptors, sigma/metabolism , Adult , Cell Proliferation , Cells, Cultured , Computer Simulation , Databases, Pharmaceutical , Drug Evaluation, Preclinical , Drug Repositioning , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Huntington Disease/drug therapy , Male , Middle Aged , Models, Molecular , Molecular Docking Simulation , Protein Conformation , Receptors, sigma/chemistry , Structure-Activity Relationship , Surface Plasmon Resonance , Sigma-1 Receptor
4.
Protein Expr Purif ; 166: 105505, 2020 02.
Article in English | MEDLINE | ID: mdl-31563543

ABSTRACT

Recombinant antibodies can be expressed as fusion constructs in combination with tags which simplify their engineering into reliable and homogeneous immunoreagents by allowing site-specific, 1:1 functionalization. Several tags and corresponding reagents for recombinant protein derivatization have been proposed but benchmarking surveys for the evaluation of their effect on the characteristics of recombinant antibodies have not been reported. In this work we evaluated the impact on expression yields, shelf-stability, thermostability and binding affinity of a set of C-terminal tags fused to the same anti-Her2 nanobody. Furthermore, we assessed the efficiency of the derivatization process. The constructs always bore a 6xHis tag plus either the controls (EGFP and C-tag) or CLIP, HALO, AviTag, the LEPTG sequence recognized by Sortase A (Sortase tag), or a free cysteine. The advantages and drawbacks of the different systems were analyzed and discussed.


Subject(s)
Recombinant Fusion Proteins/genetics , Single-Domain Antibodies/genetics , Binding, Competitive , Cysteine/metabolism , Escherichia coli , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Genetic Vectors/genetics , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Oxidoreductases/chemistry , Oxidoreductases/genetics , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/genetics , Protein Stability , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/genetics , Recombinant Fusion Proteins/chemistry , Single-Domain Antibodies/chemistry
5.
Int J Mol Sci ; 20(20)2019 Oct 21.
Article in English | MEDLINE | ID: mdl-31640223

ABSTRACT

The formation of multienzymatic complexes allows for the fine tuning of many aspects of enzymatic functions, such as efficiency, localization, stability, and moonlighting. Here, we investigated, in solution, the structure of bacterial cysteine synthase (CS) complex. CS is formed by serine acetyltransferase (CysE) and O-acetylserine sulfhydrylase isozyme A (CysK), the enzymes that catalyze the last two steps of cysteine biosynthesis in bacteria. CysK and CysE have been proposed as potential targets for antibiotics, since cysteine and related metabolites are intimately linked to protection of bacterial cells against redox damage and to antibiotic resistance. We applied a combined approach of small-angle X-ray scattering (SAXS) spectroscopy and protein painting to obtain a model for the solution structure of CS. Protein painting allowed the identification of protein-protein interaction hotspots that were then used as constrains to model the CS quaternary assembly inside the SAXS envelope. We demonstrate that the active site entrance of CysK is involved in complex formation, as suggested by site-directed mutagenesis and functional studies. Furthermore, complex formation involves a conformational change in one CysK subunit that is likely transmitted through the dimer interface to the other subunit, with a regulatory effect. Finally, SAXS data indicate that only one active site of CysK is involved in direct interaction with CysE and unambiguously unveil the quaternary arrangement of CS.


Subject(s)
Bacteria/enzymology , Cysteine Synthase/chemistry , Cysteine Synthase/metabolism , Serine O-Acetyltransferase/chemistry , Serine O-Acetyltransferase/metabolism , Bacteria/chemistry , Bacteria/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Cysteine Synthase/genetics , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Models, Molecular , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Mutagenesis, Site-Directed , Protein Interaction Maps , Scattering, Small Angle , Serine O-Acetyltransferase/genetics , X-Ray Diffraction
6.
J Struct Biol ; 203(2): 71-80, 2018 08.
Article in English | MEDLINE | ID: mdl-29545204

ABSTRACT

Baculovirus-insect cell expression system has become one of the most widely used eukaryotic expression systems for heterologous protein production in many laboratories. The availability of robust insect cell lines, serum-free media, a range of vectors and commercially-packaged kits have supported the demand for maximizing the exploitation of the baculovirus-insect cell expression system. Naturally, this resulted in varied strategies adopted by different laboratories to optimize protein production. Most laboratories have preference in using either the E. coli transposition-based recombination bacmid technology (e.g. Bac-to-Bac®) or homologous recombination transfection within insect cells (e.g. flashBAC™). Limited data is presented in the literature to benchmark the protocols used for these baculovirus vectors to facilitate the selection of a system for optimal production of target proteins. Taking advantage of the Protein Production and Purification Partnership in Europe (P4EU) scientific network, a benchmarking initiative was designed to compare the diverse protocols established in thirteen individual laboratories. This benchmarking initiative compared the expression of four selected intracellular proteins (mouse Dicer-2, 204 kDa; human ABL1 wildtype, 126 kDa; human FMRP, 68 kDa; viral vNS1-H1, 76 kDa). Here, we present the expression and purification results on these proteins and highlight the significant differences in expression yields obtained using different commercially-packaged baculovirus vectors. The highest expression level for difficult-to-express intracellular protein candidates were observed with the EmBacY baculovirus vector system.


Subject(s)
Baculoviridae/genetics , Genetic Vectors/genetics , Recombinant Proteins/metabolism , Animals , Cell Line , Escherichia coli/genetics , Escherichia coli/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Humans , Mice , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Recombinant Proteins/genetics , Ribonuclease III/genetics , Ribonuclease III/metabolism , Sf9 Cells
7.
Eur J Med Chem ; 229: 114054, 2022 Feb 05.
Article in English | MEDLINE | ID: mdl-34959172

ABSTRACT

The human kinome plays a crucial role in several pathways. Its dysregulation has been linked to diverse central nervous system (CNS)-related disorders with a drastic impact on the aging population. Among them, tauopathies, such as Alzheimer's Disease (AD) and Frontotemporal Lobar degeneration (FTLD-tau), are neurodegenerative disorders pathologically defined by the presence of hyperphosphorylated tau-positive intracellular inclusions known as neurofibrillary tangles (NFTs). Compelling evidence has reported the great potential of the simultaneous modulation of multiple protein kinases (PKs) involved in abnormal tau phosphorylation through a concerted pharmacological approach to achieve a superior therapeutic effect relative to classic "one target, one drug" approaches. Here, we report on the identification and characterization of ARN25068 (4), a low nanomolar and well-balanced dual GSK-3ß and FYN inhibitor, which also shows inhibitory activity against DYRK1A, an emerging target in AD and tauopathies. Computational and X-Ray studies highlight compound 4's typical H-bonding pattern of ATP-competitive inhibitors at the binding sites of all three PKs. In a tau phosphorylation assay on Tau0N4R-TM-tGFP U2OS cell line, 4 reduces the extent of tau phosphorylation, promoting tau-stabilized microtubule bundles. In conclusion, this compound emerges as a promising prototype for further SAR explorations to develop potent and well-balanced triple GSK-3ß/FYN/DYRK1A inhibitors to tackle tau hyperphosphorylation.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Neuroprotective Agents/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Tauopathies/drug therapy , Binding Sites , Drug Evaluation, Preclinical , Humans , Microtubules/metabolism , Models, Molecular , Neurofibrillary Tangles/metabolism , Neuroprotective Agents/pharmacology , Phosphorylation , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship , tau Proteins/metabolism , Dyrk Kinases
8.
ACS Pharmacol Transl Sci ; 4(3): 1096-1110, 2021 Jun 11.
Article in English | MEDLINE | ID: mdl-35287429

ABSTRACT

Compound repurposing is an important strategy for the identification of effective treatment options against SARS-CoV-2 infection and COVID-19 disease. In this regard, SARS-CoV-2 main protease (3CL-Pro), also termed M-Pro, is an attractive drug target as it plays a central role in viral replication by processing the viral polyproteins pp1a and pp1ab at multiple distinct cleavage sites. We here report the results of a repurposing program involving 8.7 K compounds containing marketed drugs, clinical and preclinical candidates, and small molecules regarded as safe in humans. We confirmed previously reported inhibitors of 3CL-Pro and have identified 62 additional compounds with IC50 values below 1 µM and profiled their selectivity toward chymotrypsin and 3CL-Pro from the Middle East respiratory syndrome virus. A subset of eight inhibitors showed anticytopathic effect in a Vero-E6 cell line, and the compounds thioguanosine and MG-132 were analyzed for their predicted binding characteristics to SARS-CoV-2 3CL-Pro. The X-ray crystal structure of the complex of myricetin and SARS-Cov-2 3CL-Pro was solved at a resolution of 1.77 Å, showing that myricetin is covalently bound to the catalytic Cys145 and therefore inhibiting its enzymatic activity.

