Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Phys Rev Lett ; 127(13): 135502, 2021 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-34623837

RESUMEN

Polycrystalline solids can exhibit material properties that differ significantly from those of equivalent single-crystal samples, in part, because of a spontaneous redistribution of mobile point defects into so-called space-charge regions adjacent to grain boundaries. The general analytical form of these space-charge regions is known only in the dilute limit, where defect-defect correlations can be neglected. Using kinetic Monte Carlo simulations of a three-dimensional Coulomb lattice gas, we show that grain boundary space-charge regions in nondilute solid electrolytes exhibit overscreening-damped oscillatory space-charge profiles-and underscreening-decay lengths that are longer than the corresponding Debye length and that increase with increasing defect-defect interaction strength. Overscreening and underscreening are known phenomena in concentrated liquid electrolytes, and the observation of functionally analogous behavior in solid electrolyte space-charge regions suggests that the same underlying physics drives behavior in both classes of systems. We therefore expect theoretical approaches developed to study nondilute liquid electrolytes to be equally applicable to future studies of solid electrolytes.

2.
Int J Cancer ; 146(2): 449-460, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31584195

RESUMEN

Bladder cancer is the sixth most common cancer in the United States, and it exhibits an alarming 70% recurrence rate. Thus, the development of more efficient antibladder cancer approaches is a high priority. Accordingly, this work provides the basis for a transformative anticancer strategy that takes advantage of the unique characteristics of the bladder. Unlike mucin-shielded normal bladder cells, cancer cells are exposed to the bladder lumen and overexpress EGFR. Therefore, we used an EGF-conjugated anthrax toxin that after targeting EGFR was internalized and triggered apoptosis in exposed bladder cancer cells. This unique agent presented advantages over other EGF-based technologies and other toxin-derivatives. In contrast to known agents, this EGF-toxin conjugate promoted its own uptake via receptor microclustering even in the presence of Her2 and induced cell death with a LC50 < 1 nM. Furthermore, our data showed that exposures as short as ≈3 min were enough to commit human (T24), mouse (MB49) and canine (primary) bladder cancer cells to apoptosis. Exposure of tumor-free mice and dogs with the agent resulted in no toxicity. In addition, the EGF-toxin was able to eliminate cells from human patient tumor samples. Importantly, the administration of EGF-toxin to dogs with spontaneous bladder cancer, who had failed or were not eligible for other therapies, resulted in ~30% average tumor reduction after one treatment cycle. Because of its in vitro and in vivo high efficiency, fast action (reducing treatment time from hours to minutes) and safety, we propose that this EGF-anthrax toxin conjugate provides the basis for new, transformative approaches against bladder cancer.


Asunto(s)
Antígenos Bacterianos/administración & dosificación , Antineoplásicos/administración & dosificación , Toxinas Bacterianas/administración & dosificación , Factor de Crecimiento Epidérmico/administración & dosificación , Inmunotoxinas/administración & dosificación , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Administración Intravesical , Animales , Antígenos Bacterianos/efectos adversos , Antineoplásicos/efectos adversos , Apoptosis/efectos de los fármacos , Toxinas Bacterianas/efectos adversos , Línea Celular Tumoral , Perros , Ensayos de Selección de Medicamentos Antitumorales , Factor de Crecimiento Epidérmico/efectos adversos , Femenino , Humanos , Inmunotoxinas/efectos adversos , Masculino , Ratones , Cultivo Primario de Células , Receptor ErbB-2/metabolismo , Resultado del Tratamiento , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/veterinaria
3.
Biomacromolecules ; 21(9): 3631-3643, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32706578

RESUMEN

Collagen type I is one of the major structural proteins in mammals, providing tissues such as cornea, tendon, bone, skin, and dentin with mechanical stability, strength, and toughness. Collagen fibrils are composed of collagen molecules arranged in a quarter-stagger array that gives rise to a periodicity of 67 nm along the fibril axis, with a 30 nm overlap zone and a 37 nm gap zone. The formation of such highly organized fibrils is a self-assembly process where electrostatic and hydrophobic interactions play a critical role in determining the staggering of the molecules with 67 nm periodicity. While collagen self-assembly has been extensively studied, not much is known about the mechanism, and in particular, the nature of the nuclei that initially form, the different stages of the aggregation process, and how the organization of the molecules into fibrils arises. By combining time-resolved cryo-transmission electron microscopy with molecular dynamics simulations, we show that collagen assembly is a multistep process in which the molecules first form filaments which self-organize into fibrils with a disordered structure. The appearance of the D-band periodicity is gradual and starts with the alignment of adjacent filaments at the N-terminal end of the molecules, first leading to bands with a periodicity of 67 nm and then to the formation of gap and overlap regions.


Asunto(s)
Colágeno Tipo I , Colágeno , Animales , Córnea , Microscopía por Crioelectrón , Matriz Extracelular
4.
Phys Chem Chem Phys ; 21(11): 6133-6141, 2019 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-30816891

RESUMEN

In this work, we present the first example of the self-assembly of phospholipid monolayers at the interface between air and an ionic solvent. Deep eutectic solvents are a novel class of environmentally friendly, non-aqueous, room temperature liquids with tunable properties, that have wide-ranging potential applications and are capable of promoting the self-assembly of surfactant molecules. We use a chemically-consistent Bayesian modelling of X-ray and neutron reflectometry measurements to show that these monolayers broadly behave as they do on water. This method allows for the monolayer structure to be determined, alongside the molecular volumes of the individual monolayer components, without the need for water-specific constraints to be introduced. Furthermore, using this method we are able to better understand the correlations present between parameters in the analytical model. This example of a non-aqueous phospholipid monolayer has important implications for the potential uses of these solvents and for our understanding of how biomolecules behave in the absence of water.


Asunto(s)
Lípidos/química , Modelos Químicos , Solventes/química , Teorema de Bayes , Técnicas de Química Analítica , Agua/química
5.
Proc Natl Acad Sci U S A ; 113(11): 3042-7, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26929342

RESUMEN

The bacterial type 6 secretion system (T6SS) is a dynamic apparatus that translocates proteins between cells by a mechanism analogous to phage tail contraction. T6SS sheaths are cytoplasmic tubular structures composed of stable VipA-VipB (named for ClpV-interacting protein A and B) heterodimers. Here, the structure of the VipA/B sheath was exploited to generate immunogenic multivalent particles for vaccine delivery. Sheaths composed of VipB and VipA fused to an antigen of interest were purified from Vibrio cholerae or Escherichia coli and used for immunization. Sheaths displaying heterologous antigens generated better immune responses against the antigen and different IgG subclasses compared with soluble antigen alone. Moreover, antigen-specific antibodies raised against sheaths presenting Neisseria meningitidis factor H binding protein (fHbp) antigen were functional in a serum bactericidal assay. Our results demonstrate that multivalent nanoparticles based on the T6SS sheath represent a versatile scaffold for vaccine applications.


Asunto(s)
Antígenos/administración & dosificación , Nanopartículas/química , Sistemas de Secreción Tipo VI/ultraestructura , Vacunas/administración & dosificación , Acinetobacter/química , Acinetobacter/genética , Aminoaciltransferasas/metabolismo , Animales , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Antibacterianos/inmunología , Antígenos/inmunología , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/administración & dosificación , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/inmunología , Línea Celular , Cisteína Endopeptidasas/metabolismo , Dimerización , Sistemas de Liberación de Medicamentos , Escherichia coli/química , Femenino , Genes Reporteros , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/genética , Macrófagos/fisiología , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica , Nanopartículas/administración & dosificación , Nanopartículas/ultraestructura , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/inmunología , Sistemas de Secreción Tipo VI/química , Vacunación , Vacunas/inmunología , Vibrio cholerae/química
6.
Front Immunol ; 15: 1293883, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38455057

RESUMEN

Fibrotic diseases, such as idiopathic pulmonary fibrosis (IPF) and systemic scleroderma (SSc), are commonly associated with high morbidity and mortality, thereby representing a significant unmet medical need. Interleukin 11 (IL11)-mediated cell activation has been identified as a central mechanism for promoting fibrosis downstream of TGFß. IL11 signaling has recently been reported to promote fibroblast-to-myofibroblast transition, thus leading to various pro-fibrotic phenotypic changes. We confirmed increased mRNA expression of IL11 and IL11Rα in fibrotic diseases by OMICs approaches and in situ hybridization. However, the vital role of IL11 as a driver for fibrosis was not recapitulated. While induction of IL11 secretion was observed downstream of TGFß signaling in human lung fibroblasts and epithelial cells, the cellular responses induced by IL11 was quantitatively and qualitatively inferior to that of TGFß at the transcriptional and translational levels. IL11 blocking antibodies inhibited IL11Rα-proximal STAT3 activation but failed to block TGFß-induced profibrotic signals. In summary, our results challenge the concept of IL11 blockade as a strategy for providing transformative treatment for fibrosis.


Asunto(s)
Interleucina-11 , Factor de Crecimiento Transformador beta , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal , Fibrosis , Miofibroblastos/metabolismo
7.
J Appl Crystallogr ; 56(Pt 1): 12-17, 2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36777146

RESUMEN

As a result of the availability of modern software and hardware, Bayesian analysis is becoming more popular in neutron and X-ray reflectometry analysis. The understandability and replicability of these analyses may be harmed by inconsistencies in how the probability distributions central to Bayesian methods are represented in the literature. Herein advice is provided on how to report the results of Bayesian analysis as applied to neutron and X-ray reflectometry. This includes the clear reporting of initial starting conditions, the prior probabilities, the results of any analysis and the posterior probabilities that are the Bayesian equivalent of the error bar, to enable replicability and improve understanding. It is believed that this advice, grounded in the authors' experience working in the field, will enable greater analytical reproducibility in the work of the reflectometry community, and improve the quality and usability of results.

8.
Mol Imaging Biol ; 25(2): 314-323, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35906512

RESUMEN

PURPOSE: To image colon-expressed alternatively spliced D domain of tenascin C in preclinical colitis models using near infrared (NIR)-labeled targeted molecular imaging agents. PROCEDURES: A human IgG1 with nanomolar binding affinity specific to the alternatively spliced D domain of tenascin C was generated. Immunohistochemistry identified disease-specific expression of this extracellular matrix protein in the colon of mice given dextran sulfate sodium in the drinking water. The antibody reagent was labeled with the NIR fluorophore IRDye 800CW via amine chemistry and intravenously dosed to evaluate in vivo targeting specificity. Increasing doses of imaging agent were given to estimate the saturating dose. RESULTS: The NIR-labeled proteins successfully targeted colonic lesions in a murine model of colitis. Co-administration of a molar excess competing unlabeled dose reduced normalized uptake in diseased colon by > 70%. Near infrared ex vivo images of colon resected from diseased animals showed saturation at doses exceeding 1 nmol and was confirmed with additional quantitative ex vivo biodistribution. Cellular-level specificity and protein stability were assessed via microscopy. CONCLUSIONS: Our imaging data suggest the alternatively spliced D domain of tenascin C is a promising target for delivery-based applications in inflammatory bowel diseases.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Humanos , Animales , Ratones , Tenascina , Distribución Tisular , Colitis/patología
9.
MAbs ; 14(1): 2080628, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35771588

RESUMEN

Approaches for antibody discovery have seen substantial improvement and success in recent years. Yet, advancing antibodies into the clinic remains difficult because therapeutic developability concerns are challenging to predict. We developed a computational model to simplify antibody developability assessment and enable accelerated early-stage screening. To this end, we quantified the ability of hundreds of sequence- and structure-based descriptors to differentiate clinical antibodies that have undergone rigorous screening and characterization for drug-like properties from antibodies in the human repertoire that are not natively paired. This analysis identified 144 descriptors capable of distinguishing clinical from repertoire antibodies. Five descriptors were selected and combined based on performance and orthogonality into a single model referred to as the Therapeutic Antibody Developability Analysis (TA-DA). On a hold-out test set, this tool separated clinical antibodies from repertoire antibodies with an AUC = 0.8, demonstrating the ability to identify developability attributes unique to clinical antibodies. Based on our results, the TA-DA score may serve as an approach for selecting lead antibodies for further development.Abbreviations: Affinity-Capture Self-Interaction Nanoparticle Spectroscopy (AC-SINS), Area Under the Curve (AUC), Complementary-Determining Region (CDR), Clinical-Stage Therapeutics (CST), Framework (FR), Monoclonal Antibodies (mAbs), Observed Antibody Space (OAS), Receiver Operating Characteristic (ROC), Size-Exclusion Chromatography (SEC), Structural Aggregation Propensity (SAP), Therapeutic Antibody Developability Analysis (TA-DA), Therapeutic Antibody Profiler (TAP), Therapeutic Structural Antibody Database (Thera-SAbDab), Variable Heavy (VH), Variable Light (VL).


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Monoclonales/química , Cromatografía en Gel , Humanos
10.
J Appl Crystallogr ; 52(Pt 3): 665-668, 2019 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-31236095

RESUMEN

Classical molecular dynamics simulations are a common component of multi-modal analyses of scattering measurements, such as small-angle scattering and diffraction. Users of these experimental techniques often have no formal training in the theory and practice of molecular dynamics simulation, leading to the possibility of these simulations being treated as a 'black box' analysis technique. This article describes an open educational resource (OER) designed to introduce classical molecular dynamics to users of scattering methods. This resource is available as a series of interactive web pages, which can be easily accessed by students, and as an open-source software repository, which can be freely copied, modified and redistributed by educators. The topics covered in this OER include classical atomistic modelling, parameterizing interatomic potentials, molecular dynamics simulations, typical sources of error and some of the approaches to using simulations in the analysis of scattering data.

11.
Protein Sci ; 16(12): 2726-32, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17965191

RESUMEN

A major goal in the production of therapeutic proteins, subunit vaccines, as well as recombinant proteins needed for structure determination and structural proteomics is their recovery in a pure and functional state using the simplest purification procedures. Here, we report the design and use of a novel tandem (His)(6)-calmodulin (HiCaM) fusion tag that combines two distinct purification strategies, namely, immobilized metal affinity (IMAC) and hydrophobic interaction chromatography (HIC), in a simple two-step procedure. Two model constructs were generated by fusing the HiCaM purification tag to the N terminus of either the enhanced green fluorescent protein (eGFP) or the human tumor suppressor protein p53. These fusion constructs were abundantly expressed in Escherichia coli and rapidly purified from cleared lysates by tandem IMAC/HIC to near homogeneity under native conditions. Cleavage at a thrombin recognition site between the HiCaM-tag and the constructs readily produced untagged, functional versions of eGFP and human p53 that were >97% pure. The HiCaM purification strategy is rapid, makes use of widely available, high-capacity, and inexpensive matrices, and therefore represents an excellent approach for large-scale purification of recombinant proteins as well as small-scale protein array designs.


Asunto(s)
Cromatografía de Afinidad/métodos , Proteínas Fluorescentes Verdes/aislamiento & purificación , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteína p53 Supresora de Tumor/aislamiento & purificación , Secuencia de Aminoácidos , Secuencia de Bases , Calmodulina/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Datos de Secuencia Molecular
12.
Toxins (Basel) ; 7(4): 1303-23, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25875295

RESUMEN

The pH-triggered membrane insertion of the diphtheria toxin translocation domain (T domain) results in transferring the catalytic domain into the cytosol, which is relevant to potential biomedical applications as a cargo-delivery system. Protonation of residues is suggested to play a key role in the process, and residues E349, D352 and E362 are of particular interest because of their location within the membrane insertion unit TH8-TH9. We have used various spectroscopic, computational and functional assays to characterize the properties of the T domain carrying the double mutation E349Q/D352N or the single mutation E362Q. Vesicle leakage measurements indicate that both mutants interact with the membrane under less acidic conditions than the wild-type. Thermal unfolding and fluorescence measurements, complemented with molecular dynamics simulations, suggest that the mutant E362Q is more susceptible to acid destabilization because of disruption of native intramolecular contacts. Fluorescence experiments show that removal of the charge in E362Q, and not in E349Q/D352N, is important for insertion of TH8-TH9. Both mutants adopt a final functional state upon further acidification. We conclude that these acidic residues are involved in the pH-dependent action of the T domain, and their replacements can be used for fine tuning the pH range of membrane interactions.


Asunto(s)
Toxina Diftérica/química , Toxina Diftérica/genética , Animales , Células CHO , Membrana Celular , Dicroismo Circular , Cricetinae , Cricetulus , Concentración de Iones de Hidrógeno , Simulación de Dinámica Molecular , Mutación , Estructura Terciaria de Proteína , Espectrometría de Fluorescencia
13.
J Mol Biol ; 426(3): 645-55, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24211469

RESUMEN

The catalytic moiety of Pseudomonas exotoxin A (domain III or PE3) inhibits protein synthesis by ADP-ribosylation of eukaryotic elongation factor 2. PE3 is widely used as a cytocidal payload in receptor-targeted protein toxin conjugates. We have designed and characterized catalytically inactive fragments of PE3 that are capable of structural complementation. We dissected PE3 at an extended loop and fused each fragment to one subunit of a heterospecific coiled coil. In vitro ADP-ribosylation and protein translation assays demonstrate that the resulting fusions-supplied exogenously as genetic elements or purified protein fragments-had no significant catalytic activity or effect on protein synthesis individually but, in combination, catalyzed the ADP-ribosylation of eukaryotic elongation factor 2 and inhibited protein synthesis. Although complementing PE3 fragments are catalytically less efficient than intact PE3 in cell-free systems, co-expression in live cells transfected with transgenes encoding the toxin fusions inhibits protein synthesis and causes cell death comparably as intact PE3. Complementation of split PE3 offers a direct extension of the immunotoxin approach to generate bispecific agents that may be useful to target complex phenotypes.


Asunto(s)
ADP Ribosa Transferasas/química , ADP Ribosa Transferasas/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Exotoxinas/química , Exotoxinas/metabolismo , Biosíntesis de Proteínas , Factores de Virulencia/química , Factores de Virulencia/metabolismo , Dominio Catalítico , Supervivencia Celular , Sistema Libre de Células , Cromatografía en Gel , Células HEK293 , Humanos , Immunoblotting , Inmunoprecipitación , Exotoxina A de Pseudomonas aeruginosa
14.
Mol Cancer Ther ; 12(10): 2273-81, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23945077

RESUMEN

Chimeric protein toxins that act selectively on cells expressing a designated receptor may serve as investigational probes and/or antitumor agents. Here, we report use of the enzyme sortase A (SrtA) to create four chimeric toxins designed to selectively kill cells bearing the tumor marker HER2. We first expressed and purified: (i) a receptor recognition-deficient form of diphtheria toxin that lacks its receptor-binding domain and (ii) a mutated, receptor-binding-deficient form of anthrax-protective antigen. Both proteins carried at the C terminus the sortase recognition sequence LPETGG and a H6 affinity tag. Each toxin protein was mixed with SrtA plus either of two HER2-recognition proteins--a single-chain antibody fragment or an Affibody--both carrying an N-terminal G5 tag. With wild-type SrtA, the fusion reaction between the toxin and receptor-recognition proteins approached completion only after several hours, whereas with an evolved form of the enzyme, SrtA*, the reaction was virtually complete within 5 minutes. The four fusion toxins were purified and shown to kill HER2-positive cells in culture with high specificity. Sortase-mediated ligation of binary combinations of diverse natively folded proteins offers a facile way to produce large sets of chimeric proteins for research and medicine.


Asunto(s)
Aminoaciltransferasas/genética , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Cisteína Endopeptidasas/genética , Toxina Diftérica/genética , Inmunotoxinas/genética , Proteínas Recombinantes de Fusión/genética , Secuencia de Aminoácidos , Antígenos Bacterianos/uso terapéutico , Toxinas Bacterianas/uso terapéutico , Sitios de Unión , Línea Celular Tumoral , Toxina Diftérica/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunotoxinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Unión Proteica , Receptor ErbB-2/genética , Proteínas Recombinantes de Fusión/uso terapéutico
15.
Mol Oncol ; 7(3): 440-51, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23290417

RESUMEN

Targeted therapeutics have emerged in recent years as an attractive approach to treating various types of cancer. One approach is to modify a cytocidal protein toxin to direct its action to a specific population of cancer cells. We created a targeted toxin in which the receptor-binding and pore-forming moiety of anthrax toxin, termed Protective Antigen (PA), was modified to redirect its receptor specificity to HER2, a marker expressed at the surface of a significant fraction of breast and ovarian tumors. The resulting fusion protein (mPA-ZHER2) delivered cytocidal effectors specifically into HER2-positive tumor cells, including a trastuzumab-resistant line, causing death of the cells. No off-target killing of HER2-negative cells was observed, either with homogeneous populations or with mixtures of HER2-positive and HER2-negative cells. A mixture of mPA variants targeting different receptors mediated killing of cells bearing either receptor, without affecting cells devoid of these receptors. Anthrax toxin may serve as an effective platform for developing therapeutics to ablate cells bearing HER2 or other tumor-specific cell-surface markers.


Asunto(s)
Antígenos Bacterianos/farmacología , Toxinas Bacterianas/farmacología , Neoplasias/tratamiento farmacológico , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/genética , Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/genética , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos , Receptores ErbB/metabolismo , Humanos , Neoplasias/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Trastuzumab
16.
mBio ; 3(3)2012.
Artículo en Inglés | MEDLINE | ID: mdl-22550037

RESUMEN

UNLABELLED: The actions of many bacterial toxins depend on their ability to bind to one or more cell-surface receptors. Anthrax toxin acts by a sequence of events that begins when the protective-antigen (PA) moiety of the toxin binds to either one of two cell-surface proteins, ANTXR1 and ANTXR2, and is proteolytically activated. The activated PA self-associates to form oligomeric pore precursors, which, in turn, bind the enzymatic moieties of the toxin and transport them to the cytosol. We introduced a double mutation into domain 4 of PA to ablate its native receptor-binding function and fused epidermal growth factor (EGF) to the C terminus of the mutated protein. The resulting fusion protein transported enzymatic effector proteins into a cell line that expressed the EGF receptor (A431 cells), but not into a line lacking this receptor (CHO-K1 cells). Addition of excess free EGF blocked transport of effector proteins into A431 cells via the fusion protein, but not via native PA. We also showed that fusing the diphtheria toxin receptor-binding domain to the C terminus of the mutated PA channeled effector-protein transport through the diphtheria toxin receptor. PA fusion proteins with altered receptor specificity may be useful in biological research and could have practical applications, including ablation or perturbation of selected populations of cells in vivo. IMPORTANCE: Bacterial toxins that act within mammalian cells have receptor-dependent mechanisms to transport their enzymatic components to the cytoplasmic compartment. By inactivating or otherwise modifying their respective intracellular targets, these intracellular effectors disrupt metabolic pathways and in some cases cause death of the cell. Our results show that the receptor specificity of the transport protein of anthrax toxin may be readily changed, raising the possibility that receptor-redirected forms of protective antigen (PA) and PA homologs may be useful for research and medical applications requiring modification or ablation of designated populations of cells.


Asunto(s)
Carbunco/metabolismo , Antígenos Bacterianos/metabolismo , Toxinas Bacterianas/metabolismo , Animales , Antígenos Bacterianos/química , Antígenos Bacterianos/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Línea Celular , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores de Péptidos , Especificidad de la Especie
17.
PLoS One ; 7(2): e31191, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22355345

RESUMEN

Shiga-like toxins are ribosome-inactivating proteins (RIP) produced by pathogenic E. coli strains that are responsible for hemorrhagic colitis and hemolytic uremic syndrome. The catalytic A(1) chain of Shiga-like toxin 1 (SLT-1), a representative RIP, first docks onto a conserved peptide SD[D/E]DMGFGLFD located at the C-terminus of all three eukaryotic ribosomal stalk proteins and halts protein synthesis through the depurination of an adenine base in the sarcin-ricin loop of 28S rRNA. Here, we report that the A(1) chain of SLT-1 rapidly binds to and dissociates from the C-terminal peptide with a monomeric dissociation constant of 13 µM. An alanine scan performed on the conserved peptide revealed that the SLT-1 A(1) chain interacts with the anionic tripeptide DDD and the hydrophobic tetrapeptide motif FGLF within its sequence. Based on these 2 peptide motifs, SLT-1 A(1) variants were generated that displayed decreased affinities for the stalk protein C-terminus and also correlated with reduced ribosome-inactivating activities in relation to the wild-type A(1) chain. The toxin-peptide interaction and subsequent toxicity were shown to be mediated by cationic and hydrophobic docking surfaces on the SLT-1 catalytic domain. These docking surfaces are located on the opposite face of the catalytic cleft and suggest that the docking of the A(1) chain to SDDDMGFGLFD may reorient its catalytic domain to face its RNA substrate. More importantly, both the delineated A(1) chain ribosomal docking surfaces and the ribosomal peptide itself represent a target and a scaffold, respectively, for the design of generic inhibitors to block the action of RIPs.


Asunto(s)
Interacciones Hidrofóbicas e Hidrofílicas , Oligopéptidos/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Ribosómicas/metabolismo , Ribosomas/metabolismo , Toxina Shiga I/antagonistas & inhibidores , Toxina Shiga I/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , Datos de Secuencia Molecular , Biosíntesis de Proteínas , Estructura Terciaria de Proteína , ARN Ribosómico/genética , Proteínas Recombinantes/genética , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie , Técnicas del Sistema de Dos Híbridos
18.
J Mol Biol ; 378(2): 375-86, 2008 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-18358491

RESUMEN

Shiga-like toxin 1 (SLT-1) is a type II ribosome-inactivating protein; its A(1) domain blocks protein synthesis in eukaryotic cells by catalyzing the depurination of a single adenine base in 28 S rRNA. The molecular mechanism leading to this site-specific depurination event is thought to involve interactions with eukaryotic ribosomal proteins. Here, we present evidence that the A(1) chain of SLT-1 binds to the ribosomal proteins P0, P1, and P2. These proteins were identified from a HeLa cell lysate by tandem mass spectrometry, and subsequently confirmed to bind to SLT-1 A(1) chain by yeast-two-hybrid and pull-down experiments using candidate full-length proteins. Moreover, the removal of the last 17 amino acids of either protein P1 or P2 abolishes the interaction with the A(1) chain, whereas P0, lacking this common C terminus, still binds to the A(1) domain. In vitro pull-down experiments using fusion protein-tagged C-terminal peptides corresponding to the common 7, 11, and 17 terminal residues of P1 and P2 confirmed that the A(1) chain of SLT-1 as well as the A chain of ricin bind to this shared C-terminal peptide motif. More importantly, a synthetic peptide corresponding to the 17 amino acid C terminus of P1 and P2 was shown to inhibit the ribosome-inactivating function of the A(1) chain of SLT-1 in an in vitro transcription and translation-coupled assay. These results suggest a role for the ribosomal stalk in aiding the A(1) chain of SLT-1 and other type II ribosome-inactivating proteins in localizing its catalytic domain near the site of depurination in the 28 S rRNA.


Asunto(s)
Dominio Catalítico , Fosfoproteínas/metabolismo , Proteínas Ribosómicas/metabolismo , Toxina Shiga I/antagonistas & inhibidores , Toxina Shiga I/metabolismo , Secuencia de Aminoácidos , Extractos Celulares/química , Secuencia Conservada , Células HeLa , Humanos , Datos de Secuencia Molecular , Fosfoproteínas/análisis , Fosfoproteínas/genética , Proteínas Ribosómicas/análisis , Proteínas Ribosómicas/genética , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Ricina/metabolismo , Espectrometría de Masas en Tándem , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA