Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Anal Bioanal Chem ; 411(12): 2493-2509, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30911800

RESUMEN

Inhalation of Bacillus anthracis spores can cause a rapidly progressing fatal infection. B. anthracis secretes three protein toxins: lethal factor (LF), edema factor (EF), and protective antigen (PA). EF and LF may circulate as free or PA-bound forms. Both free EF (EF) and PA-bound-EF (ETx) have adenylyl cyclase activity converting ATP to cAMP. We developed an adenylyl cyclase activity-based method for detecting and quantifying total EF (EF+ETx) in plasma. The three-step method includes magnetic immunocapture with monoclonal antibodies, reaction with ATP generating cAMP, and quantification of cAMP by isotope-dilution HPLC-MS/MS. Total EF was quantified from 5PL regression of cAMP vs ETx concentration. The detection limit was 20 fg/mL (225 zeptomoles/mL for the 89 kDa protein). Relative standard deviations for controls with 0.3, 6.0, and 90 pg/mL were 11.7-16.6% with 91.2-99.5% accuracy. The method demonstrated 100% specificity in 238 human serum/plasma samples collected from unexposed healthy individuals, and 100% sensitivity in samples from 3 human and 5 rhesus macaques with inhalation anthrax. Analysis of EF in the rhesus macaques showed that it was detected earlier post-exposure than B. anthracis by culture and PCR. Similar to LF, the kinetics of EF over the course of infection were triphasic, with an initial rise (phase-1), decline (phase-2), and final rapid rise (phase-3). EF levels were ~ 2-4 orders of magnitude lower than LF during phase-1 and phase-2 and only ~ 6-fold lower at death/euthanasia. Analysis of EF improves early diagnosis and adds to our understanding of anthrax toxemia throughout infection. The LF/EF ratio may also indicate the stage of infection and need for advanced treatments.


Asunto(s)
Carbunco/patología , Antígenos Bacterianos/sangre , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/sangre , Cromatografía Líquida de Alta Presión/métodos , Infecciones del Sistema Respiratorio/patología , Espectrometría de Masas en Tándem/métodos , Toxemia/patología , Adenosina Trifosfato/metabolismo , Animales , Carbunco/sangre , Estudios de Casos y Controles , AMP Cíclico/biosíntesis , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Límite de Detección , Macaca mulatta , Reacción en Cadena de la Polimerasa , Infecciones del Sistema Respiratorio/sangre , Toxemia/sangre , Toxemia/microbiología
2.
J Biol Chem ; 291(42): 22021-22029, 2016 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-27555325

RESUMEN

The protective antigen (PA) moiety of anthrax toxin binds to cellular receptors and mediates the translocation of the two enzymatic moieties of the toxin to the cytosol. Two PA receptors are known, with capillary morphogenesis protein 2 (CMG2) being the more important for pathogenesis and tumor endothelial marker 8 (TEM8) playing a minor role. The C-terminal PA domain 4 (PAD4) has extensive interactions with the receptors and is required for binding. Our previous study identified PAD4 variants having enhanced TEM8 binding specificity. To obtain PA variants that selectively bind to CMG2, here we performed phage display selections using magnetic beads having bound CMG2. We found that PA residue isoleucine 656 plays a critical role in PA binding to TEM8 but has a much lesser effect on PA binding to CMG2. We further characterized the role of residue 656 in distinguishing PA binding to CMG2 versus TEM8 by substituting it with the other 19 amino acids. Of the resulting variants, PA I656Q and PA I656V had significantly reduced activity on TEM8-expressing CHO cells but maintained their activity on CMG2-expressing CHO cells. The preference of these PA mutants for CMG2 over TEM8 was further demonstrated using mouse embryonic fibroblast cells and mice deficient in the CMG2 and/or the TEM8 receptors. The structural basis of the alterations in the receptor binding activities of these mutants is also discussed.


Asunto(s)
Antígenos Bacterianos/metabolismo , Bacillus anthracis/metabolismo , Toxinas Bacterianas/metabolismo , Biomarcadores de Tumor/metabolismo , Mutación Missense , Receptores de Péptidos/metabolismo , Sustitución de Aminoácidos , Animales , Antígenos Bacterianos/genética , Bacillus anthracis/genética , Toxinas Bacterianas/genética , Biomarcadores de Tumor/genética , Células CHO , Cricetinae , Cricetulus , Embrión de Mamíferos , Fibroblastos , Ratones , Proteínas de Microfilamentos , Unión Proteica , Dominios Proteicos , Receptores de Superficie Celular , Receptores de Péptidos/genética , Relación Estructura-Actividad
3.
J Biol Chem ; 290(10): 6584-95, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25564615

RESUMEN

Anthrax disease is caused by a toxin consisting of protective antigen (PA), lethal factor, and edema factor. Antibodies against PA have been shown to be protective against the disease. Variable domains of camelid heavy chain-only antibodies (VHHs) with affinity for PA were obtained from immunized alpacas and screened for anthrax neutralizing activity in macrophage toxicity assays. Two classes of neutralizing VHHs were identified recognizing distinct, non-overlapping epitopes. One class recognizes domain 4 of PA at a well characterized neutralizing site through which PA binds to its cellular receptor. A second neutralizing VHH (JKH-C7) recognizes a novel epitope. This antibody inhibits conversion of the PA oligomer from "pre-pore" to its SDS and heat-resistant "pore" conformation while not preventing cleavage of full-length 83-kDa PA (PA83) by cell surface proteases to its oligomer-competent 63-kDa form (PA63). The antibody prevents endocytosis of the cell surface-generated PA63 subunit but not preformed PA63 oligomers formed in solution. JKH-C7 and the receptor-blocking VHH class (JIK-B8) were expressed as a heterodimeric VHH-based neutralizing agent (VNA2-PA). This VNA displayed improved neutralizing potency in cell assays and protected mice from anthrax toxin challenge with much better efficacy than the separate component VHHs. The VNA protected virtually all mice when separately administered at a 1:1 ratio to toxin and protected mice against Bacillus anthracis spore infection. Thus, our studies show the potential of VNAs as anthrax therapeutics. Due to their simple and stable nature, VNAs should be amenable to genetic delivery or administration via respiratory routes.


Asunto(s)
Carbunco/inmunología , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/inmunología , Cadenas Pesadas de Inmunoglobulina/inmunología , Animales , Carbunco/microbiología , Carbunco/patología , Carbunco/terapia , Anticuerpos Antibacterianos/administración & dosificación , Bacillus anthracis/inmunología , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/antagonistas & inhibidores , Camélidos del Nuevo Mundo/inmunología , Epítopos/inmunología , Humanos , Ratones , Esporas/inmunología , Esporas/patogenicidad
4.
Sci Rep ; 4: 4754, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24755540

RESUMEN

We characterized an anti-cancer fusion protein consisting of anthrax lethal factor (LF) and the catalytic domain of Pseudomonas exotoxin A by (i) mutating the N-terminal amino acids and by (ii) reductive methylation to dimethylate all lysines. Dimethylation of lysines was achieved quantitatively and specifically without affecting binding of the fusion protein to PA or decreasing the enzymatic activity of the catalytic moiety. Ubiquitination in vitro was drastically decreased for both the N-terminally mutated and dimethylated variants, and both appeared to be slightly more stable in the cytosol of treated cells. The dimethylated variant showed greatly reduced neutralization by antibodies to LF. The two described modifications offer unique advantages such as increased cytotoxic activity and diminished antibody recognition, and thus may be applicable to other therapeutic proteins that act in the cytosol of cells.


Asunto(s)
ADP Ribosa Transferasas/genética , Antígenos Bacterianos/genética , Toxinas Bacterianas/genética , Exotoxinas/genética , Mutación , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/toxicidad , Factores de Virulencia/genética , ADP Ribosa Transferasas/química , Animales , Antígenos Bacterianos/química , Antineoplásicos , Toxinas Bacterianas/química , Línea Celular , Cricetinae , Citosol/metabolismo , Epítopos/genética , Epítopos/inmunología , Epítopos/metabolismo , Exotoxinas/química , Humanos , Cinética , Espectrometría de Masas , Metilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Ubiquitinación , Factores de Virulencia/química , Exotoxina A de Pseudomonas aeruginosa
5.
Infect Immun ; 81(12): 4592-603, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24082082

RESUMEN

Shiga toxin-producing Escherichia coli (STEC) is a major cause of severe food-borne disease worldwide, and two Shiga toxins, Stx1 and Stx2, are primarily responsible for the serious disease consequence, hemolytic-uremic syndrome (HUS). Here we report identification of a panel of heavy-chain-only antibody (Ab) V(H) (VHH) domains that neutralize Stx1 and/or Stx2 in cell-based assays. VHH heterodimer toxin-neutralizing agents containing two linked Stx1-neutralizing VHHs or two Stx2-neutralizing VHHs were generally much more potent at Stx neutralization than a pool of the two-component monomers tested in cell-based assays and in vivo mouse models. We recently reported that clearance of toxins can be promoted by coadministering a VHH-based toxin-neutralizing agent with an antitag monoclonal antibody (MAb), called the "effector Ab," that indirectly decorates each toxin molecule with four Ab molecules. Decoration occurs because the Ab binds to a common epitopic tag present at two sites on each of the two VHH heterodimer molecules that bind to each toxin molecule. Here we show that coadministration of effector Ab substantially improved the efficacy of Stx toxin-neutralizing agents to prevent death or kidney damage in mice following challenge with Stx1 or Stx2. A single toxin-neutralizing agent consisting of a double-tagged VHH heterotrimer--one Stx1-specific VHH, one Stx2-specific VHH, and one Stx1/Stx2 cross-specific VHH--was effective in preventing all symptoms of intoxication from Stx1 and Stx2 when coadministered with effector Ab. Overall, the availability of simple, defined, recombinant proteins that provide cost-effective protection against HUS opens up new therapeutic approaches to managing disease.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Síndrome Hemolítico-Urémico/inmunología , Toxina Shiga I/inmunología , Toxina Shiga II/inmunología , Anticuerpos de Dominio Único/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Escherichia coli Enteropatógena/inmunología , Escherichia coli Enteropatógena/metabolismo , Femenino , Ratones , Datos de Secuencia Molecular , Toxina Shiga I/metabolismo , Toxina Shiga II/metabolismo , Escherichia coli Shiga-Toxigénica/inmunología , Escherichia coli Shiga-Toxigénica/metabolismo
6.
PLoS One ; 8(8): e74474, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24015319

RESUMEN

Anthrax edema factor (EF) is a calmodulin-dependent adenylate cyclase that converts adenosine triphosphate (ATP) into 3'-5'-cyclic adenosine monophosphate (cAMP), contributing to the establishment of Bacillus anthracis infections and the resulting pathophysiology. We show that EF adenylate cyclase toxin activity is strongly mediated by the N-end rule, and thus is dependent on the identity of the N-terminal amino acid. EF variants having different N-terminal residues varied by more than 100-fold in potency in cultured cells and mice. EF variants having unfavorable, destabilizing N-terminal residues showed much greater activity in cells when the E1 ubiquitin ligase was inactivated or when proteasome inhibitors were present. Taken together, these results show that EF is uniquely affected by ubiquitination and/or proteasomal degradation.


Asunto(s)
Adenilil Ciclasas/metabolismo , Antígenos Bacterianos/metabolismo , Bacillus anthracis/enzimología , Toxinas Bacterianas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Adenilil Ciclasas/genética , Animales , Antígenos Bacterianos/genética , Bacillus anthracis/genética , Toxinas Bacterianas/genética , Línea Celular , Ratones
7.
J Mol Recognit ; 26(8): 376-81, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23784994

RESUMEN

The monoclonal antibody S9.6 binds DNA-RNA hybrids with high affinity, making it useful in research and diagnostic applications, such as in microarrays and in the detection of R-loops. A single-chain variable fragment (scFv) of S9.6 was produced, and its affinities for various synthetic nucleic acid hybrids were measured by surface plasmon resonance (SPR). S9.6 exhibits dissociation constants of approximately 0.6 nM for DNA-RNA and, surprisingly, 2.7 nM for RNA-RNA hybrids that are AU-rich. The affinity of the S9.6 scFv did not appear to be strongly influenced by various buffer conditions or by ionic strength below 500 mM NaCl. The smallest epitope that was strongly bound by the S9.6 scFv contained six base pairs of DNA-RNA hybrid. Published 2013. This article is a U.S. Government work and is in the public domain in the USA.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , ADN/metabolismo , Ácidos Nucleicos Heterodúplex/metabolismo , ARN/metabolismo , Anticuerpos de Cadena Única/metabolismo , Secuencia de Aminoácidos , Anticuerpos Monoclonales/aislamiento & purificación , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Tampones (Química) , Cationes Bivalentes/química , ADN/química , Epítopos/química , Epítopos/metabolismo , Concentración de Iones de Hidrógeno , Cinética , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Ácidos Nucleicos Heterodúplex/química , Concentración Osmolar , ARN/química , Anticuerpos de Cadena Única/aislamiento & purificación , Resonancia por Plasmón de Superficie
8.
J Biol Chem ; 288(13): 9058-65, 2013 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-23393143

RESUMEN

Anthrax toxin protective antigen (PA) delivers its effector proteins into the host cell cytosol through formation of an oligomeric pore, which can assume heptameric or octameric states. By screening a highly directed library of PA mutants, we identified variants that complement each other to exclusively form octamers. These PA variants were individually nontoxic and demonstrated toxicity only when combined with their complementary partner. We then engineered requirements for activation by matrix metalloproteases and urokinase plasminogen activator into two of these variants. The resulting therapeutic toxin specifically targeted cells expressing both tumor associated proteases and completely stopped tumor growth in mice when used at a dose far below that which caused toxicity. This scheme for obtaining intercomplementing subunits can be employed with other oligomeric proteins and potentially has wide application.


Asunto(s)
Antígenos Bacterianos/química , Toxinas Bacterianas/química , Neoplasias/tratamiento farmacológico , Animales , Bacillus anthracis/metabolismo , Línea Celular Tumoral , Femenino , Biblioteca de Genes , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Conformación Molecular , Mutación , Neoplasias/metabolismo , Plásmidos/metabolismo , Conformación Proteica , Ingeniería de Proteínas/métodos , Mapeo de Interacción de Proteínas/métodos , Estructura Terciaria de Proteína , Proteínas/química , Ultracentrifugación
9.
PLoS One ; 7(1): e29941, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22238680

RESUMEN

Antitoxins are needed that can be produced economically with improved safety and shelf life compared to conventional antisera-based therapeutics. Here we report a practical strategy for development of simple antitoxin therapeutics with substantial advantages over currently available treatments. The therapeutic strategy employs a single recombinant 'targeting agent' that binds a toxin at two unique sites and a 'clearing Ab' that binds two epitopes present on each targeting agent. Co-administration of the targeting agent and the clearing Ab results in decoration of the toxin with up to four Abs to promote accelerated clearance. The therapeutic strategy was applied to two Botulinum neurotoxin (BoNT) serotypes and protected mice from lethality in two different intoxication models with an efficacy equivalent to conventional antitoxin serum. Targeting agents were a single recombinant protein consisting of a heterodimer of two camelid anti-BoNT heavy-chain-only Ab V(H) (VHH) binding domains and two E-tag epitopes. The clearing mAb was an anti-E-tag mAb. By comparing the in vivo efficacy of treatments that employed neutralizing vs. non-neutralizing agents or the presence vs. absence of clearing Ab permitted unprecedented insight into the roles of toxin neutralization and clearance in antitoxin efficacy. Surprisingly, when a post-intoxication treatment model was used, a toxin-neutralizing heterodimer agent fully protected mice from intoxication even in the absence of clearing Ab. Thus a single, easy-to-produce recombinant protein was as efficacious as polyclonal antiserum in a clinically-relevant mouse model of botulism. This strategy should have widespread application in antitoxin development and other therapies in which neutralization and/or accelerated clearance of a serum biomolecule can offer therapeutic benefit.


Asunto(s)
Antitoxinas/biosíntesis , Antitoxinas/uso terapéutico , Botulismo/terapia , Inmunoterapia/tendencias , Animales , Afinidad de Anticuerpos , Antitoxinas/metabolismo , Antitoxina Botulínica/biosíntesis , Antitoxina Botulínica/metabolismo , Antitoxina Botulínica/uso terapéutico , Botulismo/inmunología , Botulismo/mortalidad , Botulismo/patología , Modelos Animales de Enfermedad , Descubrimiento de Drogas/métodos , Drogas en Investigación/metabolismo , Drogas en Investigación/uso terapéutico , Femenino , Inmunoterapia/métodos , Ratones , Modelos Biológicos , Multimerización de Proteína/fisiología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Análisis de Supervivencia , Resultado del Tratamiento
10.
Infect Immun ; 79(11): 4609-16, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21911463

RESUMEN

Bacillus anthracis is the causative agent of anthrax, and the tripartite anthrax toxin is an essential element of its pathogenesis. Edema factor (EF), a potent adenylyl cyclase, is one of the toxin components. In this work, anti-EF monoclonal antibodies (MAb) were produced following immunization of mice, and four of the antibodies were fully characterized. MAb 3F2 has an affinity of 388 pM, was most effective for EF detection, and appears to be the first antibody reported to neutralize EF by binding to the catalytic C(B) domain. MAb 7F10 shows potent neutralization of edema toxin activity in vitro and in vivo; it targets the N-terminal protective antigen binding domain. The four MAb react with three different domains of edema factor, and all were able to detect purified edema factor in Western blot analysis. None of the four MAb cross-reacted with the lethal factor toxin component. Three of the four MAb protected mice in both a systemic edema toxin challenge model and a subcutaneous spore-induced foreleg edema model. A combination of three of the MAb also significantly delayed the time to death in a third subcutaneous spore challenge model. This appears to be the first direct evidence that monoclonal antibody-mediated neutralization of EF alone is sufficient to delay anthrax disease progression.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Carbunco/prevención & control , Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/inmunología , Animales , Antígenos Bacterianos/toxicidad , Toxinas Bacterianas/toxicidad , Línea Celular , Relación Dosis-Respuesta a Droga , Edema/inducido químicamente , Edema/prevención & control , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Hibridomas , Inmunización , Inmunoglobulina G , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C
11.
Proc Natl Acad Sci U S A ; 107(32): 14070-4, 2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20660775

RESUMEN

The tripartite protein exotoxin secreted by Bacillus anthracis, a major contributor to its virulence and anthrax pathogenesis, consists of binary complexes of the protective antigen (PA) heptamer (PA63h), produced by proteolytic cleavage of PA, together with either lethal factor or edema factor. The mouse monoclonal anti-PA antibody 1G3 was previously shown to be a potent antidote that shares F(C) domain dependency with the human monoclonal antibody MDX-1303 currently under clinical development. Here we demonstrate that 1G3 instigates severe perturbation of the PA63h structure and creates a PA supercomplex as visualized by electron microscopy. This phenotype, produced by the unconventional mode of antibody action, highlights the feasibility for optimization of vaccines based on analogous structural modification of PA63h as an additional strategy for future remedies against anthrax.


Asunto(s)
Anticuerpos Neutralizantes , Complejo Antígeno-Anticuerpo/química , Antígenos Bacterianos/química , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/química , Toxinas Bacterianas/inmunología , Animales , Anticuerpos Monoclonales , Humanos , Ratones , Conformación Proteica , Multimerización de Proteína
12.
J Mol Biol ; 387(3): 680-93, 2009 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-19361425

RESUMEN

The virulence of Bacillus anthracis is critically dependent on the cytotoxic components of the anthrax toxin, lethal factor (LF) and edema factor (EF). LF and EF gain entry into host cells through interactions with the protective antigen (PA), which binds to host cellular receptors such as CMG2. Antibodies that neutralize PA have been shown to confer protection in animal models and are undergoing intense clinical development. A murine monoclonal antibody, 14B7, has been reported to interact with domain 4 of PA (PAD4) and block its binding to CMG2. More recently, the 14B7 antibody was used as the platform for the selection of very high affinity, single-chain antibodies that have tremendous potential as a combination anthrax prophylactic and treatment. Here, we report the high-resolution X-ray structures of three high-affinity, single-chain antibodies in the 14B7 family; 14B7 and two high-affinity variants 1H and M18. In addition, we present the first neutralizing antibody-PA structure, M18 in complex with PAD4 at 3.8 A resolution. These structures provide insights into the mechanism of neutralization, and the effect of various mutations on antibody affinity, and enable a comparison between the binding of the M18 antibody and CMG2 with PAD4.


Asunto(s)
Anticuerpos Monoclonales/química , Antígenos Bacterianos/química , Toxinas Bacterianas/química , Fragmentos de Péptidos/química , Estructura Terciaria de Proteína , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Afinidad de Anticuerpos , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Cristalografía por Rayos X , Región Variable de Inmunoglobulina , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo
13.
Protein Sci ; 18(2): 259-67, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19177559

RESUMEN

Disulfide bonds play a critical role in the stabilization of the immunoglobulin beta-sandwich sandwich. Under reducing conditions, such as those that prevail in the cytoplasm, disulfide bonds do not normally form and as a result most antibodies expressed in that compartment (intrabodies) accumulate in a misfolded and inactive state. We have developed a simple method for the quantitative isolation of antibody fragments that retain full activity under reducing conditions from large mutant libraries. In E. coli, inactivation of the cysteine oxidoreductase DsbA abolishes protein oxidation in the periplasm, which leads to the accumulation of scFvs and other disulfide-containing proteins in a reduced form. Libraries of mutant scFvs were tethered onto the inner membrane of dsbA cells and mutants that could bind fluorescently labeled antigen in the reducing periplasm were screened by Anchored Periplasmic Expression (APEx; Harvey et al., Proc Natl Acad Sci USA 2004;101:9193-9198.). Using this approach, we isolated scFv antibody variants that are fully active when expressed in the cytoplasm or when the four Cys residues that normally form disulfides are substituted by Ser residues.


Asunto(s)
Antígenos Bacterianos/metabolismo , Toxinas Bacterianas/metabolismo , Disulfuros/metabolismo , Región Variable de Inmunoglobulina/metabolismo , Pliegue de Proteína , Secuencia de Aminoácidos , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/inmunología , Cisteína/metabolismo , Evolución Molecular Dirigida , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Citometría de Flujo , Biblioteca de Genes , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/inmunología , Datos de Secuencia Molecular , Mutación , Oxidación-Reducción , Conformación Proteica , Proteína Disulfuro Isomerasas/genética , Ingeniería de Proteínas , Alineación de Secuencia , Serina/metabolismo
14.
Mol Cancer Ther ; 7(1): 48-58, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18202009

RESUMEN

Chronic myelogenous leukemia (CML) is driven by constitutively activated Bcr-Abl tyrosine kinase, which causes the defective adhesion of CML cells to bone marrow stroma. The overexpression of p210Bcr-Abl was reported to down-regulate CXCR4 expression, and this is associated with the cell migration defects in CML. We proposed that tyrosine kinase inhibitors, imatinib or INNO-406, may restore CXCR4 expression and cause the migration of CML cells to bone marrow microenvironment niches, which in turn results in acquisition of stroma-mediated chemoresistance of CML progenitor cells. In KBM5 and K562 cells, imatinib, INNO-406, or IFN-alpha increased CXCR4 expression and migration. This increase in CXCR4 levels on CML progenitor cells was likewise found in samples from CML patients treated with imatinib or IFN-alpha. Imatinib induced G0-G1 cell cycle block in CML cells, which was further enhanced in a mesenchymal stem cell (MSC) coculture system. MSC coculture protected KBM-5 cells from imatinib-induced cell death. These antiapoptotic effects were abrogated by the CXCR4 antagonist AMD3465 or by inhibitor of integrin-linked kinase QLT0267. Altogether, these findings suggest that the up-regulation of CXCR4 by imatinib promotes migration of CML cells to bone marrow stroma, causing the G0-G1 cell cycle arrest and hence ensuring the survival of quiescent CML progenitor cells.


Asunto(s)
Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Movimiento Celular/efectos de los fármacos , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Piperazinas/farmacología , Pirimidinas/farmacología , Receptores CXCR4/metabolismo , Antígenos CD34/metabolismo , Benzamidas , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Mesilato de Imatinib , Interferón-alfa/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Piperazinas/uso terapéutico , Pirimidinas/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos
15.
Blood ; 99(9): 3461-4, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11964319

RESUMEN

Recent studies suggest that the Bcl-2 and mitogen-activated protein kinase (MAPK) pathways together confer an aggressive, apoptosis-resistant phenotype on acute myelogenous leukemia (AML) cells. In this study, we analyzed the effects of simultaneous inhibition of these 2 pathways. In AML cell lines with constitutively activated MAPK, MAPK kinase (MEK) blockade by PD184352 strikingly potentiated the apoptosis induced by the small-molecule Bcl-2 inhibitor HA14-1 or by Bcl-2 antisense oligonucleotides. Isobologram analysis confirmed the synergistic nature of this interaction. Moreover, MEK blockade overcame Bcl-2 overexpression-mediated resistance to the proapoptotic effects of HA14-1. Most importantly, simultaneous exposure to PD184352 significantly (P =.01) potentiated HA14-1-mediated inhibition of clonogenic growth in all primary AML samples tested. These findings show that the Bcl-2 and MAPK pathways are relevant molecular targets in AML and that their concurrent inhibition could be developed into a new therapeutic strategy for this disease.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Leucemia Mieloide Aguda/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Benzamidas/farmacología , Benzopiranos/farmacología , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Humanos , Nitrilos/farmacología , Oligonucleótidos Antisentido/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Células Tumorales Cultivadas/efectos de los fármacos
16.
Blood ; 99(1): 326-35, 2002 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11756188

RESUMEN

It has been shown that the novel synthetic triterpenoid CDDO inhibits proliferation and induces differentiation and apoptosis in myeloid leukemia cells. In the current study the effects of the C-28 methyl ester of CDDO, CDDO-Me, were analyzed on cell growth and apoptosis of leukemic cell lines and primary acute myelogenous leukemia (AML). CDDO-Me decreased the viability of leukemic cell lines, including multidrug resistant (MDR)-1-overexpressing, p53(null) HL-60-Dox and of primary AML cells, and it was 3- to 5-fold more active than CDDO. CDDO-Me induced a loss of mitochondrial membrane potential, induction of caspase-3 cleavage, increase in annexin V binding and DNA fragmentation, suggesting the induction of apoptosis. CDDO-Me induced pro-apoptotic Bax protein that preceded caspase activation. Furthermore, CDDO-Me inhibited the activation of ERK1/2, as determined by the inhibition of mitochondrial ERK1/2 phosphorylation, and it blocked Bcl-2 phosphorylation, rendering Bcl-2 less anti-apoptotic. CDDO-Me induced granulo-monocytic differentiation in HL-60 cells and monocytic differentiation in primary cells. Of significance, colony formation of AML progenitors was significantly inhibited in a dose-dependent fashion, whereas normal CD34(+) progenitor cells were less affected. Combinations with ATRA or the RXR-specific ligand LG100268 enhanced the effects of CDDO-Me on cell viability and terminal differentiation of myeloid leukemic cell lines. In conclusion, CDDO-Me is an MDR-1- and a p53-independent compound that exerts strong antiproliferative, apoptotic, and differentiating effects in myeloid leukemic cell lines and in primary AML samples when given in submicromolar concentrations. Differential effects of CDDO-Me on leukemic and normal progenitor cells suggest that CDDO-Me has potential as a novel compound in the treatment of hematologic malignancies.


Asunto(s)
Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Leucemia Mieloide Aguda/patología , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/farmacología , Anexina A5/metabolismo , Crisis Blástica/patología , Caspasa 3 , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Citarabina/farmacología , Fragmentación del ADN/efectos de los fármacos , Interacciones Farmacológicas , Citometría de Flujo , Células HL-60/efectos de los fármacos , Humanos , Potenciales de la Membrana/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Retinoides/farmacología , Tretinoina/farmacología , Células Tumorales Cultivadas , Proteína X Asociada a bcl-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...