9.
J Am Chem Soc ; 132(49): 17570-7, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21082835

ABSTRACT

Conformational changes modulate macromolecular function by promoting the specific binding of ligands (such as in antigen recognition) or the stabilization of transition states in enzymatic reactions. However, quantitative characterization of the energetics underlying dynamic structural interconversions is still challenging and lacks a unified method. Here, we introduce a novel in silico approach based on the combined use of essential dynamics sampling and nonequilibrium free-energy calculations to obtain quantitative data on conformational energy landscapes. This technique allows the unbiased investigation of highly complex rearrangements, and does not require the crucial definition of user-defined collective variables. We show that free-energy values derived from profiles connecting the unliganded and ligand-bound X-ray structures of a bacterial nucleoside hydrolase match the experimental binding constant. This approach also provides first evidence for a rate-limiting character of the conformational transition in this enzyme, and an unexpected role of the protonation state of a single residue in regulating substrate binding and product release.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Imino Furanoses/metabolism , N-Glycosyl Hydrolases/chemistry , N-Glycosyl Hydrolases/metabolism , Phenylenediamines/metabolism , Crystallography, X-Ray , Escherichia coli/chemistry , Escherichia coli/metabolism , Imino Furanoses/chemistry , Ligands , Molecular Dynamics Simulation , Phenylenediamines/chemistry , Protein Binding , Protein Conformation , Thermodynamics
10.
J Chem Theory Comput ; 15(8): 4646-4659, 2019 Aug 13.
Article in English | MEDLINE | ID: mdl-31246463

ABSTRACT

It is widely accepted that drug-target association and dissociation rates directly affect drug efficacy and safety. To rationally optimize drug binding kinetics, one must know the atomic arrangement of the protein-ligand complex during the binding/unbinding process in order to detect stable and metastable states. Whereas experimental approaches can determine kinetic constants with fairly good accuracy, computational approaches based on molecular dynamics (MD) simulations can deliver the atomistic details of the unbinding process. Furthermore, they can also be utilized prospectively to predict residence time (i.e., the inverse of unbinding kinetics constant, koff) with an acceptable level of accuracy. Here, we report a novel method based on adiabatic bias MD with an electrostatics-like collective variable (dubbed elABMD) for sampling protein-ligand dissociation events in two kinases. elABMD correctly ranked a ligand series on glucokinase, in agreement with experimental data and previous calculations. Subsequently, we applied the new method prospectively to a congeneric series of GSK-3ß inhibitors. For this series, new crystal structures were generated and the residence time was experimentally measured with surface plasmon resonance (SPR). There was good agreement between computational predictions and experimental measures, suggesting that elABMD is an innovative and efficient tool for calculating residence times.


Subject(s)
Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/metabolism , Protein Kinase Inhibitors/pharmacology , Crystallography, X-Ray , Glycogen Synthase Kinase 3 beta/chemistry , Humans , Kinetics , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Protein Kinase Inhibitors/chemistry , Static Electricity , Thermodynamics
11.
Biochim Biophys Acta Proteins Proteom ; 1866(12): 1260-1270, 2018 12.
Article in English | MEDLINE | ID: mdl-30268810

ABSTRACT

Methionine deprivation of cancer cells, which are deficient in methionine biosynthesis, has been envisioned as a therapeutic strategy to reduce cancer cell viability. Methionine γ-lyase (MGL), an enzyme that degrades methionine, has been exploited to selectively remove the amino acid from cancer cell environment. In order to increase MGL catalytic activity, we performed sequence and structure conservation analysis of MGLs from various microorganisms. Whereas most of the residues in the active site and at the dimer interface were found to be conserved, residues located in the C-terminal flexible loop, forming a wall of the active site entry channel, were found to be variable. Therefore, we carried out site-saturation mutagenesis at four independent positions of the C-terminal flexible loop, P357, V358, P360 and A366 of MGL from Citrobacter freundii, generating libraries that were screened for activity. Among the active variants, V358Y exhibits a 1.9-fold increase in the catalytic rate and a 3-fold increase in KM, resulting in a catalytic efficiency similar to wild type MGL. V358Y cytotoxic activity was assessed towards a panel of cancer and nonmalignant cell lines and found to exhibit IC50 lower than the wild type. The comparison of the 3D-structure of V358Y MGL with other MGL available structures indicates that the C-terminal loop is either in an open or closed conformation that does not depend on the amino acid at position 358. Nevertheless, mutations at this position allosterically affects catalysis.


Subject(s)
Bacterial Proteins/metabolism , Carbon-Sulfur Lyases/metabolism , Citrobacter freundii/enzymology , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Carbon-Sulfur Lyases/chemistry , Carbon-Sulfur Lyases/genetics , Catalytic Domain , Cell Proliferation/drug effects , Crystallography, X-Ray , Kinetics , Mutagenesis, Site-Directed , Protein Engineering , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
12.
Nat Commun ; 8: 15772, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28598431

ABSTRACT

The prolyl isomerase PIN1, a critical modifier of multiple signalling pathways, is overexpressed in the majority of cancers and its activity strongly contributes to tumour initiation and progression. Inactivation of PIN1 function conversely curbs tumour growth and cancer stem cell expansion, restores chemosensitivity and blocks metastatic spread, thus providing the rationale for a therapeutic strategy based on PIN1 inhibition. Notwithstanding, potent PIN1 inhibitors are still missing from the arsenal of anti-cancer drugs. By a mechanism-based screening, we have identified a novel covalent PIN1 inhibitor, KPT-6566, able to selectively inhibit PIN1 and target it for degradation. We demonstrate that KPT-6566 covalently binds to the catalytic site of PIN1. This interaction results in the release of a quinone-mimicking drug that generates reactive oxygen species and DNA damage, inducing cell death specifically in cancer cells. Accordingly, KPT-6566 treatment impairs PIN1-dependent cancer phenotypes in vitro and growth of lung metastasis in vivo.


Subject(s)
Antineoplastic Agents/administration & dosage , Enzyme Inhibitors/administration & dosage , Lung Neoplasms/drug therapy , NIMA-Interacting Peptidylprolyl Isomerase/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Enzyme Inhibitors/chemistry , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/physiopathology , Mice, Nude , NIMA-Interacting Peptidylprolyl Isomerase/chemistry , NIMA-Interacting Peptidylprolyl Isomerase/genetics , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Reactive Oxygen Species/metabolism
13.
Structure ; 12(5): 739-49, 2004 May.
Article in English | MEDLINE | ID: mdl-15130467

ABSTRACT

Enzymes with nucleoside hydrolase (NH) activity are crucial for salvaging nucleic acid components in purine auxotrophic protozoan parasites, but are also present in prokaryotes and higher eukaryotes. Here we analyze the distribution of genes encoding for putative NH proteins and characterize the yeiK gene product from Escherichia coli as a pyrimidine-specific NH. The crystal structure of YeiK to 1.7 A defines the structural basis for its substrate specificity and identifies residues involved in the catalytic mechanism that differ from both nonspecific and purine-specific NHs. Large differences in the tetrameric quaternary structure compared to nonspecific protozoan NHs are brought forth by minor differences in the interacting surfaces. The first structural and functional characterization of a nonparasitic, pyrimidine nucleoside-specific NH suggests a possible role for these enzymes in the metabolism of tRNA nucleosides. The high catalytic efficiency of YeiK toward 5-fluorouridine could be exploited for suicide gene therapy in cancer treatment.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Genetic Therapy , N-Glycosyl Hydrolases/chemistry , Neoplasms/therapy , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Molecular Sequence Data , Mutation , N-Glycosyl Hydrolases/genetics , Protein Structure, Tertiary
14.
BMC Immunol ; 6: 24, 2005 Dec 05.
Article in English | MEDLINE | ID: mdl-16329759

ABSTRACT

BACKGROUND: MHC class I-peptide tetramers are currently utilised to characterize CD8+ T cell responses at single cell level. The generation and use of MHC class II tetramers to study antigen-specific CD4+ T cells appears less straightforward. Most MHC class II tetramers are produced with a homogeneously built-in peptide, reducing greatly their flexibility of use. We attempted the generation of "empty" functional HLA-DR*1101 tetramers, receptive for loading with synthetic peptides by incubation. No such reagent is in fact available for this HLA-DR allele, one of the most frequent in the Caucasian population. RESULTS: We compared soluble MHC class II-immunoglobulin fusion proteins (HLA-DR*1101-Ig) with soluble MHC class II protein fused with an optimised Bir site for enzymatic biotynilation (HLA-DR*1101-Bir), both produced in insect cells. The molecules were multimerised by binding fluorochrome-protein A or fluorochrome-streptavidin, respectively. We find that HLA-DR*1101-Bir molecules are superior to the HLA-DR*1101-Ig ones both in biochemical and functional terms. HLA-DR*1101-Bir molecules can be pulsed with at least three different promiscuous peptide epitopes, derived from Tetanus Toxoid, influenza HA and the tumour associated antigen MAGE-3 respectively, to stain specific CD4+ T cells. Both staining temperature and activation state of CD4+ T cells are critical for the binding of peptide-pulsed HLA-DR*1101-Bir to the cognate TCR. CONCLUSION: It is therefore possible to generate a soluble recombinant HLA-DR*1101 backbone that is receptive for loading with different peptides to stain specific CD4+ T cells. As shown for other HLA-DR alleles, we confirm that not all the strategies to produce soluble HLA-DR*1101 multimers are equivalent.


Subject(s)
Antigen Presentation , Antigens, Neoplasm/immunology , HLA-DR Antigens/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Tetanus Toxoid/immunology , Animals , Antigens, Neoplasm/chemistry , Biotinylation , CD4-Positive T-Lymphocytes/immunology , Cell Line , DNA, Complementary/genetics , Drosophila melanogaster/cytology , Epitopes/immunology , Genes, Immunoglobulin , Genes, MHC Class II , Genes, Synthetic , HLA-DR Antigens/chemistry , HLA-DR Antigens/genetics , HLA-DRB1 Chains , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Leucine Zippers , Neoplasm Proteins/chemistry , Peptide Fragments/chemistry , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Transfection
15.
Biochemistry ; 47(15): 4418-26, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18361502

ABSTRACT

Enzymes with nucleoside hydrolase activity (NHs) belonging to homology group I either are markedly specific for pyrimidine nucleoside substrates or hydrolyze with comparable efficiencies the N-glycosidic bond in all common nucleosides. The biochemical and structural basis for these differences in substrate specificity is still unknown. Here we characterize the binding interactions between the slowly hydrolyzed substrate inosine and the Escherichia coli pyrimidine-specific NH YeiK using cryotrapping and X-ray crystallography. Guided by the structural features of the Michaelis complex, we show the synergic effect of two specific point mutations in YeiK that increase the catalytic efficiency toward purine nucleosides to values comparable to those of natural nonspecific NHs. We demonstrate that the integrity of an active-site catalytic triad comprised of two hydroxylated amino acids and one histidine residue is a requirement for the highly efficient hydrolysis of inosine by group I NHs. Instead, cleavage of the YeiK-preferred substrate uridine is not affected by mutations at the same locations, suggesting a different fine chemical mechanism for the hydrolysis of the two nucleoside substrates. Our study provides for the first time direct evidence that distinct subsets of amino acid residues are involved in the hydrolysis of purine or pyrimidine nucleosides in group I NHs.


Subject(s)
Escherichia coli Proteins/chemistry , N-Glycosyl Hydrolases/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Inosine/chemistry , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , N-Glycosyl Hydrolases/genetics , N-Glycosyl Hydrolases/metabolism , Sequence Alignment , Substrate Specificity , Uridine/chemistry
16.
J Biol Chem ; 281(1): 383-91, 2006 Jan 06.
Article in English | MEDLINE | ID: mdl-16257969

ABSTRACT

Mammalian antimicrobial peptides provide rapid defense against infection by inactivating pathogens and by influencing the functions of cells involved in defense responses. Although the direct antibacterial properties of these peptides have been widely characterized, their multiple effects on host cells are only beginning to surface. Here we investigated the mechanistic and functional aspects of the interaction of the proline-rich antimicrobial peptide Bac7(1-35) with mammalian cells, as compared with a truncated analog, Bac7(5-35), lacking four critical N-terminal residues (RRIR) of the Bac7(1-35) sequence. By using confocal microscopy and flow cytometry, we showed that although the truncated analog Bac7(5-35) remains on the cell surface, Bac7(1-35) is rapidly taken up into 3T3 and U937 cells through a nontoxic energy- and temperature-dependent process. Cell biology-based assays using selective endocytosis inhibitors and spectroscopic and surface plasmon resonance studies of the interaction of Bac7(1-35) with phosphatidylcholine/cholesterol model membranes collectively suggest the concurrent contribution of macropinocytosis and direct membrane translocation. Structural studies with model membranes indicated that membrane-bound Bac7(5-35) is significantly more aggregated than Bac7(1-35) due to the absence of the N-terminal cationic cluster, thus providing an explanation for hampered cellular internalization of the truncated form. Further investigations aimed to reveal functional implications of intracellular uptake of Bac7(1-35) demonstrated that it correlates with enhanced S phase entry of 3T3 cells, indicating a novel function for this proline-rich peptide.


Subject(s)
Antimicrobial Cationic Peptides/pharmacokinetics , Endocytosis , Lipid Bilayers/metabolism , Animals , Antimicrobial Cationic Peptides/chemistry , Cell Membrane/metabolism , Cholesterol/metabolism , DNA/biosynthesis , Escherichia coli/drug effects , Flow Cytometry , Humans , Mammals , Mice , Microscopy, Confocal , NIH 3T3 Cells , Phosphatidylcholines/metabolism , Pinocytosis , Proline/metabolism , S Phase , Spectroscopy, Fourier Transform Infrared , Staphylococcus aureus/drug effects , Surface Plasmon Resonance , U937 Cells
17.
J Immunol ; 175(2): 977-84, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-16002697

ABSTRACT

NKT cells are immunoregulatory lymphocytes whose activation is triggered by the recognition of lipid Ags in the context of the CD1d molecules by the TCR. In this study we present the crystal structure to 2.8 A of mouse CD1d bound to phosphatidylcholine. The interactions between the ligand acyl chains and the CD1d molecule define the structural and chemical requirements for the binding of lipid Ags to CD1d. The orientation of the polar headgroup toward the C terminus of the alpha1 helix provides a rationale for the structural basis for the observed Valpha chain bias in invariant NKT cells. The contribution of the ligand to the protein surface suggests a likely mode of recognition of lipid Ags by the NKT cell TCR.


Subject(s)
Antigens, CD1/chemistry , Antigens, CD1/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Phosphatidylcholines/chemistry , Phosphatidylcholines/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigens, CD1/metabolism , Antigens, CD1d , Cell Line , Complementarity Determining Regions/metabolism , Crystallography, X-Ray , Drosophila melanogaster , Killer Cells, Natural/metabolism , Ligands , Mice , Models, Molecular , Phosphatidylcholines/metabolism , Protein Binding/immunology , Protein Isoforms/chemistry , Protein Isoforms/immunology , Protein Isoforms/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Surface Properties , T-Lymphocyte Subsets/metabolism
18.
Acta Crystallogr D Biol Crystallogr ; 60(Pt 3): 524-7, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14993681

ABSTRACT

The E. coli yeiK gene product is homologous to members of the nucleoside hydrolase family of enzymes, the physiological role of which in bacteria is still unknown. Here, the cloning, expression in milligram quantities and enzymatic characterization of YeiK as a pyrimidine-specific nucleoside hydrolase is reported. Crystals of YeiK diffract X-rays to a resolution of 1.7 A and belong to the triclinic crystal system in space group P1, with unit-cell parameters a = 44.81, b = 85.71, c = 90.68 A, alpha = 112.95, beta = 101.95, gamma = 85.92 degrees.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/isolation & purification , Escherichia coli/enzymology , N-Glycosyl Hydrolases/chemistry , N-Glycosyl Hydrolases/isolation & purification , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Gene Expression , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL