Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Proc Natl Acad Sci U S A ; 120(49): e2311240120, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38019859

ABSTRACT

High-resolution NMR spectroscopy enabled us to characterize allosteric transitions between various functional states of the dimeric Escherichia coli Lac repressor. In the absence of ligands, the dimer exists in a dynamic equilibrium between DNA-bound and inducer-bound conformations. Binding of either effector shifts this equilibrium toward either bound state. Analysis of the ternary complex between repressor, operator DNA, and inducer shows how adding the inducer results in allosteric changes that disrupt the interdomain contacts between the inducer binding and DNA binding domains and how this in turn leads to destabilization of the hinge helices and release of the Lac repressor from the operator. Based on our data, the allosteric mechanism of the induction process is in full agreement with the well-known Monod-Wyman-Changeux model.


Subject(s)
Escherichia coli Proteins , Lac Repressors/genetics , Lac Repressors/metabolism , Escherichia coli Proteins/metabolism , Allosteric Regulation/genetics , Escherichia coli/metabolism , DNA/metabolism , Protein Structure, Secondary , Lac Operon/genetics
2.
Am J Hum Genet ; 101(5): 844-855, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29100094

ABSTRACT

A series of simplex cases have been reported under various diagnoses sharing early aging, especially evident in congenitally decreased subcutaneous fat tissue and sparse hair, bone dysplasia of the skull and fingers, a distinctive facial gestalt, and prenatal and postnatal growth retardation. For historical reasons, we suggest naming the entity Fontaine syndrome. Exome sequencing of four unrelated affected individuals showed that all carried the de novo missense variant c.649C>T (p.Arg217Cys) or c.650G>A (p.Arg217His) in SLC25A24, a solute carrier 25 family member coding for calcium-binding mitochondrial carrier protein (SCaMC-1, also known as SLC25A24). SLC25A24 allows an electro-neutral and reversible exchange of ATP-Mg and phosphate between the cytosol and mitochondria, which is required for maintaining optimal adenine nucleotide levels in the mitochondrial matrix. Molecular dynamic simulation studies predict that p.Arg217Cys and p.Arg217His narrow the substrate cavity of the protein and disrupt transporter dynamics. SLC25A24-mutant fibroblasts and cells expressing p.Arg217Cys or p.Arg217His variants showed altered mitochondrial morphology, a decreased proliferation rate, increased mitochondrial membrane potential, and decreased ATP-linked mitochondrial oxygen consumption. The results suggest that the SLC25A24 mutations lead to impaired mitochondrial ATP synthesis and cause hyperpolarization and increased proton leak in association with an impaired energy metabolism. Our findings identify SLC25A24 mutations affecting codon 217 as the underlying genetic cause of human progeroid Fontaine syndrome.


Subject(s)
Aging/genetics , Antiporters/genetics , Bone Diseases, Developmental/genetics , Calcium-Binding Proteins/genetics , Mitochondrial Proteins/genetics , Mutation/genetics , Adenine/metabolism , Adenosine Triphosphate/metabolism , Cytosol/metabolism , Female , Fetal Death , Fibroblasts/metabolism , Humans , Infant , Infant, Newborn , Male , Membrane Potential, Mitochondrial/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Molecular Dynamics Simulation , Oxygen/metabolism , Phosphates/metabolism , Syndrome
3.
J Biol Chem ; 293(23): 8750-8760, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29674345

ABSTRACT

Protein-protein interactions critically regulate many biological systems, but quantifying functional assembly of multipass membrane complexes in their native context is still challenging. Here, we combined modeling-assisted protein modification and information from human disease variants with a minimal-size fusion tag, split-luciferase-based approach to probe assembly of the NADPH oxidase 4 (NOX4)-p22phox enzyme, an integral membrane complex with unresolved structure, which is required for electron transfer and generation of reactive oxygen species (ROS). Integrated analyses of heterodimerization, trafficking, and catalytic activity identified determinants for the NOX4-p22phox interaction, such as heme incorporation into NOX4 and hot spot residues in transmembrane domains 1 and 4 in p22phox Moreover, their effect on NOX4 maturation and ROS generation was analyzed. We propose that this reversible and quantitative protein-protein interaction technique with its small split-fragment approach will provide a protein engineering and discovery tool not only for NOX research, but also for other intricate membrane protein complexes, and may thereby facilitate new drug discovery strategies for managing NOX-associated diseases.


Subject(s)
NADPH Oxidase 4/metabolism , NADPH Oxidases/metabolism , Protein Interaction Mapping/methods , Protein Interaction Maps , Animals , COS Cells , Cell Membrane/chemistry , Cell Membrane/metabolism , Chlorocebus aethiops , Heme/chemistry , Heme/metabolism , Humans , Models, Molecular , NADPH Oxidase 4/chemistry , NADPH Oxidases/chemistry , Protein Domains , Protein Multimerization , Reactive Oxygen Species/metabolism
4.
Proc Natl Acad Sci U S A ; 113(37): 10406-11, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27562167

ABSTRACT

Strengthening the host immune system to fully exploit its potential as antimicrobial defense is vital in countering antibiotic resistance. Chemical compounds released during bidirectional host-pathogen cross-talk, which follows a sensing-response paradigm, can serve as protective mediators. A potent, diffusible messenger is hydrogen peroxide (H2O2), but its consequences on extracellular pathogens are unknown. Here we show that H2O2, released by the host on pathogen contact, subverts the tyrosine signaling network of a number of bacteria accustomed to low-oxygen environments. This defense mechanism uses heme-containing bacterial enzymes with peroxidase-like activity to facilitate phosphotyrosine (p-Tyr) oxidation. An intrabacterial reaction converts p-Tyr to protein-bound dopa (PB-DOPA) via a tyrosinyl radical intermediate, thereby altering antioxidant defense and inactivating enzymes involved in polysaccharide biosynthesis and metabolism. Disruption of bacterial signaling by DOPA modification reveals an infection containment strategy that weakens bacterial fitness and could be a blueprint for antivirulence approaches.


Subject(s)
Dihydroxyphenylalanine/metabolism , Host-Pathogen Interactions/drug effects , Hydrogen Peroxide/metabolism , NADPH Oxidases/metabolism , Tyrosine/metabolism , Campylobacter jejuni/metabolism , Campylobacter jejuni/pathogenicity , Cell Line , Dihydroxyphenylalanine/chemistry , Drug Resistance, Bacterial/immunology , Heme/chemistry , Heme/metabolism , Host-Pathogen Interactions/immunology , Humans , Immune System/metabolism , Immune System/microbiology , Klebsiella pneumoniae/metabolism , Klebsiella pneumoniae/pathogenicity , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , NADPH Oxidases/chemistry , Oxidation-Reduction , Oxidative Phosphorylation , Oxygen/metabolism , Peroxidase/chemistry , Peroxidase/metabolism , Phosphotyrosine/metabolism , Reactive Oxygen Species/metabolism , Salmonella enterica/metabolism , Salmonella enterica/pathogenicity
5.
J Biol Chem ; 292(7): 2842-2853, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28028171

ABSTRACT

The nucleotide excision repair protein complex ERCC1-XPF is required for incision of DNA upstream of DNA damage. Functional studies have provided insights into the binding of ERCC1-XPF to various DNA substrates. However, because no structure for the ERCC1-XPF-DNA complex has been determined, the mechanism of substrate recognition remains elusive. Here we biochemically characterize the substrate preferences of the helix-hairpin-helix (HhH) domains of XPF and ERCC-XPF and show that the binding to single-stranded DNA (ssDNA)/dsDNA junctions is dependent on joint binding to the DNA binding domain of ERCC1 and XPF. We reveal that the homodimeric XPF is able to bind various ssDNA sequences but with a clear preference for guanine-containing substrates. NMR titration experiments and in vitro DNA binding assays also show that, within the heterodimeric ERCC1-XPF complex, XPF specifically recognizes ssDNA. On the other hand, the HhH domain of ERCC1 preferentially binds dsDNA through the hairpin region. The two separate non-overlapping DNA binding domains in the ERCC1-XPF heterodimer jointly bind to an ssDNA/dsDNA substrate and, thereby, at least partially dictate the incision position during damage removal. Based on structural models, NMR titrations, DNA-binding studies, site-directed mutagenesis, charge distribution, and sequence conservation, we propose that the HhH domain of ERCC1 binds to dsDNA upstream of the damage, and XPF binds to the non-damaged strand within a repair bubble.


Subject(s)
DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Binding Sites , DNA-Binding Proteins/chemistry , Dimerization , Helix-Loop-Helix Motifs , Humans , Protein Binding , Substrate Specificity
6.
J Biol Chem ; 290(33): 20541-55, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26085086

ABSTRACT

The ERCC1-XPF heterodimer, a structure-specific DNA endonuclease, is best known for its function in the nucleotide excision repair (NER) pathway. The ERCC1 point mutation F231L, located at the hydrophobic interaction interface of ERCC1 (excision repair cross-complementation group 1) and XPF (xeroderma pigmentosum complementation group F), leads to severe NER pathway deficiencies. Here, we analyze biophysical properties and report the NMR structure of the complex of the C-terminal tandem helix-hairpin-helix domains of ERCC1-XPF that contains this mutation. The structures of wild type and the F231L mutant are very similar. The F231L mutation results in only a small disturbance of the ERCC1-XPF interface, where, in contrast to Phe(231), Leu(231) lacks interactions stabilizing the ERCC1-XPF complex. One of the two anchor points is severely distorted, and this results in a more dynamic complex, causing reduced stability and an increased dissociation rate of the mutant complex as compared with wild type. These data provide a biophysical explanation for the severe NER deficiencies caused by this mutation.


Subject(s)
Cockayne Syndrome/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , Point Mutation , Amino Acid Sequence , DNA-Binding Proteins/chemistry , Dimerization , Endonucleases/chemistry , Humans , Models, Chemical , Molecular Sequence Data , Sequence Homology, Amino Acid
7.
Bioorg Med Chem ; 23(15): 4264-4276, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26154240

ABSTRACT

Previously, we identified CYP53 as a fungal-specific target of natural phenolic antifungal compounds and discovered several inhibitors with antifungal properties. In this study, we performed similarity-based virtual screening and synthesis to obtain benzoic acid-derived compounds and assessed their antifungal activity against Cochliobolus lunatus, Aspergillus niger and Pleurotus ostreatus. In addition, we generated structural models of CYP53 enzyme and used them in docking trials with 40 selected compounds. Finally, we explored CYP53-ligand interactions and identified structural elements conferring increased antifungal activity to facilitate the development of potential new antifungal agents that specifically target CYP53 enzymes of animal and plant pathogenic fungi.


Subject(s)
Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Benzoic Acid/chemistry , Cytochromes/chemistry , Structure-Activity Relationship , Antifungal Agents/chemical synthesis , Ascomycota/drug effects , Aspergillus niger/drug effects , Cytochromes/metabolism , Drug Design , Drug Evaluation, Preclinical/methods , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Targeted Therapy/methods , Pleurotus/drug effects , Protein Conformation
8.
Nucleic Acids Res ; 41(21): 9901-10, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23965307

ABSTRACT

RecA protein is a hallmark for the bacterial response to insults inflicted on DNA. It catalyzes the strand exchange step of homologous recombination and stimulates self-inactivation of the LexA transcriptional repressor. Importantly, by these activities, RecA contributes to the antibiotic resistance of bacteria. An original way to decrease the acquisition of antibiotic resistance would be to block RecA association with LexA. To engineer inhibitors of LexA-RecA complex formation, we have mapped the interaction area between LexA and active RecA-ssDNA filament (RecA*) and generated a three-dimensional model of the complex. The model revealed that one subunit of the LexA dimer wedges into a deep helical groove of RecA*, forming multiple interaction sites along seven consecutive RecA protomers. Based on the model, we predicted that LexA in its DNA-binding conformation also forms a complex with RecA* and that the operator DNA sterically precludes interaction with RecA*, which guides the induction of SOS gene expression. Moreover, the model shows that besides the catalytic C-terminal domain of LexA, its N-terminal DNA-binding domain also interacts with RecA*. Because all the model-based predictions have been confirmed experimentally, the presented model offers a validated insight into the critical step of the bacterial DNA damage response.


Subject(s)
Bacterial Proteins/chemistry , Rec A Recombinases/chemistry , Repressor Proteins/chemistry , Serine Endopeptidases/chemistry , Amino Acid Sequence , Bacterial Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Operator Regions, Genetic , Rec A Recombinases/metabolism , Repressor Proteins/metabolism , SOS Response, Genetics , Serine Endopeptidases/metabolism
9.
Anal Bioanal Chem ; 406(1): 293-304, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24217948

ABSTRACT

In order to perform their function, proteins frequently interact with other proteins. Various methods are used to reveal protein interacting partners, and affinity chromatography is one of them. Snake venom is composed mostly of proteins, and various protein complexes in the venom have been found to exhibit higher toxicity levels than respective components separately. Complexes can modulate envenomation activity of a venom and/or potentiate its effect. Our previous data indicate that the most toxic components of the Vipera ammodytes ammodytes (Vaa) venom isolated so far-ammodytoxins (Atxs)-are contributing to the venom's toxicity only moderately; therefore, we aimed to explore whether they have some interacting partner(s) potentiating toxicity. For screening of possible interactions, immuno-affinity chromatography combined with identification by mass spectrometry was used. Various chemistries (epoxy, carbonyldiimidazole, ethylenediamine) as well as protein G functionality were used to immobilize antibodies on monolith support, a Convective Interaction Media disk. Monoliths have been demonstrated to better suit the separation of large biomolecules. Using such approach, several proteins were indicated as potential Atx-binding proteins. Among these, the interaction of Atxs with a Kunitz-type inhibitor was confirmed by far-Western dot-blot and surface plasmon resonance measurement. It can be concluded that affinity chromatography on monolithic columns combined with mass spectrometry identification is a successful approach for screening of protein interactions and it resulted with detection of the interaction of Atx with Kunitz-type inhibitor in Vaa venom for the first time.


Subject(s)
Antibodies, Immobilized/chemistry , Peptides/chemistry , Plant Proteins/chemistry , Reptilian Proteins/chemistry , Viper Venoms/chemistry , Animals , Chromatography, Affinity , Mass Spectrometry , Protein Binding , Protein Interaction Mapping , Viperidae/physiology
10.
Am J Ophthalmol ; 260: 70-83, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37460036

ABSTRACT

PURPOSE: To report the 100-week outcomes from the KESTREL and KITE trials. DESIGN: Two phase 3, double-masked, active-controlled, randomized trials. METHODS: Patients with diabetic macular edema (DME) were randomized 1:1:1 to brolucizumab 3 mg/6 mg (BRO3/BRO6) or aflibercept 2 mg (AFL) in KESTREL (N = 566) or 1:1 to BRO6 or AFL in KITE (N = 360). BRO3/BRO6 arms received 5 loading doses every 6 weeks (q6w) followed by q12w dosing, with an option to adjust to q8w at predefined disease activity assessment visits. In KITE, at week 72, based on the disease stability assessment, treatment intervals could be extended by 4 weeks in the BRO6 arm. AFL arms received 5 monthly loading doses followed by fixed q8w dosing. RESULTS: At week 100, change from baseline in BCVA (letters) was +8.8 for BRO6 and +10.6 for AFL in KESTREL; and +10.9 for BRO6 and +8.4 for AFL in KITE. In both studies, fewer BRO6 subjects had intraretinal fluid and/or subretinal fluid than AFL subjects. Results were achieved with 32.9% (KESTREL) and 47.5% (KITE) of BRO6 subjects maintained on q12w and q12w/q16w dosing, respectively. Intraocular inflammation rates for BRO6 vs AFL were 4.2% vs 1.1% (KESTREL) and 2.2% vs 1.7% (KITE), of which retinal vasculitis rates were 0.5% vs 0% in KESTREL, with no cases in KITE. Retinal vascular occlusion rates were 1.6% vs 0.5% (KESTREL) and 0.6% in both treatment arms in KITE. CONCLUSIONS: Results show the long-term efficacy and durability of brolucizumab in improving visual and anatomical outcomes in DME; the overall safety profile of brolucizumab remained unchanged through year 2.


Subject(s)
Antibodies, Monoclonal, Humanized , Diabetes Mellitus , Diabetic Retinopathy , Macular Edema , Humans , Angiogenesis Inhibitors/therapeutic use , Diabetic Retinopathy/complications , Diabetic Retinopathy/diagnosis , Diabetic Retinopathy/drug therapy , Intravitreal Injections , Macular Edema/diagnosis , Macular Edema/drug therapy , Macular Edema/etiology , Receptors, Vascular Endothelial Growth Factor/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Treatment Outcome , Visual Acuity
11.
Am J Ophthalmol ; 238: 157-172, 2022 06.
Article in English | MEDLINE | ID: mdl-35038415

ABSTRACT

PURPOSE: To compare the efficacy and safety of brolucizumab with aflibercept in patients with diabetic macular edema (DME). DESIGN: Double-masked, 100-week, multicenter, active-controlled, randomized trials. METHODS: Subjects were randomized 1:1:1 to brolucizumab 3 mg/6 mg or aflibercept 2 mg in KESTREL (n = 566) or 1:1 to brolucizumab 6 mg or aflibercept 2 mg in KITE (n = 360). Brolucizumab groups received 5 loading doses every 6 weeks (q6w) followed by 12-week (q12w) dosing, with optional adjustment to every 8 weeks (q8w) if disease activity was identified at predefined assessment visits; aflibercept groups received 5 doses every 4 weeks (q4w) followed by fixed q8w dosing. The primary endpoint was best-corrected visual acuity (BCVA) change from baseline at Week 52; secondary endpoints included the proportion of subjects maintained on q12w dosing, change in Diabetic Retinopathy Severity Scale score, and anatomical and safety outcomes. RESULTS: At Week 52, brolucizumab 6 mg was noninferior (NI margin 4 letters) to aflibercept in mean change in BCVA from baseline (KESTREL: +9.2 letters vs +10.5 letters; KITE: +10.6 letters vs +9.4 letters; P < .001), more subjects achieved central subfield thickness (CSFT) <280 µm, and fewer had persisting subretinal and/or intraretinal fluid vs aflibercept, with more than half of brolucizumab 6 mg subjects maintained on q12w dosing after loading. In KITE, brolucizumab 6 mg showed superior improvements in change of CSFT from baseline over Week 40 to Week 52 vs aflibercept (P = .001). The incidence of ocular serious adverse events was 3.7% (brolucizumab 3 mg), 1.1% (brolucizumab 6 mg), and 2.1% (aflibercept) in KESTREL; and 2.2% (brolucizumab 6 mg) and 1.7% (aflibercept) in KITE. CONCLUSION: Brolucizumab 6 mg showed robust visual gains and anatomical improvements with an overall favorable benefit/risk profile in patients with DME.


Subject(s)
Antibodies, Monoclonal, Humanized , Diabetes Mellitus , Diabetic Retinopathy , Macular Edema , Angiogenesis Inhibitors , Antibodies, Monoclonal, Humanized/therapeutic use , Diabetic Retinopathy/diagnosis , Diabetic Retinopathy/drug therapy , Humans , Intravitreal Injections , Macular Edema/diagnosis , Macular Edema/drug therapy , Receptors, Vascular Endothelial Growth Factor/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Treatment Outcome , Visual Acuity
12.
J Biomol NMR ; 51(3): 329-37, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21892794

ABSTRACT

Here we present Cys-Ph-TAHA, a new nonadentate lanthanide tag for the paramagnetic labelling of proteins. The tag can be easily synthesized and is stereochemically homogenous over a wide range of temperatures, yielding NMR spectra with a single set of peaks. Bound to ubiquitin, it induced large residual dipolar couplings and pseudocontact shifts that could be measured easily and agreed very well with the protein structure. We show that Cys-Ph-TAHA can be used to label large proteins that are biochemically challenging such as the Lac repressor in a 90 kDa ternary complex with DNA and inducer.


Subject(s)
Acetates/chemistry , Cysteine/analogs & derivatives , Cysteine/chemistry , Lanthanoid Series Elements/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Proteins/chemistry , Binding Sites , Models, Molecular , Protein Conformation , Ubiquitin/chemistry
13.
Biochem J ; 429(2): 379-89, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20450492

ABSTRACT

The human cysteine peptidase cathepsin K is a key enzyme in bone homoeostasis and other physiological functions. In the present study we investigate the mechanism of cathepsin K action at physiological plasma pH and its regulation by modifiers that bind outside of the active site. We show that at physiological plasma pH the enzyme fluctuates between multiple conformations that are differently susceptible to macromolecular inhibitors and can be manipulated by varying the ionic strength of the medium. The behaviour of the enzyme in vitro can be described by the presence of two discrete conformations with distinctive kinetic properties and different susceptibility to inhibition by the substrate benzyloxycarbonyl-Phe-Arg-7-amino-4-methylcoumarin. We identify and characterize sulfated glycosaminoglycans as natural allosteric modifiers of cathepsin K that exploit the conformational flexibility of the enzyme to regulate its activity and stability against autoproteolysis. All sulfated glycosaminoglycans act as non-essential activators in assays using low-molecular-mass substrates. Chondroitin sulfate and dermatan sulfate bind at one site on the enzyme, whereas heparin binds at an additional site and has a strongly stabilizing effect that is unique among human glycosaminoglycans. All glycosaminoglycans stimulate the elastinolytic activity of cathepsin K at physiological plasma pH, but only heparin also increases the collagenolytic activity of the enzyme under these conditions. Altogether these results provide novel insight into the mechanism of cathepsin K function at the molecular level and its regulation in the extracellular space.


Subject(s)
Cathepsin K/chemistry , Cathepsin K/metabolism , Allosteric Regulation , Animals , Cathepsin K/antagonists & inhibitors , Cathepsin K/blood , Cattle , Collagen/metabolism , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/metabolism , Cysteine Proteinase Inhibitors/physiology , Elastin/metabolism , Enzyme Activation/drug effects , Enzyme Stability , Glycosaminoglycans/chemistry , Glycosaminoglycans/metabolism , Glycosaminoglycans/pharmacology , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Kinetics , Models, Molecular , Protein Conformation/drug effects , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spectrometry, Fluorescence , Substrate Specificity , Tryptophan/chemistry
14.
Biochemistry ; 48(47): 11319-28, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19839601

ABSTRACT

Ammodytoxins are presynaptically neurotoxic snake venom group IIA secreted phospholipase A(2) enzymes that interact specifically with calmodulin in the cytosol of nerve cells. We show that calmodulin behaves as an activator of ammodytoxin under both nonreducing and reducing (cytosol-like) conditions by stimulating its enzymatic activity up to 21-fold. Kinetic analysis, using a general modifier mechanism, and surface plasmon resonance measurements reveal that calmodulin influences both the catalytic and the vesicle binding properties of the enzyme without affecting its calcium binding properties. The equilibrium dissociation constant of the ammodytoxin-calmodulin complex under cytosol-like conditions is in the low nanomolar range (3 nM), while under nonreducing conditions, the binding affinity is in the subnanomolar range (0.07-0.18 nM). Upon exposure to cytosol-like conditions, ammodytoxin undergoes a slow hysteretic transition to a less active state. Calmodulin stabilizes the conformation of ammodytoxin and thereby restores its activity. These results provide insights into the neurotoxic action of ammodytoxins and the mechanisms involved in the regulation of secreted phospholipase A(2) activity within the cytosol.


Subject(s)
Calmodulin/metabolism , Cytosol/metabolism , Phospholipases A2/metabolism , Snake Venoms/enzymology , Animals , Binding Sites , Calcium/chemistry , Calcium/metabolism , Calmodulin/chemistry , Calmodulin/toxicity , Cations, Divalent , Cytosol/chemistry , Enzyme Activation , Kinetics , Models, Molecular , Neurons/chemistry , Neurons/metabolism , Phospholipases A2/chemistry , Swine
15.
Biochim Biophys Acta ; 1783(6): 1129-39, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18261469

ABSTRACT

The molecular mechanism of the presynaptic toxicity of secreted phospholipase A2 (sPLA2) neurotoxins, including that of ammodytoxin A (AtxA), has not been resolved. Here we report the action of AtxA on mouse motoneuron-like cells, on which it induced characteristic neurotoxic effects on synaptic vesicles and on the reorganization of F-actin. AtxA also released fatty acids from the plasmalemma. Its significantly less neurotoxic V31W mutant showed similar effects on cells but with a much higher rate of hydrolysis than the wild-type, indicating that high enzymatic activity alone is not sufficient for the observed effects. The neurotoxic action was observed by confocal microscopy of a fluorescently labelled AtxA and by electron microscopy of a nanogold-labelled toxin. The Atx-binding proteins were tagged by a photo-cross-linking reagent conjugated to the toxin. AtxA was taken up rapidly by the cells, where it interacted within minutes with calmodulin and 14-3-3 proteins in the cytosol. These data demonstrate, for the first time, the translocation of an sPLA2 from the extracellular space into the cytosol of a cell. Such an event may thus be important in explaining the action of a range of homologous endogenous sPLA2 enzymes in mammals whose roles in various cellular processes are not yet completely understood.


Subject(s)
Cytosol/metabolism , Extracellular Space/metabolism , Motor Neurons/metabolism , Phospholipases A2, Secretory/metabolism , Presynaptic Terminals/metabolism , Viper Venoms/metabolism , 14-3-3 Proteins/metabolism , Actins/metabolism , Animals , Calmodulin/metabolism , Humans , Kidney/cytology , Kidney/metabolism , Mice , Microscopy, Confocal , Microscopy, Electron, Transmission , Motor Neurons/cytology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Phospholipases A2, Secretory/genetics , Protein Transport , Spinal Cord/cytology , Spinal Cord/metabolism , Viper Venoms/genetics
16.
Sci Rep ; 9(1): 283, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30670719

ABSTRACT

The ß-neurotoxic secreted phospholipases A2 (sPLA2s) block neuro-muscular transmission by poisoning nerve terminals. Damage inflicted by such sPLA2s (ß-ntx) on neuronal mitochondria is characteristic, very similar to that induced by structurally homologous endogenous group IIA sPLA2 when its activity is elevated, as, for example, in the early phase of Alzheimer's disease. Using ammodytoxin (Atx), the ß-ntx from the venom of the nose-horned viper (Vipera a. ammodytes), the sPLA2 receptor R25 has been detected in neuronal mitochondria. This receptor has been purified from porcine cerebral cortex mitochondria by a new Atx-affinity-based chromatographic procedure. Mass spectrometry analysis revealed R25 to be the subunit II of cytochrome c oxidase (CCOX), an essential constituent of the respiratory chain complex. CCOX was confirmed as being the first intracellular membrane receptor for sPLA2 by alternative Atx-affinity-labellings of purified CCOX, supported also by the encounter of Atx and CCOX in PC12 cells. This discovery suggests the explanation of the mechanism by which ß-ntx hinders production of ATP in poisoned nerve endings. It also provides a new insight into the potential function and dysfunction of endogenous GIIA sPLA2 in mitochondria.


Subject(s)
Electron Transport Complex IV/antagonists & inhibitors , Mitochondria/metabolism , Phospholipases A2, Secretory/pharmacology , Receptors, Phospholipase A2/analysis , Viper Venoms/enzymology , Animals , Cerebral Cortex/ultrastructure , Neurons/ultrastructure , Neurotoxicity Syndromes , PC12 Cells , Protein Subunits , Rats , Receptors, Phospholipase A2/isolation & purification , Swine , Viperidae
17.
Protein Expr Purif ; 62(1): 75-82, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18708147

ABSTRACT

Secreted modular calcium-binding (SMOC) proteins are little known members of the BM-40 family of matricellular proteins. SMOC-1 is localized in basement membranes, while SMOC-2 exhibits pro-angiogenic properties and stimulates cell cycle progression via integrin-linked kinase. In this work we have expressed recombinant human SMOCs in inclusion bodies in Escherichia coli. Soluble proteins were prepared by in vitro refolding with a final yield of approximately 3mg of purified SMOCs per liter of bacterial culture. The folding state of the products and their ability to reversibly bind calcium ions were verified by CD spectroscopy. The EF hands of the refolded SMOCs were both functional, one had high affinity for calcium ions (K(d) values in the 0.7-1 microM range), while the other had lower affinity (K(d) values 20-25 microM). The proteins were also examined for their ability to bind blood serum proteins. Three of the bands specifically retained on SMOC-Sepharose were identified as C-reactive protein, an acute phase protein from the pentraxin family, the basement membrane and elastic fiber-associated fibulin-1, and vitronectin, which is involved in cell adhesion, migration and proliferation and binds numerous extracellular and membrane proteins, including integrins. The interactions were additionally confirmed in solution using purified individual proteins by the method of biotin label transfer from one interacting partner to the other. Their identification is among the first pieces of information about the action of the SMOCs on molecular level and opens new possibilities for future research aimed towards elucidating the physiological roles of these versatile proteins.


Subject(s)
Blood Proteins/metabolism , C-Reactive Protein/metabolism , Calcium-Binding Proteins/biosynthesis , Osteonectin/biosynthesis , Recombinant Proteins/biosynthesis , Binding Sites , Blood Proteins/chemistry , C-Reactive Protein/chemistry , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Circular Dichroism , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Osteonectin/chemistry , Osteonectin/genetics , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
18.
Toxicon ; 51(5): 754-64, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18221975

ABSTRACT

Ammodytoxin C (AtxC) is a toxic secreted phospholipase A2 (sPLA2) from the venom of Vipera ammodytes snake. To evaluate its potential to kill cancer cells, the toxin was cross-linked to the monoclonal antibody against cathepsin B which endocytoses upon binding to cathepsin B, an antigen overexpressed on the plasma membrane of cancer cells. A photo-reactive derivative of AtxC, possessing the same biological activity as the native toxin, was reacted with antibodies to form a covalent immuno-conjugate. In conditions of the cytosolic redox potential, AtxC was gradually released from the conjugate due to reduction of the disulfide bond in the spacer arm of the cross-linker. The phospholipase activity of the preparation reached maximum in 10min and then decreased gradually. As demonstrated by fluorescence microscopy, the immuno-conjugate targeted Caco-2 colon adenocarcinoma cells but was very slowly internalized, the likely reason of only slight cytotoxicity being observed. Despite the lack of a clear cytotoxic effect of AtxC-antibody conjugate on Caco-2 cells, we demonstrated in this work a new methodology for the targeted delivery of (toxic) sPLA2 into cells, promising in research or therapy.


Subject(s)
Antibodies, Monoclonal/immunology , Antineoplastic Agents/administration & dosage , Cathepsin B/immunology , Group II Phospholipases A2/metabolism , Phospholipases A2/metabolism , Viper Venoms/metabolism , Animals , Antibodies, Monoclonal/metabolism , Antineoplastic Agents/therapeutic use , Caco-2 Cells , Drug Delivery Systems , Group II Phospholipases A2/immunology , Humans , Immunoconjugates , Neoplasms/drug therapy , Viper Venoms/immunology
19.
Thromb Haemost ; 118(10): 1713-1728, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30235482

ABSTRACT

Components of the intrinsic blood coagulation pathway, among them factor VIIIa (FVIIIa), have been recognized as suitable therapeutic targets to treat venous thromboembolism, pathological process behind two very serious cardiovascular diseases, deep vein thrombosis and pulmonary embolism. Here, we describe a unique glycoprotein from the nose-horned viper (Vipera ammodytes ammodytes [Vaa]) venom, Vaa serine proteinase homolog 1 (VaaSPH-1), structurally a serine protease but without an enzymatic activity and expressing potent anticoagulant action in human blood. We demonstrated that one of its targets in the blood coagulation system is FVIIIa of the intrinsic tenase complex, where it antagonizes the binding of FIXa. Anticoagulants with such characteristics are intensively sought, as they would be much safer for medical application as the contemporary drugs, which frequently induce excessive bleeding and other complications. VaaSPH-1 is unlikely to be orally available for chronic usage as it has molecular mass of 35 kDa. However, it represents a very promising template to design low molecular mass FVIIIa-directed anticoagulant substances, based on structural features of the interaction surface between VaaSPH-1 and FVIIIa. To this end, we constructed a three-dimensional model of VaaSPH-1 bound to FVIIIa. The model exposes the 157-loop and the preceding α-helix as the most appropriate structural elements of VaaSPH-1 to be considered as a guideline to synthesize small FVIIIa-binding molecules, potential new generation of anticoagulants.


Subject(s)
Cysteine Endopeptidases/metabolism , Neoplasm Proteins/metabolism , Reptilian Proteins/metabolism , Serine Proteases/metabolism , Venous Thromboembolism/drug therapy , Viper Venoms/metabolism , Animals , Blood Coagulation , Drug Design , Factor VIIIa/metabolism , Humans , Models, Chemical , Platelet Aggregation , Protein Binding , Structure-Activity Relationship , Viperidae/immunology
20.
J Clin Endocrinol Metab ; 103(2): 415-428, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29165578

ABSTRACT

Context: Pituitary stalk interruption syndrome (PSIS) consists of a small/absent anterior pituitary lobe, an interrupted/absent pituitary stalk, and an ectopic posterior pituitary lobe. Mendelian forms of PSIS are detected infrequently (<5%), and a polygenic etiology has been suggested. GLI2 variants have been reported at a relatively high frequency in PSIS. Objective: To provide further evidence for a non-Mendelian, polygenic etiology of PSIS. Methods: Exome sequencing (trio approach) in 20 patients with isolated PSIS. In addition to searching for (potentially) pathogenic de novo and biallelic variants, a targeted search was performed in a panel of genes associated with midline brain development (223 genes). For GLI2 variants, both (potentially) pathogenic and relatively rare variants (<5% in the general population) were studied. The frequency of GLI2 variants was compared with that of a reference population. Results: We found four additional candidate genes for isolated PSIS (DCHS1, ROBO2, CCDC88C, and KIF14) and one for syndromic PSIS (KAT6A). Eleven GLI2 variants were present in six patients. A higher frequency of a combination of two GLI2 variants (M1352V + D1520N) was found in the study group compared with a reference population (10% vs 0.68%). (Potentially) pathogenic variants were identified in genes associated with midline brain anomalies, including holoprosencephaly, hypogonadotropic hypogonadism, and absent corpus callosum and in genes involved in ciliopathies. Conclusion: Combinations of variants in genes associated with midline brain anomalies are frequently present in PSIS and sustain the hypothesis of a polygenic cause of PSIS.


Subject(s)
DNA Mutational Analysis/methods , Genetic Testing/methods , Multifactorial Inheritance , Pituitary Diseases/genetics , Pituitary Gland/abnormalities , Adolescent , Adult , Child , Child, Preschool , Exome , Female , Genetic Predisposition to Disease , Humans , Infant , Infant, Newborn , Male , Multifactorial Inheritance/genetics , Pituitary Diseases/congenital , Sequence Analysis, DNA/methods , Syndrome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL